1
|
Saini P, Kumar S, Jangra H, Solanki A, Verma R, Kumar R, Kaushik R, Grakh K, Saini G, Bangar Y, Pandey AK. The occurrence of follicular cyst affects the embryonic developmental competences of buffalo oocytes under in vitro culture conditions. Theriogenology 2025; 235:152-161. [PMID: 39826265 DOI: 10.1016/j.theriogenology.2025.01.007] [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: 10/05/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Cystic ovarian disease (COD) is a major cause of infertility in dairy cows. This study aimed to investigate the impact of follicular cysts on the invitro blastocyst developmental competence of oocytes and the relative gene expression of blastocysts developed from the subordinate follicles of ipsilateral (ovary with cyst), contralateral (ovary opposite to cyst), and normal ovaries of buffaloes. A total of 2059 ovaries were collected from slaughterhouse and classified into three categories based on the presence of follicular cysts: a) ipsilateral, b) contralateral, and c) control (absence of cysts). Oocytes of grades A, B, and C were used for invitro maturation, invitro fertilization, and invitro culture. The cleavage rates of the ipsilateral (91.54 %) and contralateral (95.71 %) categories were higher (P < 0.01) than those of the control (76.82 %). Conversely, the blastocyst development rate was higher (P < 0.01) in control (32.67 %) than in ipsilateral (8.46 %) and contralateral (7.14 %) groups. The mRNA expression levels of maturation (BMP15), steroidogenesis (STAR, CYP19), antioxidant (SOD2, HSPB1) and anti-apoptotic (BCL2) genes were decreased in arrested embryos derived from both cystic (ipsilateral and contralateral) and control category follicles when compared to blastocysts derived from their respective category follicles. Conversely, BAX expression increased in arrested embryos. Expression of SOD2 and BAX was downregulated in blastocysts from both ipsilateral and contralateral categories compared to controls. The presence of one or more follicular cysts in either ovary affected the developmental competence of oocytes derived from subordinate follicles. Therefore, the buffaloes with cysts in either ovary should be avoided when aspirating follicles for IVF.
Collapse
Affiliation(s)
- Pradeep Saini
- Veterinary Gynaecology and Obstetrics, COVS, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, HR, India
| | - Sandeep Kumar
- Veterinary Gynaecology and Obstetrics, COVS, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, HR, India
| | - Hitesh Jangra
- Veterinary Gynaecology and Obstetrics, COVS, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, HR, India
| | - Anupama Solanki
- Veterinary Gynaecology and Obstetrics, COVS, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, HR, India
| | - Rahul Verma
- Veterinary Gynaecology and Obstetrics, COVS, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, HR, India
| | - Rajesh Kumar
- Veterinary Physiology and Biochemistry, COVS, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, HR, India
| | | | - Kushal Grakh
- Veterinary Public Health and Epidemiology, COVS, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, HR, India
| | - Gitesh Saini
- Veterinary Gynaecology and Obstetrics, COVS, Rampuraphul, Guru Angad Dev University of Veterinary and Animal Sciences, Punjab, India
| | - Yogesh Bangar
- Animal Breeding and Genetics, COVS, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, HR, India
| | - Anand Kumar Pandey
- Veterinary Gynaecology and Obstetrics, COVS, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, HR, India.
| |
Collapse
|
2
|
Yang LK, Ma WJ, Wang X, Chen HR, Jiang YN, Sun H. Apoptosis in polycystic ovary syndrome: Mechanisms and therapeutic implications. Life Sci 2025; 363:123394. [PMID: 39809382 DOI: 10.1016/j.lfs.2025.123394] [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: 08/19/2024] [Revised: 12/30/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a common disorder that affects the female reproductive system, with an incidence of 8 % to 15 %. It is characterized by irregular menstruation, hyperandrogenemia, and polycystic abnormalities in the ovaries. Nevertheless, there is still much to learn about the molecular pathways underlying PCOS. Apoptosis is the process by which cells actively destroy themselves, and it is vital to an organism's ability to develop normally and maintain homeostasis. In recent years, a growing body of research has indicated a connection between the pathophysiology of PCOS and apoptosis. Therefore, it is critical to comprehend the relationship between PCOS and apoptosis in greater detail, identify the pathophysiological underpinnings of PCOS, and provide fresh perspectives and targets for its treatment. This review aims to summarize the relationship between PCOS and apoptosis, discuss how apoptosis affects normal ovarian function and how it becomes dysfunctional in the ovaries of PCOS patients, and investigate the signaling pathways associated with apoptosis in PCOS, including PI3K-Akt, TNF, NF-κB, and p53. Additionally, potential therapeutic approaches for PCOS treatment are provided by summarizing the role of apoptosis in PCOS therapy.
Collapse
Affiliation(s)
- Ling-Kun Yang
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Wan-Jing Ma
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Xiao Wang
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Huan-Ran Chen
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Ya-Nan Jiang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, State-Province Key Laboratories of Biomedicine- Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, PR China.
| | - Hui Sun
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, PR China.
| |
Collapse
|
3
|
Feng Y, Li W, Yuwen W, Xu R, Zhu C, Duan Z, Fan D. Expression, characterization and anti-colon cancer activity of recombinant ginseng peptides with amino acid tandem repeats. Protein Expr Purif 2025; 229:106663. [PMID: 39863192 DOI: 10.1016/j.pep.2025.106663] [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: 09/13/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/27/2025]
Abstract
Ginseng peptides, small molecule active ingredients in ginseng, are mainly extracted naturally or synthesized chemically, but high costs and difficulties hinder further research. In this study, a ginseng hexapeptide FKEHGY, named antitumor peptide 0601 (AT0601) and its five tandem sequence repeats AT0605, were expressed in Bacillus subtilis WB600 for the first time, and the bioactivity study showed that the anticancer activity of AT0605 was even significantly higher than that of AT0601 for colon cancer CT26 cells, with IC50s of 16.82 ± 1.3 μM (48 h) and 855.57 ± 6.04 μM (48 h), respectively, i.e. AT0605 achieves the same inhibition rate for CT26 cells at a concentration 50 times lower than that of AT0601. Similar to the ginseng peptide AT0601, recombinant AT0605 also inhibited cell growth by blocking cells in the G1 phase and activated the mitochondria-associated caspase pathway to induce apoptosis. In conclusion, all two kinds of the recombinant ginseng peptides could also inhibit cell proliferation through mechanisms such as inhibiting cell cycle arrest and inducing a decrease in mitochondrial membrane potential.
Collapse
Affiliation(s)
- Yu Feng
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710069, China
| | - Weina Li
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710069, China
| | - Weigang Yuwen
- Xi'an Gaint Biotechnology Co., Ltd., Xi'an, 710065, China
| | - Ru Xu
- Xi'an Gaint Biotechnology Co., Ltd., Xi'an, 710065, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710069, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710069, China
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710069, China.
| |
Collapse
|
4
|
Han H, Yang M, Wen Z, Mei F, Chen Q, Ma Y, Lai X, Zhang Y, Fang R, Yin T, Sun S, Wang X, Qi J, Lin H, Yang Y. Trametinib and M17, a novel small molecule inhibitor of AKT, display a synergistic antitumor effect in triple negative breast cancer cells through the AKT/mTOR and MEK/ERK pathways. Bioorg Chem 2025; 154:107981. [PMID: 39591692 DOI: 10.1016/j.bioorg.2024.107981] [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: 09/12/2024] [Revised: 11/02/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024]
Abstract
Triple negative breast cancer (TNBC) is associated with a poor prognosis and limited response to traditional chemotherapy, necessitating the exploration of novel treatment approaches. Recent researches have highlighted the interconnected roles of the PI3K/AKT pathway and MAPK pathway in TNBC cells, contributing to the efficacy of treatments. Therefore, the concurrent inhibition of both pathways presents a potential new therapeutic strategy for TNBC patients. This study aimed to evaluate the antitumor efficacy of M17, an AKT allosteric inhibitor and a new synthesized shikonin derivative, both alone and in combination with the MEK inhibitor trametinib. We applied various cellular assays and a subcutaneous 4T1 tumor bearing BALB/c mice model were utilized to assess the in vitro and in vivo antitumor effects. Computational docking and Bio-Layer Interferometry (BLI) were employed to investigate the binding of M17 with AKT. Additionally, flow cytometry, transwell assays, western blotting, and tumor xenograft assays were conducted to explore the potential synergistic mechanisms of the combined therapy. The results demonstrated that M17 exhibited moderate antitumor activity against TNBC cells, but significantly enhanced the apoptotic effects and inhibited proliferation and migration when combined with trametinib. Furthermore, the combination of M17 and trametinib showed even more pronounced antitumor activity in vivo. Mechanistically, the dual therapy synergistically suppressed TNBC by targeting the AKT/mTOR and MEK/ERK signaling pathways and inhibiting epithelial-mesenchymal transition. In conclusion, the findings suggested that the combination of M17 and trametinib holds promise as a synergistic treatment option for TNBC patients.
Collapse
Affiliation(s)
- Hongwei Han
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing 210013, China; State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Minkai Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Zhongling Wen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Feng Mei
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Qingqing Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Yudi Ma
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Xiaohui Lai
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Yahan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Rongjun Fang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Tongming Yin
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Shucun Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Xiaoming Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jinliang Qi
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Hongyan Lin
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Yonghua Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
5
|
Tian S, Goand UK, Paudel D, Le GV, Tiwari AK, Prabhu KS, Singh V. Processed Dietary Fiber Partially Hydrolyzed Guar Gum Increases Susceptibility to Colitis and Colon Tumorigenesis in Mice. RESEARCH SQUARE 2024:rs.3.rs-5522559. [PMID: 39711544 PMCID: PMC11661293 DOI: 10.21203/rs.3.rs-5522559/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The vital role of naturally occurring dietary fibers (DFs) in maintaining intestinal health has fueled the incorporation of isolated DFs into processed foods. A select group of soluble DFs, such as partially hydrolyzed guar gum (Phgg), are being promoted as dietary supplements to meet recommended DF intake. However, the potential effects of regular consumption of these processed DFs on gastrointestinal health remain largely unknown. The present study assessed the impact of Phgg on the development of intestinal inflammation and colitis-associated colon carcinogenesis (CAC). Wild-type C57BL/6 mice were fed isocaloric diets containing either 7.5% Phgg and 2.5% cellulose (Phgg group) or 10% cellulose (control) for four weeks. To induce colitis, a subgroup of mice from each group was switched to 1.4% dextran sulfate sodium (DSS) in drinking water for seven days. CAC was induced in another subgroup through a single dose of azoxymethane (AOM, 7.5 mg/kg i.p.) followed by three DSS/water cycles. To our surprise, Phgg feeding exacerbated DSS-induced colitis, as evidenced by body weight loss, disrupted colonic crypt architecture, and increased pro-inflammatory markers accompanied by a decrease in anti-inflammatory markers. Additionally, Phgg feeding led to increased colonic expression of genes promoting cell proliferation. Accordingly, extensive colon tumorigenesis was observed in Phgg-fed mice in the AOM/DSS model, whereas the control group exhibited no visible tumors. To investigate whether reducing Phgg has a distinct effect on colitis and CAC development, mice were fed a low-Phgg diet (2.5% Phgg). The low-Phgg group also exhibited increased colitis and tumorigenesis compared to the control, although the severity was markedly lower than in the regular Phgg (7.5%) group, suggesting a dose-dependent effect of Phgg in colitis and CAC development. Our study reveals that Phgg supplementation exacerbates colitis and promotes colon tumorigenesis, warranting further investigation into the potential gastrointestinal health risks associated with processed Phgg consumption.
Collapse
|
6
|
Alshehri B. Cytochrome c and cancer cell metabolism: A new perspective. Saudi Pharm J 2024; 32:102194. [PMID: 39564377 PMCID: PMC11570848 DOI: 10.1016/j.jsps.2024.102194] [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: 08/04/2024] [Accepted: 10/29/2024] [Indexed: 11/21/2024] Open
Abstract
Cytochrome c is a vital electron carrier in the mitochondrial respiratory chain. When the outer membrane of mitochondria becomes permeable, cytochrome c is discharged into the cytoplasm, where it initiates the intrinsic apoptosis pathway. The complex interaction between cytochrome c and apoptosis protease-activating factor-1 (Apaf-1) leads to the formation of the apoptosome and activation of a cascade of caspases, highlighting the critical role of cytochrome c in controlling cell death mechanisms. Additionally, cytochrome c undergoes post-translational modifications, especially phosphorylation, which intricately regulate its roles in both respiration and apoptosis. These modifications add layers of complexity to how cytochrome c effectively controls cellular functions. cytochrome c becomes a lighthouse in the intricate web of cancer, its expression patterns providing hints about prognosis and paths toward treatment. Reduced levels of cytochrome c have been observed in cancer tissues, indicating a potential inhibition of apoptosis. For instance, in glioma tissues, cytochrome c levels were lower compared to healthy tissues, and this reduction became more pronounced in advanced stages of the disease. However, the role of cytochrome c in cancer becomes more intricate as it becomes intertwined with the metabolic reprogramming of cancer cells. This suggests that cytochrome c plays a crucial role in tumor progression and resistance to treatment. Viewing cytochrome c as a molecular mosaic reveals that it is not merely a protein, but also a central player in determining cellular fate. This realization opens up exciting avenues for potential advancements in cancer diagnosis and treatment strategies. Despite the advancements made, the narrative surrounding cytochrome c remains incomplete, urging further exploration into its complexities and the biological implications linked to cancer. cytochrome c stands as a beacon of hope and a promising target for therapy in the battle against cancer, particularly due to its significant involvement in tumor metabolism. It holds the potential for a future where innovative solutions can be developed to address the intricate challenges of cellular fate. In this review, we have endeavored to illuminate the multifaceted domain of cytochrome c drawing connections among apoptosis, metabolic reprogramming, and the Warburg effect in the context of cancer.
Collapse
Affiliation(s)
- Bader Alshehri
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Almajmaah-11952, Saudi Arabia
| |
Collapse
|
7
|
Park WH. Propyl gallate induces human pulmonary fibroblast cell death through the regulation of Bax and caspase-3. Ann Med 2024; 56:2319853. [PMID: 38373208 PMCID: PMC10878342 DOI: 10.1080/07853890.2024.2319853] [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: 07/11/2023] [Accepted: 02/11/2024] [Indexed: 02/21/2024] Open
Abstract
Propyl gallate (PG) has been found to exert an inhibitory effect on the growth of different cell types, including lung cancer cells. However, little is known about the cytotoxicological effects of PG specifically on normal primary lung cells. The current study examined the cellular effects and cell death resulting from PG treatment in human pulmonary fibroblast (HPF) cells. DNA flow cytometry results demonstrated that PG (100-1,600 μM) had a significant impact on the cell cycle, leading to G1 phase arrest. Notably, 1,600 μM PG slightly increased the number of sub-G1 cells. Additionally, PG (400-1,600 μM) resulted in the initiation of cell death, a process that coincided with a loss of mitochondrial membrane potential (MMP; ΔΨm). This loss of MMP (ΔΨm) was evaluated using a FACS cytometer. In PG-treated HPF cells, inhibitors targeting pan-caspase, caspase-3, caspase-8, and caspase-9 showed no significant impact on the quantity of annexin V-positive and MMP (ΔΨm) loss cells. The administration of siRNA targeting Bax or caspase-3 demonstrated a significant attenuation of PG-induced cell death in HPF cells. However, the use of siRNAs targeting p53, Bcl-2, or caspase-8 did not exhibit any notable effect on cell death. Furthermore, none of the tested MAPK inhibitors, including MEK, c-Jun N-terminal kinase (JNK), and p38, showed any impact on PG-induced cell death or the loss of MMP (ΔΨm) in HPF cells. In conclusion, PG induces G1 phase arrest of the cell cycle and cell death in HPF cells through apoptosis and/or necrosis. The observed HPF cell death is mediated by the modulation of Bax and caspase-3. These findings offer insights into the cytotoxic and molecular effects of PG on normal HPF cells.
Collapse
Affiliation(s)
- Woo Hyun Park
- Department of Physiology, Medical School, Jeonbuk National University, Jeonju, Jeollabuk, Republic of Korea
| |
Collapse
|
8
|
Saleh DO, Abo El Nasr NME, Hussien YA, El-Baset MA, Ahmed KA. Cyclophosphamide-induced testicular injury: the role of chrysin in mitigating iron overload and ferroptosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03519-4. [PMID: 39565397 DOI: 10.1007/s00210-024-03519-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 10/06/2024] [Indexed: 11/21/2024]
Abstract
This study evaluated the beneficial effects of chrysin against cyclophosphamide (CP)-induced testicular toxicity in rats across several parameters, including hormones, oxidative stress, inflammation, apoptosis, and protein expression. Rats were pretreated with oral doses of chrysin at 25, 50, or 100 mg/kg daily for 7 days. On the 8th day, all groups except controls received CP (200 mg/kg) injection. Chrysin doses continued for 7 more days. Hormones, oxidative stress markers, inflammatory cytokines, apoptosis regulators, and iron regulatory proteins were assessed. CP decreased testosterone, inhibin B, GSH, and GPx4 and increased FSH, cholesterol, MDA, IL-6, and BAX. It also drastically reduced TfR1, liprin, and IREB2. Chrysin dose-dependently counteracted these effects. The highest 100 mg/kg chrysin dose increased testosterone, inhibin B, GSH, GPx4, BCL2, TfR1, liprin, and IREB2 while decreasing FSH, cholesterol, MDA, IL-6, and BAX close to control levels. There were also significant incremental benefits for testosterone, inhibin B, and other parameters with higher chrysin doses. Chrysin dose-dependently attenuated CP-induced hormonal dysfunction, oxidative stress, inflammation, apoptosis, and iron-regulatory protein suppression. The maximum dose showed the most optimal protective effects in restoring the testicular toxicity markers. These results validate the promising spermatoprotective properties of chrysin against chemotherapeutic germ cell damage.
Collapse
Affiliation(s)
- Dalia O Saleh
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Nesma M E Abo El Nasr
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt.
| | - Yosra A Hussien
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Marawan Abd El-Baset
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Kawkab A Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
9
|
Sun Y, Sun J, Gao X, Shi T, Wang M. Identification of Potential Biomarkers in Papillary Thyroid Carcinoma Based on Proteomics. Onco Targets Ther 2024; 17:905-923. [PMID: 39513013 PMCID: PMC11542476 DOI: 10.2147/ott.s465636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
Background To identify biomarkers of papillary thyroid carcinoma (PTC) and explore the possible pathogenic mechanism. Methods This study included five patients with PTC. Protein expression of cancer tissues and adjacent normal thyroid tissues from each patient were analyzed by TMT proteomics technology. Differentially expressed proteins were identified, and functional annotation of differentially expressed proteins was performed by bioinformatics and pathway enrichment analysis. Results A total of 639 differentially expressed proteins were identified, including 278 upregulated and 361 downregulated proteins. Six upregulated proteins were identified as potential specific markers of PTC. Conclusion Differentially expressed proteins may represent new molecular markers of PTC. These differentially expressed proteins and the related pathways may provide new insights into the pathogenic mechanisms of PTC.
Collapse
Affiliation(s)
- Yu Sun
- Department of Thyroid Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, People’s Republic of China
| | - Jiaxuan Sun
- Department of Thyroid Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, People’s Republic of China
| | - Xiaona Gao
- Department of Thyroid Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, People’s Republic of China
| | - Tiefeng Shi
- Department of Thyroid Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, People’s Republic of China
| | - Maoqing Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
10
|
Cai JL, Wang JJ, Zhang Y, Gao H, Huang W, Cai YJ, Jia WX, Chen X, Sun HY. Characterization, expression and functional analysis of Hsp40 during LPS challenge in blood parrot Amphilophus citrinellus ×Vieja melanura. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109910. [PMID: 39299406 DOI: 10.1016/j.fsi.2024.109910] [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: 07/22/2024] [Revised: 08/27/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
Heat shock protein 40 belonging to heat shock protein family plays an important role in the immune responses of organisms. In this study, the full length cDNA of Hsp40 was 2426 bp including a 1368 bp open reading frame (ORF) encoding 455 amino acids with a molecular weight of 49.16 kDa and a theoretical isoelectric point of 9.34 in blood parrot Vieja synspila ♀ × Amphilophus citrinellus ♂, an important ornamental fish in China. It had three conserved domains DnaJ, CRR and DnaJ_C. Phylogenetic analysis showed that the sequence of Hsp40 among species was conserved, and the blood parrot Hsp40 was closely related to Neolamprologus brichardi. Blood parrot Hsp40 mRNA could be detected in all of the tissues examined and mainly distributed in the cytoplasm. The expression of Hsp40 was upregulated during lipopolysaccharide (LPS) challenge. Upregulated Hsp40 inhibited the activity of nuclear factor κB (NF-κB) and activated protein 1 (AP-1) and reduced the ratio of Bax/Bcl-2 mRNA expression. This study provides a theoretical basis for further exploring the role of Hsp40 gene in the anti-bacterial immunity of blood parrot.
Collapse
Affiliation(s)
- Jie-Li Cai
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jun-Jie Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; School of Life Sciences, South China Normal University, Guangzhou, Guangdong, China
| | - Yue Zhang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Hui Gao
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wei Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yi-Jie Cai
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wei-Xin Jia
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong, China
| | - Xiao Chen
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China.
| | - Hong-Yan Sun
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China.
| |
Collapse
|
11
|
Kosanam S, Pasupula R. Cardioprotective effects of cinnamoyl imidazole on apoptosis and oxidative stress in hypoxia/reoxygenation-induced H9C2 cell lines. Life Sci 2024; 359:123189. [PMID: 39481831 DOI: 10.1016/j.lfs.2024.123189] [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: 09/28/2024] [Revised: 10/21/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND This study explored the effects of cinnamoyl imidazole on alleviating oxidative stress and apoptosis in hypoxia/reoxygenation (H/R)-induced H9C2 cells, using computational analysis with in-vitro validation. METHODS Computational techniques, including SwissADME and Swiss Target Prediction, were employed to predict the ADME properties and to identify targets of cinnamoyl imidazole. Differential gene expression (DEG) analysis was conducted on myocardial infarction (MI) datasets obtained from the Gene Expression Omnibus. Gene enrichment and molecular simulation studies were done to focus on apoptotic pathways. The computational findings were validated through In vitro experiments on H9C2 cardiomyocytes subjected to 8 h of hypoxia followed by 24 h of reoxygenation. Antioxidant enzyme levels (catalase, GST, GSH-Px, and SOD), mitochondrial membrane potential (ΔΨm), caspase-3 activity, and the expression of CASP3, MAPK8, JAK2, and BCL2L1 were assessed. RESULTS Cinnamoyl imidazole has demonstrated favourable pharmacokinetic properties, characterized by high gastrointestinal absorption and low toxicity with negative toxicity for organ endpoints. Molecular docking studies revealed the strong binding affinities for CASP3, MAPK8, and JAK2. In vitro results showed a significant increase in cell viability (94.7 % at 10 μM, p < 0.001) and antioxidant enzyme activity, along with a 64.3 % reduction in caspase-3 activity at 1000 μM (p < 0.01). Cinnamoyl imidazole treatment preserved mitochondrial membrane potential, downregulated pro-apoptotic genes CASP3 and MAPK8, and upregulated the anti-apoptotic gene BCL2L1. CONCLUSION Cinnamoyl imidazole effectively mitigates oxidative stress and apoptosis in H/R-induced H9C2 cells, enhancing cell viability and antioxidant defenses while maintaining mitochondrial integrity.
Collapse
Affiliation(s)
- Sreya Kosanam
- Department of Pharmacology, College of Pharmacy, Koneru Lakshmaiah Education Foundation, KL deemed to be University, Green Fields, Vaddeswaram, Andhra Pradesh, India
| | - Rajeshwari Pasupula
- Department of Pharmacology, College of Pharmacy, Koneru Lakshmaiah Education Foundation, KL deemed to be University, Green Fields, Vaddeswaram, Andhra Pradesh, India.
| |
Collapse
|
12
|
Tucker N, Reddien P, Hersh B, Lee D, Liu MHX, Horvitz HR. The pro-apoptotic function of the C. elegans BCL-2 homolog CED-9 requires interaction with the APAF-1 homolog CED-4. SCIENCE ADVANCES 2024; 10:eadn0325. [PMID: 39383227 DOI: 10.1126/sciadv.adn0325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 09/04/2024] [Indexed: 10/11/2024]
Abstract
In Caenorhabditis elegans, apoptosis is inhibited by the BCL-2 homolog CED-9. Although canonically anti-apoptotic, CED-9 has a poorly understood pro-apoptotic function. CED-9 is thought to inhibit apoptosis by binding to and inhibiting the pro-apoptotic C. elegans APAF-1 homolog CED-4. We show that CED-9 or CED-4 mutations located in their CED-9-CED-4 binding regions reduce apoptosis without affecting the CED-9 anti-apoptotic function. These mutant CED-9 and CED-4 proteins are defective in a CED-9-CED-4 interaction in vitro and in vivo, revealing that the known CED-9-CED-4 interaction is required for the pro-apoptotic but not for the anti-apoptotic function of CED-9. The pro-apoptotic CED-9-CED-4 interaction occurs at mitochondria. In mammals, BCL-2 family members can activate APAF-1 via cytochrome c release from mitochondria. The conserved role of mitochondria in CED-9/BCL-2-dependent CED-4/APAF-1 activation is notable and suggests that understanding how CED-9 promotes apoptosis in C. elegans could inform the understanding of mammalian apoptosis and how disruptions of apoptosis promote certain human disorders.
Collapse
Affiliation(s)
- Nolan Tucker
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter Reddien
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bradley Hersh
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dongyeop Lee
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mona H X Liu
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - H Robert Horvitz
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
13
|
Wang C, Xia W. Proanthocyanidin Regulates NETosis and Inhibits the Growth and Proliferation of Liver Cancer Cells - In Vivo, In Vitro and In Silico Investigation. Cell Biochem Biophys 2024:10.1007/s12013-024-01557-6. [PMID: 39382828 DOI: 10.1007/s12013-024-01557-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Liver cancer ranks third in global cancer-related mortality, with about 700,000 deaths recorded yearly, making it one of the most common cancers worldwide. Even though prognoses differ according to the severity of the diseases, many patients now exhibit an increased life cycle since the implementation of chemotherapy. In the current study, we investigated the effect of proanthocyanidin ‒a polyphenol molecule found in many plants‒ on the proliferation and invasion of liver cancer cells. In particular, we determined the effect of proanthocyanidin on the serum levels of four strategic liver cancer target, TNFα, IL-6, cfDNA, and IL-1β. Further molecular insight on the inhibitory mechanism of proanthocyanidin against TNFα, IL-6, and IL-1β was obtained via molecular docking, molecular dynamics simulations and binding free energy calculations. Results showed that proanthocyanidin inhibited the growth of HepG2 and HEP3B cells, and effectively reduced clonogenic survival and invasion potential when compared to control cells. Proanthocyanidin was also found to suppress the expression of Bcl-2 (26 kDa) protein in HepG2 cells, while increasing the expression of Bax (21 kDa). Molecular dynamics (MD) and thermodynamic binding free energy calculations showed that proanthocyanidin maintained stable binding within the active site of target proteins across the entire 100 ns MD simulation period, and its binding affinity outscored respective control molecules.In conclusion, the multifaceted analysis showcased in this study demonstrated promising anti-cancer effect of proanthocyanidin on HepG2 and HEP3B cancer cells, highlighting its potential as a viable liver cancer therapeutic alternative.
Collapse
Affiliation(s)
- Chenhui Wang
- Department of Pharmaceutical, Brain Hospital of Hunan Province, The Second People's Hospital of Hunan Province, No. 427, Section 3, Furong Middle Road, Changsha, 410007, China
| | - Wu Xia
- Department of Pharmaceutical, Brain Hospital of Hunan Province, The Second People's Hospital of Hunan Province, No. 427, Section 3, Furong Middle Road, Changsha, 410007, China.
| |
Collapse
|
14
|
Donadkar M, Kumar B, Singh SK, Chandra P, Dangi P, Gawai M, Jackson A, Jasrotia N, Sharma S, Chouhan V, Patra MK, Khan MH. Morpho-molecular evaluation for developmental competence of oocytes retrieved through transvaginal ovum pick-up from FSH-stimulated Tharparkar donor cows ( Bos indicus). ZYGOTE 2024; 32:376-385. [PMID: 39470002 DOI: 10.1017/s0967199424000376] [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] [Indexed: 10/30/2024]
Abstract
The study was conducted on indigenous Tharparkar cow (Bos indicus) to evaluate FSH stimulation on follicular attributes, oocyte recovery and morpho-molecular developmental competence parameters concerning oocyte quality. A total of 20 OPU sessions were performed, which included 10 sessions in each FSH stimulated at the dose of 130 µg divided into four sub-doses and non-stimulated. Findings on the size of follicles having ≥6 mm showed a significantly higher, however an opposite trend was observed in the case of smaller sized follicle (<6 mm) between stimulated and non-stimulated respectively. The stimulated cows had a significantly higher number as well as the percentage of oocytes of Grade A, having a diameter ≥120 µm and BCB+VE as compared to the non-stimulated cows. The relative mRNA expression profile of GDF9, BMP15, PCNA and BCL-2 genes was higher and BAX was lower in the FSH-stimulated cow. These results indicated that FSH stimulation before OPU in Bos indicus cows has a significant impact on follicle size, oocyte yield, recovery, and their quality with respect to COC's, diameter and BCB+VE oocytes. Further, a significant increase in the relative mRNA expression levels of GDF9, BMP15 and PCNA genes in the FSH-stimulated group suggests that FSH plays a key role in modulating the expression of these important candidate genes and thus influencing oocyte quality. The higher mRNA expression of BCL-2 genes and concomitantly lower expression of BAX gene in FSH Stimulated cows indicates the protective role of these genes and preventing programmed cell death and thus promoting cell survival, quality and embryo development.
Collapse
Affiliation(s)
- Manoj Donadkar
- Animal Reproduction Division, ICAR- Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar243122, India
| | - Brijesh Kumar
- Animal Reproduction Division, ICAR- Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar243122, India
| | - Sanjay Kumar Singh
- Animal Reproduction Division, ICAR- Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar243122, India
| | - Pradeep Chandra
- Animal Reproduction Division, ICAR- Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar243122, India
| | - Pradeep Dangi
- Animal Reproduction Division, ICAR- Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar243122, India
| | - Mohan Gawai
- Animal Reproduction Division, ICAR- Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar243122, India
| | - Amala Jackson
- Animal Reproduction Division, ICAR- Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar243122, India
| | - Nancy Jasrotia
- Animal Reproduction Division, ICAR- Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar243122, India
| | - Shweta Sharma
- Division of Physiology and Climatology, ICAR- IVRI, Izatnagar243122, India
| | - Vikrant Chouhan
- Division of Physiology and Climatology, ICAR- IVRI, Izatnagar243122, India
| | - M K Patra
- Animal Reproduction Division, ICAR- Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar243122, India
| | - Meraj Haider Khan
- Animal Reproduction Division, ICAR- Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar243122, India
| |
Collapse
|
15
|
Rajesh R U, Sangeetha D. Therapeutic potentials and targeting strategies of quercetin on cancer cells: Challenges and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155902. [PMID: 39059266 DOI: 10.1016/j.phymed.2024.155902] [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: 03/07/2024] [Revised: 07/08/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Every cell in the human body is vital because it maintains equilibrium and carries out a variety of tasks, including growth and development. These activities are carried out by a set of instructions carried by many different genes and organized into DNA. It is well recognized that some lifestyle decisions, like using tobacco, alcohol, UV, or multiple sexual partners, might increase one's risk of developing cancer. The advantages of natural products for any health issue are well known, and researchers are making attempts to separate flavonoid-containing substances from plants. Various parts of plants contain a phenolic compound called flavonoid. Quercetin, which belongs to the class of compounds known as flavones with chromone skeletal structure, has anti-cancer activity. PURPOSE The study was aimed at investigating the therapeutic action of the flavonoid quercetin on various cancer cells. METHODS The phrases quercetin, anti-cancer, nanoparticles, and cell line were used to search the data using online resources such as PubMed, and Google Scholar. Several critical previous studies have been included. RESULTS Quercetin inhibits various dysregulated signaling pathways that cause cancer cells to undergo apoptosis to exercise its anticancer effects. Numerous signaling pathways are impacted by quercetin, such as the Hedgehog system, Akt, NF-κB pathway, downregulated mutant p53, JAK/STAT, G1 phase arrest, Wnt/β-Catenin, and MAPK. There are downsides to quercetin, like hydrophobicity, first-pass effect, instability in the gastrointestinal tract, etc., because of which it is not well-established in the pharmaceutical industry. The solution to these drawbacks in the future is using bio-nanomaterials like chitosan, PLGA, liposomes, and silk fibroin as carriers, which can enhance the target specificity of quercetin. The first section of this review covers the specifics of flavonoids and quercetin; the second section covers the anti-cancer activity of quercetin; and the third section explains the drawbacks and conjugation of quercetin with nanoparticles for drug delivery by overcoming quercetin's drawback. CONCLUSIONS Overall, this review presented details about quercetin, which is a plant derivative with a promising molecular mechanism of action. They inhibit cancer by various mechanisms with little or no side effects. It is anticipated that plant-based materials will become increasingly relevant in the treatment of cancer.
Collapse
Affiliation(s)
- Udaya Rajesh R
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India
| | - Dhanaraj Sangeetha
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India.
| |
Collapse
|
16
|
Einafshar E, Javid H, Amiri H, Akbari-Zadeh H, Hashemy SI. Curcumin loaded β-cyclodextrin-magnetic graphene oxide nanoparticles decorated with folic acid receptors as a new theranostic agent to improve prostate cancer treatment. Carbohydr Polym 2024; 340:122328. [PMID: 38857995 DOI: 10.1016/j.carbpol.2024.122328] [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: 02/26/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/12/2024]
Abstract
This article presents a novel approach to treating prostate cancer using a nanocarrier composed of folic acid (FA), β-cyclodextrin (β-CD), and magnetic graphene oxide (MGO) as a theranostic agent. The carrier is designed to improve the solubility and bioavailability of curcumin, a potential therapeutic substance against prostate cancer. Folic acid receptors overexpressed on the surface of solid tumors, including prostate cancer, may facilitate targeted drug delivery to tumor cells while avoiding nonspecific effects on healthy tissues. The anticancer efficacy of Folic acid-curcumin@β-CD-MGO in vitro was also examined on LNCaP (an androgen-dependent) and PC3 (an androgen-independent) prostate cancer cells. The relaxivity of nanoparticles in MRI images was also investigated as a diagnostic factor. The results showed a concentration-dependent inhibitory effect on cell proliferation, induction of oxidative damage, and apoptotic effects. Also, nanoparticle relaxometry shows that this agent can be used as a negative contrast agent in MRI images. Overall, this study represents a promising theranostic agent to improve the delivery and trace of curcumin and enhance its therapeutic potential in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Elham Einafshar
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Javid
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | - Hamed Amiri
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hadi Akbari-Zadeh
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
Li M, Wang H, Bai Y, Xiong F, Wu S, Bi Q, Qiao Y, Zhang Y, Li X, Feng L, Guo DA. Pharmacodynamical research of extracts and compounds in traditional Chinese medicines for Parkinson's disease. Fitoterapia 2024; 177:106086. [PMID: 38897243 DOI: 10.1016/j.fitote.2024.106086] [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: 05/12/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease (AD). Currently, there is no cure for PD, and medications can only control the progression of the disease. Various experimental studies have shown the significant efficacy of TCM in treating PD, and combination with western medicine can enhance the effects and reduce toxicity. Thus, exploring effective anti-PD compounds from TCM has become a popular research fields. This review summarizes commonly used TCM extracts and natural products for the treatment of PD, both domestically and internationally. Furthermore, it delves into various mechanisms of TCM in treating PD, such as anti-oxidative stress, anti-inflammatory, anti-apoptotic, improve mitochondrial dysfunction, inhibits α-synuclein (α-Syn) misfolding and aggregation, regulating neurotransmitters, regulates intestinal flora, enhances immunity, and so on. The results reveal that most TCMs exert their neuroprotective effects through anti-inflammatory and anti-oxidative stress actions, thereby slowing down the progression of the disease. These TCM may hold the key to improving PD therapy and have tremendous potential to be developed as novel anti-PD drugs.
Collapse
Affiliation(s)
- Mengmeng Li
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hanze Wang
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuxin Bai
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fuyu Xiong
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shifei Wu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qirui Bi
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yajun Qiao
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yan Zhang
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaolan Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lin Feng
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - De-An Guo
- College of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
18
|
Lee SE, Lee HB, Yoon JW, Park HJ, Kim SH, Han DH, Lim ES, Kim EY, Park SP. Rapamycin treatment during prolonged in vitro maturation enhances the developmental competence of immature porcine oocytes. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2024; 66:905-919. [PMID: 39398303 PMCID: PMC11466741 DOI: 10.5187/jast.2023.e101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/25/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2024]
Abstract
Porcine oocytes undergo in vitro maturation (IVM) for 42-44 h. During this period, most oocytes proceed to metaphase and then to pro-metaphase if the nucleus has sufficiently matured. Forty-four hours is sufficient for oocyte nuclear maturation but not for full maturation of the oocyte cytoplasm. This study investigated the influences of extension of the IVM duration with rapamycin treatment on molecular maturation factors. The phospho-p44/42 mitogen-activated protein kinase (MAPK) level was enhanced in comparison with the total p44/42 MAPK level after 52 h of IVM. Oocytes were treated with and without 10 μM rapamycin (10 R and 0 R, respectively) and examined after 52 h of IVM, whereas control oocytes were examined after 44 h of IVM. Phospho-p44/42 MAPK activity was upregulated the 10 R and 0 R oocytes than in control oocytes. The expression levels of maternal genes were highest in 10 R oocytes and were higher in 0 R oocytes than in control oocytes. Reactive oxygen species (ROS) activity was dramatically increased in 0 R oocytes but was similar in 10 R and control oocytes. The 10 R group exhibited an increased embryo development rate, a higher total cell number per blastocyst, and decreased DNA fragmentation. The mRNA level of development-related (POU5F1 and NANOG) mRNA, oocyte-apoptotic (BCL2L1) genes were highest in 10 R blastocysts. These results suggest that prolonged IVM duration with rapamycin treatment represses ROS production and increases expression of molecular maturation factors. Therefore, this is a good strategy to enhance the developmental capacity in porcine oocytes.
Collapse
Affiliation(s)
- Seung-Eun Lee
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
- Subtropical Livestock Research Institute,
National Institute of Animal Science, RDA, Jeju 63242,
Korea
| | - Han-Bi Lee
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Jae-Wook Yoon
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
| | - Hyo-Jin Park
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
| | - So-Hee Kim
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
| | - Dong-Hun Han
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Eun-Seo Lim
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
| | - Eun-Young Kim
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Faculty of Biotechnology, College of
Applied Life Sciences, Jeju National University, Jeju 63243,
Korea
- Mirae Cell Bio, Seoul 04795,
Korea
| | - Se-Pill Park
- Stem Cell Research Center, Jeju National
University, Jeju 63243, Korea
- Mirae Cell Bio, Seoul 04795,
Korea
- Department of Bio Medical Informatic,
College of Applied Life Sciences, Jeju National University,
Jeju 63242, Korea
| |
Collapse
|
19
|
Kaur H, Carrillo O, Garcia I, Ramos I, St Vallier S, De La Torre P, Lopez A, Keniry M, Bazan D, Elizondo J, Grishma KC, Ann MacMillan-Crow L, Gilkerson R. Differentiation activates mitochondrial OPA1 processing in myoblast cell lines. Mitochondrion 2024; 78:101933. [PMID: 38986925 PMCID: PMC11390305 DOI: 10.1016/j.mito.2024.101933] [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: 09/28/2023] [Revised: 07/03/2024] [Accepted: 07/07/2024] [Indexed: 07/12/2024]
Abstract
Mitochondrial optic atrophy-1 (OPA1) plays key roles in adapting mitochondrial structure to bioenergetic function. When transmembrane potential across the inner membrane (Δψm) is intact, long (L-OPA1) isoforms shape the inner membrane through membrane fusion and the formation of cristal junctions. When Δψm is lost, however, OPA1 is cleaved to short, inactive S-OPA1 isoforms by the OMA1 metalloprotease, disrupting mitochondrial structure and priming cellular stress responses such as apoptosis. Previously, we demonstrated that L-OPA1 of H9c2 cardiomyoblasts is insensitive to loss of Δψm via challenge with the protonophore carbonyl cyanide chlorophenyl hydrazone (CCCP), but that CCCP-induced OPA1 processing is activated upon differentiation in media with low serum supplemented with all-trans retinoic acid (ATRA). Here, we show that this developmental induction of OPA1 processing in H9c2 cells is independent of ATRA; moreover, pretreatment of undifferentiated H9c2s with chloramphenicol (CAP), an inhibitor of mitochondrial protein synthesis, recapitulates the Δψm-sensitive OPA1 processing observed in differentiated H9c2s. L6.C11 and C2C12 myoblast lines display the same developmental and CAP-sensitive induction of OPA1 processing, demonstrating a general mechanism of OPA1 regulation in mammalian myoblast cell settings. Restoration of CCCP-induced OPA1 processing correlates with increased apoptotic sensitivity. Moreover, OPA1 knockdown indicates that intact OPA1 is necessary for effective myoblast differentiation. Taken together, our results indicate that a novel developmental mechanism acts to regulate OMA1-mediated OPA1 processing in myoblast cell lines, in which differentiation engages mitochondrial stress sensing.
Collapse
Affiliation(s)
- Harpreet Kaur
- School of Integrative Biological & Chemical Sciences, The University of Texas Rio Grande Valley, United States
| | - Omar Carrillo
- School of Integrative Biological & Chemical Sciences, The University of Texas Rio Grande Valley, United States
| | - Iraselia Garcia
- School of Integrative Biological & Chemical Sciences, The University of Texas Rio Grande Valley, United States; Department of Biology, South Texas College, United States
| | - Isaiah Ramos
- School of Integrative Biological & Chemical Sciences, The University of Texas Rio Grande Valley, United States
| | - Shaynah St Vallier
- School of Integrative Biological & Chemical Sciences, The University of Texas Rio Grande Valley, United States
| | - Patrick De La Torre
- School of Integrative Biological & Chemical Sciences, The University of Texas Rio Grande Valley, United States
| | - Alma Lopez
- School of Integrative Biological & Chemical Sciences, The University of Texas Rio Grande Valley, United States
| | - Megan Keniry
- School of Integrative Biological & Chemical Sciences, The University of Texas Rio Grande Valley, United States
| | - Daniel Bazan
- School of Integrative Biological & Chemical Sciences, The University of Texas Rio Grande Valley, United States
| | - Jorge Elizondo
- School of Integrative Biological & Chemical Sciences, The University of Texas Rio Grande Valley, United States
| | - K C Grishma
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, United States
| | - Lee Ann MacMillan-Crow
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, United States
| | - Robert Gilkerson
- School of Integrative Biological & Chemical Sciences, The University of Texas Rio Grande Valley, United States; Medical Laboratory Sciences/Health & Biomedical Sciences, The University of Texas Rio Grande Valley, United States.
| |
Collapse
|
20
|
Krakowczyk M, Bragoszewski P. Monitoring retro-translocation of proteins from the mitochondrial intermembrane space. Methods Enzymol 2024; 707:173-208. [PMID: 39488374 DOI: 10.1016/bs.mie.2024.07.047] [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] [Indexed: 11/04/2024]
Abstract
Mitochondria play multiple essential roles in eukaryotic cells. To perform their functions, mitochondria require an adequate supply of externally produced proteins and an intact two-membrane structure. The structure of mitochondrial membranes separates these organelles from their cytosolic environment, with proteins that make up the mitochondrial proteome either being embedded into or enveloped by these membranes. From the experimental point of view, the structural properties of mitochondria contribute to the relative ease of isolating these organelles from other cellular components. The ability to isolate intact mitochondria and analyze them in a well-controlled environment opens up the possibility of tracking any proteins that enter or escape the membrane-formed enclosure. This chapter discusses assays that monitor the movement of proteins out of mitochondria through intact membranes. These protocols provide insight into the mechanisms behind mitochondrial protein quality control. It was discovered that the retro-translocation of IMS proteins regulates the protein content of this specific sub-compartment of the organelle. Additionally, proteins can move out of the mitochondria to resolve failed import events. Assays based on isolated mitochondria precisely tackle such intricate 'dance' of proteins crossing mitochondrial membranes during import and export, maintaining the dynamics of the organellar proteome.
Collapse
Affiliation(s)
- Magda Krakowczyk
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Bragoszewski
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
21
|
Qiao M, Zeng C, Liu C, Lei Z, Liu B, Xie H. The advancement of siRNA-based nanomedicine for tumor therapy. Nanomedicine (Lond) 2024; 19:1841-1862. [PMID: 39145477 PMCID: PMC11418284 DOI: 10.1080/17435889.2024.2377062] [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: 03/05/2024] [Accepted: 07/03/2024] [Indexed: 08/16/2024] Open
Abstract
Small interfering RNA (siRNA) has been proved to be able to effectively down-regulate gene expression through the RNAi mechanism. Thus, siRNA-based drugs have become one of the hottest research directions due to their high efficiency and specificity. However, challenges such as instability, off-target effects and immune activation hinder their clinical application. This review explores the mechanisms of siRNA and the challenges in siRNA-based tumor therapy. It highlights the use of various nanomaterials - including lipid nanoparticles, polymeric nanoparticles and inorganic nanoparticles - as carriers for siRNA delivery in different therapeutic modalities. The application strategies of siRNA-based nanomedicine in chemotherapy, phototherapy and immunotherapy are discussed in detail, along with recent clinical advancements. Aiming to provide insights for future research and therapeutic approaches.
Collapse
Affiliation(s)
- Muchuan Qiao
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, Hunan, 421001, China
| | - Chenlu Zeng
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, Hunan, 421001, China
| | - Changqing Liu
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, Hunan, 421001, China
| | - Ziwei Lei
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, Hunan, 421001, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Hailong Xie
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
22
|
Odetayo AF, Akhigbe RE, Hamed MA, Balogun ME, Oluwole DT, Olayaki LA. Omega-3 fatty acids abrogates oxido-inflammatory and mitochondrial dysfunction-associated apoptotic responses in testis of tamoxifen-treated rats. Front Nutr 2024; 11:1443895. [PMID: 39149552 PMCID: PMC11324566 DOI: 10.3389/fnut.2024.1443895] [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: 06/05/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024] Open
Abstract
Background Tamoxifen (TAM) is a widely used drug in patients with gynecomastia and breast cancer. TAM exerts its anticancer effects via its antiestrogenic activities. Unfortunately, TAM has been reported to exert gonadotoxic effects on male testes. Therefore, this study was designed to explore the possible associated mechanisms involved in TAM-induced testicular dysfunction and the possible ameliorative effects of omega-3 fatty acids (O3FA). Methodology Animals were randomly divided into control, O3FA, TAM, and TAM + O3FA. All treatment lasted for 28 days. Results TAM exposure impaired sperm qualities (count, motility, and normal morphology) and decreased testicular 3β-HSD and 17β-HSD. It was accompanied by a decline in serum testosterone and an increase in estradiol, luteinizing and follicle-stimulating hormones. These observed alterations were associated with an increase in testicular injury markers, oxido-inflammatory response, and mitochondria-mediated apoptosis. These observed alterations were ameliorated by O3FA treatments. Conclusions O3FA ameliorated TAM-induced testicular dysfunction in male Wistar rats by modulating XO/UA and Nrf2/NF-kb signaling and cytochrome c-mediated apoptosis in TAM-treated rats.
Collapse
Affiliation(s)
- Adeyemi Fatai Odetayo
- Department of Physiology, Faculty of Basic Medical Sciences, Federal University of Health Sciences, Ila Orangun, Nigeria
| | - Roland Eghoghosoa Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Moses Agbomhere Hamed
- Department of Medical Laboratory Science, Afe Babalola University, Ado Ekiti, Nigeria
- The Brainwill Laboratories and Biomedical Services, Osogbo, Nigeria
| | - Morufu Eyitayo Balogun
- Department of Physiology, Faculty of Basic Medical Sciences, Federal University of Health Sciences, Ila Orangun, Nigeria
| | | | | |
Collapse
|
23
|
Wang S, Bai H, Liu T, Yang J, Wang Z. Optimization of concentrations of different n-3PUFAs on antioxidant capacity in mouse hepatocytes. Lipids Health Dis 2024; 23:214. [PMID: 38982376 PMCID: PMC11232338 DOI: 10.1186/s12944-024-02202-0] [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: 03/20/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs), mainly including α-linolenic acid (ALA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), possess antioxidant properties and play a crucial role in growth and development. However, the combined effects of ALA, EPA, and DHA at different concentrations have rarely been reported. This work explored the effects of EPA, ALA, and DHA on the viability and antioxidant capacity of mouse hepatocytes, with the objective of enhancing the antioxidant capacity. Within the appropriate concentration range, cell viability and the activity of glutathione S-transferase, superoxide dismutase, and catalase were increased, while the oxidation products of malondialdehyde and the level of intracellular reactive oxygen species were obviously reduced. Thus, oxidative stress was relieved, and cellular antioxidant levels were improved. Finally, response surface optimization was carried out for EPA, ALA, and DHA, and the model was established. The antioxidant capacity of the cells was highest at EPA, ALA, and DHA concentrations of 145.46, 405.05, and 551.52 µM, respectively. These findings lay the foundation for further exploration of the interactive mechanisms of n-3 PUFAs in the body, as well as their applications in nutraceutical food.
Collapse
Affiliation(s)
- Shuting Wang
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, China
| | - Huasong Bai
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, China
| | - Tong Liu
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, China
| | - Jiayi Yang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Zhanzhong Wang
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, China.
| |
Collapse
|
24
|
Yang Z, Fang X, Zhang Y, Bai Y, Zhao L, Zhou X. Ginkgolic acids induce liver injury in mice through cell cycle arrest and immune stress under specific condition. Toxicon 2024; 245:107788. [PMID: 38823652 DOI: 10.1016/j.toxicon.2024.107788] [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: 03/12/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Ginkgo biloba L. is a valuable plant, which can be used for medicine, food and ornamental purposes. Despite the above benefits, the components of ginkgolic acids (GA) in ginkgo are considered to cause allergies, embryotoxicity, liver damage and some other adverse reactions. However, the mechanism of GA induced liver injury is still unclear. In this study, we developed an acute liver injury model induced by GA in mice, and investigated the mechanism of GA induced liver injury from the perspectives of oxidative stress, steatosis, apoptosis, and immune response. Intraperitoneal injection of GA (400 mg/kg) can cause liver damage. The levels of serum transaminase, oxidation and triglycerides were increased, liver fibrosis, hepatocyte apoptosis, G2/M phase arrest of the hepatic cell cycle and monocyte infiltration in the liver were detected in GA-treated mice. Flow cytometry analysis of cells separated from the spleen showed that the proportion of Th1 and Th17 cells were increased, and the proportion of Th2 cells were decreased in GA-treated mice. The rise in Th1/Th2 ratio and Th17 cell ratio usually cause inflammatory problems. At the same time, cleaved Caspase-8 and Caspase-3 were detected in hepatocytes, indicating that GA may induce apoptosis through FADD pathway. Although GA is capable of causing the above problems, the inflammation and damage in liver tissue are not severe and there are certain individual differences. Our study reveals the potential hepatotoxicity of GA in ginkgo and its mechanism of action, providing a new perspective for the intervention and prevention of ginkgo toxicity.
Collapse
Affiliation(s)
- Zhiqing Yang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Xianying Fang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China.
| | - Yiwei Zhang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Yun Bai
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Linguo Zhao
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Xinhu Zhou
- Yanghe Distillery Co. Ltd, Suqian, 223800, China.
| |
Collapse
|
25
|
Nag N, Ray T, Tapader R, Gope A, Das R, Mahapatra E, Saha S, Pal A, Prasad P, Pal A. Metallo-protease Peptidase M84 from Bacillusaltitudinis induces ROS-dependent apoptosis in ovarian cancer cells by targeting PAR-1. iScience 2024; 27:109828. [PMID: 38799586 PMCID: PMC11126781 DOI: 10.1016/j.isci.2024.109828] [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: 10/05/2023] [Revised: 01/02/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
We have purified Peptidase M84 from Bacillus altitudinis in an effort to isolate anticancer proteases from environmental microbial isolates. This metallo-protease had no discernible impact on normal cell survival, but it specifically induced apoptosis in ovarian cancer cells. PAR-1, a GPCR which is reported to be overexpressed in ovarian cancer cells, was identified as a target of Peptidase M84. We observed that Peptidase M84 induced PAR-1 overexpression along with activating its downstream signaling effectors NF-κB and MAPK to promote excessive reactive oxygen species (ROS) generation. This evoked apoptotic death of the ovarian cancer cells through the intrinsic route. In in vivo set-up, weekly intraperitoneal administration of Peptidase M84 in syngeneic mice significantly diminished ascites accumulation, increasing murine survival rates by 60%. Collectively, our findings suggested that Peptidase M84 triggered PAR-1-mediated oxidative stress to act as an apoptosis inducer. This established Peptidase M84 as a drug candidate for receptor mediated targeted-therapy of ovarian cancer.
Collapse
Affiliation(s)
- Niraj Nag
- Division of Molecular Pathophysiology, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| | - Tanusree Ray
- Division of Molecular Pathophysiology, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| | - Rima Tapader
- Division of Molecular Pathophysiology, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| | - Animesh Gope
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| | - Rajdeep Das
- Molecular Cell Biology of Autophagy Lab, The Francis Crick Institute, 1, Midland Road, London NW1 1AT, UK
| | - Elizabeth Mahapatra
- Department of Environmental Carcinogenesis and Toxicology, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, West Bengal 700026, India
| | - Saibal Saha
- Division of Molecular Pathophysiology, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| | - Ananda Pal
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| | - Parash Prasad
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital and Medical Center, 3333 Burnet Avenue, Cincinnati 45229-3026, OH, USA
| | - Amit Pal
- Division of Molecular Pathophysiology, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), P-33, CIT Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| |
Collapse
|
26
|
Shanfeng Li, Zhou L, Zhao F, Wang H, Sun M. Inhibition of Liver Cancer Cell Viability by Triazole through Up-regulation of p38 Phosphorylation and Targeting the Activation of p-ERK1/2 and Akt Protein Expression. DOKL BIOCHEM BIOPHYS 2024; 516:66-72. [PMID: 38700817 DOI: 10.1134/s1607672923600525] [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: 02/20/2024] [Revised: 03/10/2024] [Accepted: 03/14/2024] [Indexed: 05/26/2024]
Abstract
The present study was aimed to explore the effect of triazole on growth and viability of liver cancer cells. Cell growth was examined using the MTT test and expression of several proteins was assessed by western blotting assay. The Matrigel-coated Transwell assay was employed to examine the infiltration of cells. The data from MTT assay showed that MHCC97H and H4TG liver cancer cell viability was inhibited by triazole in a concentration-dependent manner. After treatment with 0.5, 1.0, 2.0, 4, 8, and 16 µM doses of triazole, the rate of H4TG cell viability was decreased to 96, 73, 58, 39, 29, and 28%, respectively. Treatment of MHCC97H cells with 0.5, 1.0, 2.0, 4, 8, and 16 µM doses of triazole resulted in a reduction in cell viability to 94, 70, 53, 35, 22, and 21%, respectively. Triazole treatment also led to a significant reduction in MHCC97H cell invasiveness compared to the control cells. In MHCC97H cells treated with triazole, there was a noticeable decrease in the levels of p-ERK1/2, and p-Akt protein expression. Treatment of MHCC97H cells with triazole resulted in a prominent increase in p-p38 level. In summary, triazole inhibits growth and viability of liver cancer cells through targeting the activation of p-ERK1/2 and Akt proteins. Therefore, triazole may be investigated further as a therapeutic agent for the treatment of liver cancer.
Collapse
Affiliation(s)
- Shanfeng Li
- Interventional Vascular Surgery, Affiliated Hospital of Hebei University, 071000, Baoding, Hebei Province, China
| | - Long Zhou
- Interventional Vascular Surgery, Affiliated Hospital of Hebei University, 071000, Baoding, Hebei Province, China
| | - Feng Zhao
- Interventional Vascular Surgery, Affiliated Hospital of Hebei University, 071000, Baoding, Hebei Province, China
| | - Haisong Wang
- Interventional Vascular Surgery, Affiliated Hospital of Hebei University, 071000, Baoding, Hebei Province, China
| | - Meng Sun
- Interventional Vascular Surgery, Affiliated Hospital of Hebei University, 071000, Baoding, Hebei Province, China.
| |
Collapse
|
27
|
Murata N, Nishimura K, Harada N, Kitakaze T, Yoshihara E, Inui H, Yamaji R. Insulin reduces endoplasmic reticulum stress-induced apoptosis by decreasing mitochondrial hyperpolarization and caspase-12 in INS-1 pancreatic β-cells. Physiol Rep 2024; 12:e16106. [PMID: 38884322 PMCID: PMC11181300 DOI: 10.14814/phy2.16106] [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: 05/15/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Pancreatic β-cell mass is a critical determinant of insulin secretion. Severe endoplasmic reticulum (ER) stress causes β-cell apoptosis; however, the mechanisms of progression and suppression are not yet fully understood. Here, we report that the autocrine/paracrine function of insulin reduces ER stress-induced β-cell apoptosis. Insulin reduced the ER-stress inducer tunicamycin- and thapsigargin-induced cell viability loss due to apoptosis in INS-1 β-cells. Moreover, the effect of insulin was greater than that of insulin-like growth factor-1 at physiologically relevant concentrations. Insulin did not attenuate the ER stress-induced increase in unfolded protein response genes. ER stress did not induce cytochrome c release from mitochondria. Mitochondrial hyperpolarization was induced by ER stress and prevented by insulin. The protonophore/mitochondrial oxidative phosphorylation uncoupler, but not the antioxidants N-acetylcysteine and α-tocopherol, exhibited potential cytoprotection during ER stress. Both procaspase-12 and cleaved caspase-12 levels increased under ER stress. The caspase-12 inhibitor Z-ATAD-FMK decreased ER stress-induced apoptosis. Caspase-12 overexpression reduced cell viability, which was diminished in the presence of insulin. Insulin decreased caspase-12 levels at the post-translational stages. These results demonstrate that insulin protects against ER stress-induced β-cell apoptosis in this cell line. Furthermore, mitochondrial hyperpolarization and increased caspase-12 levels are involved in ER stress-induced and insulin-suppressed β-cell apoptosis.
Collapse
Affiliation(s)
- Nanako Murata
- Department of Applied Biological Chemistry, Graduate School of AgricultureOsaka Metropolitan UniversitySakaiOsakaJapan
| | - Kana Nishimura
- Division of Applied Life Sciences, Graduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Naoki Harada
- Department of Applied Biological Chemistry, Graduate School of AgricultureOsaka Metropolitan UniversitySakaiOsakaJapan
- Division of Applied Life Sciences, Graduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Tomoya Kitakaze
- Department of Applied Biological Chemistry, Graduate School of AgricultureOsaka Metropolitan UniversitySakaiOsakaJapan
- Division of Applied Life Sciences, Graduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
| | - Eiji Yoshihara
- The Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCaliforniaUSA
- David Geffen School of Medicine at University of California Los AngelesLos AngelesCaliforniaUSA
| | - Hiroshi Inui
- Department of Applied Biological Chemistry, Graduate School of AgricultureOsaka Metropolitan UniversitySakaiOsakaJapan
- Division of Applied Life Sciences, Graduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
- Department of Health and NutritionOtemae UniversityOsakaJapan
| | - Ryoichi Yamaji
- Department of Applied Biological Chemistry, Graduate School of AgricultureOsaka Metropolitan UniversitySakaiOsakaJapan
- Division of Applied Life Sciences, Graduate School of Life and Environmental SciencesOsaka Prefecture UniversitySakaiOsakaJapan
- Center for Research and Development of BioresourcesOsaka Metropolitan UniversitySakaiOsakaJapan
| |
Collapse
|
28
|
Zhang Z, Hou L, Liu D, Luan S, Huang M, Zhao L. Directly targeting BAX for drug discovery: Therapeutic opportunities and challenges. Acta Pharm Sin B 2024; 14:2378-2401. [PMID: 38828138 PMCID: PMC11143528 DOI: 10.1016/j.apsb.2024.02.010] [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: 11/14/2023] [Revised: 01/25/2024] [Accepted: 02/04/2024] [Indexed: 06/05/2024] Open
Abstract
For over two decades, the development of B-cell lymphoma-2 (Bcl-2) family therapeutics has primarily focused on anti-apoptotic proteins, resulting in the first-in-class drugs called BH3 mimetics, especially for Bcl-2 inhibitor Venetoclax. The pro-apoptotic protein Bcl-2-associated X protein (BAX) plays a crucial role as the executioner protein of the mitochondrial regulated cell death, contributing to organismal development, tissue homeostasis, and immunity. The dysregulation of BAX is closely associated with the onset and progression of diseases characterized by pathologic cell survival or death, such as cancer, neurodegeneration, and heart failure. In addition to conducting thorough investigations into the physiological modulation of BAX, research on the regulatory mechanisms of small molecules identified through biochemical screening approaches has prompted the identification of functional and potentially druggable binding sites on BAX, as well as diverse all-molecule BAX modulators. This review presents recent advancements in elucidating the physiological and pharmacological modulation of BAX and in identifying potentially druggable binding sites on BAX. Furthermore, it highlights the structural and mechanistic insights into small-molecule modulators targeting diverse binding surfaces or conformations of BAX, offering a promising avenue for developing next-generation apoptosis modulators to treat a wide range of diseases associated with dysregulated cell death by directly targeting BAX.
Collapse
Affiliation(s)
- Zhenwei Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linghui Hou
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shenglin Luan
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen 518000, China
| | - Min Huang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
29
|
Kim M. Mitochondria of T Lymphocytes Promote Anti-Pulmonary Tumor Immune Response. World J Oncol 2024; 15:472-481. [PMID: 38751696 PMCID: PMC11092414 DOI: 10.14740/wjon1841] [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: 02/27/2024] [Accepted: 03/30/2024] [Indexed: 05/18/2024] Open
Abstract
Background B-cell lymphoma 2 (Bcl-2), a protein involved in apoptosis, has been proven to have carcinogenic potential and is well documented. With the recent advancement in optical technology, it has become possible to observe subcellular organelles such as mitochondria in real-time without the need for staining. Consequently, we have examined the movement of mitochondria in cancer cells, correlating it with the regulation of Bcl-2. Methods Using a tomographic microscope, which can detect the internal structure of cells, we observed lung tumor cells. Cells were exposed to a laser beam (λ = 520 nm) inclined at 45°, and holographic images were recorded up to a depth of 30 µm of reconstruction. Results Intriguingly, lung tumor cells rapidly expelled mitochondria upon the attachment of Bcl-2 or B-cell lymphoma extra-large (Bcl-xL) inhibitors. On the other hand, we observed that tumor cells hijack mitochondria from T cells. The hijacked mitochondria were not immediately linked to tumor cell death, but they played a role in assisting granzyme B-induced tumor cell death. Due to lower levels of Bcl-2 and Bcl-xL on the mitochondria of T cells compared to lung tumor cells, immune cells depleted of Bcl-2 and Bcl-xL were co-cultured with the tumor cells. Conclusions As a result, a more effective tumor cell death induced by granzyme B was observed. Additionally, further enhanced anticancer immune response was observed in vivo. Together, we show that modified mitochondria of T cells can provide potential novel strategies towards tumor cell death.
Collapse
Affiliation(s)
- Minsuk Kim
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul 07804, Korea.
| |
Collapse
|
30
|
Choi YJ, Wedamulla NE, Kim SH, Oh M, Seo KS, Han JS, Lee EJ, Park YH, Park YJ, Kim EK. Salvia miltiorrhiza Bunge Ameliorates Benign Prostatic Hyperplasia through Regulation of Oxidative Stress via Nrf-2/HO-1 Activation. J Microbiol Biotechnol 2024; 34:1059-1072. [PMID: 37994101 PMCID: PMC11180924 DOI: 10.4014/jmb.2308.08053] [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: 09/02/2023] [Revised: 09/27/2023] [Accepted: 10/23/2023] [Indexed: 11/24/2023]
Abstract
Oxidative stress is a key factor in the pathogenesis of benign prostatic hyperplasia (BPH) that leads to inflammation. This study aimed to evaluate the ameliorative effects of Salvia miltiorrhiza Bunge extract (HLT-101) on BPH through the regulation of oxidative stress and inflammation. A testosterone propionate (TP)-induced BPH rat model was orally administered HLT-101 (20, 40, or 80 mg/kg), and its effects on oxidative stress- and inflammation-related gene expression were examined. Further, HLT-101 was assessed for its effect on reactive oxygen species (ROS) levels and Nrf-2/HO-1 signaling pathways in BPH-1 cells. HLT-101 decreased testosterone-induced excessive free radical production and inflammatory factor activation. Moreover, HLT-101 treatment significantly decreased the intracellular ROS level in the TNF-α and IFN-γ treated BPH-1 cells through the activation of Nrf-2. In addition, HLT-101 treatment inhibited the NF-κB pathway and androgen receptor (AR) signaling, which is highly linked to the pathogenesis of BPH. Therefore, HLT-101 has the potential to be an effective treatment reagent for BPH because of its ability to reduce inflammation and oxidative stress via Nrf-2/HO-1 signaling.
Collapse
Affiliation(s)
- Young-Jin Choi
- Department of Food Science and Nutrition, Dong-A University, Busan 49315, Republic of Korea
- Department of Health Sciences, the Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Nishala Erandi Wedamulla
- Department of Food Science and Nutrition, Dong-A University, Busan 49315, Republic of Korea
- Department of Health Sciences, the Graduate School of Dong-A University, Busan 49315, Republic of Korea
- Department of Food Science and Technology, Faculty of Animal Science and Export Agriculture, Uva Wellassa University, Badulla 90000, Sri Lanka
| | - Seok-Hee Kim
- Department of Food Science and Nutrition, Dong-A University, Busan 49315, Republic of Korea
- Department of Health Sciences, the Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Mirae Oh
- Grassland and Forages Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea
| | - Kang Sik Seo
- Curome Bioscience Co., Ltd., Suwon 16506, Republic of Korea
| | - Jeong Su Han
- Curome Bioscience Co., Ltd., Suwon 16506, Republic of Korea
| | - Eun Joo Lee
- Healthism Corporation, Cheongju 28160, Republic of Korea
| | - Young Ho Park
- Healthism Corporation, Cheongju 28160, Republic of Korea
| | - Young Jin Park
- Department of Family Medicine, Dong-A University College of Medicine, Busan 49315, Republic of Korea
| | - Eun-Kyung Kim
- Educational Major, Graduate School of Education, Dong-A University, Busan 49315, Republic of Korea
- Nutrinomics Lab. Co., Ltd., Busan 49315, Republic of Korea
| |
Collapse
|
31
|
Chang X, Li Z, Tian M, Deng Z, Zhu L, Li G. Rotenone activates the LKB1-AMPK-ULK1 signaling pathway to induce autophagy and apoptosis in rat thoracic aortic endothelial cells. BMC Pharmacol Toxicol 2024; 25:33. [PMID: 38783387 PMCID: PMC11118107 DOI: 10.1186/s40360-024-00755-5] [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: 05/05/2023] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND The specific mechanism by which rotenone impacts thoracic aortic autophagy and apoptosis is unknown. We aimed to investigate the regulatory effects of rotenone on autophagy and apoptosis in rat thoracic aortic endothelial cells (RTAEC) via activation of the LKB1-AMPK-ULK1 signaling pathway and to elucidate the molecular mechanisms of rotenone on autophagy and apoptosis in vascular endothelial cells. METHODS In vivo, 60 male SD rats were randomly selected and divided into 5 groups: control (Con), DMSO, 1, 2, and 4 mg/kg groups, respectively. After 28 days of treatment, histopathological and ultrastructural changes in each group were observed using HE and transmission electron microscopy; Autophagy, apoptosis, and LKB1-AMPK-ULK1 pathway-related proteins were detected by Western blot; Apoptosis levels in the thoracic aorta were detected by TUNEL. In vitro, RTAEC were cultured and divided into control (Con), DMSO, 20, 100, 500, and 1000 nM groups. After 24 h of intervention, autophagy, apoptosis, and LKB1-AMPK-ULK1 pathway-related factors were detected by Western blot and qRT-PCR; Flow cytometry to detect apoptosis levels; Autophagy was inhibited with 3-MA and CQ to detect apoptosis levels, and changes in autophagy, apoptosis, and downstream factors were detected by the AMPK inhibitor CC intervention. RESULTS Gavage in SD rats for 28 days, some degree of damage was observed in the thoracic aorta and heart of the rotenone group, as well as the appearance of autophagic vesicles was observed in the thoracic aorta. TUNEL analysis revealed higher apoptosis in the rotenone group's thoracic aorta; RTAEC cultured in vitro, after 24 h of rotenone intervention, showed increased ROS production and significantly decreased ATP production. The flow cytometry data suggested an increase in the number of apoptotic RTAEC. The thoracic aorta and RTAEC in the rotenone group displayed elevated levels of autophagy and apoptosis, and the LKB1-AMPK-ULK1 pathway proteins were activated and expressed at higher levels. Apoptosis and autophagy were both suppressed by the autophagy inhibitors 3-MA and CQ. The AMPK inhibitor CC reduced autophagy and apoptosis in RTAEC and suppressed the production of the AMPK downstream factors ULK1 and P-ULK1. CONCLUSIONS Rotenone may promote autophagy in the thoracic aorta and RTAEC by activating the LKB1-AMPK-ULK1 signaling pathway, thereby inducing apoptosis.
Collapse
Affiliation(s)
- Xiaoyu Chang
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China
| | - Zeyuan Li
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China
| | - Mi Tian
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China
| | - Ziwei Deng
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China
| | - Lingqin Zhu
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China.
| | - Guanghua Li
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, China.
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
32
|
Ghafoor MH, Song BL, Zhou L, Qiao ZY, Wang H. Self-Assembly of Peptides as an Alluring Approach toward Cancer Treatment and Imaging. ACS Biomater Sci Eng 2024; 10:2841-2862. [PMID: 38644736 DOI: 10.1021/acsbiomaterials.4c00491] [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] [Indexed: 04/23/2024]
Abstract
Cancer is a severe threat to humans, as it is the second leading cause of death after cardiovascular diseases and still poses the biggest challenge in the world of medicine. Due to its higher mortality rates and resistance, it requires a more focused and productive approach to provide the solution for it. Many therapies promising to deliver favorable results, such as chemotherapy and radiotherapy, have come up with more negatives than positives. Therefore, a new class of medicinal solutions and a more targeted approach is of the essence. This review highlights the alluring properties, configurations, and self-assembly of peptide molecules which benefit the traditional approach toward cancer therapy while sparing the healthy cells in the process. As targeted drug delivery systems, self-assembled peptides offer a wide spectrum of conjugation, biocompatibility, degradability-controlled responsiveness, and biomedical applications, including cancer treatment and cancer imaging.
Collapse
Affiliation(s)
- Muhammad Hamza Ghafoor
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ben-Li Song
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Lei Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Zeng-Ying Qiao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
33
|
Cappe B, Vandenabeele P, Riquet FB. A guide to the expanding field of extracellular vesicles and their release in regulated cell death programs. FEBS J 2024; 291:2068-2090. [PMID: 37872002 DOI: 10.1111/febs.16981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Homeostasis disruption is visible at the molecular and cellular levels and may often lead to cell death. This vital process allows us to maintain the more extensive system's integrity by keeping the different features (genetic, metabolic, physiologic, and individual) intact. Interestingly, while cells can die in different manners, dying cells still communicate with their environment. This communication was, for a long time, perceived as only driven by the release of soluble factors. However, it has now been reconsidered with the increasing interest in extracellular vesicles (EVs), which are discovered to be released during different regulated cell death programs, with the observation of specific effects. EVs are game changers in the paradigm of cell-cell communication with tremendous implications in fundamental research with regard to noncell autonomous functions, as well as in biomarkers research, all of which are geared toward diagnostic and therapeutic purposes. This review is composed of two main parts. The first is a comprehensive presentation of the state of the art of the EV field at large. In the second part, we focus on EVs discovered to be released during different regulated cell death programs, also known as cell death EVs (cdEVs), and EV-associated specific effects on recipient cells in the context of cell death and inflammation/inflammatory responses.
Collapse
Affiliation(s)
- Benjamin Cappe
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research (IRC), Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research (IRC), Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Franck B Riquet
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research (IRC), Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
- University of Lille, CNRS, UMR 8523 - PhLAM - Physique des Lasers Atomes et Molécules, France
| |
Collapse
|
34
|
Li X, Qian H, Ye H, Zhao H, Cui J, Fang X, Ge Z, Shi J, Qi W, Ye L. DEHP induces apoptosis and autophagy of the thyroid via Rap1 signaling pathway: In vivo and in vitro study. Food Chem Toxicol 2024; 187:114609. [PMID: 38522500 DOI: 10.1016/j.fct.2024.114609] [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: 12/08/2023] [Revised: 03/08/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
OBJECTIVE DEHP has thyroid toxicity and affects thyroid function. However, the mechanism is unclear. METHODS The offspring of SD rats were gavaged with different doses of DEHP from in utero to 8 or 12 weeks old. We observed the thyroid morphology with HE and autophagosomes with TEM. The THs levels were tested with ELISA. The apoptosis level was tested by flow cytometry. The levels of apoptosis-related genes, autophagy-related genes and Rap1 pathway genes, were measured with qRT-PCR and Western blot. We established an MEHP-treated Nthy-ori 3-1 cell model and inhibited the Rap1 to verify the mechanism. RESULTS DEHP could cause pathological damage and ultrastructure damage of thyroids in offspring rats. After DEHP exposure, the THs levels were altered, the apoptosis levels increased, and autophagosomes appeared. DEHP significantly affected the levels of apoptosis-related genes and autophagy-related genes. DEHP also affected the levels of Rap1 pathway, which was correlated with the levels of apoptosis and autophagy. After inhibiting Rap1 in Nthy-ori 3-1 cells, the THs levels were altered. Rap1 pathway was inhibited and the levels of apoptosis and autophagy were down-regulated. CONCLUSION DEHP could induce the apoptosis and autophagy of the thyroid, and Rap1 signaling pathway may play a significant role.
Collapse
Affiliation(s)
- Xu Li
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, 130021, China
| | - Honghao Qian
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, 130021, China
| | - Hui Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, 130021, China
| | - Haotang Zhao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, 130021, China
| | - Jianwei Cui
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, 130021, China
| | - Xiaoqi Fang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, 130021, China
| | - Zhili Ge
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, 130021, China
| | - Jingjing Shi
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, 130021, China
| | - Wen Qi
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, 130021, China.
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, 130021, China.
| |
Collapse
|
35
|
Egorova KS, Kibardin AV, Posvyatenko AV, Ananikov VP. Mechanisms of Biological Effects of Ionic Liquids: From Single Cells to Multicellular Organisms. Chem Rev 2024; 124:4679-4733. [PMID: 38621413 DOI: 10.1021/acs.chemrev.3c00420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
The review presents a detailed discussion of the evolving field studying interactions between ionic liquids (ILs) and biological systems. Originating from molten salt electrolytes to present multiapplication substances, ILs have found usage across various fields due to their exceptional physicochemical properties, including excellent tunability. However, their interactions with biological systems and potential influence on living organisms remain largely unexplored. This review examines the cytotoxic effects of ILs on cell cultures, biomolecules, and vertebrate and invertebrate organisms. Our understanding of IL toxicity, while growing in recent years, is yet nascent. The established findings include correlations between harmful effects of ILs and their ability to disturb cellular membranes, their potential to trigger oxidative stress in cells, and their ability to cause cell death via apoptosis. Future research directions proposed in the review include studying the distribution of various ILs within cellular compartments and organelles, investigating metabolic transformations of ILs in cells and organisms, detailed analysis of IL effects on proteins involved in oxidative stress and apoptosis, correlation studies between IL doses, exposure times and resulting adverse effects, and examination of effects of subtoxic concentrations of ILs on various biological objects. This review aims to serve as a critical analysis of the current body of knowledge on IL-related toxicity mechanisms. Furthermore, it can guide researchers toward the design of less toxic ILs and the informed use of ILs in drug development and medicine.
Collapse
Affiliation(s)
- Ksenia S Egorova
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow 119991, Russia
| | - Alexey V Kibardin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russian Federation, Moscow 117198, Russia
| | - Alexandra V Posvyatenko
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow 119991, Russia
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russian Federation, Moscow 117198, Russia
| | - Valentine P Ananikov
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow 119991, Russia
| |
Collapse
|
36
|
Saha P, Hegde M, Chakraborty K, Singha A, Mukerjee N, Ghosh D, Kunnumakkara AB, Khan MS, Ahmad MI, Ghosh A, Kumer A, Sil SK. Targeted inhibition of colorectal cancer proliferation: The dual-modulatory role of 2,4-DTBP on anti-apoptotic Bcl-2 and Survivin proteins. J Cell Mol Med 2024; 28:e18150. [PMID: 38494866 PMCID: PMC10945088 DOI: 10.1111/jcmm.18150] [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: 09/20/2023] [Revised: 12/29/2023] [Accepted: 01/16/2024] [Indexed: 03/19/2024] Open
Abstract
The anti-apoptotic proteins, Bcl-2 and Survivin, are consistently overexpressed in numerous human malignancies, notably in colorectal cancer. 2,4-Di-tert-butylphenol (2,4-DTBP) is a naturally occurring phenolic compound known for its diverse biological activities, including anti-cancer properties. The mechanism behind 2,4-DTBP-induced inhibition of cell proliferation and apoptosis in human colorectal cancer cells, specifically regarding Bcl-2 and Survivin, remains to be elucidated. In this study, we employed both in silico and in vitro methodologies to underpin this interaction at the molecular level. Molecular docking demonstrated a substantial binding affinity of 2,4-DTBP towards Bcl-2 (ΔG = -9.8 kcal/mol) and Survivin (ΔG = -5.6 kcal/mol), suggesting a potential inhibitory effect. Further, molecular dynamic simulations complemented by MM-GBSA calculations confirmed the significant binding of 2,4-DTBP with Bcl-2 (dGbind = -54.85 ± 6.79 kcal/mol) and Survivin (dGbind = -32.36 ± 1.29 kcal/mol). In vitro assays using HCT116 colorectal cancer cells revealed that 2,4-DTBP inhibited proliferation and promoted apoptosis in both a dose- and time-dependent manner. Fluorescence imaging and scanning electron microscopy illustrated the classical features associated with apoptosis upon 2,4-DTBP exposure. Cell cycle analysis through flow cytometry highlighted a G1 phase arrest and apoptosis assay demonstrated increased apoptotic cell population. Notably, western blotting results indicated a decreased expression of Bcl-2 and Survivin post-treatment. Considering the cytoprotective roles of Bcl-2 and Survivin through the inhibition of mitochondrial dysfunction, our findings of disrupted mitochondrial bioenergetics, characterized by reduced ATP production and oxygen consumption, further accentuate the functional impairment of these proteins. Overall, the integration of in silico and in vitro data suggests that 2,4-DTBP holds promise as a therapeutic agent targeting Bcl-2 and Survivin in colorectal cancer.
Collapse
Affiliation(s)
- Partha Saha
- Molecular Genetics and Cell Physiology Laboratory, Department of Human PhysiologyTripura UniversitySuryamaninagarTripuraIndia
| | - Mangala Hegde
- Cancer Biology Laboratory and DBT‐AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and BioengineeringIndian Institute of Technology (IIT) GuwahatiGuwahatiAssamIndia
| | - Kanak Chakraborty
- Molecular Genetics and Cell Physiology Laboratory, Department of Human PhysiologyTripura UniversitySuryamaninagarTripuraIndia
| | - Achinta Singha
- Molecular Genetics and Cell Physiology Laboratory, Department of Human PhysiologyTripura UniversitySuryamaninagarTripuraIndia
| | - Nobendu Mukerjee
- Center for Global Health ResearchSaveetha Medical College and Hospital, Saveetha Institute of Medical and Technical SciencesChennaiTamil NaduIndia
- Department of Health SciencesNovel Global Community Educational FoundationHebershamNew South WalesAustralia
| | - Deepshikha Ghosh
- Cell Biology and Physiology DivisionCSIR‐Indian Institute of Chemical BiologyKolkataWest BengalIndia
| | - Ajaikumar B. Kunnumakkara
- Cancer Biology Laboratory and DBT‐AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and BioengineeringIndian Institute of Technology (IIT) GuwahatiGuwahatiAssamIndia
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of ScienceKing Saud UniversityRiyadhSaudi Arabia
| | - Md Irshad Ahmad
- Department of Structural Biology, School of MedicineUTHEALTH Science CenterSan AntonioTexasUSA
| | - Arabinda Ghosh
- Department of Computational Biology and BiotechnologyMahapurusha Srimanta Sankaradeva ViswavidalayaGuwahatiAssamIndia
| | - Ajoy Kumer
- Department of Chemistry, College of Arts and SciencesIUBAT‐International University of Business Agriculture and TechnologyDhakaBangladesh
| | - Samir Kumar Sil
- Molecular Genetics and Cell Physiology Laboratory, Department of Human PhysiologyTripura UniversitySuryamaninagarTripuraIndia
| |
Collapse
|
37
|
Erazo-Oliveras A, Muñoz-Vega M, Salinas ML, Wang X, Chapkin RS. Dysregulation of cellular membrane homeostasis as a crucial modulator of cancer risk. FEBS J 2024; 291:1299-1352. [PMID: 36282100 PMCID: PMC10126207 DOI: 10.1111/febs.16665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/09/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
Abstract
Cellular membranes serve as an epicentre combining extracellular and cytosolic components with membranous effectors, which together support numerous fundamental cellular signalling pathways that mediate biological responses. To execute their functions, membrane proteins, lipids and carbohydrates arrange, in a highly coordinated manner, into well-defined assemblies displaying diverse biological and biophysical characteristics that modulate several signalling events. The loss of membrane homeostasis can trigger oncogenic signalling. More recently, it has been documented that select membrane active dietaries (MADs) can reshape biological membranes and subsequently decrease cancer risk. In this review, we emphasize the significance of membrane domain structure, organization and their signalling functionalities as well as how loss of membrane homeostasis can steer aberrant signalling. Moreover, we describe in detail the complexities associated with the examination of these membrane domains and their association with cancer. Finally, we summarize the current literature on MADs and their effects on cellular membranes, including various mechanisms of dietary chemoprevention/interception and the functional links between nutritional bioactives, membrane homeostasis and cancer biology.
Collapse
Affiliation(s)
- Alfredo Erazo-Oliveras
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Mónica Muñoz-Vega
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Michael L. Salinas
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Xiaoli Wang
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Robert S. Chapkin
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
- Center for Environmental Health Research; Texas A&M University; College Station, Texas, 77843; USA
| |
Collapse
|
38
|
Habib TN, Altonsy MO, Ghanem SA, Salama MS, Hosny MA. Optimizing combination therapy in prostate cancer: mechanistic insights into the synergistic effects of Paclitaxel and Sulforaphane-induced apoptosis. BMC Mol Cell Biol 2024; 25:5. [PMID: 38438917 PMCID: PMC10910811 DOI: 10.1186/s12860-024-00501-z] [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: 08/02/2023] [Accepted: 02/12/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Combination therapies in cancer treatment have demonstrated synergistic or additive outcomes while also reducing the development of drug resistance compared to monotherapy. This study explores the potential of combining the chemotherapeutic agent Paclitaxel (PTX) with Sulforaphane (SFN), a natural compound primarily found in cruciferous vegetables, to enhance treatment efficacy in prostate cancer. METHODS Two prostate cancer cell lines, PC-3 and LNCaP, were treated with varying concentrations of PTX, SFN, and their combination. Cell viability was assessed using the thiazolyl blue tetrazolium bromide (MTT) assay to determine the EC50 values. Western blot analysis was conducted to evaluate the expression of Bax, Bcl2, and Caspase-3 activation proteins in response to individual and combined treatments of PTX and SFN. Fluorescent microscopy was employed to observe morphological changes indicative of apoptotic stress in cell nuclei. Flow cytometry analysis was utilized to assess alterations in cell cycle phases, such as redistribution and arrest. Statistical analyses, including Student's t-tests and one-way analysis of variance with Tukey's correction, were performed to determine significant differences between mono- and combination treatments. RESULTS The impact of PTX, SFN, and their combination on cell viability reduction was evaluated in a dose-dependent manner. The combined treatment enhanced PTX's effects and decreased the EC50 values of both drugs compared to individual treatments. PTX and SFN treatments differentially regulated the expression of Bax and Bcl2 proteins in PC-3 and LNCaP cell lines, favoring apoptosis over cell survival. Our data indicated that combination therapy significantly increased Bax protein expression and the Bax/Bcl2 ratio compared to PTX or SFN alone. Flow cytometry analysis revealed alterations in cell cycle phases, including S-phase arrest and an increased population of apoptotic cells. Notably, the combination treatments did not have a discernible impact on necrotic cells. Signs of apoptotic cell death were confirmed through Caspase-3 cleavage, and morphological changes in cell nuclei were assessed via western blot and fluorescent microscopy. CONCLUSION This combination therapy of PTX and SFN has the potential to improve prostate cancer treatment by minimizing side effects while maintaining efficacy. Mechanistic investigations revealed that SFN enhances PTX efficacy by promoting apoptosis, activating caspase-3, inducing nuclear morphology changes, modulating the cell cycle, and altering Bax and Bcl2 protein expression. These findings offer valuable insights into the synergistic effects of PTX and SFN, supporting the optimization of combination therapy and providing efficient therapeutic strategies in preclinical research.
Collapse
Affiliation(s)
- Tito N Habib
- Molecular Genetics Lab, Department of Zoology, Faculty of Science, Sohag University, Sohag, Egypt.
| | - Mohammed O Altonsy
- Molecular Genetics Lab, Department of Zoology, Faculty of Science, Sohag University, Sohag, Egypt
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Salah A Ghanem
- Molecular Genetics Lab, Department of Zoology, Faculty of Science, Sohag University, Sohag, Egypt
| | - Mohamed S Salama
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mai A Hosny
- Molecular Genetics Lab, Department of Zoology, Faculty of Science, Sohag University, Sohag, Egypt
| |
Collapse
|
39
|
Şingar E, Akbulut A, Koca G, Yazihan N, Atilgan Hİ, Yumuşak N, Demir A, Burcu A, Korkmaz M. The influencer effect of Dexmedetomidine on radioiodine relevant to lacrimal gland impairment. Int Ophthalmol 2024; 44:115. [PMID: 38411703 DOI: 10.1007/s10792-024-03052-6] [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] [Received: 05/11/2023] [Accepted: 02/16/2024] [Indexed: 02/28/2024]
Abstract
PURPOSE To assess the potential influencing effects of Dexmedetomidine on impaired lacrimal glands after high-dose radioiodine treatment (RAI). METHODS Thirty-six rats were arbitrarily separated into 3 groups: Sham, RAI, and Dexmedetomidine. Dexmedetomidine group was given Dexmedetomidine and RAI, the Sham group was given the same millimeters of saline, and the RAI group was given RAI only. All forms of lacrimal glands, including harderian glands (HG), extraorbital (EG), and intraorbital (IG) lacrimal glands, were evaluated for immunohistochemical, histopathologic assessments and also for tissue cytokines, oxidant and antioxidant levels. RESULTS Dexmedetomidine significantly ameliorated histopathologic changes such as periacinar fibrosis, acinar atrophy, lymphocytic infiltration, ductal proliferation, lipofuscin-like accumulation, and nucleus changes caused by RAI in all lacrimal gland forms (p < 0.05 for all of the parameters). However, periductal fibrosis was improved significantly only in EG (p = 0.049), and mast cell infiltration was improved significantly only in IG (p = 0.038) in Dexmedetomidine groups. There was a significant decrease in the elevated caspase-3 and TUNEL levels after RAI administration in the Dexmedetomidine group in all lacrimal gland forms (p < 0.05 for all parameters). Dexmedetomidine attenuated NF-kb, TNF-α, and IL-6 levels significantly diminished total oxidant status and raised total antioxidant status levels (p < 0.05 for all parameters). CONCLUSIONS The results of this study demonstrated that following RAI, Dexmedetomidine diminished inflammation, tissue cytokine levels, and apoptosis and ameliorated impaired histopathologic patterns of the lacrimal glands.
Collapse
Affiliation(s)
- Evin Şingar
- Department of Ophthalmology, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey.
| | - Aylin Akbulut
- Department of Nuclear Medicine, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| | - Gökhan Koca
- Department of Nuclear Medicine, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| | - Nuray Yazihan
- Department of Physiopathology, Ankara University, Ankara, Turkey
| | - Hasan İkbal Atilgan
- Department of Nuclear Medicine, Hatay Mustafa Kemal University, Hatay, Turkey
| | - Nihat Yumuşak
- Veterinary Faculty, Department of Pathology, Harran University, Sanliurfa, Turkey
| | - Ayten Demir
- Faculty of Nursing, Ankara University, Ankara, Turkey
| | - Ayse Burcu
- Department of Ophthalmology, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| | - Meliha Korkmaz
- Department of Nuclear Medicine, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
40
|
Geng H, Zhang H, Cheng L, Dong S. Sivelestat ameliorates sepsis-induced myocardial dysfunction by activating the PI3K/AKT/mTOR signaling pathway. Int Immunopharmacol 2024; 128:111466. [PMID: 38176345 DOI: 10.1016/j.intimp.2023.111466] [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: 04/02/2023] [Revised: 12/07/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
The cardioprotective role of sivelestat, a neutrophil elastase inhibitor, has already been demonstrated, but the underlying molecular mechanism remains unclear. This study aimed to explore the mechanism underlying the role of sivelestat in sepsis-induced myocardial dysfunction (SIMD). We found that sivelestat treatment remarkably improved the viability and suppressed the apoptosis of lipopolysaccharide (LPS)-stimulated H9c2 cells. In vivo, sivelestat treatment was associated with an improved survival rate; reduced serum cTnT, TNF-α, IL-1β levels and myocardial TNF-α and IL-1β levels; ameliorated cardiac function and structure; and reduced cardiomyocyte apoptosis. Moreover, sivelestat treatment substantially increased Bcl-2 expression and suppressed caspase-3 and Bax expression in LPS-induced H9c2 cells and in the heart tissues of septic rats. Furthermore, the phosphatidylinositol 3-kinase/protein kinase B/mechanistic target of rapamycin (PI3K/AKT/mTOR) signaling pathway was activated both in vitro and in vivo. The protective effect of sivelestat against SIMD was reversed by the PI3K inhibitor LY294002. In summary, sivelestat can protect against SIMD by activating the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hongyu Geng
- Department of Intensive Care Unit, Baoding First Central Hospital, Baoding, China
| | - Hongbo Zhang
- Department of General Surgery, Baoding First Central Hospital, Baoding, China
| | - Lianfang Cheng
- Department of Intensive Care Unit, Baoding First Central Hospital, Baoding, China
| | - Shimin Dong
- Department of Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
41
|
Liu LY, He SJ, Chen Z, Ge M, Lyu CY, Gao D, Yu JP, Cai MH, Yuan JX, Zhang JL. The Role of Regulatory Cell Death in Vitiligo. DNA Cell Biol 2024; 43:61-73. [PMID: 38153369 DOI: 10.1089/dna.2023.0188] [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] [Indexed: 12/29/2023] Open
Abstract
Vitiligo is one of the common chronic autoimmune skin diseases in clinic, which is characterized by localized or generalized depigmentation and seriously affects the physical and mental health of patients. At present, the pathogenesis of vitiligo is not clear; mainly, heredity, autoimmunity, oxidative stress, melanocyte (MC) self-destruction, and the destruction, death, or dysfunction of MCs caused by various reasons are always the core of vitiligo. Regulatory cell death (RCD) is an active and orderly death mode of cells regulated by genes, which widely exists in various life activities, plays a pivotal role in maintaining the homeostasis of the organism, and is closely related to the occurrence and development of many diseases. With the deepening of the research and understanding of RCD, people gradually found that there are many different forms of RCD in the lesions and perilesional skin of vitiligo patients, such as apoptosis, autophagy, pyroptosis, ferroptosis, and so on. Different cell death modes have different mechanisms in vitiligo, and different RCDs can interact and regulate each other. In this article, the mechanism related to RCD in the pathogenesis of vitiligo is reviewed, which provides new ideas for exploring the pathogenesis and targeted treatment of vitiligo.
Collapse
Affiliation(s)
- Lyu-Ye Liu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Si-Jia He
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, People's Republic of China
| | - Zhao Chen
- First Clinical Medical College Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Man Ge
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Chun-Yi Lyu
- First Clinical Medical College Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Dandan Gao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Ji-Peng Yu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Meng-Han Cai
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Jin-Xiang Yuan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Jun-Ling Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, People's Republic of China
| |
Collapse
|
42
|
Zalewski J, Szajna M, Stępień K, Nowak K, Karcińska A, Yika ADC, Krawczyk K, Karwat K, Zalewska M, Pierzchalski P. Endothelial Cell Apoptosis but Not Necrosis Is Inhibited by Ischemic Preconditioning. Int J Mol Sci 2024; 25:1238. [PMID: 38279236 PMCID: PMC10816637 DOI: 10.3390/ijms25021238] [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: 12/16/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
This study aimed to assess the influence of ischemic preconditioning (IP) on hypoxia/reoxygenation (HR)-induced endothelial cell (EC) death. Human umbilical vein endothelial cells (HUVECs) were subjected to 2 or 6 h hypoxia with subsequent reoxygenation. IP was induced by 20 min of hypoxia followed by 20 min of reoxygenation. Necrosis was assessed by the release of lactate dehydrogenase (LDH) and apoptosis by double staining with propidium iodide/annexin V (PI/AV), using TUNEL test, and Bcl-2 and Bax gene expression measured using RT-PCR. In PI/AV staining, after 24 h of reoxygenation, 30-33% of EC were necrotic and 16-21% were apoptotic. In comparison to HR cells, IP reduced membrane apoptosis after 24 h of reoxygenation by 50% but did not influence EC necrosis. Nuclear EC apoptosis affected about 15-17% of EC after 24 h of reoxygenation and was reduced with IP by 55-60%. IP was associated with a significantly higher Bcl-2/Bax ratio, at 8 h 2-4 times and at 24 h 2-3 times as compared to HR. Longer hypoxia was associated with lower values of Bcl-2/Bax ratio in EC subjected to HR or IP. IP delays, without reducing, the extent of HR-induced EC necrosis but significantly inhibits their multi-level evaluated apoptosis.
Collapse
Affiliation(s)
- Jarosław Zalewski
- Department of Coronary Artery Disease and Heart Failure, Jagiellonian University Medical College, 31-202 Krakow, Poland; (K.S.); (K.N.); (K.K.)
| | | | - Konrad Stępień
- Department of Coronary Artery Disease and Heart Failure, Jagiellonian University Medical College, 31-202 Krakow, Poland; (K.S.); (K.N.); (K.K.)
- Department of Thromboembolic Disorders, Jagiellonian University Medical College, 31-202 Krakow, Poland
| | - Karol Nowak
- Department of Coronary Artery Disease and Heart Failure, Jagiellonian University Medical College, 31-202 Krakow, Poland; (K.S.); (K.N.); (K.K.)
- Department of Thromboembolic Disorders, Jagiellonian University Medical College, 31-202 Krakow, Poland
| | - Aleksandra Karcińska
- Student Research Group, Department of Coronary Artery Disease and Heart Failure, Jagiellonian University Medical College, 31-202 Krakow, Poland; (A.K.); (A.d.C.Y.); (K.K.)
| | - Alicia del Carmen Yika
- Student Research Group, Department of Coronary Artery Disease and Heart Failure, Jagiellonian University Medical College, 31-202 Krakow, Poland; (A.K.); (A.d.C.Y.); (K.K.)
| | - Kornelia Krawczyk
- Student Research Group, Department of Coronary Artery Disease and Heart Failure, Jagiellonian University Medical College, 31-202 Krakow, Poland; (A.K.); (A.d.C.Y.); (K.K.)
| | - Krzysztof Karwat
- Department of Coronary Artery Disease and Heart Failure, Jagiellonian University Medical College, 31-202 Krakow, Poland; (K.S.); (K.N.); (K.K.)
| | - Magdalena Zalewska
- Department of Emergency Medicine, Faculty of Health Science, Jagiellonian University Medical College, 31-126 Krakow, Poland;
| | - Piotr Pierzchalski
- Faculty of Health Science, Jagiellonian University Medical College, 31-126 Krakow, Poland;
| |
Collapse
|
43
|
Li L, Lin Z, Yuan J, Li P, Wang Q, Cho N, Wang Y, Lin Z. The neuroprotective mechanisms of naringenin: Inhibition of apoptosis through the PI3K/AKT pathway after hypoxic-ischemic brain damage. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116941. [PMID: 37480970 DOI: 10.1016/j.jep.2023.116941] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/04/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Naringenin (NGN) is a widely distributed flavonoid with potent antioxidant and neuroprotective properties. Neuroprotective agents play a crucial role in the treatment of hypoxic-ischemic encephalopathy (HIE). It has shown potential therapeutic effects for neurological disorders. However, its efficacy on HIE is yet to be investigated. AIM OF THE STUDY This study aims to investigate the potential neuroprotective effect of naringenin and its underlying molecular mechanisms in reducing oxidative stress, apoptosis, and improving brain outcomes following HIE. Additionally, the study aims to identify the potential targets, mechanisms, and functions of naringenin using network pharmacology analysis. MATERIALS AND METHODS Neonatal mice were exposed to the hypoxic-ischemic brain damage (HIBD) model to determine brain water content, and brain tissue was subjected to hematoxylin and eosin (HE), immunohistochemistry (IHC), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and Nissl staining to investigate its neuroprotective effects. Furthermore, the neonatal mouse primary neuron oxygen-glucose deprivation (OGD) model to measure reactive oxygen species (ROS) production in vitro. The protein levels were characterized by Western Blot, and mRNA levels were evaluated by a real-time quantitative PCR detecting system (qPCR). Transmission electron microscopy (TEM) and mitochondrial fluorescent staining were used to observe mitochondrial morphology. Neuronal nuclei (NeuN) and microtubule-associated protein 2 (MAP2) were detected by Immunofluorescence (IF). Finally, network pharmacology was employed to determine the common target of naringenin and HIE. The core genes were obtained via protein-protein interaction networks (PPI) analysis and molecular docking was examined, and the mechanism of action was explored through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Additionally, small interfering RNA (siRNA) was constructed for verification. RESULTS Naringenin has a neuroprotective effect in HIBD by modulating Vegfa expression and activating the PI3K/AKT pathway to inhibit apoptosis. Furthermore, molecular docking results suggest that Vegfa is a potential binding target of naringenin, and silencing Vegfa partially reverses the pharmacological effects of NGN. CONCLUSION Our findings suggest that naringenin demonstrates potential clinical application for treating HIE as a novel neuroprotective agent.
Collapse
Affiliation(s)
- Luyao Li
- Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; College of Pharmacy, Chonnam National University, Gwangju, South Korea
| | - Zhen Lin
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Junhui Yuan
- Wenling Maternal and Child Health Care Hospital, Xiabao Road, Chengdong Street of Wenling City, Zhejiang Province, 317500, China
| | - Pingping Li
- Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China
| | - Qi Wang
- Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China
| | - Namki Cho
- College of Pharmacy, Chonnam National University, Gwangju, South Korea.
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Zhenlang Lin
- Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
44
|
Xu AP, Xu LB, Smith ER, Fleishman JS, Chen ZS, Xu XX. Cell death in cancer chemotherapy using taxanes. Front Pharmacol 2024; 14:1338633. [PMID: 38249350 PMCID: PMC10796453 DOI: 10.3389/fphar.2023.1338633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024] Open
Abstract
Cancer cells evolve to be refractory to the intrinsic programmed cell death mechanisms, which ensure cellular tissue homeostasis in physiological conditions. Chemotherapy using cytotoxic drugs seeks to eliminate cancer cells but spare non-cancerous host cells by exploring a likely subtle difference between malignant and benign cells. Presumably, chemotherapy agents achieve efficacy by triggering programmed cell death machineries in cancer cells. Currently, many major solid tumors are treated with chemotherapy composed of a combination of platinum agents and taxanes. Platinum agents, largely cis-platin, carboplatin, and oxaliplatin, are DNA damaging agents that covalently form DNA addicts, triggering DNA repair response pathways. Taxanes, including paclitaxel, docetaxel, and cabazitaxel, are microtubule stabilizing drugs which are often very effective in purging cancer cells in clinical settings. Generally, it is thought that the stabilization of microtubules by taxanes leads to mitotic arrest, mitotic catastrophe, and the triggering of apoptotic programmed cell death. However, the precise mechanism(s) of how mitotic arrest and catastrophe activate the caspase pathway has not been established. Here, we briefly review literature on the involvement of potential cell death mechanisms in cancer therapy. These include the classical caspase-mediated apoptotic programmed cell death, necroptosis mediated by MLKL, and pore forming mechanisms in immune cells, etc. In particular, we discuss a newly recognized mechanism of cell death in taxane-treatment of cancer cells that involves micronucleation and the irreversible rupture of the nuclear membrane. Since cancer cells are commonly retarded in responding to programmed cell death signaling, stabilized microtubule bundle-induced micronucleation and nuclear membrane rupture, rather than triggering apoptosis, may be a key mechanism accounting for the success of taxanes as anti-cancer agents.
Collapse
Affiliation(s)
- Ana P. Xu
- Department of Biology, University of Miami, Coral Gables, FL, United States
| | - Lucy B. Xu
- Department of Biology, University of Miami, Coral Gables, FL, United States
| | - Elizabeth R. Smith
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Joshua S. Fleishman
- College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Xiang-Xi Xu
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
45
|
Rajan RK, Kumar RP, Ramanathan M. Piceatannol improved cerebral blood flow and attenuated JNK3 and mitochondrial apoptotic pathway in a global ischemic model to produce neuroprotection. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:479-496. [PMID: 37470802 DOI: 10.1007/s00210-023-02616-0] [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: 12/26/2022] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Abstract
Cerebral ischemia is one of the leading causes of death and disability worldwide. The only FDA-approved treatment is recanalization with systemic tissue plasminogen activators like alteplase, although reperfusion caused by recanalization can result in neuroinflammation, which can cause brain cell apoptosis. Therefore, after an ischemic/reperfusion injury, interventions are needed to minimize the neuroinflammatory cascade. In the present study, piceatannol (PCT) was studied for its neuroprotective efficacy in a rat model of global ischemic injury by attenuating c-Jun N-terminal kinase 3 (JNK3) downstream signaling. PCT is a resveratrol analog and a polyphenolic stilbenoid naturally occurring in passion fruit and grapes. The neuroprotective efficacy of PCT (1, 5, 10 mg/kg) in ischemic conditions was assessed through pre- and post-treatment. Cerebral blood flow (CBF) and tests for functional recovery were assessed. Protein and gene expression were done for JNK3 and other inflammatory markers. A docking study was performed to identify the amino acid interaction. The results showed that PCT improved motor and memory function as measured by a functional recovery test believed to be due to an increase in cerebral blood flow. Also, the caspase signaling which promotes apoptosis was found to be down-regulated; however, nitric oxide synthase expression was up-regulated, which could explain the enhanced cerebral blood flow (CBF). According to our findings, PCT impeded c-Jun N-terminal kinase 3 (JNK3) signaling by suppressing phosphorylation and disrupting the mitochondrial apoptotic pathway, which resulted in the neuroprotective effect. Molecular docking analysis was performed to investigate the atomic-level interaction of JNK3 and PCT, which reveals that Met149, Leu206, and Lys93 amino acid residues are critical for the interaction of PCT and JNK3. According to our current research, JNK3 downstream signaling and the mitochondrial apoptosis pathway are both inhibited by PCT, which results in neuroprotection under conditions of global brain ischemia. Piceatannol attenuated JNK3 phosphorylation during the ischemic condition and prevented neuronal apoptosis.
Collapse
Affiliation(s)
- Ravi Kumar Rajan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, 641004, Tamilnadu, India.
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science, Girijananda Chowdhury University, Dekargaon, Tezpur, 784501, Assam, India.
| | - Ram Pravin Kumar
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, 641004, Tamilnadu, India
| | - M Ramanathan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, 641004, Tamilnadu, India
| |
Collapse
|
46
|
Zhou Y, Yang D, Qiang Z, Meng Y, Li R, Fan X, Zhao W, Meng Y. Ribosome-inactivating Protein MAP30 Isolated from Momordica Charantia L. Induces Apoptosis in Hepatocellular Carcinoma Cells. Recent Pat Anticancer Drug Discov 2024; 19:223-232. [PMID: 36330636 DOI: 10.2174/1574892818666221103114649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Ribosome-inactivating proteins (RIPs) have been reported to exert antitumor and anti-virus activities. A recent patent CN202011568116.7 has developed a new method to prepare Momordica anti-HIV protein of 30 kDa (MAP30). MAP30 is a type I RIP, which kills various tumor cells through the N-glycosidase activity and irreversibly inhibits protein synthesis. OBJECTIVE To assess the potential role of MAP30 in inducing apoptosis of human hepatocellular carcinoma HCC-LM3 cells and elucidate the molecular mechanism of MAP30. METHODS CCK-8 assay was used to assess the proliferation of HCC-LM3 cells. Flow cytometry was used to measure the cycle, the level of ROS and apoptosis in HCC-LM3 cells. Western blots was used to measure protein levels. RESULTS Treatment with MAP30 reduced survival and proliferation of human liver cancer HCCLM3 cells in a dose-dependent manner. PI staining showed cell cycle arrest in G0/G1 phase. Furthermore, MAP30 increased the level of ROS in HCC-LM3 cells in 24 h treatment. To further confirm the role of MAP30 in inducing cell apoptosis, immunoblotting was carried out to detect the change of apoptosis-related proteins including PARP poly (ADP-ribose) polymerase (PARP- 1), Casepase3 and Cleaved-Caspase9. We found that PARP-1 and Caspase-3 were downregulated, whereas Cleaved-Caspase9 was up-regulated in HCC-LM3 cells treated with MAP30. CONCLUSION This study indicated that MAP30 has the potential to be a novel therapeutic agent for human hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yiping Zhou
- School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu 610500, Sichuan, China
| | - Di Yang
- School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu 610500, Sichuan, China
| | - Zihao Qiang
- School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu 610500, Sichuan, China
| | - Yanfa Meng
- Key Laboratory of Bio-resources and Eco-environment Ministry of Education/Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu 610064, China
| | - Ruigang Li
- Key Laboratory of Bio-resources and Eco-environment Ministry of Education/Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu 610064, China
| | - Xiang Fan
- Key Laboratory of Bio-resources and Eco-environment Ministry of Education/Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Science, Sichuan University, Chengdu 610064, China
| | - Wei Zhao
- School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu 610500, Sichuan, China
| | - Yao Meng
- School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu 610500, Sichuan, China
| |
Collapse
|
47
|
Liu H, Yao Q, Wang X, Xie H, Yang C, Gao H, Xie C. The research progress of crosstalk mechanism of autophagy and apoptosis in diabetic vascular endothelial injury. Biomed Pharmacother 2024; 170:116072. [PMID: 38147739 DOI: 10.1016/j.biopha.2023.116072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/08/2023] [Accepted: 12/21/2023] [Indexed: 12/28/2023] Open
Abstract
In recent years, the widespread prevalence of diabetes has become a major killer that threatens the health of people worldwide. Of particular concern is hyperglycemia-induced vascular endothelial injury, which is one of the factors that aggravate diabetic vascular disease. During the process of diabetic vascular endothelial injury, apoptosis is an important pathological manifestation and autophagy is a key regulatory mechanism. Autophagy and apoptosis interact with each other. Hence, the crosstalk mechanism between the two processes is an important means of regulating diabetic vascular endothelial injury. This article reviews the research progress in apoptosis in the context of diabetic vascular endothelial injury and discusses the crosstalk mechanism of autophagy and apoptosis and its role in this injury. The purpose is to guide the prevention and treatment of diabetic vascular endothelial injury in the future.
Collapse
Affiliation(s)
- Hanyu Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, PR China
| | - Qiyuan Yao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, PR China
| | - Xueru Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, PR China
| | - Hongyan Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan 610075, PR China; Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, PR China
| | - Chan Yang
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, PR China.
| | - Hong Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan 610075, PR China; Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, PR China.
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan 610075, PR China; Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, PR China.
| |
Collapse
|
48
|
Chi W, Fu J, Martyniuk CJ, Wang J, Zhou L. Post-Subfunctionalization Functions of HIF-1αA and HIF-1αB in Cyprinid Fish: Fine-Tuning Mitophagy and Apoptosis Regulation Under Hypoxic Stress. J Mol Evol 2023; 91:780-792. [PMID: 37924420 DOI: 10.1007/s00239-023-10138-9] [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: 03/31/2023] [Accepted: 10/22/2023] [Indexed: 11/06/2023]
Abstract
Hypoxia-inducible factor 1 (HIF-1) is a crucial transcriptional factor that can restore oxygen balance in the body by regulating multiple vital activities. Two HIF-1α copies were retained in cyprinid fish after experiencing a teleost-specific genome duplication. How the "divergent collaboration" of HIF-1αA and HIF-1αB proceeds in regulating mitophagy and apoptosis under hypoxic stress in cells of cyprinid fish remains unclear. In this study, zebrafish HIF-1αA/B expression plasmids were constructed and transfected into the epithelioma papulosum cyprini cells and were subjected to hypoxic stress. HIF-1αA induced apoptosis through promoting ROS generation and mitochondrial depolarization when cells were subjected to oxygen deficiency. Conversely, HIF-1αB was primarily responsible for mitophagy induction, prompting ATP production to mitigate apoptosis. HIF-1αA did not induce mitophagy in the mitochondria and lysosomes co-localization assay but it was involved in the regulation of different mitophagy pathways. Over-expression of HIF-1αA increased the expression of bnip3, fundc1, Beclin1, and foxo3, suggesting it has a dual role in mitochondrial autophagy and cell death. Each duplicated copy also experienced functional divergence and target shifting in the regulation of complexes in the mitochondrial electron transport chain (ETC). Our findings shed light on the post-subfunctionalization function of HIF-1αA and HIF-1αB in zebrafish to fine-tune regulation of mitophagy and apoptosis following hypoxia exposure.
Collapse
Affiliation(s)
- Wei Chi
- School of Life Sciences, Huizhou University, Huizhou, 510607, China.
| | | | - Chris J Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Jiangyong Wang
- School of Life Sciences, Huizhou University, Huizhou, 510607, China
| | - Libin Zhou
- School of Life Sciences, Huizhou University, Huizhou, 510607, China
| |
Collapse
|
49
|
Liu P, Zhao L, Zitvogel L, Kepp O, Kroemer G. The BCL2 inhibitor venetoclax mediates anticancer effects through dendritic cell activation. Cell Death Differ 2023; 30:2447-2451. [PMID: 37845384 PMCID: PMC10733328 DOI: 10.1038/s41418-023-01232-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 10/18/2023] Open
Abstract
BCL2 is an apoptosis-inhibitory oncoprotein that also possesses apoptosis-unrelated activities. Pharmacological BCL2 inhibitors have been developed with the scope of driving BCL2-dependent cancer cells into apoptosis, and one BCL2 antagonist, venetoclax, has been clinically approved for the treatment of specific leukemias and lymphomas. Nonetheless, it appears that venetoclax, as well as genetic BCL2 inhibition, can mediate anticancer effects through an indirect action. Such an indirect effect relies on the enhancement of the immunostimulatory function of dendritic cells, hence increasing tumor immunosurveillance. Mechanistically, BCL2 inhibition involves improved antigen presentation by conventional type-1 dendritic cells (cDC1s) due to the activation of an interferon response, leading to a T cell-mediated anticancer immune response that can be further enhanced by PD-1 blockade. These findings support the emerging hypothesis that successful antineoplastic drugs generally mediate their effects indirectly, through the immune system, rather via merely cell-autonomous effects on malignant cells.
Collapse
Affiliation(s)
- Peng Liu
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Liwei Zhao
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, ClinicObiome, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France.
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
50
|
Ahamed A, Hasan M, Samanta A, Alam SSM, Jamil Z, Ali S, Hoque M. Prospective pharmacological potential of cryptotanshinone in cancer therapy. PHARMACOLOGICAL RESEARCH - MODERN CHINESE MEDICINE 2023; 9:100308. [DOI: 10.1016/j.prmcm.2023.100308] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|