Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.989
Filter
1.
Oncol Res ; 32(9): 1389-1399, 2024.
Article in English | MEDLINE | ID: mdl-39220125

ABSTRACT

Resveratrol (RSV), the primary polyphenol found in grapes, has been revealed to have anti-inflammatory properties by reducing the capacity of the peripheral blood mononuclear cells to produce pro-inflammatory cytokines, including IL-1ß, IL-6, IL-1ra and TNFα. Considering the close association between chronic inflammation and cancer development, RSV's immunomodulatory properties are one way by which the polyphenol may inhibit cancer initiation, proliferation, neovascularization, and migration. Resveratrol influences the generation of microtumor environment which is one of the key factors in cancer progress. In addition to immunomodulation, RSV inhibits cancer development by expressing anti-oxidant effects, causing cell cycle arrest, stimulating the function of certain enzymes, and activating cell signaling pathways. The end outcome is one of the various forms of cell death, including apoptosis, pyroptosis, necroptosis, and more, as it has been observed in vitro. RSV has been shown to act against cancer in practically every organ, while its effects on colon cancer have been documented more frequently. It is remarkable that longer-term clinical studies that may have established the potential for this natural substance to serve as a therapeutic adjuvant to traditional anti-cancer medications were not prompted by the encouraging outcomes seen with cancer cells treated with non-toxic doses of resveratrol. The current review aims to assess the recent findings about the immunological and anti-cancer characteristics of RSV, with a particular emphasis on cancers of the digestive tract, as a challenge for future clinical research that may contribute to the better prognosis of cancer.


Subject(s)
Digestive System Neoplasms , Resveratrol , Resveratrol/pharmacology , Resveratrol/therapeutic use , Humans , Digestive System Neoplasms/drug therapy , Digestive System Neoplasms/prevention & control , Animals , Immunomodulating Agents/pharmacology , Immunomodulating Agents/therapeutic use , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use
2.
Hum Exp Toxicol ; 43: 9603271241266106, 2024.
Article in English | MEDLINE | ID: mdl-39291655

ABSTRACT

OBJECTIVE: This study aimed to explore the expression and biological functions of SIRT3 in colorectal cancer cells (HCT-116), the impacts of sulforaphane on the ferroptosis of HCT-116 cells and the involvement of the SIRT3/AMPK/mTOR axis in those effects. METHODS: SIRT3-overexpressing (OE) and SIRT3-knockout (KO) cell lines were treated with different concentrations of sulforaphane, RSL-3, and IKE. Cell viability, intracellular ROS, MDA, iron levels, as well as mRNA and protein expressions of target genes were measured. RESULTS: SIRT3 expression in HCT-116 cells was increased by ferroptosis inducers and decreased by ferroptosis inhibitors. SIRT3 overexpression reduced cell viability and increased intracellular levels of ROS, MDA, and iron, whereas SIRT3 knockdown achieved the opposite effects. SIRT3 overexpression suppressed SLC7A11 expression and promoted the activation of AMPK/mTOR pathway. Restoration of SLC7A11 expression blocked the effects of SIRT3 on ferroptosis induction and cell viability inhibition. SIRT3 effects on cell viability and ferroptosis were antagonized by inhibitors of AMPK or mTOR. Moreover, sulforaphane triggered the ferroptosis of HCT-116 cells by activating the SIRT3/AMPK/mTOR axis. CONCLUSIONS: SIRT3 triggered SLC7A11-mediated ferroptosis in HCT-116 cells, reducing cell viability by activating the AMPK/mTOR pathway, and sulforaphane targets it to inhibit colorectal cancer.


Subject(s)
AMP-Activated Protein Kinases , Colorectal Neoplasms , Ferroptosis , Isothiocyanates , Signal Transduction , Sirtuin 3 , Sulfoxides , TOR Serine-Threonine Kinases , Humans , Isothiocyanates/pharmacology , Sirtuin 3/metabolism , Sirtuin 3/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Ferroptosis/drug effects , AMP-Activated Protein Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , HCT116 Cells , Anticarcinogenic Agents/pharmacology , Cell Survival/drug effects
3.
Nutrients ; 16(16)2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39203892

ABSTRACT

The olive tree (Olea europaea) and olive oil hold significant cultural and historical importance in Europe. The health benefits associated with olive oil consumption have been well documented. This paper explores the mechanisms of the anti-cancer effects of olive oil and olive leaf, focusing on their key bioactive compounds, namely oleocanthal, oleacein, and oleuropein. The chemopreventive potential of oleocanthal, oleacein, and oleuropein is comprehensively examined through this systematic review. We conducted a systematic literature search to identify eligible articles from Scopus, PubMed, and Web of Science databases published up to 10 October 2023. Among 4037 identified articles, there were 88 eligible articles describing mechanisms of chemopreventive effects of oleocanthal, oleacein, and oleuropein. These compounds have the ability to inhibit cell proliferation, induce cell death (apoptosis, autophagy, and necrosis), inhibit angiogenesis, suppress tumor metastasis, and modulate cancer-associated signalling pathways. Additionally, oleocanthal and oleuropein were also reported to disrupt redox hemostasis. This review provides insights into the chemopreventive mechanisms of O. europaea-derived secoiridoids, shedding light on their role in chemoprevention. The bioactivities summarized in the paper support the epidemiological evidence demonstrating a negative correlation between olive oil consumption and cancer risk. Furthermore, the mapped and summarized secondary signalling pathways may provide information to elucidate new synergies with other chemopreventive agents to complement chemotherapies and develop novel nutrition-based anti-cancer approaches.


Subject(s)
Aldehydes , Cyclopentane Monoterpenes , Iridoid Glucosides , Neoplasms , Olea , Olive Oil , Phenols , Animals , Humans , Aldehydes/pharmacology , Aldehydes/therapeutic use , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Cell Proliferation/drug effects , Cyclopentane Monoterpenes/pharmacology , Cyclopentane Monoterpenes/therapeutic use , Iridoid Glucosides/pharmacology , Iridoid Glucosides/therapeutic use , Neoplasms/drug therapy , Neoplasms/prevention & control , Olea/chemistry , Olive Oil/chemistry , Olive Oil/therapeutic use , Phenols/pharmacology , Phenols/therapeutic use , Plant Leaves/chemistry
4.
Molecules ; 29(14)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39064880

ABSTRACT

Cancer is a complicated and ever-evolving disease that remains a significant global cause of disease and mortality. Its complexity, which is evident at the genetic and phenotypic levels, contributes to its diversity and resistance to treatment. Numerous scientific investigations on human and animal models demonstrate the potential of phytochemicals in cancer prevention. Coffee has been shown to possess potent anti-carcinogenic properties, and studies have documented the consumption of coffee as a beverage reduces the risk of cancer occurrence. The major secondary metabolites of coffee, named caffeine and chlorogenic acid, have been linked to anti-inflammatory and antineoplastic effects through various signaling. In light of this, this review article provides a comprehensive analysis based on studies in anticancer effects of coffee, chlorogenic acid, and caffeine published between 2010 and 2023, sourced from Scopus, Pubmed, and Google Scholar databases. We summarize recent advances and scientific evidence on the association of phytochemicals found in coffee with a special emphasis on their biological activities against cancer and their molecular mechanism deemed potential to be used as a novel therapeutic target for cancer prevention and therapy.


Subject(s)
Caffeine , Chlorogenic Acid , Coffee , Neoplasms , Chlorogenic Acid/pharmacology , Chlorogenic Acid/chemistry , Humans , Caffeine/pharmacology , Caffeine/chemistry , Coffee/chemistry , Neoplasms/prevention & control , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Phytochemicals/pharmacology , Phytochemicals/chemistry , Phytochemicals/therapeutic use , Chemoprevention , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry
5.
Sci Rep ; 14(1): 16016, 2024 07 11.
Article in English | MEDLINE | ID: mdl-38992004

ABSTRACT

Triple-negative breast cancer (TNBC) represents aggressive phenotype with limited treatment options due to the lack of drug targets. Natural compounds are extensively studied regarding their potential to alter the efficacy of cancer treatment Among them sulforaphane - an isothiocyanate of natural origin, was shown to be a hormetic compound, that may exert divergent effects: cytoprotective or cytotoxic depending on its concentrations. Thus, the aim of this study was to determine the effect of its low, dietary concentrations on the proliferation and migration of the TNBC cells in the in vivo and in vitro 2D and 3D model. Results of the in vivo experiment showed up to 31% tumor growth inhibition after sulforaphane treatment associated with lowered proliferating potential of cancer cells, reduced areas of necrosis, and changed immune cell type infiltration, showing less malignant type of tumor in contrast to the non-treated group. Also, the study revealed that sulforaphane decreased the number of lung metastases. The in vitro study confirmed that SFN inhibited cell migration, but only in cells derived from 3D spheroids, not from 2D in vitro cultures. The results show a specific role of sulforaphane in the case of cells released from the TNBC primary tumor and its environment.


Subject(s)
Cell Movement , Cell Proliferation , Isothiocyanates , Sulfoxides , Triple Negative Breast Neoplasms , Isothiocyanates/pharmacology , Isothiocyanates/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Sulfoxides/pharmacology , Female , Humans , Cell Movement/drug effects , Cell Line, Tumor , Animals , Cell Proliferation/drug effects , Mice , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Xenograft Model Antitumor Assays
6.
Mol Med ; 30(1): 94, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38902597

ABSTRACT

Despite recent advances in tumor diagnosis and treatment technologies, the number of cancer cases and deaths worldwide continues to increase yearly, creating an urgent need to find new methods to prevent or treat cancer. Sulforaphane (SFN), as a member of the isothiocyanates (ITCs) family, which is the hydrolysis product of glucosinolates (GLs), has been shown to have significant preventive and therapeutic cancer effects in different human cancers. Early studies have shown that SFN scavenges oxygen radicals by increasing cellular defenses against oxidative damage, mainly through the induction of phase II detoxification enzymes by nuclear factor erythroid 2-related factor 2 (Nrf2). More and more studies have shown that the anticancer mechanism of SFN also includes induction of apoptotic pathway in tumor cells, inhibition of cell cycle progression, and suppression of tumor stem cells. Therefore, the application of SFN is expected to be a necessary new approach to treating cancer. In this paper, we review the multiple molecular mechanisms of SFN in cancer prevention and treatment in recent years, which can provide a new vision for cancer treatment.


Subject(s)
Anticarcinogenic Agents , Isothiocyanates , Neoplasms , Sulfoxides , Isothiocyanates/pharmacology , Isothiocyanates/therapeutic use , Sulfoxides/pharmacology , Sulfoxides/therapeutic use , Humans , Neoplasms/prevention & control , Neoplasms/drug therapy , Neoplasms/metabolism , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Animals , Apoptosis/drug effects , NF-E2-Related Factor 2/metabolism
7.
Biomed Pharmacother ; 176: 116852, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38834007

ABSTRACT

The incidence of breast cancer is increasing annually, making it a major health threat for women. Chemoprevention using natural, dietary, or synthetic products has emerged as a promising approach to address this growing burden. Atractylenolide-III (AT-III), a sesquiterpenoid present in various medicinal herbs, has demonstrated potential therapeutic effects against several diseases, including tumors, nonalcoholic fatty liver disease, and cerebral ischemic injury. However, its impact on breast cancer chemoprevention remains unexplored. In this study, we used an N-methyl-N-nitrosourea (NMU)-induced rat breast cancer model and 17ß-estradiol (E2)-treated MCF-10A cells to evaluate the chemopreventive potential of AT-III on mammary tumorigenesis. AT-III inhibited mammary tumor progression, evidenced by reduced tumor volume and multiplicity, prolonged tumor latency, and the reversal of NMU-induced weight loss. Furthermore, AT-III suppressed NMU-induced inflammation and oxidative stress through the Nrf2/ARE pathway in breast cancer tissues. In vitro, AT-III effectively suppressed E2-induced anchorage-independent growth and cell migration in MCF-10A cells. Nrf2 knockdown attenuated the protective effects of AT-III, highlighting the pivotal role of Nrf2 in AT-III-mediated suppression of tumorigenesis. The mechanism involves the induction of Nrf2 expression by AT-III through the autophagic degradation of Kelch-like ECH-associated protein 1 (Keap1). Overall, the results of this study indicate that AT-III is a promising candidate for breast cancer chemoprevention and provide valuable insights into its molecular interactions and signaling pathways.


Subject(s)
Autophagy , Kelch-Like ECH-Associated Protein 1 , Lactones , NF-E2-Related Factor 2 , Sesquiterpenes , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Sesquiterpenes/pharmacology , Female , Kelch-Like ECH-Associated Protein 1/metabolism , Lactones/pharmacology , Autophagy/drug effects , Signal Transduction/drug effects , Rats , Humans , Cell Line, Tumor , Rats, Sprague-Dawley , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Mammary Neoplasms, Experimental/chemically induced , Oxidative Stress/drug effects , Methylnitrosourea/toxicity , Carcinogenesis/drug effects , Anticarcinogenic Agents/pharmacology , Estradiol/pharmacology
8.
Sci Rep ; 14(1): 12091, 2024 05 27.
Article in English | MEDLINE | ID: mdl-38802425

ABSTRACT

Estrogen receptor-negative [ER(-)] mammary cancer is the most aggressive type of breast cancer (BC) with higher rate of metastasis and recurrence. In recent years, dietary prevention of BC with epigenetically active phytochemicals has received increased attention due to its feasibility, effectiveness, and ease of implementation. In this regard, combinatorial phytochemical intervention enables more efficacious BC inhibition by simultaneously targeting multiple tumorigenic pathways. We, therefore, focused on investigation of the effect of sulforaphane (SFN)-rich broccoli sprouts (BSp) and withaferin A (WA)-rich Ashwagandha (Ash) combination on BC prevention in estrogen receptor-negative [ER(-)] mammary cancer using transgenic mice. Our results indicated that combinatorial BSp + Ash treatment significantly reduced tumor incidence and tumor growth (~ 75%) as well as delayed (~ 21%) tumor latency when compared to the control treatment and combinatorial BSp + Ash treatment was statistically more effective in suppressing BC compared to single BSp or Ash intervention. At the molecular level, the BSp and Ash combination upregulated tumor suppressors (p53, p57) along with apoptosis associated proteins (BAX, PUMA) and BAX:BCL-2 ratio. Furthermore, our result indicated an expressional decline of epigenetic machinery HDAC1 and DNMT3A in mammary tumor tissue because of combinatorial treatment. Interestingly, we have reported multiple synergistic interactions between BSp and Ash that have impacted both tumor phenotype and molecular expression due to combinatorial BSp and Ash treatment. Our RNA-seq analysis results also demonstrated a transcriptome-wide expressional reshuffling of genes associated with multiple cell-signaling pathways, transcription factor activity and epigenetic regulations due to combined BSp and Ash administration. In addition, we discovered an alteration of gut microbial composition change because of combinatorial treatment. Overall, combinatorial BSp and Ash supplementation can prevent ER(-) BC through enhanced tumor suppression, apoptosis induction and transcriptome-wide reshuffling of gene expression possibly influencing multiple cell signaling pathways, epigenetic regulation and reshaping gut microbiota.


Subject(s)
Breast Neoplasms , Epigenesis, Genetic , Gastrointestinal Microbiome , Isothiocyanates , Sulfoxides , Withanolides , Isothiocyanates/pharmacology , Animals , Withanolides/pharmacology , Sulfoxides/pharmacology , Female , Mice , Epigenesis, Genetic/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Gastrointestinal Microbiome/drug effects , Mice, Transgenic , Plant Extracts/pharmacology , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Humans , Brassica/chemistry , Histone Deacetylase 1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Anticarcinogenic Agents/pharmacology
9.
Adv Nutr ; 15(7): 100240, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734077

ABSTRACT

The vitamin E family contains α-tocopherol (αT), ßT, γT, and δT and α-tocotrienol (TE), ßTE, γTE, and δTE. Research has revealed distinct roles of these vitamin E forms in prostate cancer (PCa). The ATBC trial showed that αT at a modest dose significantly decreased PCa mortality among heavy smokers. However, other randomized controlled trials including the Selenium and Vitamin E Cancer Prevention Trial (SELECT) indicate that supplementation of high-dose αT (≥400 IU) does not prevent PCa among nonsmokers. Preclinical cell and animal studies also do not support chemopreventive roles of high-dose αT and offer explanations for increased incidence of early-stage PCa reported in the SELECT. In contrast, accumulating animal studies have demonstrated that γT, δT, γTE, and δTE appear to be effective for preventing early-stage PCa from progression to adenocarcinoma in various PCa models. Existing evidence also support therapeutic roles of γTE and its related combinations against advanced PCa. Mechanistic and cell-based studies show that different forms of vitamin E display varied efficacy, that is, δTE ≥ γTE > δT ≥ γT >> αT, in inhibiting cancer hallmarks and enabling characteristics, including uncontrolled cell proliferation, angiogenesis, and inflammation possibly via blocking 5-lipoxygenase, nuclear factor κB, hypoxia-inducible factor-1α, modulating sphingolipids, and targeting PCa stem cells. Overall, existing evidence suggests that modest αT supplement may be beneficial to smokers and γT, δT, γTE, and δTE are promising agents for PCa prevention for modest-risk to relatively high-risk population. Despite encouraging preclinical evidence, clinical research testing γT, δT, γTE, and δTE for PCa prevention is sparse and should be considered.


Subject(s)
Prostatic Neoplasms , Tocopherols , Tocotrienols , Male , Humans , Prostatic Neoplasms/prevention & control , Tocotrienols/pharmacology , Tocotrienols/therapeutic use , Tocopherols/pharmacology , Tocopherols/therapeutic use , Animals , Dietary Supplements , Chemoprevention/methods , Randomized Controlled Trials as Topic , Vitamin E/pharmacology , Vitamin E/therapeutic use , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use
10.
Nutrients ; 16(8)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38674816

ABSTRACT

Colorectal cancer (CRC) accounts for 30% of all cancer cases worldwide and is the second leading cause of cancer-related deaths. CRC develops over a long period of time, and in the early stages, pathological changes can be mitigated through nutritional interventions using bioactive plant compounds. Our study aims to determine the effect of highly purified oat beta-glucan on an animal CRC model. The study was performed on forty-five male Sprague-Dawley rats with azoxymethane-induced early-stage CRC, which consumed feed containing 1% or 3% low molar mass oat beta-glucan (OBG) for 8 weeks. In the large intestine, morphological changes, CRC signaling pathway genes (RT-PCR), and proteins (Western blot, immunohistochemistry) expression were analyzed. Whole blood hematology and blood redox status were also performed. Results indicated that the histologically confirmed CRC condition led to a downregulation of the WNT/ß-catenin pathway, along with alterations in oncogenic and tumor suppressor gene expression. However, OBG significantly modulated these effects, with the 3% OBG showing a more pronounced impact. Furthermore, CRC rats exhibited elevated levels of oxidative stress and antioxidant enzyme activity in the blood, along with decreased white blood cell and lymphocyte counts. Consumption of OBG at any dose normalized these parameters. The minimal effect of OBG in the physiological intestine and the high activity in the pathological condition suggest that OBG is both safe and effective in early-stage CRC.


Subject(s)
Avena , Dietary Supplements , Oxidative Stress , Rats, Sprague-Dawley , beta-Glucans , Animals , Male , beta-Glucans/pharmacology , beta-Glucans/administration & dosage , Avena/chemistry , Rats , Oxidative Stress/drug effects , Colonic Neoplasms/prevention & control , Anticarcinogenic Agents/pharmacology , Azoxymethane , Wnt Signaling Pathway/drug effects , Disease Models, Animal , Animal Feed , Colon/pathology , Colon/drug effects , Colon/metabolism , Colorectal Neoplasms/prevention & control , Antioxidants/pharmacology
11.
Nutrients ; 16(6)2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38542669

ABSTRACT

Isothiocyanates are biologically active products resulting from the hydrolysis of glucosinolates predominantly present in cruciferous vegetables belonging to the Brassicaceae family. Numerous studies have demonstrated the diverse bioactivities of various isothiocyanates, encompassing anticarcinogenic, anti-inflammatory, and antioxidative properties. Nature harbors distinct isothiocyanate precursors, glucosinolates such as glucoraphanin and gluconastrin, each characterized by unique structures, physical properties, and pharmacological potentials. This comprehensive review aims to consolidate the current understanding of Moringa isothiocyanates, mainly 4-[(α-L-rhamnosyloxy) benzyl] isothiocyanate), comparing this compound with other well-studied isothiocyanates such as sulforaphane and phenyl ethyl isothiocyanates. The focus is directed toward elucidating differences and similarities in the efficacy of these compounds as agents with anticancer, anti-inflammatory, and antioxidative properties.


Subject(s)
Anticarcinogenic Agents , Brassicaceae , Glucosinolates/pharmacology , Brassicaceae/chemistry , Isothiocyanates/pharmacology , Antioxidants/pharmacology , Anti-Inflammatory Agents , Anticarcinogenic Agents/pharmacology
12.
Toxicol Appl Pharmacol ; 485: 116916, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38537874

ABSTRACT

This study aims to explore the impact and underlying mechanism of sulforaphane (SFN) intervention on the migration and invasion of lung adenocarcinoma induced by 7, 8-dihydroxy-9, 10-epoxy-benzo (a) pyrene (BPDE). Human lung adenocarcinoma A549 cells were exposed to varying concentrations of BPDE (0.25, 0.50, and 1.00 µM) and subsequently treated with 5 µM SFN. Cell viability was determined using CCK8 assay, while migration and invasion were assessed using Transwell assays. Lentivirus transfection was employed to establish NLRP12 overexpressing A549 cells. ELISA was utilized to quantify IL-33, CXCL12, and CXCL13 levels in the supernatant, while quantitative real-time PCR (qRT-PCR) and Western Blot were used to analyze the expression of NLRP12 and key factors associated with canonical and non-canonical NF-κB pathways. Results indicated an increase in migratory and invasive capabilities, concurrent with heightened expression of IL-33, CXCL12, CXCL13, and factors associated with both canonical and non-canonical NF-κB pathways. Moreover, mRNA and protein levels of NLRP12 were decreased in BPDE-stimulated A549 cells. Subsequent SFN intervention attenuated BPDE-induced migration and invasion of A549 cells. Lentivirus-mediated NLRP12 overexpression not only reversed the observed phenotype in BPDE-induced cells but also led to a reduction in the expression of critical factors associated with both canonical and non-canonical NF-κB pathways. Collectively, we found that SFN could inhibit BPDE-induced migration and invasion of A549 cells by upregulating NLRP12, thereby influencing both canonical and non-canonical NF-κB pathways.


Subject(s)
Adenocarcinoma of Lung , Cell Movement , Isothiocyanates , Lung Neoplasms , Neoplasm Invasiveness , Sulfoxides , Humans , Isothiocyanates/pharmacology , Sulfoxides/pharmacology , Cell Movement/drug effects , A549 Cells , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Anticarcinogenic Agents/pharmacology , NF-kappa B/metabolism , Cell Survival/drug effects , Signal Transduction/drug effects , Gene Expression Regulation, Neoplastic/drug effects
13.
Biochem Pharmacol ; 228: 116165, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38527559

ABSTRACT

In this comprehensive review we tried to reassess the role of phytochemicals in cancer chemoprevention. The exploration of the "synergistic effect" concept, advocating combined chemopreventive agents, faces challenges like low bioavailability. The review incorporates personal, occasionally controversial, viewpoints on natural compounds' cancer preventive capabilities, delving into mechanisms. Prioritizing significant contributions within the vast research domain, we aim stimulating discussion to provide a comprehensive insight into the evolving role of phytochemicals in cancer prevention. While early years downplayed the role of phytochemicals, the late nineties witnessed a shift, with leaders exploring their potential alongside synthetic compounds. Challenges faced by chemoprevention, such as limited pharmaceutical interest and cost-effectiveness issues, persist despite successful drugs. Recent studies, including the EPIC study, provide nuanced insights, indicating a modest risk reduction for increased fruit and vegetable intake. Phytochemicals, once attributed to antioxidant effects, face scrutiny due to low bioavailability and conflicting evidence. The Nrf2-EpRE signaling pathway and microbiota-mediated metabolism emerge as potential mechanisms, highlighting the complexity of understanding phytochemical mechanisms in cancer chemoprevention.


Subject(s)
Neoplasms , Phytochemicals , Humans , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Neoplasms/prevention & control , Neoplasms/metabolism , Animals , Chemoprevention/methods , Chemoprevention/trends , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use
14.
Toxicol Mech Methods ; 34(5): 507-516, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38221767

ABSTRACT

This study aimed to examine the expression and biological functions of ACTL6A in glioma cells (U251), the effects of sulforaphane on the growth of U251 cells and the involvement of the ACTL6A/PGK1 pathway in those effects. The U251 cell line was transfected with ACTL6A over-expression plasmids to upregulate the protein, or with ACTL6A inhibitor to underexpress it, then treated with different concentrations of sulforaphane. Cell viability, proliferation, and apoptosis were assessed using standard assays, and levels of mRNAs encoding ACTL6A, PGK1, cyclin D1, Myc, Bax or Bcl-2 were measured using quantitative real-time polymerase chain reaction (qRT-PCR). ACTL6A and PGK1 were expressed at higher levels in glioma cell lines than in normal HEB cells. ACTL6A overexpression upregulated PGK1, whereas ACTL6A inhibition had the opposite effect. ACTL6A overexpression induced proliferation, whereas its inhibition repressed proliferation, enhanced apoptosis, and halted the cell cycle. Moreover, sulforaphane suppressed the growth of U251 cells by inactivating the ACTL6A/PGK1 axis. ACTL6A acts via PGK1 to play a critical role in glioma cell survival and proliferation, and sulforaphane targets it to inhibit glioma.


Subject(s)
Anticarcinogenic Agents , Apoptosis , Cell Proliferation , Glioma , Isothiocyanates , Phosphoglycerate Kinase , Sulfoxides , Humans , Apoptosis/drug effects , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioma/pathology , Glioma/metabolism , Glioma/drug therapy , Glioma/genetics , Isothiocyanates/pharmacology , Phosphoglycerate Kinase/genetics , Phosphoglycerate Kinase/metabolism , Signal Transduction/drug effects , Anticarcinogenic Agents/pharmacology
15.
Nutrients ; 16(2)2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38276538

ABSTRACT

Exposure to B[a]P, the most characterized polycyclic aromatic hydrocarbon, significantly increases breast cancer risk. Our lab has previously reported that diallyl trisulfide (DATS), a garlic organosulfur compound (OSC) with chemopreventive and cell cycle arrest properties, reduces lipid peroxides and DNA damage in normal breast epithelial (MCF-10A) cells. In this study, we evaluated the ability of DATS to block the B[a]P-induced initiation of carcinogenesis in MCF-10A cells by examining changes in proliferation, clonogenic formation, reactive oxygen species (ROS) formation, 8-hydroxy-2-deoxyguanosine (8-OHdG) levels, and protein expression of ARNT/HIF-1ß, CYP1A1, and DNA POLß. The study results indicate that B[a]P increased proliferation, clonogenic formation, ROS formation, and 8-OHdG levels, as well as increasing the protein expression of ARNT/HIF-1ß and CYP1A1 compared to the control. Conversely, DATS/B[a]P co-treatment (CoTx) inhibited cell proliferation, clonogenic formation, ROS formation, and 8-OHdG levels compared to B[a]P alone. Treatment with DATS significantly inhibited (p < 0.0001) AhR expression, implicated in the development and progression of breast cancer. The CoTx also attenuated all the above-mentioned B[a]P-induced changes in protein expression. At the same time, it increased DNA POLß protein expression, which indicates increased DNA repair, thus causing a chemopreventive effect. These results provide evidence for the chemopreventive effects of DATS in breast cancer prevention.


Subject(s)
Allyl Compounds , Anticarcinogenic Agents , Breast Neoplasms , Garlic , Precancerous Conditions , Humans , Female , Garlic/metabolism , Antioxidants/pharmacology , Benzo(a)pyrene/toxicity , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Apoptosis , Sulfides/pharmacology , Epithelial Cells/metabolism , Anticarcinogenic Agents/pharmacology , DNA Repair , Breast Neoplasms/chemically induced , Breast Neoplasms/drug therapy , Breast Neoplasms/prevention & control , DNA
16.
Curr Med Chem ; 31(32): 5149-5164, 2024.
Article in English | MEDLINE | ID: mdl-38173069

ABSTRACT

Cancer is a general term for a group of similar diseases. It is a combined process that results from an accumulation of abnormalities at different biological levels, which involves changes at both genetic and biochemical levels in the cells. Several modifiable risk factors for each type of cancer include heredity, age, and institutional screening guidelines, including colonoscopy, mammograms, prostate-specific antigen testing, etc., which an individual cannot modify. Although a wide range of resources is available for cancer drugs and developmental studies, the cases are supposed to increase by about 70% in the next two decades due to environmental factors commonly driven by the way of living. The drugs used in cancer prevention are not entirely safe, have potential side effects and are generally unsuitable owing to substantial monetary costs. Interventions during the initiation and progression of cancer can prevent, diminish, or stop the transformation of healthy cells on the way to malignancy. Diet modifications are one of the most promising lifestyle changes that can decrease the threat of cancer development by nearly 40%. Neoxanthin is a xanthophyll pigment found in many microalgae and macroalgae, having significant anti-cancer, antioxidant and chemo-preventive activity. In this review, we have focused on the anti-cancer activity of neoxanthin on different cell lines and its cancer-preventive activity concerning obesity and oxidative stress. In addition to this, the preclinical studies and future perspectives are also discussed in this review.


Subject(s)
Neoplasms , Xanthophylls , Humans , Xanthophylls/chemistry , Xanthophylls/pharmacology , Xanthophylls/therapeutic use , Neoplasms/prevention & control , Neoplasms/drug therapy , Animals , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Anticarcinogenic Agents/chemistry
17.
Cancer Metastasis Rev ; 43(1): 55-85, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37507626

ABSTRACT

Despite tremendous medical treatment successes, colorectal cancer (CRC) remains a leading cause of cancer deaths worldwide. Chemotherapy as monotherapy can lead to significant side effects and chemoresistance that can be linked to several resistance-activating biological processes, including an increase in inflammation, cellular plasticity, multidrug resistance (MDR), inhibition of the sentinel gene p53, and apoptosis. As a consequence, tumor cells can escape the effectiveness of chemotherapeutic agents. This underscores the need for cross-target therapeutic approaches that are not only pharmacologically safe but also modulate multiple potent signaling pathways and sensitize cancer cells to overcome resistance to standard drugs. In recent years, scientists have been searching for natural compounds that can be used as chemosensitizers in addition to conventional medications for the synergistic treatment of CRC. Resveratrol, a natural polyphenolic phytoalexin found in various fruits and vegetables such as peanuts, berries, and red grapes, is one of the most effective natural chemopreventive agents. Abundant in vitro and in vivo studies have shown that resveratrol, in interaction with standard drugs, is an effective chemosensitizer for CRC cells to chemotherapeutic agents and thus prevents drug resistance by modulating multiple pathways, including transcription factors, epithelial-to-mesenchymal transition-plasticity, proliferation, metastasis, angiogenesis, cell cycle, and apoptosis. The ability of resveratrol to modify multiple subcellular pathways that may suppress cancer cell plasticity and reversal of chemoresistance are critical parameters for understanding its anti-cancer effects. In this review, we focus on the chemosensitizing properties of resveratrol in CRC and, thus, its potential importance as an additive to ongoing treatments.


Subject(s)
Anticarcinogenic Agents , Colorectal Neoplasms , Stilbenes , Humans , Resveratrol/pharmacology , Resveratrol/therapeutic use , Signal Transduction , Transcription Factors , Anticarcinogenic Agents/pharmacology , Colorectal Neoplasms/pathology , Stilbenes/pharmacology , Stilbenes/therapeutic use
18.
Int J Biol Macromol ; 254(Pt 1): 127718, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37918594

ABSTRACT

In this study, the nano-assemblies of bovine casein hydrolyzed peptides (HP) modified by fatty acids with various alkyl chain lengths (C8, C10, C12 and C14) were synthesized. The physicochemical properties of HP-C8-HP-C14 nano-assemblies were characterized using spectra, laser particle size analyzer, contact angle meter, scanning electron microscope (SEM) and cryo-transmission electron microscope (Cryo-TEM). HP-C8 and HP-C10 self-assembled into a hollow cube cage with an average size of ~500 nm, and the assembly of HP-C12 showed a flower-shaped morphology with more dispersed behavior, and droplet size was observed as ~20 nm. The in vitro cytotoxicity against human breast cancer cells MCF-7 was tested using CCK-8 assay and flow cytometry analysis. HP-C12 showed the highest cytotoxicity for MCF-7 cells with an inhibition rate of 66.03 % ± 0.35 % with an IC50 value of 7.4 µM among HP-Cn. HP-C8, HP-C10 and HP-C12 significantly affected on the migration, invasion and apoptosis of MCF-7 cells. The apoptosis mechanism may depend on the upregulation of anti-apoptotic protein Bcl-2 as well as pro-apoptotic proteins Bax and caspase-8. The dead MCF-7 cells were analyzed with UHPLC-MS/MS using untargeted metabolomics, revealing key metabolic pathways.


Subject(s)
Anticarcinogenic Agents , Fatty Acids , Animals , Cattle , Humans , Fatty Acids/chemistry , Anticarcinogenic Agents/pharmacology , Caseins/pharmacology , Tandem Mass Spectrometry , Apoptosis , MCF-7 Cells
19.
Phytother Res ; 38(1): 331-348, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37882581

ABSTRACT

The isothiocyanates (ITCs) derived from the precursor glucosinolate molecules present in Brassica vegetables are bioactive organo-sulfur compounds with numerous pharmacologically important properties such as antioxidant, antiinflammatory, antimicrobial, and anticancer. Over the years, ITCs have been the focus of several research investigations associated with cancer treatment. Due to their potent chemo-preventive action, ITCs have been considered to be promising therapeutics for cancer therapy in place of the already existing conventional anticancer drugs. However, their wide spread use at the clinical stage is greatly restricted due to several factors such as low solubility in an aqueous medium, low bioavailability, low stability, and hormetic effect. To overcome these hindrances, nanotechnology can be exploited to develop nano-scale delivery systems that have the potential to enhance stability, and bioavailability and minimize the hermetic effect of ITCs.


Subject(s)
Anticarcinogenic Agents , Antineoplastic Agents , Brassica , Isothiocyanates/pharmacology , Vegetables , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/pharmacology
20.
Nutrients ; 15(22)2023 Nov 17.
Article in English | MEDLINE | ID: mdl-38004214

ABSTRACT

Okra flowers are a good source of polysaccharides and flavonoids, with biological activities of anti-inflammatory action and modulation of the gut microbiota. Previously, we reported that flavonoid-rich extracts from okra flowers (AFE) presented effective anti-colorectal cancer (CRC) activity in CRC cells as well as xenograft models, but their role in colitis-associated cancer (CAC) is unidentified. In this study, we aimed to evaluate the effects of AFE and APE (polysaccharides extracted from okra flowers) on the CAC symptoms of azoxymethane (AOM)/dextran sodium sulfate (DSS)-intervened mice. The results showed that APE and AFE exert potent efficacy in inhibiting colitis and colorectal tumorigenesis stimulated by AOM/DSS, characterized by decreased colonic shortening, DAI score, and tumor numbers. Compared with the control group, APE/AFE alleviated the microbiota dysbiosis driven by AOM/DSS. In addition, AFE elicited its anticancer activity through regulation of NFκB/IL-6/Stat3, JAK2/Stat3, MAPKs, PI3K/AKT, and Wnt/ß-catenin signal transductions in AOM/DSS mice, which was consistent with a vitro model of CT26 cells, while APE treatment exhibited anticancer activity through regulation of Nrf2/IL-6, MAPKs, PI3K/AKT, and Wnt/ß-catenin signal transductions in the AOM/DSS mouse model. Collectively, our studies revealed, for the first time, that flavonoids and polysaccharides from okra flowers possess the ability to attenuate colitis and colorectal tumorigenesis, with them having great potential to become promising candidates against CRC.


Subject(s)
Abelmoschus , Anticarcinogenic Agents , Colitis-Associated Neoplasms , Colitis , Colorectal Neoplasms , Hominidae , Humans , Mice , Animals , Flavonoids/adverse effects , Dextran Sulfate/adverse effects , Interleukin-6 , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , beta Catenin , Colitis/chemically induced , Colitis/complications , Colitis/drug therapy , Azoxymethane , Carcinogenesis , Cell Transformation, Neoplastic , Anticarcinogenic Agents/pharmacology , Disease Models, Animal , Mice, Inbred C57BL , Colorectal Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL