1
|
Adegoke A, Oduola AB, Idowu K, Abiona J. Fatty acid composition, lipid profile and oxidative stability of meat of broiler chickens fed diet containing bird eye pepper of varying inclusion level and sieve size. Trop Anim Health Prod 2024; 56:342. [PMID: 39400759 DOI: 10.1007/s11250-024-04185-7] [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: 03/07/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Nutritional modifications to improve meat quality is targeted by farmers. Bird eye pepper (BEP) contains bio-compounds of physiological significance. The potency of BEP of varying inclusion level and sieve size on meat quality [fatty acid (FA), lipid profile and oxidative stability] of broiler chickens was investigated. A total of 246 birds fed diet-containing BEP [inclusion level (0, 0.15 and 0.3%), sieve size (0.05 and 0.1 mm)] were randomized to six treatments replicated 4 times in a 2 by 3 factorial layout. After feeding (31 days), forty-eight birds (two per replicate) were slaughtered and breast muscles harvested. Meat lipid profile and 2-thiobarbituric acid reactive substance (TBARs) were determined on day (d) 0, while TBARs was further assessed on d 3 and 5, but FA on d 10 of refrigeration storage. BEP diet (0.15%) increased (p < 0.05) total monounsaturated FA (MUFA), unsaturated FA (UFA) and n-3 FA, while 0.05 mm BEP lowered (p < 0.05) meat index of thrombogenicity (TI) but increased meat hypocholesteromic: hypercholesteromic (HH) value. Dietary 0.15% (0.05 mm) BEP yielded low (p < 0.05) SFA but high MUFA: SFA, UFA: SFA and NVI, while 0.15% (0.1 mm) BEP diet resulted in high total MUFA and higher (p < 0.05) UFA, n-3 and n-3: n-6 FA. Control, 0.15% and 0.05 mm BEP diets reduced (p < 0.05) meat cholesterol value. This study has shown that 0.15% (0.05 mm) BEP diet had no deleterious effect on the growth of broiler chickens but improved the NVI, IA, TI, HH, TBARs and cholesterol of the meat - a significance to health-conscious consumers.
Collapse
Affiliation(s)
- Adeola Adegoke
- Department of Animal Production and Health, Federal University of Agriculture, P.M.B. 2240, Abeokuta, Ogun State, Nigeria.
| | - Abdul-Basit Oduola
- Department of Animal Production and Health, Federal University of Agriculture, P.M.B. 2240, Abeokuta, Ogun State, Nigeria
| | - Kemi Idowu
- Department of Animal Production and Health, Federal University of Agriculture, P.M.B. 2240, Abeokuta, Ogun State, Nigeria
| | - John Abiona
- Department of Animal Physiology, Federal University of Agriculture, P.M.B. 2240, Abeokuta, Ogun State, Nigeria
| |
Collapse
|
2
|
Grunt TW, Wagner R, Ries A, Berghoff AS, Preusser M, Grusch M, Valent P. Targeting endogenous fatty acid synthesis stimulates the migration of ovarian cancer cells to adipocytes and promotes the transport of fatty acids from adipocytes to cancer cells. Int J Oncol 2024; 64:24. [PMID: 38214315 PMCID: PMC10807641 DOI: 10.3892/ijo.2024.5612] [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: 06/09/2023] [Accepted: 12/08/2023] [Indexed: 01/13/2024] Open
Abstract
Despite significant advances in oncology, 1 of 108 female patients succumb to ovarian cancer (OC) each year. Improved novel treatments against this aggressive disease would be a major improvement. The growth of OC cells has been demonstrated to be highly dependent on lipids. OC cells are abundantly present in the abdominal cavity and omentum, the main sites of OC expansion. Accordingly, it has been attempted not only to block the hyperactive synthesis of fatty acids (FAs) in cancer cells, but also to disrupt lipid supply. While either strategy has yielded promising results as monotherapy, the induction of resistance pathways diminishing the anticancer effects is yet conceivable. The endogenous regulation of lipid biosynthesis in OC has been extensively studied. However, the role of stromal cells in the modulation of the effects of anti‑lipogenic drugs has not yet been well documented. The present study thus examined the interaction between OC cells and associated stromal cells, when de novo FA synthesis was blocked. It has recently been revealed by the authors that when FA are provided to OC cells in monoculture, the lipid deficiency induced by pharmacological inhibition of FA synthase (FASN), the key enzyme of endogenous FA synthesis, cannot be compensated through an increased FA uptake by OC cells. In the present study, OC cells were co‑cultured with adipocytes preloaded with fluorescent FA and the effects of FASN‑inhibition on OC homing to adipocytes and the transcellular delivery of fluorescent FA from adipocytes to OC cells were examined. The FASN inhibitors, G28UCM and Fasnall, stimulated the spontaneous migration of A2780 OC cells in a concentration‑dependent manner and stimulated the transfer of FA from adipocytes to OC cells. Similar effects were observed with all types of adipocytes tested. The models applied in the present study demonstrated that co‑cultured cancer‑associated adipocytes may attenuate the anticancer effects of FASN inhibitors by attracting tumor cells and by supplying the cells with FA. This lipid‑mediated dependency may provide a rationale for the design of new treatment approaches for the treatment of OC.
Collapse
Affiliation(s)
- Thomas W. Grunt
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Renate Wagner
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Alexander Ries
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - Anna S. Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, A-1090 Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, A-1090 Vienna, Austria
| | - Michael Grusch
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - Peter Valent
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, A-1090 Vienna, Austria
- Division of Hematology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
3
|
Thapliyal A, Tomar AK, Chandra KB, Naglot S, Dhiman S, Singh N, Sharma JB, Yadav S. Differential Sperm Proteomics Reveals the Significance of Fatty Acid Synthase and Clusterin in Idiopathic Recurrent Pregnancy Loss. Reprod Sci 2023; 30:3456-3468. [PMID: 37378824 DOI: 10.1007/s43032-023-01288-8] [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: 04/17/2023] [Accepted: 06/22/2023] [Indexed: 06/29/2023]
Abstract
Recurrent pregnancy loss (RPL) is a pervasive health issue affecting a large number of couples globally, which leads to increased emotional and financial strain on the affected families. While female factors have been extensively studied and are well known, the contribution of male factors to RPL remains largely unknown. As high as 40% of RPL cases are unexplained, which are termed as idiopathic RPL (iRPL), necessitating the investigation of male factors. The role of spermatozoa in early embryonic development is now well established, and recent research studies have shown that oxidative stress and DNA fragmentation in sperm cells are linked to RPL. The aim of this study was to identify proteomic markers of iRPL in human spermatozoa using tandem mass spectrometry. A label-free method quantified a total of 1820 proteins, and statistical analysis identified 359 differentially expressed proteins, the majority of which were downregulated in iRPL samples (344). Bioinformatics analysis revealed that proteomic alterations were mainly associated with biological processes such as response to stress, protein folding, chromatin organization, DNA conformation change, oxidative phosphorylation, and electron transport chain. In coherence with past studies, we determined fatty acid synthase (FASN) and clusterin (CLU) to be the most potential sperm markers for iRPL and confirmed their expression changes in iRPL by western blotting. Conclusively, we believe that FASN and CLU might serve as potential markers of iRPL and suggest exploratory functional studies to identify their specific role in pregnancy loss.
Collapse
Affiliation(s)
- Ayushi Thapliyal
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Anil Kumar Tomar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Kumari Binita Chandra
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sarla Naglot
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
- Medical Device and Diagnostics Mission Secretariat (MDMS), ICMR, New Delhi, 110029, India
| | - Soniya Dhiman
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Neeta Singh
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Jai Bhagwan Sharma
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Savita Yadav
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
4
|
Luján-Méndez F, Roldán-Padrón O, Castro-Ruíz JE, López-Martínez J, García-Gasca T. Capsaicinoids and Their Effects on Cancer: The "Double-Edged Sword" Postulate from the Molecular Scale. Cells 2023; 12:2573. [PMID: 37947651 PMCID: PMC10650825 DOI: 10.3390/cells12212573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
Capsaicinoids are a unique chemical species resulting from a particular biosynthesis pathway of hot chilies (Capsicum spp.) that gives rise to 22 analogous compounds, all of which are TRPV1 agonists and, therefore, responsible for the pungency of Capsicum fruits. In addition to their human consumption, numerous ethnopharmacological uses of chili have emerged throughout history. Today, more than 25 years of basic research accredit a multifaceted bioactivity mainly to capsaicin, highlighting its antitumor properties mediated by cytotoxicity and immunological adjuvancy against at least 74 varieties of cancer, while non-cancer cells tend to have greater tolerance. However, despite the progress regarding the understanding of its mechanisms of action, the benefit and safety of capsaicinoids' pharmacological use remain subjects of discussion, since CAP also promotes epithelial-mesenchymal transition, in an ambivalence that has been referred to as "the double-edge sword". Here, we update the comparative discussion of relevant reports about capsaicinoids' bioactivity in a plethora of experimental models of cancer in terms of selectivity, efficacy, and safety. Through an integration of the underlying mechanisms, as well as inherent aspects of cancer biology, we propose mechanistic models regarding the dichotomy of their effects. Finally, we discuss a selection of in vivo evidence concerning capsaicinoids' immunomodulatory properties against cancer.
Collapse
Affiliation(s)
- Francisco Luján-Méndez
- Laboratorio de Biología Celular y Molecular, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. De las Ciencias s/n, Juriquilla, Querétaro 76230, Querétaro, Mexico; (F.L.-M.); (O.R.-P.); (J.L.-M.)
| | - Octavio Roldán-Padrón
- Laboratorio de Biología Celular y Molecular, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. De las Ciencias s/n, Juriquilla, Querétaro 76230, Querétaro, Mexico; (F.L.-M.); (O.R.-P.); (J.L.-M.)
| | - J. Eduardo Castro-Ruíz
- Escuela de Odontología, Facultad de Medicina, Universidad Autónoma de Querétaro, Querétaro 76176, Querétaro, Mexico;
| | - Josué López-Martínez
- Laboratorio de Biología Celular y Molecular, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. De las Ciencias s/n, Juriquilla, Querétaro 76230, Querétaro, Mexico; (F.L.-M.); (O.R.-P.); (J.L.-M.)
| | - Teresa García-Gasca
- Laboratorio de Biología Celular y Molecular, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. De las Ciencias s/n, Juriquilla, Querétaro 76230, Querétaro, Mexico; (F.L.-M.); (O.R.-P.); (J.L.-M.)
| |
Collapse
|
5
|
Sun Z, Wu K, Feng C, Lei XG. Selenium-dependent glutathione peroxidase 1 regulates transcription of elongase 3 in murine tissues. Free Radic Biol Med 2023; 208:708-717. [PMID: 37726091 DOI: 10.1016/j.freeradbiomed.2023.09.010] [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: 08/03/2023] [Revised: 08/26/2023] [Accepted: 09/11/2023] [Indexed: 09/21/2023]
Abstract
We have previously shown dysregulated lipid metabolism in tissues of glutathione peroxidase 1 (GPX1) overexpressing (OE) or deficient (KO) mice. This study explored underlying mechanisms of GPX1 in regulating tissue fatty acid (FA) biosynthesis. GPX1 OE, KO, and wild-type (WT) mice (n = 5, male, 3-6 months old) were fed a Se-adequate diet (0.3 mg/kg) and assayed for liver and adipose tissue FA profiles and mRNA levels of key enzymes of FA biosynthesis and redox-responsive transcriptional factors (TFs). These three genotypes of mice (n = 5) were injected intraperitoneally with diquat, ebselen, and N-acetylcysteine (NAC) at 10, 50, and 50 mg/kg of body weight, respectively, and killed at 0 and 12 h after the injections to detect mRNA levels of FA elongases and desaturases and the TFs in the liver and adipose tissue. A luciferase reporter assay with targeted deletions of mouse Elovl3 promoter was performed to determine transcriptional regulations of the gene by GPX1 mimic ebselen in HEK293T cells. Compared with WT, GPX1 OE and KO mice had 9-42% lower (p < 0.05) and 36-161% higher (p < 0.05) concentrations of C20:0, C22:0, and C24:0 in these two tissues, respectively, along with reciprocal increases and decreases (p < 0.05) of Elovl3 transcripts. Ebselen and NAC decreased (p < 0.05), whereas diquat decreased (p < 0.05), Elovl3 transcripts in the two tissues. Overexpression and knockout of GPX1 decreased (p < 0.05) and increased (p < 0.05) ELOVL3 levels in the two tissues, respectively. Three TFs (GABP, SP1, and DBP) were identified to bind the Elovl3 promoter (-1164/+33 base pairs). Deletion of DBP (-98/-86 base pairs) binding domain in the promoter attenuated (13%, p < 0.05) inhibition of ebselen on Elovl3 promoter activation. In summary, GPX1 overexpression down-regulated very long-chain FA biosynthesis via transcriptional inhibition of the Elovl3 promoter activation.
Collapse
Affiliation(s)
- Ziqiao Sun
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
| | - Kun Wu
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
| | - Chenhan Feng
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
| | - Xin Gen Lei
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
6
|
Muangpat P, Meesil W, Ngoenkam J, Teethaisong Y, Thummeepak R, Sitthisak S, Tandhavanant S, Chantratita N, Bode HB, Vitta A, Thanwisai A. Genome analysis of secondary metabolite‑biosynthetic gene clusters of Photorhabdus akhurstii subsp. akhurstii and its antibacterial activity against antibiotic-resistant bacteria. PLoS One 2022; 17:e0274956. [PMID: 36129957 PMCID: PMC9491552 DOI: 10.1371/journal.pone.0274956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/08/2022] [Indexed: 11/27/2022] Open
Abstract
Xenorhabdus and Photorhabdus can produce a variety of secondary metabolites with broad spectrum bioactivity against microorganisms. We investigated the antibacterial activity of Xenorhabdus and Photorhabdus against 15 antibiotic-resistant bacteria strains. Photorhabdus extracts had strong inhibitory the growth of Methicillin-resistant Staphylococcus aureus (MRSA) by disk diffusion. The P. akhurstii s subsp. akhurstii (bNN168.5_TH) extract showed lower minimum inhibitory concentrations (MIC) and minimal bactericidal concentrations (MBC). The interaction between either P. akhurstii subsp. akhurstii (bNN141.3_TH) or P. akhurstii subsp. akhurstii (bNN168.5_TH) or P. hainanensis (bNN163.3_TH) extract in combination with oxacillin determined by checkerboard assay exhibited partially synergistic interaction with fractional inhibitory concentration index (FICI) of 0.53. Time-killing assay for P. akhurstii subsp. akhurstii (bNN168.5_TH) extract against S. aureus strain PB36 significantly decreased cell viability from 105 CFU/ml to 103 CFU/ml within 30 min (P < 0.001, t-test). Transmission electron microscopic investigation elucidated that the bNN168.5_TH extract caused treated S. aureus strain PB36 (MRSA) cell membrane damage. The biosynthetic gene clusters of the bNN168.5_TH contained non-ribosomal peptide synthetase cluster (NRPS), hybrid NRPS-type l polyketide synthase (PKS) and siderophore, which identified potentially interesting bioactive products: xenematide, luminmide, xenortide A-D, luminmycin A, putrebactin/avaroferrin and rhizomide A-C. This study demonstrates that bNN168.5_TH showed antibacterial activity by disrupting bacterial cytoplasmic membrane and the draft genome provided insights into the classes of bioactive products. This also provides a potential approach in developing a novel antibacterial agent.
Collapse
Affiliation(s)
- Paramaporn Muangpat
- Faculty of Medical Science, Department of Microbiology and Parasitology, Naresuan University, Phitsanulok, Thailand
| | - Wipanee Meesil
- Faculty of Medical Science, Department of Microbiology and Parasitology, Naresuan University, Phitsanulok, Thailand
| | - Jatuporn Ngoenkam
- Faculty of Medical Science, Department of Microbiology and Parasitology, Naresuan University, Phitsanulok, Thailand
| | - Yothin Teethaisong
- Faculty of Allied Health Sciences, Department of Biomedical Sciences, Burapha University, Chonburi, Thailand
- Research Unit for Sensor Inovation (RUSI), Burapha University, Chon Buri, Thailand
| | - Rapee Thummeepak
- Faculty of Medical Science, Department of Microbiology and Parasitology, Naresuan University, Phitsanulok, Thailand
| | - Sutthirat Sitthisak
- Faculty of Medical Science, Department of Microbiology and Parasitology, Naresuan University, Phitsanulok, Thailand
| | - Sarunporn Tandhavanant
- Faculty of Tropical Medicine, Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand
| | - Narisara Chantratita
- Faculty of Tropical Medicine, Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand
| | - Helge B. Bode
- Molekulare Biotechnologie, Goethe Universität Frankfurt, Frankfurt am Main, Germany
- Department of Natural Products in Organismic Interactions, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Apichat Vitta
- Faculty of Medical Science, Department of Microbiology and Parasitology, Naresuan University, Phitsanulok, Thailand
- Faculty of Sciences, Center of Excellence for Biodiversity, Naresuan University, Phitsanulok, Thailand
- Faculty of Medical Science, Centre of Excellence in Medical Biotechnology (CEMB), Naresuan University, Phitsanulok, Thailand
| | - Aunchalee Thanwisai
- Faculty of Medical Science, Department of Microbiology and Parasitology, Naresuan University, Phitsanulok, Thailand
- Faculty of Sciences, Center of Excellence for Biodiversity, Naresuan University, Phitsanulok, Thailand
- Faculty of Medical Science, Centre of Excellence in Medical Biotechnology (CEMB), Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
7
|
Khan A, Siddiqui S, Husain SA, Mazurek S, Iqbal MA. Phytocompounds Targeting Metabolic Reprogramming in Cancer: An Assessment of Role, Mechanisms, Pathways, and Therapeutic Relevance. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:6897-6928. [PMID: 34133161 DOI: 10.1021/acs.jafc.1c01173] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The metabolism of cancer is remarkably different from that of normal cells and confers a variety of benefits, including the promotion of other cancer hallmarks. As the rewired metabolism is a near-universal property of cancer cells, efforts are underway to exploit metabolic vulnerabilities for therapeutic benefits. In the continued search for safer and effective ways of cancer treatment, structurally diverse plant-based compounds have gained substantial attention. Here, we present an extensive assessment of the role of phytocompounds in modulating cancer metabolism and attempt to make a case for the use of plant-based compounds in targeting metabolic vulnerabilities of cancer. We discuss the pharmacological interactions of phytocompounds with major metabolic pathways and evaluate the role of phytocompounds in the regulation of growth signaling and transcriptional programs involved in the metabolic transformation of cancer. Lastly, we examine the potential of these compounds in the clinical management of cancer along with limitations and challenges.
Collapse
Affiliation(s)
- Asifa Khan
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Shumaila Siddiqui
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Syed Akhtar Husain
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Sybille Mazurek
- Institute of Veterinary-Physiology and Biochemistry, University of Giessen, Giessen 35392, Germany
| | - Mohammad Askandar Iqbal
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| |
Collapse
|
8
|
Najafi Dorcheh S, Rahgozar S, Talei D. 6-Shogaol induces apoptosis in acute lymphoblastic leukaemia cells by targeting p53 signalling pathway and generation of reactive oxygen species. J Cell Mol Med 2021; 25:6148-6160. [PMID: 33939282 PMCID: PMC8406487 DOI: 10.1111/jcmm.16528] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 12/22/2022] Open
Abstract
Combination therapies, using medicinal herbs, are broadly recommended to attenuate the chemotherapy adverse effects. Based on our previous findings considering the anti-leukaemic effects of ginger extract on acute lymphoblastic leukaemia (ALL) cells, the present study was aimed to investigate the anti-cancer role of this pharmaceutical plant on ALL mice models. Moreover, we worked towards identifying the most anti-leukaemic derivative of ginger and the mechanism through which it may exert its cytotoxic impact. In vivo experiments were performed using five groups of six C57BL/6 nude mice, and the anti-leukaemic activity of ginger extract alone or in combination with methotrexate (MTX) was examined. Results showed increased survival rate and reduced damages in mice brain and liver tissues. Subsequently, MTT assay demonstrated synergistic growth inhibitory effect of 6-shogaol (6Sh) and MTX on ALL cell lines and patients primary cells. Eventually, the molecular anti-neoplastic mechanism of 6Sh was evaluated using Bioinformatics. Flow cytometry illustrated 6Sh-mediated apoptosis in Nalm-6 cells confirmed by Western blotting and RT-PCR assays. Further analyses exhibited the generation of reactive oxygen species (ROS) through 6Sh. The current study revealed the in vivo novel anti-leukaemic role of ginger extract, promoted by MTX. Moreover, 6-shogaol was introduced as the major player of ginger cytotoxicity through inducing p53 activity and ROS generation.
Collapse
Affiliation(s)
| | | | - Daryush Talei
- Medicinal Plants Research CenterShahed UniversityTehranIran
| |
Collapse
|
9
|
Li R, Lan Y, Chen C, Cao Y, Huang Q, Ho CT, Lu M. Anti-obesity effects of capsaicin and the underlying mechanisms: a review. Food Funct 2020; 11:7356-7370. [PMID: 32820787 DOI: 10.1039/d0fo01467b] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Obesity and overweight have become serious health problems in the world and are linked to a variety of metabolic disorders. Phytochemicals with a weight-loss effect have been widely studied for the past few decades. Capsaicin is the major bioactive component in red chili peppers with many beneficial functions. Its anti-obesity effects have been evaluated extensively using different model systems, including cell models, animal models and human subjects. In this paper, anti-obesity effects of capsaicin are reviewed and the underlying mechanisms are characterized.
Collapse
Affiliation(s)
- Run Li
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Yaqi Lan
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Chengyu Chen
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Qingrong Huang
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA.
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA.
| | - Muwen Lu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
10
|
A review on anti-cancer properties of Quercetin in breast cancer. Life Sci 2020; 248:117463. [DOI: 10.1016/j.lfs.2020.117463] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/10/2020] [Accepted: 02/21/2020] [Indexed: 12/19/2022]
|
11
|
Ahmad R, Khan MA, Srivastava A, Gupta A, Srivastava A, Jafri TR, Siddiqui Z, Chaubey S, Khan T, Srivastava AK. Anticancer Potential of Dietary Natural Products: A Comprehensive Review. Anticancer Agents Med Chem 2020; 20:122-236. [DOI: 10.2174/1871520619666191015103712] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 06/21/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
Abstract
Nature is a rich source of natural drug-like compounds with minimal side effects. Phytochemicals
better known as “Natural Products” are found abundantly in a number of plants. Since time immemorial, spices
have been widely used in Indian cuisine as flavoring and coloring agents. Most of these spices and condiments
are derived from various biodiversity hotspots in India (which contribute 75% of global spice production) and
form the crux of India’s multidiverse and multicultural cuisine. Apart from their aroma, flavor and taste, these
spices and condiments are known to possess several medicinal properties also. Most of these spices are mentioned
in the Ayurveda, the indigenous system of medicine. The antimicrobial, antioxidant, antiproliferative,
antihypertensive and antidiabetic properties of several of these natural products are well documented in
Ayurveda. These phytoconstituemts are known to act as functional immunoboosters, immunomodulators as well
as anti-inflammatory agents. As anticancer agents, their mechanistic action involves cancer cell death via induction
of apoptosis, necrosis and autophagy. The present review provides a comprehensive and collective update
on the potential of 66 commonly used spices as well as their bioactive constituents as anticancer agents. The
review also provides an in-depth update of all major in vitro, in vivo, clinical and pharmacological studies done
on these spices with special emphasis on the potential of these spices and their bioactive constituents as potential
functional foods for prevention, treatment and management of cancer.
Collapse
Affiliation(s)
- Rumana Ahmad
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Mohsin A. Khan
- Chancellor, Era University, Sarfarazganj, Hardoi Road, Lucknow-226003, UP, India
| | - A.N. Srivastava
- Department of Pathology, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Anamika Gupta
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Aditi Srivastava
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Tanvir R. Jafri
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Zainab Siddiqui
- Department of Pathology, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Sunaina Chaubey
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Tahmeena Khan
- Department of Chemistry, Integral University, Dasauli, P.O. Bas-ha, Kursi Road, Lucknow 226026, UP, India
| | - Arvind K. Srivastava
- Department of Food and Nutrition, Era University, Sarfarazganj, Lucknow-226003, UP, India
| |
Collapse
|
12
|
Fakhri S, Moradi SZ, Farzaei MH, Bishayee A. Modulation of dysregulated cancer metabolism by plant secondary metabolites: A mechanistic review. Semin Cancer Biol 2020; 80:276-305. [PMID: 32081639 DOI: 10.1016/j.semcancer.2020.02.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/08/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Abstract
Several signaling pathways and basic metabolites are responsible for the control of metabolism in both normal and cancer cells. As emerging hallmarks of cancer metabolism, the abnormal activities of these pathways are of the most noticeable events in cancer. This altered metabolism expedites the survival and proliferation of cancer cells, which have attracted a substantial amount of interest in cancer metabolism. Nowadays, targeting metabolism and cross-linked signaling pathways in cancer has been a hot topic to investigate novel drugs against cancer. Despite the efficiency of conventional drugs in cancer therapy, their associated toxicity, resistance, and high-cost cause limitations in their application. Besides, considering the numerous signaling pathways cross-linked with cancer metabolism, discovery, and development of multi-targeted and safe natural compounds has been a high priority. Natural secondary metabolites have exhibited promising anticancer effects by targeting dysregulated signaling pathways linked to cancer metabolism. The present review reveals the metabolism and cross-linked dysregulated signaling pathways in cancer. The promising therapeutic targets in cancer, as well as the critical role of natural secondary metabolites for significant anticancer enhancements, have also been highlighted to find novel/potential therapeutic agents for cancer treatment.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
13
|
Fu J, Tan YXR, Gong Z, Bae S. The toxic effect of triclosan and methyl-triclosan on biological pathways revealed by metabolomics and gene expression in zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 189:110039. [PMID: 31830605 DOI: 10.1016/j.ecoenv.2019.110039] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/17/2019] [Accepted: 11/30/2019] [Indexed: 05/23/2023]
Abstract
The omnipresence of antimicrobial triclosan (TCS) and by-products in aquatic environments is a threat to aquatic organisms. Traditionally, the adverse effects of TCS and its by-products have been evaluated by examining the phenotypic output relevant to predicting acute toxicity rather than studying the perturbation of biological pathways. Identifying alterations in the key pathways and molecular mechanisms caused by toxic chemicals helps researchers assess the ecological risks of TCS and its by-products to aquatic environments. In this study, we used metabolomics and reverse transcription qPCR to investigate the adverse effects of a wide range of concentrations of triclosan and its derivative methyl-triclosan (MTCS), ranging from relatively low environmentally relevant levels (ng/L) to high-dose concentrations (sublethal concentration), on zebrafish (Danio rerio) embryos. The metabolism and transcriptome analysis revealed changes in the metabolite and transcripts expression of zebrafish embryos after 96 h exposure at 30 μg/L and 300 μg/L of TCS, 400 μg/L of MTCS and the TCS/MTCS mixture (30 μg/L TCS + 3 μg/L MTCS and 300 μg/L TCS + 30 μg/L MTCS). Significant dysregulations in the expression of the urea transporter (UT), glucose-6-phosphate dehydrogenase (G6PD), alanine transaminase (ALT), glutamate dehydrogenase (GDH), phosphoglucomutase (PGM), and fatty acid synthase (FASN), together with changes in alanine, urea, glucose, 6-phosphogluconalactone, and palmitic acid were observed in the TCS, MTCS, and TCS/MTCS treatments. Particularly, the MTCS treatment group showed fold changes in the mRNA expression of nitrogen metabolism, energy metabolism, and fatty acid synthesis, indicating a disruption of the zebrafish embryos' biological pathways. The changes in the metabolites and gene expressions induced by the TCS, MTCS and the TCS/MTCS mixture treatment demonstrate the pathway changes in starch and sucrose metabolism, nitrogen metabolism, fatty acid synthesis, and phenylalanine, tyrosine and tryptophan biosynthesis. Therefore, our study provides better insights into the risks of the parental compound (TCS) and its by-product (MTCS), as well as the perturbation in biological pathways induced by these two compounds in aquatic environments.
Collapse
Affiliation(s)
- Jing Fu
- Department of Civil and Environmental Engineering, National University of Singapore, Singapore
| | - Yue Xuan Rochelle Tan
- Department of Civil and Environmental Engineering, National University of Singapore, Singapore
| | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Sungwoo Bae
- Department of Civil and Environmental Engineering, National University of Singapore, Singapore.
| |
Collapse
|
14
|
Zhang Z, Liu S, Ma H, Nie D, Wen F, Zhao J, Sun A, Yuan G, Su S, Xiang X, Hu P, Tang G. Validation of R-2-[18F]Fluoropropionic Acid as a Potential Tracer for PET Imaging of Liver Cancer. Mol Imaging Biol 2019; 21:1127-1137. [PMID: 30847820 DOI: 10.1007/s11307-019-01346-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
15
|
Maruca A, Catalano R, Bagetta D, Mesiti F, Ambrosio FA, Romeo I, Moraca F, Rocca R, Ortuso F, Artese A, Costa G, Alcaro S, Lupia A. The Mediterranean Diet as source of bioactive compounds with multi-targeting anti-cancer profile. Eur J Med Chem 2019; 181:111579. [PMID: 31398616 DOI: 10.1016/j.ejmech.2019.111579] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 12/20/2022]
Abstract
Many bioactive agents have been extracted from plants or belong to functional foods and have been considered in the treatment of serious and multifactorial diseases, such as cancer. In particular, this review is focused on the anti-cancer properties owned by several natural products typically from the Mediterranean area. In some regions of the South of Italy, a lower cancer incidence has been observed. There is increasing evidence that adherence to a Mediterranean dietary pattern correlates with reduced risk of several cancer types. This could be mainly attributed to the typical lifestyle aspects of the Mediterranean diet, such as high consumption of fruit and vegetables. In this review, the main natural products of the Mediterranean area are discussed, with particular attention on their anti-cancer properties endowed with multi-target profiles.
Collapse
Affiliation(s)
- Annalisa Maruca
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Raffaella Catalano
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Donatella Bagetta
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Francesco Mesiti
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Francesca Alessandra Ambrosio
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Isabella Romeo
- Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Department of Chemistry and Chemical Technology, University of Calabria, Via Pietro Bucci, 87036, Arcavacata di Rende, Cosenza, Italy
| | - Federica Moraca
- Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, 80131, Naples, Italy
| | - Roberta Rocca
- Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Department of Experimental and Clinical Medicine "Magna Græcia" University, Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy.
| | - Francesco Ortuso
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Anna Artese
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Giosuè Costa
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Antonio Lupia
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| |
Collapse
|
16
|
ROS-Mediated Cancer Cell Killing through Dietary Phytochemicals. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9051542. [PMID: 31217841 PMCID: PMC6536988 DOI: 10.1155/2019/9051542] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) promote carcinogenesis by inducing genetic mutations, activating oncogenes, and raising oxidative stress, which all influence cell proliferation, survival, and apoptosis. Cancer cells display redox imbalance due to increased ROS level compared to normal cells. This unique feature in cancer cells may, therefore, be exploited for targeted therapy. Over the past few decades, natural compounds have attracted attention as potential cancer therapies because of their ability to maintain cellular redox homeostasis with minimal toxicity. Preclinical studies show that bioactive dietary polyphenols exert antitumor effects by inducing ROS-mediated cytotoxicity in cancer cells. These bioactive compounds also regulate cell proliferation, survival, and apoptotic and antiapoptotic signalling pathways. In this review, we discuss (i) how ROS is generated and (ii) regulated and (iii) the cell signalling pathways affected by ROS. We also discuss (iv) the various dietary phytochemicals that have been implicated to have cancer therapeutic effects through their ROS-related functions.
Collapse
|
17
|
Guerra AR, Duarte MF, Duarte IF. Targeting Tumor Metabolism with Plant-Derived Natural Products: Emerging Trends in Cancer Therapy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:10663-10685. [PMID: 30227704 DOI: 10.1021/acs.jafc.8b04104] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Recognition of neoplastic metabolic reprogramming as one of cancer's hallmarks has paved the way for developing novel metabolism-targeted therapeutic approaches. The use of plant-derived natural bioactive compounds for this endeavor is especially promising, due to their diverse structures and multiple targets. Hence, over the past decade, a growing number of studies have assessed the impact of phytochemicals on tumor cell metabolism, aiming at improving current knowledge on their mechanisms of action and, at the same time, evaluating their potential as anti-cancer metabolic modulators. In this Review, we focus on three classes of plant-derived compounds with promising anti-cancer activity-phenolic compounds, isoprenoids, and alkaloids-to describe their effects on major energetic and biosynthetic pathways of human tumor cells. Such a comprehensive and integrated account of the ability of these compounds to hit different metabolic targets is expected to contribute to the rational design and critical assessment of novel anti-cancer therapies based on natural-product-mediated metabolic reprogramming.
Collapse
Affiliation(s)
- Angela R Guerra
- Centro de Biotecnologia Agrícola e Agro-Alimentar do Alentejo (CEBAL), Instituto Politécnico de Beja , Apartado 6158 , 7801-908 Beja , Portugal
- CICECO - Instituto de Materiais de Aveiro, Departamento de Quı́mica , Universidade de Aveiro , Campus de Santiago , 3810-193 Aveiro , Portugal
| | - Maria F Duarte
- Centro de Biotecnologia Agrícola e Agro-Alimentar do Alentejo (CEBAL), Instituto Politécnico de Beja , Apartado 6158 , 7801-908 Beja , Portugal
- ICAAM - Instituto de Ciências Agrárias e Ambientais Mediterrânicas , Universidade de Évora , Pólo da Mitra, 7006-554 Évora , Portugal
| | - Iola F Duarte
- CICECO - Instituto de Materiais de Aveiro, Departamento de Quı́mica , Universidade de Aveiro , Campus de Santiago , 3810-193 Aveiro , Portugal
| |
Collapse
|
18
|
Phokrai P, Poolsri W, Suwankulanan S, Phakdeeto N, Kaewkong W, Pekthong D, Richert L, Srisawang P. Suppressed de novo lipogenesis by plasma membrane citrate transporter inhibitor promotes apoptosis in HepG2 cells. FEBS Open Bio 2018; 8:986-1000. [PMID: 29928578 PMCID: PMC5986055 DOI: 10.1002/2211-5463.12435] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 04/03/2018] [Accepted: 04/18/2018] [Indexed: 01/02/2023] Open
Abstract
Suppression of the expression or activities of enzymes that are involved in the synthesis of de novo lipogenesis (DNL) in cancer cells triggers cell death via apoptosis. The plasma membrane citrate transporter (PMCT) is the initial step that translocates citrate from blood circulation into the cytoplasm for de novo long-chain fatty acids synthesis. This study investigated the antitumor effect of the PMCT inhibitor (PMCTi) in inducing apoptosis by inhibiting the DNL pathway in HepG2 cells. The present findings showed that PMCTi reduced cell viability and enhanced apoptosis through decreased intracellular citrate levels, which consequently caused inhibition of fatty acid and triacylglycerol productions. Thus, as a result of the reduction in fatty acid synthesis, the activity of carnitine palmitoyl transferase-1 (CPT-1) was suppressed. Decreased CPT-1 activity also facilitated the disruption of mitochondrial membrane potential (ΔΨm) leading to stimulation of apoptosis. Surprisingly, primary human hepatocytes were not affected by PMCTi. Increased caspase-8 activity as a consequence of reduction in fatty acid synthesis was also found to cause disruption of ΔΨm. In addition, apoptosis induction by PMCTi was associated with an enhanced reactive oxygen species generation. Taken together, we suggest that inhibition of the DNL pathway following reduction in citrate levels is an important regulator of apoptosis in HepG2 cells via suppression of CPT-1 activity. Thus, targeting the DNL pathway mediating CPT-1 activity by PMCTi may be a selective potential anticancer therapy.
Collapse
Affiliation(s)
- Phornpun Phokrai
- Department of Medical TechnologyFaculty of Science and TechnologyBansomdejchaopraya Rajabhat UniversityBangkokThailand
| | - Wan‐angkan Poolsri
- Department of PhysiologyFaculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
| | - Somrudee Suwankulanan
- Department of PhysiologyFaculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
| | - Narinthorn Phakdeeto
- Department of PhysiologyFaculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
| | - Worasak Kaewkong
- Department of BiochemistryFaculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
| | - Dumrongsak Pekthong
- Department of Pharmacy PracticeFaculty of Pharmaceutical SciencesNaresuan UniversityPhitsanulokThailand
| | | | - Piyarat Srisawang
- Department of PhysiologyFaculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
| |
Collapse
|
19
|
Cui Y, Xing P, Wang Y, Liu M, Qiu L, Ying G, Li B. NADPH accumulation is responsible for apoptosis in breast cancer cells induced by fatty acid synthase inhibition. Oncotarget 2018; 8:32576-32585. [PMID: 28427229 PMCID: PMC5464810 DOI: 10.18632/oncotarget.15936] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/22/2017] [Indexed: 01/09/2023] Open
Abstract
Fatty acid synthase (FAS), as a key enzyme involved in de novo lipogenesis, is highly expressed in many cancers. FAS inhibition induces cell death in vivo and in vitro, rendering FAS as an attractive target for cancer therapy, but the defined mechanism is still not well understood. Herein, we confirmed that FAS was highly expressed in breast cancers and FAS inhibition by its inhibitors or knockdown induced apoptosis in breast cancer cells. Our results showed that a significantly high level of reactive oxygen species was induced but not responsible for apoptosis in breast cancer cells by FAS inhibition. Instead, NADPH accumulation resulting from FAS inhibition was found to stimulate NADPH oxidase to generate reactive oxygen species and highly associated with apoptosis induction. Suppression of NADPH oxidase almost totally blocked reactive oxygen species generation while significantly potentiated the in vitro and in vivo killing of breast cancers by FAS inhibition. Taken together, these data suggest that FAS plays a critical role in maintaining cellular redox homeostasis and its inhibition leads to NADPH accumulation-mediated apoptosis. Our finding may provide new insights into cancer metabolism and aid in designing effective anticancer treatments.
Collapse
Affiliation(s)
- Yanfen Cui
- Laboratory of Cancer Cell Biology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Pan Xing
- Laboratory of Cancer Cell Biology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yuanyuan Wang
- Laboratory of Cancer Cell Biology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Miao Liu
- Laboratory of Cancer Cell Biology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Li Qiu
- Laboratory of Cancer Cell Biology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Guoguang Ying
- Laboratory of Cancer Cell Biology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Binghui Li
- Laboratory of Cancer Cell Biology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| |
Collapse
|
20
|
Khiewkamrop P, Phunsomboon P, Richert L, Pekthong D, Srisawang P. Epistructured catechins, EGCG and EC facilitate apoptosis induction through targeting de novo lipogenesis pathway in HepG2 cells. Cancer Cell Int 2018; 18:46. [PMID: 29588626 PMCID: PMC5863485 DOI: 10.1186/s12935-018-0539-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/13/2018] [Indexed: 02/06/2023] Open
Abstract
Background Abnormally high expression of the mammalian de novo lipogenesis (DNL) pathway in various cancer cells promotes cell over-proliferation and resistance to apoptosis. Inhibition of key enzymes in the DNL pathway, namely, ATP citrate lyase, acetyl-CoA carboxylase, and fatty acid synthase (FASN) can increase apoptosis without cytotoxicity to non-cancerous cells, leading to the search for and presentation of novel selective and powerful targets for cancer therapy. Previous studies reported that epistructured catechins, epigallocatechin gallate (EGCG) and epicatechin (EC) exhibit different mechanisms regarding a strong inducer of apoptosis in various cancer cell lines. Thus, the current study investigated the growth inhibitory effect of EGCG and EC, on the enzyme expression and activity of the DNL pathway, which leads to the prominent activity of carnitine palmitoyl transferase-1 (CPT-1) mediating apoptosis in HepG2 cells. Methods The cytotoxicity on HepG2 cells of EGCG and EC was determined by MTT assay. Cell death caused by apoptosis, the dissipation of mitochondrial membrane potential (MMP), and cell cycle arrest were then detected by flow cytometry. We further investigated the decrease of fatty acid levels associated with DNL retardation, followed by evaluation of DNL protein expression. Then, the negative inhibitory effect of depleted fatty acid synthesis on malonyl-CoA synthesis followed by regulating of CPT-1 activity was investigated. Thereafter, we inspected the enhanced reactive oxygen species (ROS) generation, which is recognized as one of the causes of apoptosis in HepG2 cells. Results We found that EGCG and EC decreased cancer cell viability by increasing apoptosis as well as causing cell cycle arrest in HepG2 cells. Apoptosis was associated with MMP dissipation. Herein, EGCG and EC inhibited the expression of FASN enzymes contributing to decreasing fatty acid levels. Notably, this decrease consequently showed a suppressing effect on the CPT-1 activity. We suggest that epistructured catechin-induced apoptosis targets CPT-1 activity suppression mediated through diminishing the DNL pathway in HepG2 cells. In addition, increased ROS production was found after treatment with EGCG and EC, indicating oxidative stress mechanism-induced apoptosis. The strong apoptotic effect of EGCG and EC was specifically absent in primary human hepatocytes. Conclusion Our supportive evidence confirms potential alternative cancer treatments by EGCG and EC that selectively target the DNL pathway.
Collapse
Affiliation(s)
- Phuriwat Khiewkamrop
- 1Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000 Thailand
| | - Pattamaphron Phunsomboon
- 2Clinical Research Unit Floor 5 His Majesty's 7th Cycle Birthday Anniversary 2, Faculty of Medicine, Naresuan University, Phitsanulok, 65000 Thailand
| | - Lysiane Richert
- KaLy-Cell, 20A rue du Général Leclerc, 67115 Plobsheim, France.,Laboratoire de Toxicologie Cellulaire, Université de Bourgogne Franche-Comté, EA 4267, Besançon, France
| | - Dumrongsak Pekthong
- 5Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, 65000 Thailand
| | - Piyarat Srisawang
- 1Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000 Thailand
| |
Collapse
|
21
|
Poolsri WA, Phokrai P, Suwankulanan S, Phakdeeto N, Phunsomboon P, Pekthong D, Richert L, Pongcharoen S, Srisawang P. Combination of Mitochondrial and Plasma Membrane Citrate Transporter Inhibitors Inhibits De Novo Lipogenesis Pathway and Triggers Apoptosis in Hepatocellular Carcinoma Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3683026. [PMID: 29546056 PMCID: PMC5818947 DOI: 10.1155/2018/3683026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/23/2017] [Accepted: 12/03/2017] [Indexed: 12/27/2022]
Abstract
Increased expression levels of both mitochondrial citrate transporter (CTP) and plasma membrane citrate transporter (PMCT) proteins have been found in various cancers. The transported citrates by these two transporter proteins provide acetyl-CoA precursors for the de novo lipogenesis (DNL) pathway to support a high rate of cancer cell viability and development. Inhibition of the DNL pathway promotes cancer cell apoptosis without apparent cytotoxic to normal cells, leading to the representation of selective and powerful targets for cancer therapy. The present study demonstrates that treatments with CTP inhibitor (CTPi), PMCT inhibitor (PMCTi), and the combination of CTPi and PMCTi resulted in decreased cell viability in two hepatocellular carcinoma cell lines (HepG2 and HuH-7). Treatment with citrate transporter inhibitors caused a greater cytotoxic effect in HepG2 cells than in HuH-7 cells. A lower concentration of combined CTPi and PMCTi promotes cytotoxic effect compared with either of a single compound. An increased cell apoptosis and an induced cell cycle arrest in both cell lines were reported after administration of the combined inhibitors. A combination treatment exhibits an enhanced apoptosis through decreased intracellular citrate levels, which consequently cause inhibition of fatty acid production in HepG2 cells. Apoptosis induction through the mitochondrial-dependent pathway was found as a consequence of suppressed carnitine palmitoyl transferase-1 (CPT-1) activity and enhanced ROS generation by combined CTPi and PMCTi treatment. We showed that accumulation of malonyl-CoA did not correlate with decreasing CPT-1 activity. The present study showed that elevated ROS levels served as an inhibition on Bcl-2 activity that is at least in part responsible for apoptosis. Moreover, inhibition of the citrate transporter is selectively cytotoxic to HepG2 cells but not in primary human hepatocytes, supporting citrate-mediating fatty acid synthesis as a promising cancer therapy.
Collapse
Affiliation(s)
- Wan-angkan Poolsri
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Phornpun Phokrai
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Somrudee Suwankulanan
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Narinthorn Phakdeeto
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | | | - Dumrongsak Pekthong
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Lysiane Richert
- KaLy-Cell, 20A rue du Général Leclerc, 67115 Plobsheim, France
- Laboratoire de Toxicologie Cellulaire, Université de Bourgogne Franche-Comté, EA 4267, Besançon, France
| | - Sutatip Pongcharoen
- Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok 65000, Thailand
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
- Center of Excellence in Petroleum, Petrochemicals and Advanced Materials, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Piyarat Srisawang
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
22
|
Li CF, Fang FM, Chen YY, Liu TT, Chan TC, Yu SC, Chen LT, Huang HY. Overexpressed Fatty Acid Synthase in Gastrointestinal Stromal Tumors: Targeting a Progression-Associated Metabolic Driver Enhances the Antitumor Effect of Imatinib. Clin Cancer Res 2017; 23:4908-4918. [PMID: 28442505 DOI: 10.1158/1078-0432.ccr-16-2770] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 03/02/2017] [Accepted: 04/19/2017] [Indexed: 11/16/2022]
Abstract
Purpose: In gastrointestinal stromal tumors (GIST), lipid-metabolizing enzymes remain underexplored, including fatty acid synthase (FASN).Experimental Design: Forty GISTs were quantitated for FASN mRNA abundance. FASN immunoexpression was informative in 350 GISTs, including 213 with known KIT/PDGFRA/BRAF genotypes. In imatinib-resistant FASN-overexpressing GIST cells, the roles of overexpressed FASN and FASN-targeting C75 in tumor phenotypes, apoptosis and autophagy, KIT transcription, PI3K/AKT/mTOR activation, and imatinib resistance were analyzed by RNAi or myristoylated-AKT transfection. The therapeutic relevance of dual blockade of FASN and KIT was evaluated in vivoResults:FASN mRNA abundance significantly increased from very low/low-risk to high-risk levels of NCCN guidelines (P < 0.0001). FASN overexpression was associated with a nongastric location (P = 0.05), unfavorable genotype (P = 0.005), and increased risk level (P < 0.001) and independently predicted shorter disease-free survival (P < 0.001). In vitro, FASN knockdown inhibited cell growth and migration, inactivated the PI3K/AKT/mTOR pathway, and resensitized resistant GIST cells to imatinib. C75 transcriptionally repressed the KIT promoter, downregulated KIT expression and phosphorylation, induced LC3-II and myristoylated AKT-suppressible activity of caspases 3 and 7, attenuated the PI3K/AKT/mTOR/RPS6/4E-BP1 pathway activation, and exhibited dose-dependent therapeutic additivism with imatinib. Compared with both monotherapies, the C75/imatinib combination more effectively suppressed the growth of xenografts, exhibiting decreased KIT phosphorylation, Ki-67, and phosphorylated PI3K/AKT/mTOR levels and increased TUNEL labeling.Conclusions: We have characterized the prognostic, biological, and therapeutic implications of overexpressed FASN in GISTs. C75 represses KIT transactivation, abrogates PI3K/AKT/mTOR activation, and provides a rationale for dual blockade of KIT and FASN in treating imatinib-resistant GISTs. Clin Cancer Res; 23(16); 4908-18. ©2017 AACR.
Collapse
Affiliation(s)
- Chien-Feng Li
- Department of Pathology, Chi-Mei Medical Center, Tainan, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fu-Min Fang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yen-Yang Chen
- Division of Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ting-Ting Liu
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ti-Chun Chan
- Department of Pathology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Shih-Chen Yu
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Hsuan-Ying Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
23
|
Galadari S, Rahman A, Pallichankandy S, Thayyullathil F. Reactive oxygen species and cancer paradox: To promote or to suppress? Free Radic Biol Med 2017; 104:144-164. [PMID: 28088622 DOI: 10.1016/j.freeradbiomed.2017.01.004] [Citation(s) in RCA: 657] [Impact Index Per Article: 82.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/16/2016] [Accepted: 01/03/2017] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS), a group of highly reactive ions and molecules, are increasingly being appreciated as powerful signaling molecules involved in the regulation of a variety of biological processes. Indeed, their role is continuously being delineated in a variety of pathophysiological conditions. For instance, cancer cells are shown to have increased ROS levels in comparison to their normal counterparts. This is partly due to an enhanced metabolism and mitochondrial dysfunction in cancer cells. The escalated ROS generation in cancer cells contributes to the biochemical and molecular changes necessary for the tumor initiation, promotion and progression, as well as, tumor resistance to chemotherapy. Therefore, increased ROS in cancer cells may provide a unique opportunity to eliminate cancer cells via elevating ROS to highly toxic levels intracellularly, thereby, activating various ROS-induced cell death pathways, or inhibiting cancer cell resistance to chemotherapy. Such results can be achieved by using agents that either increase ROS generation, or inhibit antioxidant defense, or even a combination of both. In fact, a large variety of anticancer drugs, and some of those currently under clinical trials, effectively kill cancer cells and overcome drug resistance via enhancing ROS generation and/or impeding the antioxidant defense mechanism. This review focuses on our current understanding of the tumor promoting (tumorigenesis, angiogenesis, invasion and metastasis, and chemoresistance) and the tumor suppressive (apoptosis, autophagy, and necroptosis) functions of ROS, and highlights the potential mechanism(s) involved. It also sheds light on a very novel and an actively growing field of ROS-dependent cell death mechanism referred to as ferroptosis.
Collapse
Affiliation(s)
- Sehamuddin Galadari
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE; Al Jalila Foundation Research Centre, P.O. Box 300100, Dubai, UAE.
| | - Anees Rahman
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| | - Siraj Pallichankandy
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| | - Faisal Thayyullathil
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| |
Collapse
|
24
|
Yasueda Y, Tamura T, Fujisawa A, Kuwata K, Tsukiji S, Kiyonaka S, Hamachi I. A Set of Organelle-Localizable Reactive Molecules for Mitochondrial Chemical Proteomics in Living Cells and Brain Tissues. J Am Chem Soc 2016; 138:7592-602. [PMID: 27228550 DOI: 10.1021/jacs.6b02254] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein functions are tightly regulated by their subcellular localization in live cells, and quantitative evaluation of dynamically altered proteomes in each organelle should provide valuable information. Here, we describe a novel method for organelle-focused chemical proteomics using spatially limited reactions. In this work, mitochondria-localizable reactive molecules (MRMs) were designed that penetrate biomembranes and spontaneously concentrate in mitochondria, where protein labeling is facilitated by the condensation effect. The combination of this selective labeling and liquid chromatography-mass spectrometry (LC-MS) based proteomics technology facilitated identification of mitochondrial proteomes and the profile of the intrinsic reactivity of amino acids tethered to proteins expressed in live cultured cells, primary neurons and brain slices. Furthermore, quantitative profiling of mitochondrial proteins whose expression levels change significantly during an oxidant-induced apoptotic process was performed by combination of this MRMs-based method with a standard quantitative MS technique (SILAC: stable isotope labeling by amino acids in cell culture). The use of a set of MRMs represents a powerful tool for chemical proteomics to elucidate mitochondria-associated biological events and diseases.
Collapse
Affiliation(s)
- Yuki Yasueda
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University , Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Tomonori Tamura
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University , Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Alma Fujisawa
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University , Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Keiko Kuwata
- Institute of Transformative Bio-Molecules (ITbM), Nagoya University , Chikusa, Nagoya, 464-8602, Japan
| | - Shinya Tsukiji
- Frontier Research Institute for Materials Science (FRIMS), Nagoya Institute of Technology , Gokiso-cho, Showa-ku, Nagoya, 466-8555, Japan.,Department of Life Science and Applied Chemistry, Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology , Gokiso-cho, Showa-ku, Nagoya, 466-8555, Japan
| | - Shigeki Kiyonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University , Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University , Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan.,CREST (Core Research for Evolutional Science and Technology, JST) , Sanbancho, Chiyodaku, Tokyo, 102-0075, Japan
| |
Collapse
|
25
|
Chen X, Tan M, Xie Z, Feng B, Zhao Z, Yang K, Hu C, Liao N, Wang T, Chen D, Xie F, Tang C. Inhibiting ROS-STAT3-dependent autophagy enhanced capsaicin–induced apoptosis in human hepatocellular carcinoma cells. Free Radic Res 2016; 50:744-55. [DOI: 10.3109/10715762.2016.1173689] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
26
|
Downregulation of fatty acid synthase complex suppresses cell migration by targeting phospho-AKT in bladder cancer. Mol Med Rep 2015; 13:1845-50. [DOI: 10.3892/mmr.2015.4746] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 11/19/2015] [Indexed: 11/05/2022] Open
|
27
|
Rae C, Haberkorn U, Babich JW, Mairs RJ. Inhibition of Fatty Acid Synthase Sensitizes Prostate Cancer Cells to Radiotherapy. Radiat Res 2015; 184:482-93. [DOI: 10.1667/rr14173.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
28
|
Impheng H, Richert L, Pekthong D, Scholfield CN, Pongcharoen S, Pungpetchara I, Srisawang P. [6]-Gingerol inhibits de novo fatty acid synthesis and carnitine palmitoyltransferase-1 activity which triggers apoptosis in HepG2. Am J Cancer Res 2015; 5:1319-1336. [PMID: 26101700 PMCID: PMC4473313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/15/2015] [Indexed: 06/04/2023] Open
Abstract
The de novo fatty acid synthesis catalyzed by key lipogenic enzymes, including fatty acid synthase (FASN) has emerged as one of the novel targets of anti-cancer approaches. The present study explored the possible inhibitory efficacy of [6]-gingerol on de novo fatty acid synthesis associated with mitochondrial-dependent apoptotic induction in HepG2 cells. We observed a dissipation of mitochondrial membrane potential accompanied by a reduction of fatty acid levels. [6]-gingerol administration manifested inhibition of FASN expression, indicating FASN is a major target of [6]-gingerol inducing apoptosis in HepG2 cells. Indeed, we found that increased ROS generation could likely be a mediator of the anti-cancer effect of [6]-gingerol. A reduction of fatty acid levels and induction of apoptosis were restored by inhibition of acetyl-CoA carboxylase (ACC) activity, suggesting an accumulation of malonyl-CoA level could be the major cause of apoptotic induction of [6]-gingerol in HepG2 cells. The present study also showed that depletion of fatty acid following [6]-gingerol treatment caused an inhibitory effect on carnitine palmitoyltransferase-1 activity (CPT-1), whereas C75 augmented CPT-1 activity, indicating that [6]-gingerol exhibits the therapeutic benefit on suppression of fatty acid β-oxidation.
Collapse
Affiliation(s)
- Hathaichanok Impheng
- Department of Physiology, Faculty of Medical Science, Naresuan UniversityPhitsanulok, Thailand 65000
| | - Lysiane Richert
- Laboratoire de Toxicologie Cellulaire, Faculté de Médecine et de Pharmacie, Université de Franche-ComtéBesançon, France
| | - Dumrongsak Pekthong
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan UniversityPhitsanulok, Thailand 6500
| | - C Norman Scholfield
- Faculty of Pharmaceutical Sciences, Naresuan UniversityPhitsanulok, Thailand 65000
| | - Sutatip Pongcharoen
- Department of Internal Medicine, Faculty of Medicine, Naresuan UniversityPhitsanulok, Thailand 65000
- Centre of Excellence in Medical Biotechnology (CEMB), Faculty of Medical Science, Naresuan UniversityPhitsanulok, Thailand 65000
| | - Ittipon Pungpetchara
- Department of Anatomy, Faculty of Medical Science, Naresuan UniversityPhitsanulok, Thailand 65000
| | - Piyarat Srisawang
- Department of Physiology, Faculty of Medical Science, Naresuan UniversityPhitsanulok, Thailand 65000
| |
Collapse
|