1
|
Guo C, Kumar A, Liao C. Pterostilbene Exhibited the Anticancer Effect Against 1, 2-Dimethylhydrazine (DMH)-Induced Colorectal Cancer via Alteration of Oxidative Stress, Inflammation and Gut Microbiota. J Biochem Mol Toxicol 2025; 39:e70123. [PMID: 40084940 DOI: 10.1002/jbt.70123] [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: 10/16/2024] [Revised: 12/06/2024] [Accepted: 12/21/2024] [Indexed: 03/16/2025]
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related deaths worldwide. The current investigation aimed to assess the chemoprotective effects of pterostilbene against 1,2-dimethylhydrazine (DMH)-induced colorectal cancer in mice. An in-silico study was conducted to perform docking studies against nuclear factor kappa factor (NF-κB). CRC was induced in mice by administering DMH (20 mg/kg) subcutaneously, and the mice were subsequently administered various dosages of pterostilbene (5, 10, and 15 mg/kg). At the end of the study, various biochemical parameters, including inflammatory cytokines, inflammatory markers, and antioxidant enzymes, were examined. Additionally, the mice's stools were collected for the analysis of intestinal microbiota. A total of 5 hydrogen bonds were identified between NF-κB and pterostilbene using LigPlot+. Pterostilbene significantly (p < 0.001) reduced tumor incidence, tumor weight, and increased body weight. Pterostilbene significantly (p < 0.001) altered the levels of lipid peroxidation, reduced glutathione, superoxide dismutase, glutathione peroxidase, and catalase as well as the activity of both phase I and phase II enzymes. Furthermore, pterostilbene significantly (p < 0.001) decreased the levels of proinflammatory cytokines such as tumor necrosis factor-α, interleukin-6, interferon-γ, and interleukin-1β, while increasing the levels of anti-inflammatory cytokines like interleukin-4 and interleukin-10. Pterostilbene considerably suppressed the levels of cyclooxygenase-2 and prostaglandin E2, as well as inducible nitric oxide synthase and simultaneously elevated the levels of apoptosis-related parameters, including caspase-3, caspase-8, and caspase-9. Moreover, pterostilbene significantly reduced the abundance of Staphylococcus in the intestinal microbiota and enhanced the levels of beneficial bacteria, such as Bifidobacterium, Akkermansia, and Lactobacillus. Pterostilbene demonstrated a chemoprotective effect against CRC by effectively reducing oxidative stress, mitigating inflammatory responses, and inducing alterations in gut microbiota levels.
Collapse
Affiliation(s)
- Chengyun Guo
- Department of Anorectal, Sanmenxia Central Hospital, Henan, China
| | - Ankit Kumar
- Department of Pharmacology, Venkateshwara College of Pharmacy, Meerut, India
| | - Chao Liao
- Department of Anorectal, Ankang Hospital of Traditional Chinese Medicine, Ankang, China
| |
Collapse
|
2
|
Amintas S, Dupin C, Derieppe MA, Moranvillier I, Lamrissi I, Bourdié C, Feurer Z, Fernandez B, Heng-Pradère T, Moreau-Gaudry F, Bedel A, Vendrely V, Dabernat S. Resveratrol and capsaicin as safer radiosensitizers for colorectal cancer compared to 5-fluorouracil. Biomed Pharmacother 2025; 183:117799. [PMID: 39755023 DOI: 10.1016/j.biopha.2024.117799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/14/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND AND AIM Standard rectal cancer treatment includes neoadjuvant radiotherapy sensitized by 5-fluorouracil (5-FU) chemotherapy. However, 5-FU increased chemoradiotherapy response rate comes with significant toxicity, especially in older, frail patients. The development of alternatives to chemotherapy enabling radiosensitization with limited systemic toxicity is therefore needed to improve patient management. Bioactive food components (BFCs) can exhibit chemo or radio-sensitizing properties against cancer cells. Moreover, the cytotoxic action of BFCs may be tumor-specific, with reduced impact on healthy cells. We hypothesized that BFCs, in particular resveratrol and capsaicin, alone or in association, could lead to specific radio-sensitization of colorectal tumors while offering reduced toxicity compared to 5-FU. EXPERIMENTAL PROCEDURE Colorectal tumor and non-tumor cell lines were treated with resveratrol, capsaicin, or 5-FU, alone or in combination, then irradiated; survival, cell cycle, and apoptosis were analyzed. RAGγ2C-/- mice with xenografts received oral resveratrol, resveratrol + capsaicin, or 5-FU, followed by radiotherapy, with tumor growth and systemic toxicity evaluated. KEY RESULTS Resveratrol alone or in association with capsaicin radio-potentiates colorectal tumor cells in vitro, impacting both cell cycle and apoptosis. In a preclinical mouse model, the oral administration of resveratrol and capsaicin, but not resveratrol alone, allowed the radio-sensitization of subcutaneous colorectal tumors with similar efficiency to 5-FU. Moreover, the global as well as the hematological toxicity of the BFC association was lower than those of 5-FU. CONCLUSION This work establishes BFCs as effective enhancers of radiotherapy, offering a safer alternative to traditional radiosensitization with chemotherapy.
Collapse
Affiliation(s)
- Samuel Amintas
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Tumor Biology and Tumor Bank Department, Bordeaux University Hospital, Bordeaux, France
| | - Charles Dupin
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Radiotherapy Department, Bordeaux University Hospital, Bordeaux, France
| | - Marie-Alix Derieppe
- University of Bordeaux, Bordeaux, France; Shared Animal Facility, Common Animal Facilities Service, University of Bordeaux, Bordeaux, France
| | - Isabelle Moranvillier
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France
| | - Isabelle Lamrissi
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France
| | - Corine Bourdié
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France
| | - Zoe Feurer
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France
| | - Benjamin Fernandez
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Colorectal Digestive Surgery Department, Bordeaux University Hospital (CHU de Bordeaux), France
| | - Tyty Heng-Pradère
- Pathology Department, Bordeaux University Hospital (CHU de Bordeaux), Bordeaux, France
| | - François Moreau-Gaudry
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Biochemistry Department, Bordeaux University Hospital (CHU de Bordeaux), Bordeaux, France
| | - Aurélie Bedel
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Biochemistry Department, Bordeaux University Hospital (CHU de Bordeaux), Bordeaux, France
| | - Véronique Vendrely
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Radiotherapy Department, Bordeaux University Hospital, Bordeaux, France
| | - Sandrine Dabernat
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Biochemistry Department, Bordeaux University Hospital (CHU de Bordeaux), Bordeaux, France.
| |
Collapse
|
3
|
Jakimiuk K, Kruk A, Lemieszek MK, Strawa JW, Granica S, Wiater A, Tomczyk M. Ex vivo biotransformation of lady's mantle extracts via the human gut microbiota: the formation of phenolic metabolites and their impact on human normal and colon cancer cell lines. Front Pharmacol 2025; 16:1504787. [PMID: 39911846 PMCID: PMC11794788 DOI: 10.3389/fphar.2025.1504787] [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: 10/01/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025] Open
Abstract
Introduction For centuries, various species from the genus Alchemilla have been utilized in traditional medicine worldwide. Among them, Alchemilla vulgaris L. (Rosaceae) stands out as a promising herbal drug candidate due to its phytochemicals displaying anti-inflammatory and antioxidant properties. Methods In our study, we investigated the interaction between the human gut microbiota and lady's mantle herb extract (AV) following the biotransformation of the extract's constituents and their impact on colorectal cancer cells (HT-29) and normal CCD 841 CoN epithelial cells. The A. vulgaris herb metabolites were obtained by incubating the extract (AV) with human fecal slurries from three healthy donors (D1, D2, and D3). Results After incubating the AV extract with the human gut microbiota (AVD1-AVD3 samples), thirty-three metabolites were detected and characterized by LC-MS. Among them, one was identified as urolithin C. The AV and AVD1-AVD3 extracts and their metabolites exhibit various levels of antiproliferative and cytotoxic activities against cancer cells. Their biological effect might be linked to the changes and direct activity of bioavailable metabolites. Samples from AVD1, AVD2, and AVD3 increase the lactate dehydrogenase (LDH) released from damaged colon cancer cells in a dose-dependent manner. At 250 μg/mL, AVD1, AVD2, and AVD3 elevated the LDH level by 12.6%, 25.3%, and 30.0%, respectively. The biotransformed samples also showed significantly higher antiproliferative activity than the AV extract. The most active sample from donor 3 (AVD3) reached IC50 = 471 μg/mL. Discussion The differences in anticancer effect might be linked to the changes and direct activity of bioavailable metabolites. The non-transformed AV extract affected neither normal nor cancer colon cells, indicating the beneficial effect of the biotransformation procedure on the anticancer properties of the evaluated extracts. The above results clearly indicate that microbial metabolism is a crucial factor that is potent in altering the biological activity of lady's mantle extract.
Collapse
Affiliation(s)
- Katarzyna Jakimiuk
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, Białystok, Poland
| | - Aleksandra Kruk
- Microbiota Lab, Department of Pharmaceutical Biology, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | | | - Jakub W. Strawa
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, Białystok, Poland
| | - Sebastian Granica
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Adrian Wiater
- Department of Industrial and Environmental Microbiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Michał Tomczyk
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
4
|
Sharma U, Sahni PK, Sharma B, Gupta M, Kaur D, Mathkor DM, Haque S, Khatoon S, Tuli HS, Mishra A, Ahmad F. Silymarin: a promising modulator of apoptosis and survival signaling in cancer. Discov Oncol 2025; 16:66. [PMID: 39836338 PMCID: PMC11751200 DOI: 10.1007/s12672-025-01800-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Cancer, one of the deadliest diseases, has remained the epicenter of biological research for more than seven decades. Yet all the efforts for a perfect therapeutic cure come with certain limitations. The use of medicinal plants and their phytochemicals as therapeutics has received much attention in recent years. Silymarin, a polyphenolic flavonoid with a variety of anti-cancerous properties, was isolated from the plant Silybum marianum. The present review centres on the function of silymarin in controlling important signalling pathways related to apoptosis and survival, such as the JAK/STAT pathway, PI3K/Akt/mTOR, Bcl-2/Bax, and Fas/FasL. It is emphasised that silymarin's capacity to target these pathways is a key mechanism behind its anticancer effects against a variety of malignancies. By upregulating pro-apoptotic and downregulating anti-apoptotic proteins, silymarin controls a series of events that result in tumor suppression and cell death in a variety of cancer types. The low bioavailability and limited therapeutic efficacy of silymarin are improved by the application of various nano-delivery systems. As efficient carriers, liposomes, polymeric micelles, lipid- and metal-based nanoparticles, increase the solubility and distribution of silymarin in target tissues. Lastly, a number of preclinical studies that provide a basis for upcoming therapeutic interventions are highlighted in the review, providing encouraging directions for additional research and advancement.
Collapse
Affiliation(s)
- Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda, 151001, India
| | - Praveen Kumar Sahni
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda, 151001, India
| | - Bunty Sharma
- Department of Biotechnology, Graphic Era (Deemed to Be University), Dehradun, Uttarakhand, India
| | - Madhu Gupta
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Damandeep Kaur
- University Center for Research & Development (UCRD), Chandigarh University, Gharuan, Mohali, Punjab, 140413, India
| | - Darin Mansor Mathkor
- Department of Nursing, College of Nursing and Health Sciences, Jazan University, 45142, Jazan, Saudi Arabia
| | - Shafiul Haque
- Department of Nursing, College of Nursing and Health Sciences, Jazan University, 45142, Jazan, Saudi Arabia
- Universidad Espiritu Santo, Samborondon, Ecuador
| | - Sabiha Khatoon
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India
| | - Astha Mishra
- Department of Optometry, Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab, India
| | - Faraz Ahmad
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, India.
| |
Collapse
|
5
|
Alrumaihi F. Exploring the Chemopreventive Potential of Artemisia annua Methanolic Extract in Colorectal Cancer Induced by Azoxymethane in Mice. Pharmaceuticals (Basel) 2024; 18:34. [PMID: 39861099 PMCID: PMC11769478 DOI: 10.3390/ph18010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Colorectal cancer (CRC) remains a major global health burden, necessitating innovative preventive approaches. Artemisia annua (A. annua), known for its extensive pharmacological properties, has shown potential in cancer therapy. This study investigates the chemopreventive efficacy of methanolic extract of A. annua (MEA) in an azoxymethane (AOM)-induced murine model of CRC, with a focus on its antioxidant, biomarker modulation, and pro-apoptotic activities. Methods: MEA was obtained via cold solvent extraction, yielding 39%, and demonstrated potent in vitro cytotoxicity against HCT116 and RKO colon cancer cell lines, with IC50 values of 20 µg/mL and 15 µg/mL, respectively. Swiss albino mice were treated with MEA beginning two weeks before AOM induction, with treatment continuing for 21 weeks. Survival was monitored for 40 weeks. Key outcomes included serum biomarker levels (ADA, GGT, CD73, LDH), antioxidant enzyme activities (SOD, CAT, GPx1, MDA), reactive oxygen species (ROS) modulation, apoptosis induction, and histopathological evaluation. Results: MEA significantly improved survival rates, reduced AOM-induced weight loss, and modulated cancer biomarkers, with marked reductions in ADA, GGT, CD73, and LDH levels. Antioxidant defenses were restored, as evidenced by increased SOD, CAT, and GPx1 activities and decreased MDA levels. ROS levels were significantly reduced, and apoptosis in colonic cells was effectively induced. Histopathological analysis revealed substantial mitigation of CRC-associated morphological abnormalities. Conclusions: MEA exhibits robust chemopreventive properties, demonstrating its potential to reduce oxidative stress, modulate key biomarkers, and induce apoptosis in CRC. These findings position MEA as a promising natural candidate for CRC prevention and therapy, warranting further exploration for clinical application.
Collapse
Affiliation(s)
- Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
6
|
Aktar A, Bhuia S, Chowdhury R, Ferdous J, Khatun M, Hasan SA, Mia E, Hasan R, Islam MT. An Insight of Plant Source, Toxicological Profile, and Pharmacological Activities of Iridoid Loganic Acid: A ComprehensiveReview. Chem Biodivers 2024; 21:e202400874. [PMID: 39113595 DOI: 10.1002/cbdv.202400874] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 08/08/2024] [Indexed: 10/20/2024]
Abstract
This study evaluates the pharmacological effects of iridoid glucoside loganic acid, a plant constituent with diverse properties, based on literature, and explores the underlying cellular mechanisms for treating several ailments. Data were collected from reliable electronic databases, including PubMed, Scopus, Web of Science, and Google Scholar, etc. The results demonstrated the anti-inflammatory, anti-oxidant, and other protective effects of loganic acid on metabolic diseases and disorders such as atherosclerosis, diabetes, and obesity, in addition to its osteoprotective and anticancer properties. The antioxidant activity of loganic acid demonstrates its capacity to protect cells from oxidative damage and mitigates inflammation by reducing the activity of inflammatory cytokines involving TNF-α and IL-6, substantially upregulating the expression of PPAR-γ/α, and decreasing the clinical signs of inflammation-related conditions related to hypertriglyceridemia and atherosclerosis. Meanwhile, loganic acid inhibits bone loss, exhibits osteoprotective properties by increasing mRNA expression levels of bone synthesizing genes such as Alpl, Bglap, and Sp7, and significantly increases osteoblastic proliferation in preosteoblast cells. Loganic acid is an anti-metastatic drug that reduces MnSOD expression, inhibits EMT and metastasis, and prevents cellular migration, proliferation, and invasion in hepatocellular carcinoma cells. However, additional clinical trials are required to assess its safety, efficacy, and human dose.
Collapse
Affiliation(s)
- Asma Aktar
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Dhaka, Bangladesh
| | - Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Dhaka, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Dhaka, Bangladesh
| | - Jannatul Ferdous
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Muslima Khatun
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Dhaka, Bangladesh
| | - Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Dhaka, Bangladesh
| | - Emon Mia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Dhaka, Bangladesh
| | - Rubel Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Dhaka, Bangladesh
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Dhaka, Bangladesh
| |
Collapse
|
7
|
Beyaztas H, Bozali K, Koc S, Ozdemir M, Yalcin B, Guler EM. Synthesis and characterization of 7-diethylamino-4-Chloromethyl coumarin: Spectroscopic analysis, molecular docking, and anticancer activity on large intestine carcinoma cells. Chem Biol Interact 2024; 404:111287. [PMID: 39442679 DOI: 10.1016/j.cbi.2024.111287] [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: 08/20/2024] [Revised: 10/09/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024]
Abstract
Cancer, characterized by uncontrolled cell growth and metastasis, poses a significant global health burden, ranking as a leading cause of mortality worldwide. Colorectal cancer (CRC) specifically accounts for a substantial portion of cancer cases, with increasing incidence projected over the coming decades. While conventional treatments exist, they often entail adverse effects and limited efficacy, driving interest in natural remedies like coumarin derivatives due to their diverse biological activities and perceived safety profile. This study focuses on the synthesis and characterization of a novel compound, 7-diethylamino-4-chloromethyl coumarin (referred to as 7D4C), derived from coumarin. Structural elucidation employed Fourier transform infrared spectroscopy (FT-IR), proton and carbon-13 nuclear magnetic resonance spectroscopy (1H and 13C NMR), and mass spectrometry (MALDI-TOF-MS). Molecular docking studies were conducted to explore potential biological interactions. Furthermore, the anti-cancer potential of 7D4C was assessed using human epithelial adenocarcinom (LoVo) and healthy fibroblast (CCD-18Co) cell lines. Viability analysis, comet assay for DNA damage, and evaluation of cancer biomarkers including apoptosis, intracellular reactive oxygen species (iROS) levels, mitochondrial membrane potential (MMP), intracellular glutathione (GSH) concentration, and intracellular calcium (iCa2+) levels were performed. The synthesis of 7D4C was successfully completed, and its structure was confirmed. Molecular docking results indicate that 7D4C exhibits strong binding affinity to the p53 protein, highlighting its potential as a novel modulator of p53 activity. Subsequent investigations revealed that the synthesized compound induced apoptosis in cancer cells by reducing MMP and triggering DNA damage through the production of iROS. The promising anti-cancer activity of 7D4C in the LoVo cell line highlights its importance in coumarin-based therapies. Introducing 7D4C could significantly enhance future research in this area, leveraging insights from in vitro coumarin studies.
Collapse
Affiliation(s)
- Hakan Beyaztas
- Department of Medical Biochemistry, Hamidiye Institute of Health Sciences, University of Health Sciences Turkey, Istanbul, Turkiye; Department of Medical Biochemistry, Hamidiye Faculty of Medicine, University of Health Sciences Turkey, Istanbul, Turkiye
| | - Kubra Bozali
- Department of Medical Biochemistry, Hamidiye Institute of Health Sciences, University of Health Sciences Turkey, Istanbul, Turkiye; Department of Medical Biochemistry, Hamidiye Faculty of Medicine, University of Health Sciences Turkey, Istanbul, Turkiye
| | - Sumeyye Koc
- Department of Medical Biochemistry, Hamidiye Institute of Health Sciences, University of Health Sciences Turkey, Istanbul, Turkiye; Department of Medical Biochemistry, Hamidiye Faculty of Medicine, University of Health Sciences Turkey, Istanbul, Turkiye
| | - Mucahit Ozdemir
- Department of Chemistry, Marmara University, Istanbul, Turkiye
| | - Bahattin Yalcin
- Department of Chemistry, Marmara University, Istanbul, Turkiye
| | - Eray Metin Guler
- Department of Medical Biochemistry, Hamidiye Faculty of Medicine, University of Health Sciences Turkey, Istanbul, Turkiye; Department of Medical Biochemistry, University of Health Sciences Turkey, Hamidiye Faculty of Medicine, Haydarpasa Numune Health Application and Research Center, Istanbul, Turkiye.
| |
Collapse
|
8
|
El Hajj J, Karam L, Jaber A, Cheble E, Akoury E, Kobeissy PH, Ibrahim JN, Yassin A. Evaluation of Antiproliferative Potentials Associated with the Volatile Compounds of Lantana camara Flowers: Selective In Vitro Activity. Molecules 2024; 29:5431. [PMID: 39598820 PMCID: PMC11597665 DOI: 10.3390/molecules29225431] [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: 09/16/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Probing the chemical profiles and biological activities of medicinal plants is important for the discovery of new potent therapeutic products. Our study deciphers the chemical composition of the essential oils (EOs) obtained from three different flowers of Lantana camara and evaluates their antioxidant and anticancer activities. This work represents the first study of EOs obtained from this plant and is based particularly on the difference in flower color. In addition, no other reports dealing specifically with the antitumor effects of such flower-derived EOs have been described in the literature. The collected flowers, white, pink, and orange, were extracted by hydrodistillation to yield EO1, EO2, and EO3 respectively. Gas chromatography-mass spectroscopy was primarily employed to identify the existing volatile compounds in the samples. Their antioxidant activities were screened through both DPPH (2,2-diphenyl-1-picrylhydrazyl) scavenging assays and FRAP (ferric-reducing antioxidant power) assays. The antiproliferative effects were evaluated on two distinct breast cancer cell lines, MCF-7 and MDA-MB-231, and compared to a normal human breast cell line, MCF-10A, using an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium) assay. All EOs showed notable antioxidant potential attributed to the active phytochemical compounds, with results being supported by a positive correlation between such activity and the total phenolic and flavonoid content. The most eminent, EO1, revealed a selective dose-dependent antiproliferative effect in both breast cancer cell lines, thus reflecting its potent role as an anticancer agent. We suggest that this highly selective activity is associated with the presence of bicyclogermacrene and epi-bicyclosesquiphellandrene in its chemical composition.
Collapse
Affiliation(s)
- Jennifer El Hajj
- RDMPN Laboratory, Faculty of Pharmacy, Lebanese University, Beirut BP 14/6573, Lebanon; (J.E.H.); (E.C.)
| | - Louna Karam
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102-2801, Lebanon; (L.K.); (P.H.K.); (J.-N.I.)
| | - Ali Jaber
- Department of Physical Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102-2801, Lebanon;
| | - Edmond Cheble
- RDMPN Laboratory, Faculty of Pharmacy, Lebanese University, Beirut BP 14/6573, Lebanon; (J.E.H.); (E.C.)
| | - Elias Akoury
- Department of Physical Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102-2801, Lebanon;
| | - Philippe Hussein Kobeissy
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102-2801, Lebanon; (L.K.); (P.H.K.); (J.-N.I.)
| | - José-Noel Ibrahim
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102-2801, Lebanon; (L.K.); (P.H.K.); (J.-N.I.)
| | - Ali Yassin
- Department of Physical Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102-2801, Lebanon;
| |
Collapse
|
9
|
Abdelaziz MA, Alalawy AI, Sobhi M, Alatawi OM, Alaysuy O, Alshehri MG, Mohamed ELI, Abdelaziz MM, Algrfan IA, Mohareb RM. Elaboration of chitosan nanoparticles loaded with star anise extract as a therapeutic system for lung cancer: Physicochemical and biological evaluation. Int J Biol Macromol 2024; 279:135099. [PMID: 39197631 DOI: 10.1016/j.ijbiomac.2024.135099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/13/2024] [Accepted: 08/24/2024] [Indexed: 09/01/2024]
Abstract
The research study aimed to maximize the important medical role of star anise extract (SAE) through its loading on a widely available natural polymer (chitosan, Cs). Thus, SAE loaded chitosan nanoparticles (CsNPs) was prepared. The finding illustrated the formation of spherical particles of SAE loaded CsNPs as proved by transmission electron microscope (TEM). In addition, the average particle size of CsNPs and SAE loaded CsNPs are 131.8 ± 24.63 and 318.5 ± 73.94 nm, respectively. Scanning electron microscope (SEM) showed the presence of many spherical particles deposited on the surface of CsNPs owing to the deposition of SAE on the surface and encapsulated into pores of CsNPs. It also showed the presence of elements such as sodium, potassium, copper, magnesium, zinc, calcium, and iron, as well as the elements that accompanied with CsNPs: carbon, oxygen, nitrogen, and phosphorus. The extract was rich in bioactive components, such as anethole, shikimic acid, and different flavonoids, contributing to its medicinal qualities. The bioactive molecules in SAE were assessed by chromatographic analysis. Using the agar well diffusion test, the antibacterial qualities of CsNPs and SAE loaded CsNPs were evaluated against pathogenic bacteria linked to lung illnesses. The most significant inhibition zones showed that the SAE loaded CsNPs had the most antibacterial activity. The anticancer activity using MTT assay was used in the biological assessments to determine the cytotoxicity against the NCl-H460 lung cancer cell line. The results showed that CsNPs loaded with SAE considerably decreased cell viability in a dose-dependent manner, with the most significant anticancer impact by SAE loaded CsNPs. Furthermore, in vivo tests on lung cancer therapy revealed that when compared to other treatment groups, the SAE loaded CsNPs group showed the greatest reduction in tumor biomarkers and inflammation, as seen by decreased levels of Plasma malondialdehyde (MDA), tumor protein 53 (p53), Tumor necrosis factor-alpha (TNF- alpha), and fibronectin. Results concluded that these thorough characterizations, biological assessments, and antibacterial tests have confirmed the effective integration of SAE into CsNPs. Further, SAE loaded CsNPs could be a suitable option for various biomedical applications in tackling lung cancer and the inactivation of bacterial infection.
Collapse
Affiliation(s)
- Mahmoud A Abdelaziz
- Organic Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia.
| | - Adel I Alalawy
- Department of Biochemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Mohamed Sobhi
- Organic Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Omar M Alatawi
- Organic Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Omaymah Alaysuy
- Organic Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Maryam G Alshehri
- Department of Mathematics, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - ELsiddig Idriss Mohamed
- Department of Statistics, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Maiar M Abdelaziz
- Department of Mathematics, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Ibrahim A Algrfan
- Organic Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Rafat M Mohareb
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
10
|
Ye W, Zhao Q, Li P, Zhou T. Scleromitrion diffusum (Willd.) R. J. Wang Inhibits Gastric Cancer via ERBB2/ERBB3/PI3K/AKT Pathway. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2024; 35:831-838. [PMID: 39549017 PMCID: PMC11562600 DOI: 10.5152/tjg.2024.24152] [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: 03/20/2024] [Accepted: 06/26/2024] [Indexed: 11/18/2024]
Abstract
Background/Aims This study aimed to evaluate the anticarcinogenic potential of Scleromitrion diffusum (Willd.) R. J. Wang (SD) extracts in vitro, along with exploring the underlying compatibility mechanisms. Materials and Methods Scleromitrion diffusum (Willd.) R. J. Wang extract was prepared and gastric cancer (GC) cells were treated to detect the half maximal inhibitory concentration (IC50)/proliferation and migration/invasion by MTS method and transwell assay. The compatibility mechanisms of SD were analyzed by systems pharmacology strategy, combined with cellular experimental validation. Results Scleromitrion diffusum (Willd.) R. J. Wang extract showed inhibitory ability on the proliferation of the GC cell lines dose- and time-dependently. A total of 3 active ingredients are involved in anti-gastric cancer effects of SD, based on the top 50 pathways. The "herb-composition-target-pathway" network showed the multi-target and multi-pathway characteristics of SD. There were 52 related targets shared by SD and GC. The cellular experiments supported that SD significantly reduced ERBB2 and ERBB3 expression levels in GC cells. The overexpression of ERBB2 or ERBB3 partially offset the anti-tumor effects of SD. Conclusion Scleromitrion diffusum (Willd.) R. J. Wang inhibited gastric cancer growth and metastasis in vitro, which may be related to the inhibition of the ERBB2/ERBB3/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Wei Ye
- Department of Oncology, Changzhou Cancer Hospital, Changzhou, China
| | - Qiu Zhao
- Department of Oncology, Changzhou Cancer Hospital, Changzhou, China
| | - Peng Li
- Department of Oncology, Changzhou Cancer Hospital, Changzhou, China
| | - Tong Zhou
- Department of Oncology, Changzhou Cancer Hospital, Changzhou, China
| |
Collapse
|
11
|
Pricop MA, Lukinich-Gruia AT, Cristea IM, Păunescu V, Tatu CA. Aristolochia clematitis L. Ethanolic Extracts: In Vitro Evaluation of Antioxidant Activity and Cytotoxicity on Caco-2 Cell Line. PLANTS (BASEL, SWITZERLAND) 2024; 13:2987. [PMID: 39519906 PMCID: PMC11547917 DOI: 10.3390/plants13212987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Aristolochia sp. plants are used in traditional medicine because of their immunostimulatory and anticarcinogenic properties, despite their content of aristolochic acids (AAs), carcinogenic and nephrotoxic agents. Therefore, ethanolic extracts of Aristolochia clematitis leaves, a specie growing in Western Romania, were obtained to study antioxidant and cytotoxic effects. The antioxidant capacity of the extract was evaluated by five in vitro chemical-based assays, proving that ABTS assay was a better method for this type of evaluation showing an IC50 of 160.89 ± 0.21 µg/mL. Furthermore, the cytotoxic effects of the extract were established by an IC50 of 216 µg/mL for 24 h by MTT assay, followed by a cell-based assay on Caco-2 cells by the ABTS method. The antioxidant effects of the A. clematitis extract demonstrate potential therapeutic applications in complementary medicine.
Collapse
Affiliation(s)
- Maria-Alexandra Pricop
- OncoGen Centre, County Hospital Pius Branzeu, 156 Liviu Rebreanu Blvd., 300736 Timisoara, Romania; (M.-A.P.); (I.-M.C.); (V.P.)
- Department of Applied Chemistry and Environmental Engeneering and Inorganic Compounds, Faculty of Industrial Chemistry, Biotechnology and Environmental Engeneering, Polytechnic University of Timisoara, Vasile Pârvan 6, 300223 Timisoara, Romania
| | | | - Iustina-Mirabela Cristea
- OncoGen Centre, County Hospital Pius Branzeu, 156 Liviu Rebreanu Blvd., 300736 Timisoara, Romania; (M.-A.P.); (I.-M.C.); (V.P.)
| | - Virgil Păunescu
- OncoGen Centre, County Hospital Pius Branzeu, 156 Liviu Rebreanu Blvd., 300736 Timisoara, Romania; (M.-A.P.); (I.-M.C.); (V.P.)
- Department of Functional Sciences, Center of Immuno-Physiology (CIFBIOTEH), University of Medicine and Pharmacy “Victor Babes”, Eftimie Murgu Sq. 2, 300041 Timisoara, Romania
| | - Călin Adrian Tatu
- OncoGen Centre, County Hospital Pius Branzeu, 156 Liviu Rebreanu Blvd., 300736 Timisoara, Romania; (M.-A.P.); (I.-M.C.); (V.P.)
- Department of Functional Sciences, Center of Immuno-Physiology (CIFBIOTEH), University of Medicine and Pharmacy “Victor Babes”, Eftimie Murgu Sq. 2, 300041 Timisoara, Romania
| |
Collapse
|
12
|
Kim D, Go SH, Song Y, Lee DK, Park JR. Decursin Induces G1 Cell Cycle Arrest and Apoptosis through Reactive Oxygen Species-Mediated Endoplasmic Reticulum Stress in Human Colorectal Cancer Cells in In Vitro and Xenograft Models. Int J Mol Sci 2024; 25:9939. [PMID: 39337425 PMCID: PMC11432441 DOI: 10.3390/ijms25189939] [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: 08/26/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Decursin, a coumarin isolated from Angelica gigas Nakai, possesses anti-inflammatory and anti-cancer properties. However, the molecular mechanisms underlying its anti-cancer effects against human colorectal cancer (CRC) are unclear. Therefore, this study aimed to evaluate the biological activities of decursin in CRC in vitro and in vivo and to determine its underlying mechanism of action. Decursin exhibited anti-tumor activity in vitro, accompanied by an increase in G1 cell cycle arrest and apoptosis in HCT-116 and HCT-8 CRC cells. Decursin also induced the production of reactive oxygen species (ROS), thereby activating the endoplasmic reticulum (ER) stress apoptotic pathway in CRC cells. Furthermore, the role of ROS in decursin-induced apoptosis was investigated using the antioxidant N-acetyl-L-cysteine. Inhibiting ROS production reversed decursin-induced ER stress. Moreover, decursin significantly suppressed tumor growth in a subcutaneous xenograft mouse model of HCT-116 and HCT-8 CRC cells without causing host toxicity. Decursin also decreased cell proliferation, as documented by Ki-67, and partly increased cleaved caspase 3 expression in tumor tissues by activating ER stress apoptotic pathways. These findings suggest that decursin induces cell cycle arrest and apoptosis in human CRC cells via ROS-mediated ER stress, suggesting that decursin could be a therapeutic agent for CRC.
Collapse
Affiliation(s)
| | | | | | - Dong-Keon Lee
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea; (D.K.); (S.-H.G.); (Y.S.)
| | - Jeong-Ran Park
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea; (D.K.); (S.-H.G.); (Y.S.)
| |
Collapse
|
13
|
Di Carlo E, Sorrentino C. Oxidative Stress and Age-Related Tumors. Antioxidants (Basel) 2024; 13:1109. [PMID: 39334768 PMCID: PMC11428699 DOI: 10.3390/antiox13091109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Oxidative stress is the result of the imbalance between reactive oxygen and nitrogen species (RONS), which are produced by several endogenous and exogenous processes, and antioxidant defenses consisting of exogenous and endogenous molecules that protect biological systems from free radical toxicity. Oxidative stress is a major factor in the aging process, contributing to the accumulation of cellular damage over time. Oxidative damage to cellular biomolecules, leads to DNA alterations, lipid peroxidation, protein oxidation, and mitochondrial dysfunction resulting in cellular senescence, immune system and tissue dysfunctions, and increased susceptibility to age-related pathologies, such as inflammatory disorders, cardiovascular and neurodegenerative diseases, diabetes, and cancer. Oxidative stress-driven DNA damage and mutations, or methylation and histone modification, which alter gene expression, are key determinants of tumor initiation, angiogenesis, metastasis, and therapy resistance. Accumulation of genetic and epigenetic damage, to which oxidative stress contributes, eventually leads to unrestrained cell proliferation, the inhibition of cell differentiation, and the evasion of cell death, providing favorable conditions for tumorigenesis. Colorectal, breast, lung, prostate, and skin cancers are the most frequent aging-associated malignancies, and oxidative stress is implicated in their pathogenesis and biological behavior. Our aim is to shed light on the molecular and cellular mechanisms that link oxidative stress, aging, and cancers, highlighting the impact of both RONS and antioxidants, provided by diet and exercise, on cellular senescence, immunity, and development of an antitumor response. The dual role of ROS as physiological regulators of cell signaling responsible for cell damage and diseases, as well as its use for anti-tumor therapeutic purposes, will also be discussed. Managing oxidative stress is crucial for promoting healthy aging and reducing the risk of age-related tumors.
Collapse
Affiliation(s)
- Emma Di Carlo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Carlo Sorrentino
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
14
|
Zhang J, Huang X, Zhang T, Gu C, Zuo W, Fu L, Dong Y, Liu H. Antitumorigenic potential of Lactobacillus-derived extracellular vesicles: p53 succinylation and glycolytic reprogramming in intestinal epithelial cells via SIRT5 modulation. Cell Biol Toxicol 2024; 40:66. [PMID: 39110260 PMCID: PMC11306434 DOI: 10.1007/s10565-024-09897-y] [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: 12/22/2023] [Accepted: 06/21/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE Colorectal cancer progression involves complex cellular mechanisms. This study examines the effects of Lactobacillus plantarum-derived extracellular vesicles (LEVs) on the SIRT5/p53 axis, focusing on glycolytic metabolic reprogramming and abnormal proliferation in intestinal epithelial cells. METHODS LEVs were isolated from Lactobacillus plantarum and incubated with Caco-2 cells. Differential gene expression was analyzed through RNA sequencing and compared with TCGA-COAD data. Key target genes and pathways were identified using PPI network and pathway enrichment analysis. Various assays, including RT-qPCR, EdU staining, colony formation, flow cytometry, and Western blotting, were used to assess gene expression, cell proliferation, and metabolic changes. Co-immunoprecipitation confirmed the interaction between SIRT5 and p53, and animal models were employed to validate in vivo effects. RESULTS Bioinformatics analysis indicated the SIRT5/p53 axis as a critical pathway in LEVs' modulation of colorectal cancer. LEVs were found to inhibit colorectal cancer cell proliferation and glycolytic metabolism by downregulating SIRT5, influencing p53 desuccinylation. In vivo, LEVs regulated this axis, reducing tumor formation in mice. Clinical sample analysis showed that SIRT5 and p53 succinylation levels correlated with patient prognosis. CONCLUSION Lactobacillus-derived extracellular vesicles play a pivotal role in suppressing colonic tumor formation by modulating the SIRT5/p53 axis. This results in decreased glycolytic metabolic reprogramming and reduced proliferation in intestinal epithelial cells.
Collapse
Affiliation(s)
- Jingbo Zhang
- Department of Spleen and Stomach Disease, Yubei District Hospital of Traditional Chinese Medicine, Chongqing, 401120, China
| | - Xiumei Huang
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.3, North Guangchang Road, Changyuan Street, Rongchang District, Chongqing, 402460, China
| | - Tingting Zhang
- Department of Digestion, Rongchang District People's Hospital of Chongqing, No.3, North Guangchang Road, Changyuan Street, Rongchang District, Chongqing, 402460, China
| | - Chongqi Gu
- Department of Pediatrics, Rongchang District People's Hospital, Chongqing, 402460, China
| | - Wei Zuo
- Department of Herbal Medicine, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
- Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha, 410219, China
| | - Lijuan Fu
- Department of Herbal Medicine, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
- Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha, 410219, China
| | - Yiping Dong
- Department of Digital Medicine, Department of Bioengineering and Imaging, Army Medical University, Chongqing, 400038, China
| | - Hao Liu
- Department of Pediatrics, Rongchang District People's Hospital, Chongqing, 402460, China.
| |
Collapse
|
15
|
Hu Y, Wu X, Tan X, Zhang J. Hsa_circRNA_007630 knockdown delays colon cancer progression by modulation of ferroptosis via miR-506-3p/AURKA axis. J Biochem Mol Toxicol 2024; 38:e23771. [PMID: 39015057 DOI: 10.1002/jbt.23771] [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: 02/04/2024] [Revised: 06/06/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024]
Abstract
Colon cancer contributes to high mortality rates internationally that has seriously endangered human health. Aurora kinase A (AURKA) served as a key molecule in colon cancer. However, its role of AURKA on regulating ferroptosis in colon cancer and their possible interactions with miRNAs and circRNAs remain still elusive. Comprehensive bioinformatics analysis after RNA-sequencing was conducted to determine the differentially expressed genes (DEGs), ferroptosis-related DEGs and hub genes. The direct relationship between miR-506-3p and hsa_circRNA_007630 or AURKA was predicted, then verified by dual luciferase reporter and quantitative real-time polymerase chain reaction. The rescue experiments were conducted by cotransfection with si-hsa_circRNA_007630, miR-506-3p inhibitor or pcDNA-AURKA in HT29 cells. Erastin was used to induce ferroptosis in HT29 cells and validated by detecting levels of intracellular Fe2+, lipid reactive oxygen species, glutathione, malondialdehyde and ferroptosis markers expression. We screened a total of 331 DEGs, 26 ferroptosis-related genes, among which 3 hub genes were identified through PPI network analysis. Therein, AURKA expression was elevated in colon cancer cells. Moreover, AURKA was targeted by miR-506-3p, and hsa_circRNA_007630 operated as miR-506-3p sponge. The effect of hsa_circRNA_007630 depletion on the inhibiting malignant phenotypes of HT29 cells was rescued by inhibition of miR-506-3p or AURKA overexpression. Additionally, AURKA reduced erastin-induced ferroptosis in HT29 cells. Depletion of circRNA_007630 exerts as a suppressive role in colon cancer through a novel miR-506-3p/AURKA pathway related to ferroptosis, and might become a novel marker for colon cancer.
Collapse
Affiliation(s)
- Ying Hu
- Department of Gastroenterology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Xiongjian Wu
- Department of Gastroenterology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Xiaobin Tan
- Department of Clinical Laboratory, First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Jingzhi Zhang
- Department of Gastroenterology, Ganzhou People's Hospital (Ganzhou Hospital Affiliated to Nanchang University), Ganzhou City, China
| |
Collapse
|
16
|
Yadav P, Singh SK, Datta S, Verma S, Verma A, Rakshit A, Bali A, Bhatti JS, Khurana A, Navik U. Therapeutic potential and pharmacological mechanism of visnagin. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:399-412. [PMID: 38797603 DOI: 10.1016/j.joim.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 04/10/2024] [Indexed: 05/29/2024]
Abstract
Visnagin is a furanochromone and one of the most important compound in the Ammi visnaga (L.) Lam (a synonym of Visnaga daucoides Gaertn.) plant, which is used to cure various ailments. Many investigations into the bioactive properties of visnagin have been studied to date. The literature on visnagin demonstrates its biological properties, including anti-inflammatory, anti-diabetic, and beneficial effects in cardiovascular and renal diseases. Moreover, visnagin improves sperm quality parameters, stimulates steroidogenesis, and increases serum gonadotropins and testosterone levels, while decreasing pro-inflammatory cytokines, oxidative damage, genomic instability, and it modulates apoptosis. Thus, visnagin has emerged as an exciting lead for further research, owing to its potential in various unmet clinical needs. The current review summarized its basic structure, pharmacokinetics, and pharmacological effects, focusing on its mechanisms of action. The review will help to understand the potential of visnagin as an alternative treatment strategy for several diseases and provide insight into research topics that need further exploration for visnagin's safe clinical use. Please cite this article as: Yadav P, Singh SK, Datta S, Verma S, Verma A, Rakshit A, Bali A, Bhatti JS, Khurana A, Navik U. Therapeutic potential and pharmacological mechanism of visnagin. J Integr Med. 2024; 22(4): 399-412.
Collapse
Affiliation(s)
- Poonam Yadav
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Sumeet Kumar Singh
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5000, USA
| | - Saloni Verma
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Aarti Verma
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Arnab Rakshit
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Anjana Bali
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Jasvinder Singh Bhatti
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Amit Khurana
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India.
| | - Umashanker Navik
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India.
| |
Collapse
|
17
|
Rao PP, Shenoy Belle V, Nayak AG, Kumar N, Rao V, Cheruku SP, Prabhu K. Evaluation of the merit of ethanolic extract of Annona reticulata as an anti-cancer agent in human colon cancer cell lines (HCT-116). F1000Res 2024; 12:1571. [PMID: 39866203 PMCID: PMC11759889 DOI: 10.12688/f1000research.141542.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 01/28/2025] Open
Abstract
Background Colon cancer is the third most common cancer type worldwide. Novel alternative therapeutic anti-cancer drugs against colon cancer with less toxicity are to be explored . This study was aimed to explore the anti-proliferative and anti-migratory activity of various fractions of Annona reticulata ethanolic leaf extract on human colon cancer cell lines (HCT-116) and to explore the potential molecular targets from the most potent plant extract fraction. Methods After obtaining ethical clearance from the institutional ethics committee, the extract and fractions were prepared and a preliminary analysis of the phytochemical was done qualitatively. Total phenolic and flavonoids were determined. Ethanolic leaf extract and its fractions were subjected to cytotoxicity analysis using the sulforhodamine B assay and the most promising fraction which showed the highest viability was selected to study anti-migratory activity. The anti-migratory effect was studied using a scratch wound healing assay. Gas chromatography-mass spectrometry (GC-MS) was done to identify the major phytocompounds present in the fraction. The major five phytocompounds identified from the GC-MS were subjected to bioinformatics analysis. Result Among the four fractions, the petroleum ether fraction exhibited the highest anti-proliferative activity. The migration of colon cancer cells was significantly inhibited by the extract and petroleum ether fraction. The major phytocompounds identified from GC-MS were phytol (13.03%), 2,6-bis (3,4-methylenedioxyphenyl)-3,7-dioxabicyclo (3.3.0) octane (11.95%), gamma.-sitosterol (10.45%), alpha.-tocopherol-beta.-D-mannoside (7.50%) and 3-amino-4-piperonyl-5-pyrazolone (5.84%). The bioinformatics analysis of these phytochemicals showed a high potential to affect the levels of key proteins driving colon cancer progression, inhibiting the enzymes and proteins overexpressed in cancer. Conclusion The outcome of this study endorses the potential of phytochemicals of the petroleum ether fraction of ethanolic leaf extract of Annona reticulata for the development of a new chemotherapeutic agent in the treatment of colon cancer.
Collapse
Affiliation(s)
- Pooja Prakash Rao
- Department of Biochemistry, Kastubra Medical College Manipal, Maniapl Academy of Higher Education, Manipal, Karnataka, India
| | - Vijetha Shenoy Belle
- Department of Biochemistry, Kastubra Medical College Manipal, Maniapl Academy of Higher Education, Manipal, Karnataka, India
| | - Akshatha G Nayak
- Division of Biochemistry, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Export Promotions Industrial Park (EPIP), Industrial Area Hajipur, Vaishali, Bihar, India
| | - Vanishree Rao
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sri Pragnya Cheruku
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Krishnananda Prabhu
- Department of Biochemistry, Kastubra Medical College Manipal, Maniapl Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
18
|
Nejad FS, Alizade-Harakiyan M, Haghi M, Ebrahimi R, Zangeneh MM, Farajollahi A, Fathi R, Mohammadi R, Miandoab SS, Asl MH, Asgharian P, Divband B, Ahmadi A. Investigating the effectiveness of iron nanoparticles synthesized by green synthesis method in chemoradiotherapy of colon cancer. Heliyon 2024; 10:e28343. [PMID: 38560153 PMCID: PMC10981048 DOI: 10.1016/j.heliyon.2024.e28343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Current methods of colon cancer treatment, especially chemotherapy, require new treatment methods due to adverse side effects. One important area of interest in recent years is the use of nanoparticles as drug delivery vehicles since several studies have revealed that they can improve the target specificity of the treatment thus lowering the dosage of the drugs while preserving the effectiveness of the treatment thus reducing the side effects. The use of traditional medicine has also been a favorite topic of interest in recent years in medical research, especially cancer research. In this research work, the green synthesis of Fe nanoparticles was carried out using Mentha spicata extract and the synthesized nanoparticles were identified using FT-IR, XRD, FE-SEM and EDS techniques. Then the effect of Mentha spicata, Fe nanoparticles, and Mentha spicata -loaded Fe nanoparticles on LS174t colon cancer cells, and our result concluded that all three, especially Mentha spicata -loaded Fe nanoparticles, have great cytotoxic effects against LS174t cells, and exposure to radiotherapy just further intensified these results. The in vitro condition revealed alterations in the expression of pro-apoptotic BAX and anti-apoptotic Bcl2, suggesting a pro-apoptotic effect from all three components, particularly the Mentha spicata-loaded Fe nanoparticles. After further clinical trials, these nanoparticles can be used to treat colon cancer.
Collapse
Affiliation(s)
- Farshad Seyed Nejad
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Radiation Oncology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Alizade-Harakiyan
- Department of Radiation Oncology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Medical Physics Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Haghi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Rokhsareh Ebrahimi
- Medicinal Chemistry Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Mahdi Zangeneh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Alireza Farajollahi
- Department of Radiation Oncology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Medical Physics Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Medical Radiation Science Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roghayeh Fathi
- Polymer Research Laboratory, Department of Organic and Biochemistry, University of Tabriz, Tabriz, Iran
| | - Reza Mohammadi
- Polymer Research Laboratory, Department of Organic and Biochemistry, University of Tabriz, Tabriz, Iran
| | | | | | - Parina Asgharian
- Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Baharak Divband
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Amin Ahmadi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
19
|
Yu M, Fan Y, Zhao Y, Tang Y. MicroRNA-140-3p inhibits proliferation and promotes apoptosis in non-small cell lung cancer by targeting MDIG. ENVIRONMENTAL TOXICOLOGY 2024; 39:1521-1530. [PMID: 38009637 DOI: 10.1002/tox.24026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 08/29/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND MicroRNAs (miRNAs) are associated with cancer progression. MiR-140-3p is a tumor suppressor. Nevertheless, its function in non-small cell lung cancer (NSCLC) is unclear. METHODS MiR-140-3p expression in NSCLC clinical specimens was examined using the TCGA database and real-time PCR. NSCLC cell proliferation and apoptosis were investigated after the miRNA overexpression. Then, mineral dust-induced gene (MDIG) levels in NSCLC clinical specimens were monitored by real-time PCR and western blotting. Bioinformatics predicated the binding of miR-140-3p to MDIG, and their relationship was validated by luciferase reporter assay. The miR-140-3p/MDIG axis was further validated through rescue experiments. The involvement of STAT3 signaling in the actions of miR-140-3p/MDIG axis was investigated. RESULTS MiR-140-3p was decreased in NSCLC tissues and negatively correlated with MDIG expression. Additionally, it was also lower in high-grade specimens than in low-grade ones. MiR-140-3p restrained cell proliferation, facilitated apoptosis, and inhibited STAT3 signaling in NSCLC. Interestingly, MDIG was a target of this miRNA. Furthermore, MDIG upregulation abolished miR-140-3p's effect on cell proliferation, apoptosis, and STAT3 pathway in NSCLC cells. CONCLUSION MiR-140-3p restrained NSCLC development through the regulation of the STAT3 pathway by targeting MDIG. This axis may be a promising target for NSCLC treatment.
Collapse
Affiliation(s)
- Miaomiao Yu
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Yueren Fan
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Yihang Zhao
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Yu Tang
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, China
| |
Collapse
|
20
|
Cong S, Fu Y, Zhao X, Guo Q, Liang T, Wu D, Wang J, Zhang G. KIF26B and CREB3L1 Derived from Immunoscore Could Inhibit the Progression of Ovarian Cancer. J Immunol Res 2024; 2024:4817924. [PMID: 38380081 PMCID: PMC10878761 DOI: 10.1155/2024/4817924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/07/2024] [Accepted: 01/28/2024] [Indexed: 02/22/2024] Open
Abstract
Background Ovarian cancer (OV) is characteristic of high incidence rate and fatality rate in the malignant tumors of female reproductive system. Researches on pathogenesis and therapeutic targets for OV need to be continued. This study mainly analyzed the immune-related pathogenesis and discovered the key immunotherapy targets for OV. Methods WGCNA was used for excavating hub gene modules and hub genes related to the immunity of OV. Enrichment analysis was aimed to analyze the related pathways of hub gene modules. Biological experiments were used for exploring the effect of hub genes on SKOV3 cells. Results We identified two hub gene modules related to the immunoscore of OV and found that these genes in the modules were related to the extracellular matrix and viral infections. At the same time, we also discovered six hub genes related to the immunity of OV. Among them, KIF26B and CREB3L1 can affect the proliferation, migration, and invasion of SKOV3 cells by the Wnt/β-catenin pathway. Conclusions The local infection or inflammation of ovarian may affect the immunity of OV. KIF26B and CREB3L1 are expected to be potential targets for the immunotherapy of OV.
Collapse
Affiliation(s)
- Shanshan Cong
- Department of Gynecology, Affiliated Women's Hospital of Jiangnan University, Wuxi, China
| | - Yao Fu
- Department of Pharmacy, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Xibo Zhao
- Department of Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiuyan Guo
- Department of Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tian Liang
- Department of Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Di Wu
- Department of Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Wang
- Department of Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guangmei Zhang
- Department of Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
21
|
Li J, Liu F, Mo K, Ni H, Yin Y. Effects of weaning on intestinal longitudinal muscle-myenteric plexus function in piglets. SCIENCE CHINA. LIFE SCIENCES 2024; 67:379-390. [PMID: 37824029 DOI: 10.1007/s11427-022-2391-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/18/2023] [Indexed: 10/13/2023]
Abstract
Weaning piglets usually suffer from severe diarrhea (commonly known as postweaning diarrhea, PWD) along with intestinal motility disorder. Intestinal peristalsis is mainly regulated by the longitudinal muscle-myenteric plexus (LM-MP). To understand the relationship between intestinal LM-MP function and the development of PWD, we compared the intestinal electrical activity, and the transcriptional profile of the LM-MP between 21-day-old piglets (just weaned, n=7) and 24-day-old piglets (suffered the most severe weaning stress, n=7). The results showed that 24-day-old piglets exhibited different degrees of diarrhea. A significant increase in the slow-wave frequency in the ileum and colon was observed in 24-day-old piglets, while c-kit expression in the intestinal LM-MPs was significantly decreased, indicating that PWD caused by elevated slow-wave frequency may be associated with loss of c-kit. The real-time quantitative PCR (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA) showed that intestinal LM-MPs in 24-day-old piglets may undergo inflammation and oxidative stress. Significant increases in 8-hydroxy-2'-deoxyguanosine and decreases in thioredoxin suggest that weaning may lead to DNA damage in the LM-MP of 24-day-old piglets. In addition, activating transcription factor 3 was significantly upregulated, indicating nerve damage in the LM-MP of 24-day-old piglets. The transcriptomic results showed that most of the differentially expressed genes in the ileal LM-MP after weaning were downregulated and closely related to the cell cycle process. Subsequent RT-qPCR analysis showed that the relative expression of p21 was upregulated, while the expression of cyclin A2, cyclin B1, and proliferating cell nuclear antigen was downregulated in the ileal and colonic LM-MP of 24-day-old piglets, suggesting that weaning may inhibit cell proliferation and cause G1/S cell cycle arrest in ileal and colonic LM-MP. In conclusion, weaning may lead to cell cycle arrest by causing DNA damage in the LM-MP, impairing intestinal motility regulation, and ultimately leading to diarrhea in piglets.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Fenfen Liu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100008, China
| | - Kaibin Mo
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Hengjia Ni
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 100008, China.
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 100008, China.
| |
Collapse
|
22
|
Laiso RAN, Ferreira JC, Rici REG, Cabral LGDS, Maria DA. Cytotoxic Effect of Andrographis paniculata Associated with 2-Aminoethyl Dihydrogen Phosphate in Triple-Negative Breast Cells. Curr Issues Mol Biol 2024; 46:527-541. [PMID: 38248336 PMCID: PMC10814097 DOI: 10.3390/cimb46010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/23/2024] Open
Abstract
Cancer stands out as a major global public health concern and a significant impediment to increasing life expectancy worldwide. Natural bioactives derived from plants are renowned for their efficacy in treating various types of cancer. Andrographis paniculata (Burm.f.) is a well-known plant traditionally employed in diverse medical systems across the globe. The 2-AEH2P monophosphoester, a molecule intricately involved in phospholipid turnover, demonstrates antiproliferative effects across a broad spectrum of cancer types. This study aims to assess the antitumor, antiproliferative, and pharmacological effects of andrographolide at different concentrations, both individually and in conjunction with 2-aminoethyl dihydrogen phosphate. The cytotoxicity of the treatments was evaluated using the colorimetric MTT method, cell cycle phases, mitochondrial electrical potential, and markers expression via flow cytometry, while the pharmacological effects were assessed using SynergyFinder software 3.0. Treatments with A. paniculata, isolated at concentrations of 10%, 30%, and 50% of andrographolide, induced cell death in tumor cells, resulting in a reduction in mitochondrial electrical potential and alterations in cell cycle phases, particularly a decrease in the population of MDA MB-231 cells in the G0/G1 phase. The combination treatments exhibited significant cytotoxicity toward tumor cells, with minimal toxicity observed in normal fibroblast cells FN1. This led to a reduction in mitochondrial electrical potential and cell cycle arrest in the S phase for MDA MB-231 cells. Across all concentrations, the combined treatments demonstrated a synergistic pharmacological effect, underscoring the efficacy of the association. There was a change in the markers involved in cell death, such as p53, caspase 3, Bcl-2, and cytochrome c, suggesting the induction of regulated cell death. Markers associated with progression and proliferation, such as cyclin D1 and p21, corroborate the findings for cytotoxicity and cell cycle arrest.
Collapse
Affiliation(s)
| | - Julia Carolina Ferreira
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia, UNIMAR, Marilia 17525-902, Brazil; (J.C.F.); (R.E.G.R.)
| | - Rose Eli Grassi Rici
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia, UNIMAR, Marilia 17525-902, Brazil; (J.C.F.); (R.E.G.R.)
- Postgraduate Program in Domestic and Wild Animals, Faculty of Veterinary Medicine and Zootechnics, FMVZUSP, University of São Paulo, Sao Paulo 14049-900, Brazil
| | - Laertty Garcia de Sousa Cabral
- Laboratory of Development and Innovation, Butantan Institute, Sao Paulo 05585-000, Brazil;
- Faculty of Medicine, FMUSP, University of Sao Paulo, Sao Paulo 14049-900, Brazil
| | - Durvanei Augusto Maria
- Laboratory of Development and Innovation, Butantan Institute, Sao Paulo 05585-000, Brazil;
- Faculty of Medicine, FMUSP, University of Sao Paulo, Sao Paulo 14049-900, Brazil
| |
Collapse
|
23
|
Nxasana TH, Mangoato IM, Masiu PM, Mishra AP, Matsabisa MG. Investigating the combinatory effect of Sclerocarya birrea with doxorubicin against selected colorectal cancer cell lines. Drug Target Insights 2024; 18:94-104. [PMID: 39677854 PMCID: PMC11638844 DOI: 10.33393/dti.2024.3219] [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: 07/23/2024] [Accepted: 09/19/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction Colorectal cancer incidences continue to increase annually, worldwide. Herbal plants with antiproliferative properties received research interest as agents that can be adjuvant therapies with chemotherapy drugs to enhance their efficacy and reverse drug resistance. Methods Sclerocarya birrea ethanolic (SBE) and aqueous (SBW) extracts combined with doxorubicin (DOX) against drug-sensitive and drug-resistant colorectal cancer cells were investigated for their potential adjuvant and drug resistance reversal. The extracts were assessed for their potential anticancer activities on HCT15 and HT29 cell lines as well as their doxorubicin potentiating and drug resistance reversal effects respectively. The extracts were assessed for their cytotoxicity on normal 3T3-L1 fibroblast cells. Results Both SBE and SBW extracts exhibited no toxicity against normal 3T3 cells and showed low activity on the HT29 cell line. Contrarily, resistant HCT15 cells showed moderate to low activity with significantly higher inhibitory concentration (IC)50 values. The combination of SBE with DOX and SBW with DOX resulted in antagonistic interactions, causing an increase in IC50 values for HT29 and HCT15 cells. In contrast, the combination of DOX and verapamil (VER) produced an additive effect, with no change in their IC50 values. Conclusion Based on the findings from the combination treatment, the SBE and SBW extracts demonstrated higher efficacy and synergistic effects combined with DOX at IC75 compared to the combination of DOX and VER, suggesting their potential as anticancer agents. However, further research on both the SBE and SBW extracts' mechanisms of action and in vivo effects is warranted.
Collapse
Affiliation(s)
- Thembelihle H. Nxasana
- Department of Pharmacology, University of the Free State, Faculty of Health Sciences, School of Medicine, Bloemfontein - South Africa
| | - Innocensia M. Mangoato
- Department of Pharmacology, University of the Free State, Faculty of Health Sciences, School of Medicine, Bloemfontein - South Africa
| | - Patriciah M. Masiu
- Department of Pharmacology, University of the Free State, Faculty of Health Sciences, School of Medicine, Bloemfontein - South Africa
| | - Abhay P. Mishra
- Department of Pharmacology, University of the Free State, Faculty of Health Sciences, School of Medicine, Bloemfontein - South Africa
- Cosmetics and Natural Products Research Center (CosNat), Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok - Thailand
| | - Motlalepula G. Matsabisa
- Department of Pharmacology, University of the Free State, Faculty of Health Sciences, School of Medicine, Bloemfontein - South Africa
| |
Collapse
|
24
|
Tripathi PK, Jain CK. Medicinal Plant-rich Diet: A Potential Therapeutic Role in Colorectal Cancer. Cardiovasc Hematol Agents Med Chem 2024; 22:308-318. [PMID: 37724674 DOI: 10.2174/1871525722666230915103747] [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: 05/19/2023] [Revised: 07/22/2023] [Accepted: 08/18/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Colorectal cancer is estimated to become the leading cause of cancer death worldwide. Since most of the available therapies affect vital organs such as heart and liver, herbal remedies as a substitute therapy have been reported in several evidence-based studies. OBJECTIVE Medicinal plants exhibit a diverse range of bioactive elements known for their medicinal properties, such as anti-inflammatory, anticancer, antioxidant, and antimicrobial effects. Phytochemicals present in medicinal plants significantly trigger different signaling pathways, contributing to their therapeutic activities. This review covers a comprehensive summary of the therapeutic potential of an herbal diet in treating colorectal cancer and other ailments. Special attention will be given to exploring the interactions of medicinal plants with the microbiota and their associations with cancer pathways. CONCLUSION A medicinal plant rich in bioactive compounds is a therapeutic option for colorectal cancer and potent cardioprotective and hepatoprotective agents. These bioactive compounds have demonstrated the ability to impede the growth of cancerous cells and trigger apoptosis. Our findings suggest that pomegranate, garlic, soybean, olive, green tea, papaya, and grapes are potential medicinal plants for combating cancer and related side effects. Bioactive compounds can modulate the gut microbiota's metabolism, and short-chain fatty acid production shows cardioprotective effects and reduces the risk of colorectal cancer. Hence, it can be stated that the interaction between a medicinal plant-rich diet and the gut microbiota plays a crucial role in preventing colorectal cancer and cardiac arrest.
Collapse
Affiliation(s)
- Pankaj Kumar Tripathi
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, NOIDA, Uttar Pradesh, 201309, India
| | - Chakresh Kumar Jain
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, NOIDA, Uttar Pradesh, 201309, India
| |
Collapse
|
25
|
Deshmukh R, Prajapati M, Harwansh RK. Management of Colorectal Cancer Using Nanocarriers-based Drug Delivery for Herbal Bioactives: Current and Emerging Approaches. Curr Pharm Biotechnol 2024; 25:599-622. [PMID: 38807329 DOI: 10.2174/0113892010242028231002075512] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 05/30/2024]
Abstract
Colorectal cancer (CRC) is a complex and multifactorial disorder in middle-aged people. Several modern medicines are available for treating and preventing it. However, their therapeutic uses are limited due to drawbacks, such as gastric perforation, diarrhea, intestinal bleeding, abdominal cramps, hair loss, nausea, vomiting, weight loss, and adverse reactions. Hence, there is a continuous quest for safe and effective medicines to manage human health problems, like CRC. In this context, herbal medicines are considered an alternative disease control system. It has become popular in countries, like American, European, and Asian, due to its safety and effectiveness, which has been practiced for 1000 years. During the last few decades, herbal medicines have been widely explored through multidisciplinary fields for getting active compounds against human diseases. Several herbal bioactives, like curcumin, glycyrrhizin, paclitaxel, chlorogenic acid, gallic acid, catechin, berberine, ursolic acid, betulinic acid, chrysin, resveratrol, quercetin, etc., have been found to be effective against CRC. However, their pharmacological applications are limited due to low bioavailability and therapeutic efficacy apart from their several health benefits. An effective delivery system is required to increase their bioavailability and efficacy. Therefore, targeted novel drug delivery approaches are promising for improving these substances' solubility, bioavailability, and therapeutic effects. Novel carrier systems, such as liposomes, nanoparticles, micelles, microspheres, dendrimers, microbeads, and hydrogels, are promising for delivering poorly soluble drugs to the target site, i.e., the colon. Thus, the present review is focused on the pathophysiology, molecular pathways, and diagnostic and treatment approaches for CRC. Moreover, an emphasis has been laid especially on herbal bioactive-based novel delivery systems and their clinical updates.
Collapse
Affiliation(s)
- Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Mahendra Prajapati
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| |
Collapse
|
26
|
Bhardwaj K, Sharma A, Kumar R, Tyagi V, Kumar R. Improving Oral Bioavailability of Herbal Drugs: A Focused Review of Self-Emulsifying Drug Delivery System for Colon Cancer. Curr Drug Deliv 2024; 21:389-402. [PMID: 37151062 DOI: 10.2174/1567201820666230505113108] [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: 11/08/2022] [Revised: 03/19/2023] [Accepted: 03/24/2023] [Indexed: 05/09/2023]
Abstract
One of the most frequent malignancies in the world is colon cancer. Both men and women are affected in the same way. The colon, which makes up the last part of the digestive system and is where water and minerals from food waste are absorbed, is vulnerable to cancer. The most suitable technique of drug administration is oral administration. Aqueous solubility is low in more than 40% of novel chemical entities, resulting in poor oral drug administration. In the formulation of oral medications, low inconsistent bioavailability is a major challenge. Increasing medication bioavailability is one of the most difficult aspects of pharmacological development. Self-nano-emulsifying drug delivery systems (SNEDDS) have been a potential platform for biopharmaceutical classification system class II and IV drugs for oral delivery. Enhanced bioavailability and solubility, control of toxicity, pharmacological effects, improved stability, improved tissue macrophage dispersion, prolonged delivery, and resistance to physical and chemical degradation are just a few benefits of SNEDDS for herbal drugs. To increase activity and address problems associated with herbal drugs, nanosized modern drug delivery technologies are expected to have a promising future. Improved patient compliance, fewer problems with liquid SNEDDS filled in capsules, and enhanced stability SNEDDS are all benefits of converting liquid SNEDDS to solid oral dosage forms or solid SNEDDS. SNEDDS differs from previous solubility augmentation methods due to its biodegradable components, simplicity of large-scale production, and range of drug-targeting possibilities.
Collapse
Affiliation(s)
- Khushboo Bhardwaj
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Arun Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Rajan Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Varnit Tyagi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Rajesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| |
Collapse
|
27
|
Mohan S, Hakami MA, Dailah HG, Khalid A, Najmi A, Zoghebi K, Halawi MA, Alotaibi TM. From inflammation to metastasis: The central role of miR-155 in modulating NF-κB in cancer. Pathol Res Pract 2024; 253:154962. [PMID: 38006837 DOI: 10.1016/j.prp.2023.154962] [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: 10/24/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 11/27/2023]
Abstract
Cancer is a multifaceted, complex disease characterized by unchecked cell growth, genetic mutations, and dysregulated signalling pathways. These factors eventually cause evasion of apoptosis, sustained angiogenesis, tissue invasion, and metastasis, which makes it difficult for targeted therapeutic interventions to be effective. MicroRNAs (miRNAs) are essential gene expression regulators linked to several biological processes, including cancer and inflammation. The NF-κB signalling pathway, a critical regulator of inflammatory reactions and oncogenesis, has identified miR-155 as a significant participant in its modulation. An intricate network of transcription factors known as the NF-κB pathway regulates the expression of genes related to inflammation, cell survival, and immunological responses. The NF-κB pathway's dysregulation contributes to many cancer types' development, progression, and therapeutic resistance. In numerous cancer models, the well-studied miRNA miR-155 has been identified as a crucial regulator of NF-κB signalling. The p65 subunit and regulatory molecules like IκB are among the primary targets that miR-155 directly targets to alter NF-κB activity. The molecular processes by which miR-155 affects the NF-κB pathway are discussed in this paper. It also emphasizes the miR-155's direct and indirect interactions with important NF-κB cascade elements to control the expression of NF-κB subunits. We also investigate how miR-155 affects NF-κB downstream effectors in cancer, including inflammatory cytokines and anti-apoptotic proteins.
Collapse
Affiliation(s)
- Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al, Quwayiyah, Shaqra University, Riyadh, Saudi Arabia.
| | - Hamad Ghaleb Dailah
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan 45142, Saudi Arabia
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Khalid Zoghebi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Maryam A Halawi
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | | |
Collapse
|
28
|
El Khatabi K, Kumar S, El-Mernissi R, Singh AK, Ajana MA, Lakhlifi T, Bouachrine M. Novel Eubacterium rectale inhibitor from Coriandrum sativum L. for possible prevention of colorectal cancer: a computational approach. J Biomol Struct Dyn 2023; 41:8402-8416. [PMID: 36264092 DOI: 10.1080/07391102.2022.2134210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/03/2022] [Indexed: 10/24/2022]
Abstract
This research aims to screen out the effective bioactive compounds from Coriander (Coriandrum sativum L.), which may be novel potential inhibitors of Eubacterium rectale for the prevention of colorectal cancer (CRC). A series of 8 coriander-derived chemical compounds previously assessed for their anti-inflammatory, antioxidant, and antidiabetic activities were tested against Carbohydrate ABC transporter substrate-binding protein and compared to the standard inhibitor Acarbose, to support their use as novel Eubacterium rectale inhibitors. Herein, these derivatives were submitted to a thorough analysis of docking studies, in which detailed interactions of the selected phytocompounds with carbohydrate ABC transporter substrate-binding protein were revealed. Molecular docking analysis recommends Rutin, Gallocatechin, and Epigallocatechin as the most potential Eubacterium rectale inhibitors among the eight selected phytochemical compounds. Subsequently, the stability of the three selected phytochemical complexes was checked using molecular dynamics (MD) simulation at 100 ns and Molecular Mechanics combined with Poisson-Boltzmann Surface Area (MM-PBSA). The results show quite good stability for Rutin and Gallocatechin. In silico ADMET prediction was performed on the selected compounds, and the findings revealed a reasonably good ADMET profile for both Rutin and Gallocatechin. The current findings predict that Gallocatechin could be a better CRC preventive natural compound, and, further in vitro, in vivo and clinical studies may confirm its therapeutic potential.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Khalil El Khatabi
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University of Meknes, Meknes, Morocco
| | - Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, India
| | - Reda El-Mernissi
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University of Meknes, Meknes, Morocco
| | - Atul Kumar Singh
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, India
| | - Mohammed Aziz Ajana
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University of Meknes, Meknes, Morocco
| | - Tahar Lakhlifi
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University of Meknes, Meknes, Morocco
| | - Mohammed Bouachrine
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University of Meknes, Meknes, Morocco
- EST Khenifra, Sultan Moulay Sliman University, Beni-Mellal, Morocco
| |
Collapse
|
29
|
Liu B, Li Y, Xu Y, Xue W, Jin Z. Jian Yun Qing Hua Decoction inhibits malignant behaviors of gastric carcinoma cells via COL12A1 mediated ferroptosis signal pathway. Chin Med 2023; 18:118. [PMID: 37700383 PMCID: PMC10496189 DOI: 10.1186/s13020-023-00799-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/12/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Jian Yun Qing Hua Decoction (JYQHD), a traditional Chinese medicine decoction, which has been applied in the treatment of gastric cancer (GC). We attempt to confirm the anti-gastric cancer effect of JYQHD and explore the mechanism of JYQHD. METHODS Acute toxicity test was used to understand the toxicity of JYQHD. We studied the expression and prognostic outcome of COL12A1 within GC tissues through the network databases. Using several web-based databases, we analyzed the major components and targets of JYQHD, as well as known therapeutic targets in gastric cancer. The Venn diagram was utilized to obtain the overlapped genes. Lentiviral vector, shRNAs and plasmids, were used to transfect GC cells. Cell counting kit-8 (CCK8), sphere formation, malondialdehyde (MDA), glutathione (GSH), reactive oxygen species (ROS), Fe2+, transmission electron microscopy (TEM), quantitative Real-Time Polymerase Chain Reaction (qRT-PCR), Western-Blot (WB), and immunohistochemical (IHC) assays were employed to investigate the role and mechanism of COL12A1 and JYQHD in GC. RESULTS The results showed that JYQHD was non-toxic and safe. JYQHD inhibited growth and sphere formation ability through inducing the ferroptosis of GC cells, and suppressed the GC cells induced subcutaneous xenograft tumor growth. COL12A1 was highly expressed in gastric cancer tissues, indicating poor prognosis. COL12A1 specifically enhanced GC cell progression and stemness via suppressing ferroptosis. JYQHD down-regulated COL12A1 in order to suppress the stemness of GC cells via inducing ferroptosis. CONCLUSION COL12A1 inhibited ferroptosis and enhanced stemness in GC cells. JYQHD inhibited the development of GC cells by inhibiting cancer cell stemness via the ferroptosis pathway mediated by COL12A1.
Collapse
Affiliation(s)
- Baoxinzi Liu
- Department of Medical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yu Li
- Department of Medical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yuanyuan Xu
- Department of Medical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Weiwei Xue
- Department of Medical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Zhichao Jin
- Department of Medical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
30
|
Viana AR, Bottari NB, Oviedo VR, Santos D, Londero JEL, Schetinger MRC, Flores EMM, Pigatto A, Schuch AP, Krause A, Krause LMF. Phytochemical and biological characterization of aqueous extract of Vassobia breviflora on proliferation and viability of melanoma cells: involvement of purinergic pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2023; 86:632-652. [PMID: 37434435 DOI: 10.1080/15287394.2023.2233989] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Vassobia breviflora belongs to the Solanaceae family, possessing biological activity against tumor cells and is a promising alternative for therapy. The aim of this investigation was to determine the phytochemical properties V. breviflora using ESI-ToF-MS. The cytotoxic effects of this extract were examined in B16-F10 melanoma cells and the relationship if any to purinergic signaling was involved. The antioxidant activity of total phenols, (2,2-diphenyl-1-picrylhydrazyl (DPPH), 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid (ABTS) was analyzed, as well as production of reactive oxygen species (ROS) and nitric oxide (NO) was determined. Genotoxicity was assessed by DNA damage assay. Subsequently, the structural bioactive compounds were docked against purinoceptors P2X7 and P2Y1 receptors. The bioactive compounds found in V. breviflora were N-methyl-(2S,4 R)-trans-4-hydroxy-L-proline, calystegine B, 12-O-benzoyl- tenacigenin A and bungoside B. In vitro cytotoxicity was demonstrated at concentration ranges of 0.1-10 mg/ml, and plasmid DNA breaks only at the concentration of 10 mg/ml. V. breviflora extracts affected hydrolysis by ectoenzymes, such as ectonucleoside triphosphate diphosphohydrolase (E-NTPDase) and ectoadenosine deaminase (E-ADA) which control levels of degradation and formation of nucleosides and nucleotides. In the presence of substrates ATP, ADP, AMP and adenosine, the activities of E-NTPDase, 5´-NT or E-ADA were significantly modulated by V. breviflora. N-methyl-(2S,4 R)-trans-4-hydroxy-L-proline presented higher binding affinity (according to receptor-ligand complex estimated binding affinity as evidenced by ∆G values) to bind to both P2X7 and P2Y1purinergic receptors.Our results suggest a putative interaction of V. breviflora bioactive compounds with growth inhibitory potential in B16-F10 melanoma and suggest that may be considered as promising compounds in melanoma and cancer treatment.
Collapse
Affiliation(s)
- Altevir Rossato Viana
- Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Brazil
| | | | | | - Daniel Santos
- Chemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | | | | | | | - Aline Pigatto
- Postgraduate Program in Teaching Science and Mathematics, Franciscan University, Santa Maria, Brazil
| | - André Passaglia Schuch
- Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Alexandre Krause
- Veterinary Medicine, Federal University of Santa Maria, Santa Maria, Brazil
| | | |
Collapse
|
31
|
Guo S, Chen M, Li S, Geng Z, Jin Y, Liu D. Natural Products Treat Colorectal Cancer by Regulating miRNA. Pharmaceuticals (Basel) 2023; 16:1122. [PMID: 37631037 PMCID: PMC10459054 DOI: 10.3390/ph16081122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Diseases are evolving as living standards continue to improve. Cancer is the main cause of death and a major public health problem that seriously threatens human life. Colorectal cancer is one of the top ten most common malignant tumors in China, ranking second after gastric cancer among gastrointestinal malignant tumors, and its incidence rate is increasing dramatically each year due to changes in the dietary habits and lifestyle of the world's population. Although conventional therapies, such as surgery, chemotherapy, and radiotherapy, have profoundly impacted the treatment of colorectal cancer (CRC), drug resistance and toxicity remain substantial challenges. Natural products, such as dietary therapeutic agents, are considered the safest alternative for treating CRC. In addition, there is substantial evidence that natural products can induce apoptosis, inhibit cell cycle arrest, and reduce the invasion and migration of colon cancer cells by targeting and regulating the expression and function of miRNAs. Here, we summarize the recent research findings on the miRNA-regulation-based antitumor mechanisms of various active ingredients in natural products, highlighting how natural products target miRNA regulation in colon cancer prevention and treatment. The application of natural drug delivery systems and predictive disease biomarkers in cancer prevention and treatment is also discussed. Such approaches will contribute to the discovery of new regulatory mechanisms associated with disease pathways and provide a new theoretical basis for developing novel colon cancer drugs and compounds and identifying new therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.G.); (M.C.); (S.L.); (Z.G.)
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.G.); (M.C.); (S.L.); (Z.G.)
| |
Collapse
|
32
|
Huang S, Nan Y, Chen G, Ning N, Du Y, Lu D, Yang Y, Meng F, Yuan L. The Role and Mechanism of Perilla frutescens in Cancer Treatment. Molecules 2023; 28:5883. [PMID: 37570851 PMCID: PMC10421205 DOI: 10.3390/molecules28155883] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Perilla frutescens is an annual herb of the Labiatae family and is widely grown in several countries in Asia. Perilla frutescens is a plant that is used medicinally in its entirety, as seen in its subdivision into perilla seeds, perilla stalks, and perilla leaves, which vary more markedly in their chemical composition. Several studies have shown that Perilla frutescens has a variety of pharmacological effects, including anti-inflammatory, antibacterial, detoxifying, antioxidant, and hepatoprotective. In the absence of a review of Perilla frutescens for the treatment of cancer. This review provides an overview of the chemical composition and molecular mechanisms of Perilla frutescens for cancer treatment. It was found that the main active components of Perilla frutescens producing cancer therapeutic effects were perilla aldehyde (PAH), rosmarinic acid (Ros A), lignan, and isoestrogen (IK). In addition to these, extracts of the leaves and fruits of Perilla frutescens are also included. Among these, perilla seed oil (PSO) has a preventive effect against colorectal cancer due to the presence of omega-3 polyunsaturated fatty acids. This review also provides new ideas and thoughts for scientific innovation and clinical applications related to Perilla frutescens.
Collapse
Affiliation(s)
- Shicong Huang
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; (S.H.); (Y.N.); (G.C.); (N.N.); (Y.D.)
| | - Yi Nan
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; (S.H.); (Y.N.); (G.C.); (N.N.); (Y.D.)
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Guoqing Chen
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; (S.H.); (Y.N.); (G.C.); (N.N.); (Y.D.)
| | - Na Ning
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; (S.H.); (Y.N.); (G.C.); (N.N.); (Y.D.)
| | - Yuhua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; (S.H.); (Y.N.); (G.C.); (N.N.); (Y.D.)
| | - Doudou Lu
- Clinical Medical School, Ningxia Medical University, Yinchuan 750004, China;
| | - Yating Yang
- Institute of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004, China; (Y.Y.); (F.M.)
| | - Fandi Meng
- Institute of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004, China; (Y.Y.); (F.M.)
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; (S.H.); (Y.N.); (G.C.); (N.N.); (Y.D.)
| |
Collapse
|
33
|
Shahabi Nejad F, Karami H, Darvish M. Triggering of Endoplasmic Reticulum Stress by Tannic Acid Inhibits the Proliferation and Migration of Colorectal Cancer Cells. Asian Pac J Cancer Prev 2023; 24:2705-2711. [PMID: 37642057 PMCID: PMC10685243 DOI: 10.31557/apjcp.2023.24.8.2705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 08/07/2023] [Indexed: 08/31/2023] Open
Abstract
INTRODUCTION Due to the pivotal role of endoplasmic reticulum (ER) stress in cancers, interfering with its function can cause the accumulation of unfolded proteins, which ultimately leads to the activation of the unfolded protein response (UPR) signaling pathway and apoptosis. Therefore, the use of plant compounds such as tannic acid with UPR-inducing properties can be proposed as a possible treatment method for cancer. In this study, we investigated the effect of tannic acid on cell migration, colony formation, growth, and UPR-induced apoptosis in the SW48 colorectal cancer cell line. METHODS The MTT assay was performed to investigate the cytotoxic effect of tannic acid. We performed the qPCR method to elucidate the effect of tannic acid on the expression of Bim, MMP-9, Bcl-xL, cyclin D1, CHOP, and ATF4 genes. We also used the colony formation and migration experiments to investigate the effect of this compound on the colony formation and migration ability of tumor cells. Finally, we used Hoechst staining to measure cell apoptosis. RESULTS Tannic acid inhibited the cell survival, clonogenic, and migration of colon cancer cells. This compound increased the expression of ER stress-mediated UPR genes, ATF4 and CHOP. Moreover; tannic acid increased the expression of pro-apoptotic proteins like Bim, while at the same time causing a sharp decline in the expression of anti-apoptotic protein Bcl-xL. A decline in MMP-9 expression confirmed the anti-metastatic role of this compound. CONCLUSION Taken together, tannic acid can induce apoptosis via ER stress-mediated UPR pathway, and has a suppressive effect on cell viability, growth, migration, colony formation, and metastasis, suggesting it may be a potential drug in colorectal cancer treatment.
Collapse
Affiliation(s)
- Fatemeh Shahabi Nejad
- Department of Molecular Medicine and Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran.
| | - Hadi Karami
- Department of Molecular Medicine and Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran.
- Traditional and Complementary Medicine Research Center, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran.
| | - Maryam Darvish
- Department of Molecular Medicine and Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran.
| |
Collapse
|
34
|
Li W, Xu X. Advances in mitophagy and mitochondrial apoptosis pathway-related drugs in glioblastoma treatment. Front Pharmacol 2023; 14:1211719. [PMID: 37456742 PMCID: PMC10347406 DOI: 10.3389/fphar.2023.1211719] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant tumor of the central nervous system (CNS). It is a leading cause of death among patients with intracranial malignant tumors. GBM exhibits intra- and inter-tumor heterogeneity, leading to drug resistance and eventual tumor recurrence. Conventional treatments for GBM include maximum surgical resection of glioma tissue, temozolomide administration, and radiotherapy, but these methods do not effectively halt cancer progression. Therefore, development of novel methods for the treatment of GBM and identification of new therapeutic targets are urgently required. In recent years, studies have shown that drugs related to mitophagy and mitochondrial apoptosis pathways can promote the death of glioblastoma cells by inducing mitochondrial damage, impairing adenosine triphosphate (ATP) synthesis, and depleting large amounts of ATP. Some studies have also shown that modern nano-drug delivery technology targeting mitochondria can achieve better drug release and deeper tissue penetration, suggesting that mitochondria could be a new target for intervention and therapy. The combination of drugs targeting mitochondrial apoptosis and autophagy pathways with nanotechnology is a promising novel approach for treating GBM.This article reviews the current status of drug therapy for GBM, drugs targeting mitophagy and mitochondrial apoptosis pathways, the potential of mitochondria as a new target for GBM treatment, the latest developments pertaining to GBM treatment, and promising directions for future research.
Collapse
|
35
|
Herrera-Calderon O, Herrera-Ramírez A, Cardona-G W, Melgar-Merino EJ, Chávez H, Pari-Olarte JB, Loyola-Gonzales E, Kong-Chirinos JF, Almeida-Galindo JS, Peña-Rojas G, Andía-Ayme V. Dodonaea viscosa Jacq. induces cytotoxicity, antiproliferative activity, and cell death in colorectal cancer cells via regulation of caspase 3 and p53. Front Pharmacol 2023; 14:1197569. [PMID: 37426815 PMCID: PMC10326442 DOI: 10.3389/fphar.2023.1197569] [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: 03/31/2023] [Accepted: 06/13/2023] [Indexed: 07/11/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer diagnosed worldwide and is the second leading cause of cancer-related death due to an insufficiency prognosis and is generally diagnosed in the last step of development. The Peruvian flora has a wide variety of medicinal plants with therapeutic potential in several diseases. Dodonaea viscosa Jacq. is a plant used to treat inflammatory process as well as gastrointestinal diseases. The aim of this study was to examine the cytotoxic, antiproliferative, and cell death-inducing effects of D. viscosa on colorectal cancer cells (SW480 and SW620). The hydroethanolic extract was obtained by maceration at 70% ethanol, the phytochemical constituents were identified by LC-ESI-MS. D. viscosa revealed 57 compounds some of them are: isorhamnetin, kaempferol, quercetin, methyl dodovisate B, hardwickiic acid, viscosol, and dodonic acid. Regarding the antitumoral activity, D. viscosa induced cytotoxic and antiproliferative activity in both SW480 and SW620 cancer cells, accompanied with, important changes in mitochondrial membrane potential, formation of the Sub G0/G1 population and increasing levels of apoptotic biomarkers (caspase 3 and the tumor suppressor protein p53) in the metastatic derivative cell line (SW620), suggesting an intrinsic apoptotic process after the treatment with the hydroethanolic extract of D. viscosa.
Collapse
Affiliation(s)
- Oscar Herrera-Calderon
- Department of Pharmacology, Bromatology, and Toxicology, Faculty of Pharmacy and Biochemistry, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Angie Herrera-Ramírez
- Chemistry of Colombian Plants Group, Institute of Chemistry, Faculty of Exact and Natural Sciences, University of Antioquia (UdeA), Medellín, Colombia
| | - Wilson Cardona-G
- Chemistry of Colombian Plants Group, Institute of Chemistry, Faculty of Exact and Natural Sciences, University of Antioquia (UdeA), Medellín, Colombia
| | - Elizabeth Julia Melgar-Merino
- Department of Chemistry Sciences, Faculty of Pharmacy and Biochemistry, Universidad Nacional San Luis Gonzaga, Ica, Peru
| | - Haydee Chávez
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biochemistry, Universidad Nacional San Luis Gonzaga, Ica, Peru
| | - Josefa Bertha Pari-Olarte
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biochemistry, Universidad Nacional San Luis Gonzaga, Ica, Peru
| | - Eddie Loyola-Gonzales
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Biochemistry, Universidad Nacional San Luis Gonzaga, Ica, Peru
| | - José Francisco Kong-Chirinos
- Department of Surgical Clinical Sciences, Faculty of Human Medicine, Universidad Nacional San Luis Gonzaga, Ica, Peru
| | | | - Gilmar Peña-Rojas
- Laboratory of Cellular and Molecular Biology, Biological Sciences Faculty, Universidad Nacional de San Cristóbal de Huamanga, Ayacucho, Peru
| | - Vidalina Andía-Ayme
- Food Microbiology Laboratory, Biological Sciences Faculty, Universidad Nacional de San Cristóbal de Huamanga, Ayacucho, Peru
| |
Collapse
|
36
|
Liu T, Zhuang Z, Wang D. Paeoniflorin mitigates high glucose-induced lifespan reduction by inhibiting insulin signaling in Caenorhabditis elegans. Front Pharmacol 2023; 14:1202379. [PMID: 37405055 PMCID: PMC10315627 DOI: 10.3389/fphar.2023.1202379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/12/2023] [Indexed: 07/06/2023] Open
Abstract
In organisms, high glucose can cause several aspects of toxicity, including the lifespan reduction. Paeoniflorin is the major component of Paeoniaceae plants. Nevertheless, the possible effect of paeoniflorin to suppress high glucose toxicity in reducing lifespan and underlying mechanism are largely unclear. Thus, in this study, we examined the possible effect of paeoniflorin in suppressing high glucose (50 mM)-induced lifespan reduction and the underlying mechanism in Caenorhabditis elegans. Administration with 16-64 mg/L paeoniflorin could prolong the lifespan in glucose treated nematodes. Accompanied with this beneficial effect, in glucose treated nematodes, expressions of daf-2 encoding insulin receptor and its downstream kinase genes (age-1, akt-1, and akt-2) were decreased and expression of daf-16 encoding FOXO transcriptional factor was increased by 16-64 mg/L paeoniflorin administration. Meanwhile, the effect of paeoniflorin in extending lifespan in glucose treated nematodes was enhanced by RNAi of daf-2, age-1, akt-1, and akt-2 and inhibited by RNAi of daf-16. In glucose treated nematodes followed by paeoniflorin administration, the increased lifespan caused by daf-2 RNAi could be suppressed by RNAi of daf-16, suggesting that DAF-2 acted upstream of DAF-16 to regulate pharmacological effect of paeoniflorin. Moreover, in glucose treated nematodes followed by paeoniflorin administration, expression of sod-3 encoding mitochondrial Mn-SOD was inhibited by daf-16 RNAi, and the effect of paeoniflorin in extending lifespan in glucose treated nematodes could be suppressed by sod-3 RNAi. Molecular docking analysis indicated the binding potential of paeoniflorin with DAF-2, AGE-1, AKT-1, and AKT-2. Therefore, our results demonstrated the beneficial effect of paeoniflorin administration in inhibiting glucose-induced lifespan reduction by suppressing signaling cascade of DAF-2-AGE-1-AKT-1/2-DAF-16-SOD-3 in insulin signaling pathway.
Collapse
Affiliation(s)
- Tianwen Liu
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, China
- Medical School, Southeast University, Nanjing, China
| | - Ziheng Zhuang
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, China
| | - Dayong Wang
- Medical School, Southeast University, Nanjing, China
| |
Collapse
|
37
|
Chen F, Cui JJ, Jiang DC, Wang HZ, Zhuang W, Feng YN, Lin XL, Xi SY. Antitumor mechanism of kangliu pill on gliomas in mice through PI3K-Akt signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116252. [PMID: 36775078 DOI: 10.1016/j.jep.2023.116252] [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: 12/12/2022] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gliomas are common malignant intracranial tumors that have worse prognosis and pose a serious threat to human health. The Kangliu pill (KLP) is an innovative herbal compound from Xuanwu Hospital of Capital Medical University that has been clinically used for the treatment of gliomas for more than 40 years, and is one of the few drugs for primary treatment of this disorder. But the fundamental molecular mechanisms and pathways of KLP are not clear. AIM OF THE STUDY To investigate the therapeutic mechanism of KLP in the treatment of gliomas. MATERIALS AND METHODS An in situ xenograft model of red fluorescent protein-labeled human glioma cell line (U87-RFP) in BALB/c-nu mouse was established, and the therapeutic effect of KLP on gliomas was assessed by tumor weights and fluorescence areas. A quantitative proteomics approach using tandem mass tags combined with liquid chromatography-tandem mass spectrometry was performed to explore differentially expressed proteins (DEPs) in glioma tissues, and bioinformatics analyses including Gene Ontology analysis, pathway analysis, and network analysis were performed to analyze the proteins involved in the network therapeutic mechanisms responsible for key metabolic pathways. Cytological experiments corroborated the above analysis results. RESULTS Network pharmacology approach screened 246 bioactive compounds contained in KLP, targeting 724 proteins and 173 potential targets of KLP for glioma treatment. The important targets obtained after visualizing the PPI network were AKT1, INS, GAPDH, SRC, TP53, etc. The KEGG enrichment results showed that 9 proteins were related to cancer, including Pathways in cancer, PI3K/AKT signaling pathway, etc. KLP had antitumor activity in gliomas, which reduced tumor weights and fluorescence areas. A number of DEPs possibly associated with gliomas were identified through quantitative proteomic techniques. Among these DEPs, 50 (25 upregulated and 25 downregulated) were identified that might be associated with KLP action. Bioinformatics showed that these 50 DEPs were mainly focused on focal adhesion, extracellular matrix (ECM)-receptor interactions, and the PI3K-Akt signaling pathway. Cytological experiments revealed that KLP significantly inhibited the proliferation and promoted apoptosis of U87-MG human glioma cells, and its mechanism was through the inhibition of PI3K/AKT signaling pathway. CONCLUSION Therapeutic effect of KLP was regulation of multiple pathways in the treatment of gliomas. In specific, it interacts through the PI3K-Akt signaling pathway. This work may contribute proteomic insights for further research on the medical treatment of glioma using KLP.
Collapse
Affiliation(s)
- Fei Chen
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Gerontic Disease Clinical Research Center, Beijing, 100053, China.
| | - Jing-Jing Cui
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Gerontic Disease Clinical Research Center, Beijing, 100053, China.
| | - De-Chun Jiang
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| | - Hai-Zheng Wang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Gerontic Disease Clinical Research Center, Beijing, 100053, China.
| | - Wei Zhuang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Gerontic Disease Clinical Research Center, Beijing, 100053, China.
| | - Ying-Nan Feng
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Gerontic Disease Clinical Research Center, Beijing, 100053, China.
| | - Xiao-Lan Lin
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Gerontic Disease Clinical Research Center, Beijing, 100053, China.
| | - Sheng-Yan Xi
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
| |
Collapse
|
38
|
Subarmaniam T, Mahmad Rusli RN, Perumal KV, Yong YK, Hadizah S, Othman F, Salem K, Shafie NH, Hasham R, Yin KB, Abdul Kadir KK, Bahari H, Zakaria ZA. The Potential Chemopreventive Effect of Andrographis paniculata on 1,2-Dimethylhydrazine and High-Fat-Diet-Induced Colorectal Cancer in Sprague Dawley Rats. Int J Mol Sci 2023; 24:ijms24065224. [PMID: 36982300 PMCID: PMC10049149 DOI: 10.3390/ijms24065224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 03/11/2023] Open
Abstract
Colorectal cancer (CRC) is responsible for a notable rise in the overall mortality rate. Obesity is found to be one of the main factors behind CRC development. Andrographis paniculata is a herbaceous plant famous for its medicinal properties, particularly in Southeast Asia for its anti-cancer properties. This study examines the chemopreventive impact of A. paniculata ethanolic extract (APEE) against a high-fat diet and 1,2-dimethylhydrazine-induced colon cancer in Sprague Dawley rats. Sprague Dawley rats were administered 1,2-dimethylhydrazine (40 mg/kg, i.p. once a week for 10 weeks) and a high-fat diet (HFD) for 20 weeks to induce colorectal cancer. APEE was administered at 125 mg/kg, 250 mg/kg, and 500 mg/kg for 20 weeks. At the end of the experiment, blood serum and organs were collected. DMH/HFD-induced rats had abnormal crypts and more aberrant crypt foci (ACF). APEE at a dose of 500 mg/kg improved the dysplastic state of the colon tissue and caused a 32% reduction in the total ACF. HFD increased adipocyte cell size, while 500 mg/kg APEE reduced it. HFD and DMH/HFD rats had elevated serum insulin and leptin levels. Moreover, UHPLC-QTOF-MS analysis revealed that APEE was rich in anti-cancer phytochemicals. This finding suggests that APEE has anti-cancer potential against HFD/DMH-induced CRC and anti-adipogenic and anti-obesity properties.
Collapse
Affiliation(s)
- Tharani Subarmaniam
- Borneo Research on Algesia, Inflammation and Neurodegeneration (BRAIN) Group, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Jalan UMS, Kota Kinabalu 88400, Sabah, Malaysia (Z.A.Z.)
- Department of Human Anatomy, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | | | - Kokila Vani Perumal
- Department of Human Anatomy, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Yoke Keong Yong
- Department of Human Anatomy, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Siti Hadizah
- Department of Human Anatomy, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Fezah Othman
- Department Biomedical Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Khaled Salem
- Department Biomedical Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Nurul Husna Shafie
- Department of Nutrition, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Laboratory of UPM-MAKNA Cancer Research, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Rosnani Hasham
- Department of Bioprocess and Polymer Engineering, School of Chemical and Energy Engineering, Faculty of Engineering, Universiti Teknologi Malaysia, Johor Bahru 81310, Johor, Malaysia
| | - Khoo Boon Yin
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Penang, Malaysia
| | - Khairul Kamilah Abdul Kadir
- Department of Innovation and Commercialization, Forest Research Institution Malaysia, Kepong 52109, Selangor, Malaysia
| | - Hasnah Bahari
- Department of Human Anatomy, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Correspondence: (H.B.); (Z.A.Z.)
| | - Zainul Amiruddin Zakaria
- Borneo Research on Algesia, Inflammation and Neurodegeneration (BRAIN) Group, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Jalan UMS, Kota Kinabalu 88400, Sabah, Malaysia (Z.A.Z.)
- Correspondence: (H.B.); (Z.A.Z.)
| |
Collapse
|
39
|
Laurindo LF, Direito R, Bueno Otoboni AMM, Goulart RA, Quesada K, Barbalho SM. Grape Processing Waste: Effects on Inflammatory Bowel Disease and Colorectal Cancer. FOOD REVIEWS INTERNATIONAL 2023. [DOI: 10.1080/87559129.2023.2168281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology/School of Medicine, UNIMAR, Marília, São Paulo, Brazil
| | - Rosa Direito
- Department of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | | | - Ricardo Alvares Goulart
- Postgraduate Program (Structural and Functional Interactions in Rehabilitation), UNIMAR, Marília, São Paulo, Brazil
| | - Karina Quesada
- Department of Biochemistry and Pharmacology/School of Medicine, UNIMAR, Marília, São Paulo, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology/School of Medicine, UNIMAR, Marília, São Paulo, Brazil
- Department of Biochemistry and Nutrition, FATEC, Avenida Castro Alves, São Paulo, Brazil
- Postgraduate Program (Structural and Functional Interactions in Rehabilitation), UNIMAR, Marília, São Paulo, Brazil
| |
Collapse
|
40
|
De S, Paul S, Manna A, Majumder C, Pal K, Casarcia N, Mondal A, Banerjee S, Nelson VK, Ghosh S, Hazra J, Bhattacharjee A, Mandal SC, Pal M, Bishayee A. Phenolic Phytochemicals for Prevention and Treatment of Colorectal Cancer: A Critical Evaluation of In Vivo Studies. Cancers (Basel) 2023; 15:993. [PMID: 36765950 PMCID: PMC9913554 DOI: 10.3390/cancers15030993] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer (CRC) is the third most diagnosed and second leading cause of cancer-related death worldwide. Limitations with existing treatment regimens have demanded the search for better treatment options. Different phytochemicals with promising anti-CRC activities have been reported, with the molecular mechanism of actions still emerging. This review aims to summarize recent progress on the study of natural phenolic compounds in ameliorating CRC using in vivo models. This review followed the guidelines of the Preferred Reporting Items for Systematic Reporting and Meta-Analysis. Information on the relevant topic was gathered by searching the PubMed, Scopus, ScienceDirect, and Web of Science databases using keywords, such as "colorectal cancer" AND "phenolic compounds", "colorectal cancer" AND "polyphenol", "colorectal cancer" AND "phenolic acids", "colorectal cancer" AND "flavonoids", "colorectal cancer" AND "stilbene", and "colorectal cancer" AND "lignan" from the reputed peer-reviewed journals published over the last 20 years. Publications that incorporated in vivo experimental designs and produced statistically significant results were considered for this review. Many of these polyphenols demonstrate anti-CRC activities by inhibiting key cellular factors. This inhibition has been demonstrated by antiapoptotic effects, antiproliferative effects, or by upregulating factors responsible for cell cycle arrest or cell death in various in vivo CRC models. Numerous studies from independent laboratories have highlighted different plant phenolic compounds for their anti-CRC activities. While promising anti-CRC activity in many of these agents has created interest in this area, in-depth mechanistic and well-designed clinical studies are needed to support the therapeutic use of these compounds for the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Samhita De
- Division of Molecular Medicine, Bose Institute, Kolkata 700 054, India
| | - Sourav Paul
- Department of Biotechnology, National Institute of Technology, Durgapur 713 209, India
| | - Anirban Manna
- Division of Molecular Medicine, Bose Institute, Kolkata 700 054, India
| | | | - Koustav Pal
- Jawaharlal Institute Post Graduate Medical Education and Research, Puducherry 605 006, India
| | - Nicolette Casarcia
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Arijit Mondal
- Department of Pharmaceutical Chemistry, M.R. College of Pharmaceutical Sciences and Research, Balisha 743 234, India
| | - Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol 713 301, India
| | - Vinod Kumar Nelson
- Department of Pharmacology, Raghavendra Institute of Pharmaceutical Education and Research, Anantapur 515 721, India
| | - Suvranil Ghosh
- Division of Molecular Medicine, Bose Institute, Kolkata 700 054, India
| | - Joyita Hazra
- Department of Biotechnology, Indian Institute of Technology, Chennai 600 036, India
| | - Ashish Bhattacharjee
- Department of Biotechnology, National Institute of Technology, Durgapur 713 209, India
| | | | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, Kolkata 700 054, India
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| |
Collapse
|
41
|
Sadat SN, Bagheri KP, Maghsoudi H, Shahbazzadeh D. Oxineur, a novel peptide from Caspian cobra Naja naja oxiana against HT-29 colon cancer. Biochim Biophys Acta Gen Subj 2023; 1867:130285. [PMID: 36462597 DOI: 10.1016/j.bbagen.2022.130285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 10/09/2022] [Accepted: 11/20/2022] [Indexed: 12/02/2022]
Abstract
Colon cancer ranks fourth in mortality. This cancer is still an important clinical challenge worldwide due to its high prevalence and poor prognosis. Proteomic studies revealed that snake venom is a diverse and variable mixture of enzymatic and non-enzymatic proteins and peptides. Despite the toxic effects of these molecules, several proteins and peptides have been isolated that have practical applications and appear to induce apoptosis and prevent cell metastasis. In this study, we worked on cytotoxic effects and anticancer activity of Naja naja oxiana (Iranian Caspian cobra) snake venom components on HT-29 cell line colon cancer. Separated Fraction-5 by FPLC indicated the high cytotoxicity on HT-29 cell line colon cancer by MTT test. Further isolation of F5 by HPLC showed that the purified peak 2, nominated as Oxineur that contains a cytotoxic effect on HT-29 cells and reduces cell viability at 8 μg/ml to 4% in 24 h. Oxineur has the least cytotoxic effect on HEK-293 normal cells. Further studies on Oxineur peptide confirmed the apoptotic effects with high expression of CASP9 gene and DNA fragmentation in cancerous cells. The partial sequence of Oxineur revealed 71% homology with the neurotoxin II from Naja naja oxiana. Since our target molecule is a peptide in the molecular weight range of 7 kDa, it has potentially a therapeutic value.
Collapse
Affiliation(s)
- Seyedeh Narjes Sadat
- Biotechnology Research Center, Medical Biotechnology Department, Venom and Biotherapeutics Molecules Lab., Pasteur Institute of Iran, Tehran, Iran
| | - Kamran Pooshang Bagheri
- Biotechnology Research Center, Medical Biotechnology Department, Venom and Biotherapeutics Molecules Lab., Pasteur Institute of Iran, Tehran, Iran
| | - Hosein Maghsoudi
- Department of Biology, Faculty of Basic Sciences, Payame Noor University, PNU Rey, Tehran, Iran
| | - Delavar Shahbazzadeh
- Biotechnology Research Center, Medical Biotechnology Department, Venom and Biotherapeutics Molecules Lab., Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
42
|
Wang J, Huang P, Pan X, Xia C, Zhang H, Zhao H, Yuan Z, Liu J, Meng C, Liu F. Resveratrol reverses TGF-β1-mediated invasion and metastasis of breast cancer cells via the SIRT3/AMPK/autophagy signal axis. Phytother Res 2023; 37:211-230. [PMID: 36086852 DOI: 10.1002/ptr.7608] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 01/19/2023]
Abstract
Resveratrol (Resv) has antitumorigenic and antimetastatic activities; however, the molecular mechanisms underlying the inhibitory effects of Resv on the invasion and metastasis of breast cancer cells are still a subject of debate. In our study, we demonstrated that Resv inhibited tumor cell proliferation and tumor growth. It also suppressed invasion and pulmonary metastasis of breast cancer by reversing the transforming growth factor beta 1 (TGF-β1)-mediated EMT process. Meanwhile, the anticarcinogenic effects of Resv were abolished by the autophagy blocker 3-methyladenine (3-MA) or Beclin 1 small interfering RNA. Moreover, Resv upregulated autophagy-related genes and protein levels and induced the formation of autophagosomes in 4T1 breast cancer cells and xenograft mice, suggesting that autophagy was involved in the anticarcinogenic activities of Resv in both models. In addition, Resv-induced autophagy by increasing the expression of SIRT3 and phosphorylated AMPK. SIRT3 knockdown reduced AMPK phosphorylation and autophagy-related proteins levels, and suppressed the anticancer effects of Resv, demonstrating that the inhibitory effects of Resv on tumor progression were mediated via the SIRT3/AMPK/autophagy pathway. Taken together, our study provided novel insight into the anticancer effects of Resv and revealed that targeting the SIRT3/AMPK/autophagy pathway can serve as a new therapeutic target against breast cancer.
Collapse
Affiliation(s)
- Jia Wang
- Clinical Pharmacology Institute, Nanchang University, Nanchang, People's Republic of China
| | - Ping Huang
- Clinical Pharmacology Institute, Nanchang University, Nanchang, People's Republic of China
| | - Xiafang Pan
- Clinical Pharmacology Institute, Nanchang University, Nanchang, People's Republic of China
| | - Chunhua Xia
- Clinical Pharmacology Institute, Nanchang University, Nanchang, People's Republic of China
| | - Hong Zhang
- Clinical Pharmacology Institute, Nanchang University, Nanchang, People's Republic of China
| | - Han Zhao
- Clinical Pharmacology Institute, Nanchang University, Nanchang, People's Republic of China
| | - Zhao Yuan
- Clinical Trial Research Center, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Jianming Liu
- Clinical Pharmacology Institute, Nanchang University, Nanchang, People's Republic of China
| | - Chao Meng
- Clinical Pharmacology Institute, Nanchang University, Nanchang, People's Republic of China
| | - Fanglan Liu
- Clinical Pharmacology Institute, Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
43
|
Youn BY, Kim JH, Jo YK, Yoon S, Im JY, Kim HJ, Lee JD, Ko SG. Current Characteristics of Herbal Medicine Interventions for Cancer on Clinical Databases: A Cross-Sectional Study. Integr Cancer Ther 2023; 22:15347354231218255. [PMID: 38099482 PMCID: PMC10725141 DOI: 10.1177/15347354231218255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 09/09/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The utilization of herbal medicine has been noteworthy for treating cancer; however, there is not enough information regarding the characteristics of clinical trials of herbal medicine interventions. This study aimed to evaluate the characteristic of registered trials using herbal medicine interventions for cancer. METHODS A cross-sectional study was performed via the website ClinicalTrials.gov, ISRCTN registry, Chinese clinical trial registry, and international clinical trials registry platform to gather associated registered clinical trials using an advanced search with the developed keyword strategy as of March 26, 2023. All obtainable information from the trials was collected without any restrictions to conduct a comprehensive review. RESULTS A total of 169 registered trials were included for evaluation. Of all trials, 102 trials were eligible for this study. Countries from Asia registered the most trials (62.75%), and hospitals sponsored most of the trials (42.16%). Randomized, Phase 2, interventional trials were dominant, and approximately 64.71% of the trials anticipated recruiting less than 100 participants. More than half of the trials were from 2016 to 2023 (53.92%). While 45 trials were completed, only 16 trials had results for further analysis. According to the completed results, the types of herbal medicines from the trials mainly focused on lung, breast, and colorectal cancer. CONCLUSION This study is the first to explore the characteristics of clinical trials of herbal medicine for cancer registered in large clinical databases. The acquired trials had relatively informative data; however, better-designed trials may be needed for health professionals to consider herbal medicine as an option when treating cancer patients.
Collapse
Affiliation(s)
- Bo-Young Youn
- Hwasung Medi-Science University, Hwaseong-si, Gyeonggi-do, South Korea
| | - Ji-Hyun Kim
- Kyung Hee University, Seoul, Republic of Korea
| | - Yong-Kyu Jo
- Kyung Hee University, Seoul, Republic of Korea
| | | | - Ji-Yeong Im
- Kyung Hee University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
44
|
Khalid A, Nadeem T, Khan MA, Ali Q, Zubair M. In vitro evaluation of immunomodulatory, anti-diabetic, and anti-cancer molecular mechanisms of Tribulus terrestris extracts. Sci Rep 2022; 12:22478. [PMID: 36577761 PMCID: PMC9797551 DOI: 10.1038/s41598-022-26742-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Dampened immunity and impaired wound healing in diabetic patients may lead to diabetic foot ulcer disease, which is the leading cause of limb amputations and hospitalization. On the other hand, cancer is the most significant cause of mortality globally, accounting for over 10 million fatalities in 2020, or nearly one in every six deaths. Plants and herbs have been used to treat chronic diseases due to their essential pharmaceutical attributes, such as mitigating drug resistance, ameliorating systemic toxicities, reducing the need for synthetic chemotherapeutic agents,and strengthening the immune system. The present study has been designed to evaluate the effects of Tribulus terrestris on wound healing, cytotoxic and anti-inflammatory responses against HepG-2 liver cancer cell line. Two solvents (methanol and ethanol) were used for root extraction of T. terrestris. The wound healing potential of the extracts was studied on diabetic cell culture line by scratch assay. The anti-oxidant and cytotoxic potentials were evaluated by in vitro assays against HepG2 cell line. The methanolic root extract resulted in the coverage of robust radical scavenging or maximum inhibition of 66.72%,potent cytotoxic activity or reduced cell viability of 40.98%, and anti-diabetic activity having mighty α-glucosidase inhibition of 50.16% at a concentration of 80 μg/ml. Significant reduction in the levels of LDH leakage (56.38%), substantial ROS (48.45%) and SOD (72.13%) activities were recorededMoreover, gene expression analysis demonstrated the down-regulation of inflammatory markers (TNF-α, MMP-9, Bcl-2, and AFP) in HepG-2 cells when treated with T. terresteris methanolic extract as compared to stress. Furthermore, the down-regulation of inflammatory markers was validated through ELISA-mediated protein estimation of IL-1β and TNF-α. It is expected that this study will lay a foundation and lead to the development of efficient but low-cost, natural herbs extract-based dressing/ointment for diabetic patients and identify potential drug metabolites to treat out-of-whack inflammatory responses involved in cancer onset, progression, and metastasis.
Collapse
Affiliation(s)
- Abdullah Khalid
- grid.411786.d0000 0004 0637 891XDepartment of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Tariq Nadeem
- grid.11173.350000 0001 0670 519XCentre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Asaf Khan
- grid.412298.40000 0000 8577 8102Institute of Plant Breeding and Biotechnology, MNS-University of Agriculture, Multan, Pakistan
| | - Qurban Ali
- grid.11173.350000 0001 0670 519XDepartment of Plant Breeding and Genetics, Faculty of Agricultural Sciences, University of the Punjab Lahore, Lahore, Pakistan
| | - Muhammad Zubair
- grid.411786.d0000 0004 0637 891XDepartment of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
45
|
Aloliqi AA. Therapeutic Potential of 6-Gingerol in Prevention of Colon Cancer Induced by Azoxymethane through the Modulation of Antioxidant Potential and Inflammation. Curr Issues Mol Biol 2022; 44:6218-6228. [PMID: 36547085 PMCID: PMC9776754 DOI: 10.3390/cimb44120424] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
A polyphenolic component of ginger, 6-gingerol, is widely reported to possess antioxidant, anti-inflammatory and anticancer activities. In the current study, it was aimed to investigate the anticancer effects of 6-gingerol (6-Gin) on azoxymethane (AOM)-induced colon cancer in rats. The results reveal that 6-Gin treatment significantly improves the antioxidant status disturbed by AOM intoxication. The 6-Gin treatment animal group showed enhanced activity of catalase (CAT) (46.6 ± 6.4 vs. 23.3 ± 4.3 U/mg protein), superoxide dismutase (SOD) (81.3 ± 7.6 vs. 60.4 ± 3.5 U/mg protein) and glutathione-S-transferase (GST) (90.3 ± 9.4 vs. 53.8 ± 10 mU/mg protein) (p < 0.05) as compared to the disease control group. Furthermore, the results reveal that AOM significantly enhances the inflammatory response and 6-gingerol potentially attenuates this response, estimated by markers, such as tumor necrosis factor-α (TNF-α) (1346 ± 67 vs. 1023 ± 58 pg/g), C-reactive protein (CRP) (1.12 ± 0.08 vs. 0.92 ± 0.7 ng/mL) and interleukin-6 (IL-6) (945 ± 67 vs. 653 ± 33 pg/g). In addition, the lipid peroxidation estimated in terms of malondialdehyde (MDA) provoked by AOM exposure is significantly reduced by 6-gingerol treatment (167 ± 7.5 vs. 128.3 nmol/g). Furthermore, 6-gingerol significantly maintains the colon tissue architecture disturbed by the AOM treatment. Loss of tumor suppressor protein, phosphatase and tensin homolog (PTEN) expression was noticed in the AOM treated group, whereas in the animals treated with 6-gingerol, the positivity of PTEN expression was high. In conclusion, the current findings advocate the health-promoting effects of 6-gingerol on colon cancer, which might be due to its antioxidant and anti-inflammatory potential.
Collapse
Affiliation(s)
- Abdulaziz A Aloliqi
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia
| |
Collapse
|
46
|
Pan X, Hou X, Zhang F, Tang P, Wan W, Su Z, Yang Y, Wei W, Du Z, Deng J, Hao E. Gnetum montanum extract induces apoptosis by inhibiting the activation of AKT in SW480 human colon cancer cells. PHARMACEUTICAL BIOLOGY 2022; 60:915-930. [PMID: 35587342 PMCID: PMC9122364 DOI: 10.1080/13880209.2022.2063340] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 04/01/2022] [Accepted: 04/02/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Gnetum montanum Markgr. (Gnetaceae) is used to treat rheumatic arthralgia and bruises in the clinic. OBJECTIVE To exam the activity and mechanism of G. montanum extract (GME) against colon cancer cells SW480. MATERIALS AND METHODS The anti-proliferative activity of GME (0-120 μg/mL) on SW480 cells was determined using MTS assay at 24, 48, and 72 h. The in vitro activity of GME (0-120 μg/mL) on SW480 cells was investigated using flow cytometry and western blotting analysis. The in vivo activity of GME was evaluated using xenograft tumour model of zebrafish and nude mice. The chemical composition of GME was detected by using HPLC-MS/MS. RESULTS The IC50 value SW480 cells viability by GME were 126.50, 78.25, and 50.77 μg/mL, respectively, for 24, 48, and 72 h. The experiments showed that apoptotic cells and G2/M phase cells increased from 20.81 to 61.53% (p < 0.01) and 25.76 to 34.93% with 120 μg/mL GME, respectively. GME also down-regulated the protein expression of P-AKT, P-GSK-3β, P-PDK1, P-c-Raf, caspase-3, and Bcl-2, and up-regulated the expression cleaved caspase-3, cleaved PARP, and Bax. In vivo study found that GME can significantly inhibit the growth and migration of SW480 cells in xenograft zebrafish. GME reduced the nude mice tumour weight to approximately 32.19% at 28 mg/kg/day and to 53.17% (p < 0.01) at 56 mg/kg/day. Forty-two compounds were identified from the GME. DISCUSSION AND CONCLUSIONS GME has a significant antitumor effect on colon cancer cells SW480, and it has the potential to be developed as an anticancer agent.
Collapse
Affiliation(s)
- Xianglong Pan
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Xiaotao Hou
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Fan Zhang
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Peiling Tang
- Department of Bioscience, Faculty of Applied Sciences, Tunku Abdul Rahman University College, Kuala Lumpur, Malaysia
| | - Wanruo Wan
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Zixia Su
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Yeguo Yang
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Wei Wei
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Zhengcai Du
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Jiagang Deng
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Erwei Hao
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| |
Collapse
|
47
|
Recinella L, Chiavaroli A, Veschi S, Cama A, Acquaviva A, Libero ML, Leone S, Di Simone SC, Pagano E, Zengin G, Menghini L, Brunetti L, Izzo AA, Orlando G, Ferrante C. A grape (Vitis vinifera L.) pomace water extract modulates inflammatory and immune response in SW-480 cells and isolated mouse colon. Phytother Res 2022; 36:4620-4630. [PMID: 36069605 PMCID: PMC10087928 DOI: 10.1002/ptr.7581] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/07/2022] [Accepted: 07/07/2022] [Indexed: 12/13/2022]
Abstract
Grape (Vitis vinifera L.) pomace is a residue derived from the winemaking process, which contains bioactive compounds displaying noteworthy health-promoting properties. The aim of the present study was to investigate the phenolic composition and protective effects of a water extract of grape pomace (WEGP) in colorectal cancer cell line SW480 and in isolated mouse colon exposed to Escherichia coli lipopolysaccharide (LPS). The extract decreased SW-480 cell viability, as well as vascular endothelial factor A (VEGFA), hypoxia-induced factor 1α (HIF1α), and transient receptor potential M8 (TRPM8) LPS-induced gene expression. Moreover, the extract inhibited mRNA levels of nuclear factor kB (NFkB), cyclooxygenase (COX)-2, tumor necrosis factor (TNF)α, interleukin (IL)-6, IL-1β, IL-10, inducible nitric oxide synthase (iNOS), and interferon (IFN)γ, in isolated colon. Conversely, WEGP increased the gene expression of antioxidant catalase (CAT) and superoxide dismutase (SOD), in the same model. The modulatory effects exerted by WEGP could be related, at least in part, to the phenolic composition, with particular regards to the catechin level. Docking calculations also predicted the interactions of catechin toward TRPM8 receptor, deeply involved in colon cancer; thus further suggesting the grape pomace as a valuable source of bioactive extracts and phytochemicals with protective effects in the colon.
Collapse
Affiliation(s)
- Lucia Recinella
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Annalisa Chiavaroli
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Serena Veschi
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Alessandro Cama
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Alessandra Acquaviva
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy.,Veridia Italia Srl, Pescara, Italy
| | - Maria Loreta Libero
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Sheila Leone
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | | | - Ester Pagano
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy.,Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Gokhan Zengin
- Physiology and Biochemistry Laboratory, Department of Biology, Science Faculty, Selcuk University, Konya, Turkey
| | - Luigi Menghini
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Luigi Brunetti
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Angelo Antonio Izzo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Giustino Orlando
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Claudio Ferrante
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
48
|
Pharmacological Activities and Characterization of Phenolic and Flavonoid Compounds in Solenostemma argel Extract. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238118. [PMID: 36500211 PMCID: PMC9741047 DOI: 10.3390/molecules27238118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/05/2022] [Accepted: 07/09/2022] [Indexed: 11/23/2022]
Abstract
Solenostemma argel is a desert medicinal plant indigenous to African countries. This research aims to study the pharmacological properties of Solenostemma argel plant. Aerial parts (leaves and flowers) of Solenostemma argel (Delile) Hayane were tested for antibacterial activity, antioxidant activity, anticancer, and anti-inflammatory activity. Phenolic and flavonoid contents of the plant were characterized. There was an increase in the antioxidant activity of Solenostemma argel extract from 12.16% to 94.37% by increasing concentration from10 µg/mL to 1280 µg/mL. The most sensitive organism was S. epidermidis with chloroform extract. The MTT assay revealed that methanolic extracts of Solenostemma argel showed potent cytotoxic effects on the A549, Caco-2, and MDAMB-231 cell lines, respectively. The anti-inflammatory activity increased by increasing the concentration of methanolic extract of Solenostemma argel, using indomethacin as a standard. Gallic acid was the most abundant phenolic acid, followed by synergic acid and p-coumaric acid, respectively. Catechin, quercetin, luteolin, kaempferol and rutin flavonoids were also found in the methanolic extract. GC-mass analysis showed that aerial parts of Solenostemma argel were rich in 2-(5-methyl-5 vinyl tetrahydro-2-furanyl)-2-propanol (11.63%), hexanoic acid methyl ester (10.93%), 3-dioxolane,4-methyl-2-pentadecyl (9.69%), phenol, 2-(1,1-dimethylethyl) (8.50%). It can be concluded that Solenostemma argel methanolic extract contain natural bioactive constituents with potential medicinal importance such as antioxidants, antimicrobial, anti-inflammatory, and anticancer activities.
Collapse
|
49
|
Alfaleh MA, Fahmy O, Al-Rabia MW, Abourehab MAS, Ahmed OAA, Fahmy UA, H Alsulimani H, Badr-Eldin SM, Aldawsari HM, Aldhabi BM, Alharbi AS, Alhakamy NA. Hybrid nanoparticulate system of Fluvastatin loaded phospholipid, alpha lipoic acid and melittin for the management of colon cancer. Sci Rep 2022; 12:19446. [PMID: 36376469 PMCID: PMC9663543 DOI: 10.1038/s41598-022-24151-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
As a hydroxymethylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, Fluvastatin (FLV) is used for reducing low-density lipoprotein (LDL) cholesterol as well as to prevent cardiovascular problems. FLV showed cell line cytotoxicity and antitumor effect. Melittin (MEL) exhibits antineoplastic activity and is known to be promising as a therapeutic option for cancer patients. The aim of this work was to investigate the combination of FLV with MEL loaded hybrid formula of phospholipid (PL) with alpha lipoic acid (ALA) nanoparticles to maximize anticancer tendencies. This study examines the optimization of the prepared formulation in order to minimize nanoparticles size and maximize zeta potential to potentiate cytotoxic potentialities in colon cancer cells (Caco2), cell viability, cell cycle analysis and annexin V were tested. In addition to biological markers as P53, Bax, bcl2 and Caspase 3 evaluation The combination involving FLV PL ALA MEL showed enhanced cytotoxic potentiality (IC50 = 9.242 ± 0.35 µg/mL), about twofold lower, compared to the raw FLV (IC50 = 21.74 ± 0.82 µg/mL). According to studies analyzing cell cycle, optimized FLV PL ALA MEL was found to inhibit Caco2 colon cancer cells more significantly than other therapeutic treatments, wherein a higher number of cells were found to accumulate over G2/M and pre-G1 phases, whereas G0/G1/S phases witnessed the accumulation of a lower number of cells. The optimized formulation may pave the way for a novel and more efficacious treatment for colon cancer.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Omar Fahmy
- Department of Urology, Universiti Putra Malaysia, 43400, Selangor, Malaysia
| | - Mohammed W Al-Rabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, and Industrial Pharmacy, Faculty, of Pharmacy, Minia University, Minia, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Osama A A Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Usama A Fahmy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Helal H Alsulimani
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shaimaa M Badr-Eldin
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Hibah M Aldawsari
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Bander M Aldhabi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Awaad S Alharbi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
50
|
Almutairi MH, Alrubie TM, Almutairi BO, Alamri AM, Alrefaei AF, Arafah MM, Alanazi M, Semlali A. The Expression Patterns of Human Cancer-Testis Genes Are Induced through Epigenetic Drugs in Colon Cancer Cells. Pharmaceuticals (Basel) 2022; 15:1319. [PMID: 36355490 PMCID: PMC9692864 DOI: 10.3390/ph15111319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/28/2022] [Accepted: 10/21/2022] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND The expression of human germline genes is restricted to the germ cells of the gonads, which produce sperm and eggs. The germline genes involved in testis development and potentially activated in cancer cells are known as cancer-testis (CT) genes. These genes are potential therapeutic targets and biomarkers, as well as drivers of the oncogenic process. CT genes can be reactivated by treatment with drugs that demethylate DNA. The majority of the existing literature on CT gene activation focuses on X-chromosome-produced CT genes. We tested the hypothesis that epigenetic landscape changes, such as DNA methylation, can alter several CT gene expression profiles in cancer and germ cells. METHODS Colon cancer (CC) cell lines were treated with the DNA methyltransferase inhibitor (DNMTi) 5-aza-2'-deoxycytidine, or with the histone deacetylase inhibitor (HDACi) trichostatin A (TSA). The effects of these epigenetic treatments on the transcriptional activation of previously published CT genes (CTAG1A, SCP2D1, TKTL2, LYZL6, TEX33, and ACTRT1) and testis-specific genes (NUTM1, ASB17, ZSWIM2, ADAM2, and C10orf82) were investigated. RESULTS We found that treatment of CC cell lines with 5-aza-2'-deoxycytidine or TSA correlated with activation of X-encoded CT genes and non-X-encoded CT genes in somatic (non-germline) cells. CONCLUSION These findings confirm that a subset of CT genes can be regulated by hypomethylating drugs and subsequently provide a potential therapeutic target for cancer.
Collapse
Affiliation(s)
- Mikhlid H. Almutairi
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Turki M. Alrubie
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Bader O. Almutairi
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah M. Alamri
- Genome Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdulwahed F. Alrefaei
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Maha M. Arafah
- Pathology Department, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Alanazi
- Genome Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdelhabib Semlali
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, 2420 Rue de la Terrasse, Local 1758, Québec, QC G1V 0A6, Canada
| |
Collapse
|