1
|
Haghir-Sharif-Zamini Y, Khosravi A, Hassan M, Zarrabi A, Vosough M. c-FLIP/Ku70 complex; A potential molecular target for apoptosis induction in hepatocellular carcinoma. Arch Biochem Biophys 2025; 765:110306. [PMID: 39818348 DOI: 10.1016/j.abb.2025.110306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignancies worldwide and the most common form of liver cancer. Despite global efforts toward early diagnosis and effective treatments, HCC is often diagnosed at advanced stages, where conventional therapies frequently lead to resistance and/or high recurrence rates. Therefore, novel biomarkers and promising medications are urgently required. Epi-drugs, or epigenetic-based medicines, have recently emerged as a promising therapeutic modality. Since the epigenome of the cancer cells is always dysregulated and this is followed by apoptosis-resistance, reprogramming the epigenome of cancer cells by epi-drugs (such as HDAC inhibitors (HDACis), and DNMT inhibitors (DNMTis)) could be an alternative approach to use in concert with established treatment protocols. C-FLIP, an anti-apoptotic protein, and Ku70, a member of the DNA repair system, bind together and make a cytoplasmic complex in certain cancers and induce resistance to apoptosis. Many epi-drugs, such as HDACis, can dissociate this complex through Ku70 acetylation and activate cellular apoptosis. The novel compounds for dissociating this complex could provide an innovative insight into molecular targeted HCC treatments. In this review, we address the innovative therapeutic potential of targeting c-FLIP/Ku70 complex by epi-drugs, particularly HDACis, to overcome apoptosis resistance of HCC cells. This review will cover the mechanisms by which the c-FLIP/Ku70 complex facilitates cancer cell survival, the impact of epigenetic alterations on the complex dissociation, and highlight HDACis potential in combination therapies, biomarker developments and mechanistic overviews. This review highlights c-FLIP ubiquitination and Ku70 acetylation levels as diagnostic and prognostic tools in HCC management.
Collapse
Affiliation(s)
- Yasamin Haghir-Sharif-Zamini
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul, 34959, Turkiye
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai - 600 077, India.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
2
|
Michalkova R, Mirossay L, Kello M, Mojzisova G, Baloghova J, Podracka A, Mojzis J. Anticancer Potential of Natural Chalcones: In Vitro and In Vivo Evidence. Int J Mol Sci 2023; 24:10354. [PMID: 37373500 DOI: 10.3390/ijms241210354] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/12/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
There is no doubt that significant progress has been made in tumor therapy in the past decades. However, the discovery of new molecules with potential antitumor properties still remains one of the most significant challenges in the field of anticancer therapy. Nature, especially plants, is a rich source of phytochemicals with pleiotropic biological activities. Among a plethora of phytochemicals, chalcones, the bioprecursors of flavonoid and isoflavonoids synthesis in higher plants, have attracted attention due to the broad spectrum of biological activities with potential clinical applications. Regarding the antiproliferative and anticancer effects of chalcones, multiple mechanisms of action including cell cycle arrest, induction of different forms of cell death and modulation of various signaling pathways have been documented. This review summarizes current knowledge related to mechanisms of antiproliferative and anticancer effects of natural chalcones in different types of malignancies including breast cancers, cancers of the gastrointestinal tract, lung cancers, renal and bladder cancers, and melanoma.
Collapse
Affiliation(s)
- Radka Michalkova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Gabriela Mojzisova
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Janette Baloghova
- Department of Dermatovenerology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Anna Podracka
- Department of Dermatovenerology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Jan Mojzis
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| |
Collapse
|
3
|
Garcinia spp: Products and by-products with potential pharmacological application in cancer. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
4
|
Desai S, Sharma P, Kashyap P, Choudhary B, Kaur J. Bioactive compounds, bio‐functional properties, and food applications of
Garcinia indica
: A review. J Food Biochem 2022; 46:e14344. [DOI: 10.1111/jfbc.14344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Sahil Desai
- Department of Food Technology and Nutrition School of Agriculture, Lovely Professional University Phagwara India
| | - Poorva Sharma
- Department of Food Technology and Nutrition School of Agriculture, Lovely Professional University Phagwara India
| | - Piyush Kashyap
- Department of Food Technology and Nutrition School of Agriculture, Lovely Professional University Phagwara India
| | | | - Jasleen Kaur
- Department of Food Technology and Nutrition School of Agriculture, Lovely Professional University Phagwara India
| |
Collapse
|
5
|
Garcinol-A Natural Histone Acetyltransferase Inhibitor and New Anti-Cancer Epigenetic Drug. Int J Mol Sci 2021; 22:ijms22062828. [PMID: 33799504 PMCID: PMC8001519 DOI: 10.3390/ijms22062828] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/04/2021] [Accepted: 03/06/2021] [Indexed: 12/11/2022] Open
Abstract
Garcinol extracted from Garcinia indica fruit peel and leaves is a polyisoprenylated benzophenone. In traditional medicine it was used for its antioxidant and anti-inflammatory properties. Several studies have shown anti-cancer properties of garcinol in cancer cell lines and experimental animal models. Garcinol action in cancer cells is based on its antioxidant and anti-inflammatory properties, but also on its potency to inhibit histone acetyltransferases (HATs). Recent studies indicate that garcinol may also deregulate expression of miRNAs involved in tumour development and progression. This paper focuses on the latest research concerning garcinol as a HAT inhibitor and miRNA deregulator in the development and progression of various cancers. Garcinol may be considered as a candidate for next generation epigenetic drugs, but further studies are needed to establish the precise toxicity, dosages, routes of administration, and safety for patients.
Collapse
|
6
|
Yoo JD, Bae SM, Seo J, Jeon IS, Vadevoo SMP, Kim SY, Kim IS, Lee B, Kim S. Designed ferritin nanocages displaying trimeric TRAIL and tumor-targeting peptides confer superior anti-tumor efficacy. Sci Rep 2020; 10:19997. [PMID: 33203916 PMCID: PMC7672110 DOI: 10.1038/s41598-020-77095-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022] Open
Abstract
TRAIL is considered a promising target for cancer therapy because it mediates activation of the extrinsic apoptosis pathway in a tumor-specific manner by binding to and trimerizing its functional receptors, DR4 or DR5. Although recombinant human TRAIL has shown high potency and specificity for killing cancer cells in preclinical studies, it has failed in multiple clinical trials for several reasons, including a very short half-life mainly caused by instability of the monomeric form of TRAIL and rapid renal clearance of the off-targeted TRAIL. To overcome such obstacles, we developed a TRAIL-active trimer nanocage (TRAIL-ATNC) that presents the TRAIL ligand in its trimer-like conformation by connecting it to a triple helix sequence that links to the threefold axis of the ferritin nanocage. We also ligated the tumor-targeting peptide, IL4rP, to TRAIL-ATNC to enhance tumor targeting. The developed TRAIL-ATNCIL4rP showed enhanced agonistic activity compared with monomeric TRAIL. The in vivo serum half-life of TRAIL-ATNCIL4rP was ~ 16-times longer than that of native TRAIL. As a consequence of these properties, TRAIL-ATNCIL4rP exhibited efficacy as an anti-tumor agent in vivo against xenograft breast cancer as well as orthotopic pancreatic cancer models, highlighting the promise of this system for development as novel therapeutics against cancer.
Collapse
Affiliation(s)
- Jae Do Yoo
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Sang Mun Bae
- PrismCDX, Inc., 593-16, Dongtangiheung-ro, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Junyoung Seo
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - In Seon Jeon
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Sri Murugan Poongkavithai Vadevoo
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Soyoun Kim
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
| |
Collapse
|
7
|
Bicyclic polyprenylated acylphloroglucinols and their derivatives: structural modification, structure-activity relationship, biological activity and mechanism of action. Eur J Med Chem 2020; 205:112646. [PMID: 32791400 DOI: 10.1016/j.ejmech.2020.112646] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/02/2020] [Accepted: 07/04/2020] [Indexed: 12/22/2022]
Abstract
Bicyclic polyprenylated acylphloroglucinols (BPAPs), the principal bioactive benzophenone products isolated from plants of genera Garcinia and Hypericum, have attracted noticeable attention from the synthetic and biological communities due to their fascinating chemical structures and promising biological activities. However, the potential drug interaction, undesired physiochemical properties and toxicity have limited their potential use and development. In the last decade, pharmaceutical research on the structural modifications, structure-activity relationships (SARs) and mechanisms of action of BPAPs has been greatly developed to overcome the challenges. A comprehensive review of these scientific literature is extremely needed to give an overview of the rapidly emerging area and facilitate research related to BPAPs. This review, containing over 226 references, covers the progress made in the chemical synthesis-based structure modifications, SARs and the mechanism of action of BPAPs in vivo and vitro. The most relevant articles will focus on the discovery of lead compounds via synthetic modifications and the important BPAPs for which the direct targets have been deciphered. From this review, several key points of the SARs and mode of actions of this novel class of compounds have been summarized. The perspective and future direction of the research on BPAPs are concluded. This review would be helpful to get a better grasp of medicinal research of BPAPs and become a compelling guide for chemists dedicated to the synthesis of these compounds.
Collapse
|
8
|
Lee SH, Moon HJ, Lee YS, Kang CD, Kim SH. Potentiation of TRAIL‑induced cell death by nonsteroidal anti‑inflammatory drug in human hepatocellular carcinoma cells through the ER stress‑dependent autophagy pathway. Oncol Rep 2020; 44:1136-1148. [PMID: 32705218 PMCID: PMC7388578 DOI: 10.3892/or.2020.7662] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most commonly diagnosed primary liver malignancy. The limited success with relapse of the disease in HCC therapy is frequently associated with the acquired resistance to anticancer drugs. To develop a strategy and design for overcoming the resistance of HCC cells to TNF-related apoptosis inducing ligand (TRAIL)-induced cell death, we evaluated the efficacy of a non-steroidal anti-inflammatory drug (NSAID) in combination with TRAIL against TRAIL-resistant HCC cells expressing a high level of CD44. We revealed by MTT and western blotting, respectively, that celecoxib (CCB), an NSAID, and 2,5-dimethyl celecoxib (DMC), a non-cyclooxygenase (COX)-2 inhibitor analog of CCB, were able to sensitize TRAIL-resistant HCC cells to TRAIL, implicating a COX-independent mechanism. CCB dose-dependently enhanced LC3-II and reduced p62 levels through AMPK activation and inhibition of the Akt/mTOR pathway and upregulated expression of ATF4/CHOP, leading to activation of endoplasmic reticulum (ER) stress-dependent autophagy. The TRAIL sensitization capacity of CCB in TRAIL-resistant HCC cells was abrogated by an ER stress inhibitor. In addition, we also revealed by flow cytometry and western blotting, respectively, that accelerated downregulation of TRAIL-mediated c-FLIP expression, DR5 activation and CD44 degradation/downregulation by NSAID resulted in activation of caspases and poly(ADP-ribose) polymerase (PARP), leading to the sensitization of TRAIL-resistant HCC cells to TRAIL and thereby reversal of TRAIL resistance. From these results, we propose that NSAID in combination with TRAIL may improve the antitumor activity of TRAIL in TRAIL-resistant HCC, and this approach may serve as a novel strategy that maximizes the therapeutic efficacy of TRAIL for clinical application.
Collapse
Affiliation(s)
- Su-Hoon Lee
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, Gyeongsangnam‑do 626‑870, Republic of Korea
| | - Hyun-Jung Moon
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, Gyeongsangnam‑do 626‑870, Republic of Korea
| | - Young-Shin Lee
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, Gyeongsangnam‑do 626‑870, Republic of Korea
| | - Chi-Dug Kang
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, Gyeongsangnam‑do 626‑870, Republic of Korea
| | - Sun-Hee Kim
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, Gyeongsangnam‑do 626‑870, Republic of Korea
| |
Collapse
|
9
|
Aggarwal V, Tuli HS, Kaur J, Aggarwal D, Parashar G, Chaturvedi Parashar N, Kulkarni S, Kaur G, Sak K, Kumar M, Ahn KS. Garcinol Exhibits Anti-Neoplastic Effects by Targeting Diverse Oncogenic Factors in Tumor Cells. Biomedicines 2020; 8:103. [PMID: 32365899 PMCID: PMC7277375 DOI: 10.3390/biomedicines8050103] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
Garcinol, a polyisoprenylated benzophenone, is the medicinal component obtained from fruits and leaves of Garcinia indica (G. indica) and has traditionally been extensively used for its antioxidant and anti-inflammatory properties. In addition, it has been also been experimentally illustrated to elicit anti-cancer properties. Several in vitro and in vivo studies have illustrated the potential therapeutic efficiency of garcinol in management of different malignancies. It mainly acts as an inhibitor of cellular processes via regulation of transcription factors NF-κB and JAK/STAT3 in tumor cells and have been demonstrated to effectively inhibit growth of malignant cell population. Numerous studies have highlighted the anti-neoplastic potential of garcinol in different oncological transformations including colon cancer, breast cancer, prostate cancer, head and neck cancer, hepatocellular carcinoma, etc. However, use of garcinol is still in its pre-clinical stage and this is mainly attributed to the limitations of conclusive evaluation of pharmacological parameters. This necessitates evaluation of garcinol pharmacokinetics to precisely identify an appropriate dose and route of administration, tolerability, and potency under physiological conditions along with characterization of a therapeutic index. Hence, the research is presently ongoing in the dimension of exploring the precise metabolic mechanism of garcinol. Despite various lacunae, garcinol has presented with promising anti-cancer effects. Hence, this review is motivated by the constantly emerging and promising positive anti-cancerous effects of garcinol. This review is the first effort to summarize the mechanism of action of garcinol in modulation of anti-cancer effect via regulation of different cellular processes.
Collapse
Affiliation(s)
- Vaishali Aggarwal
- Department of Histopathology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab 160012, India;
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Jagjit Kaur
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Faculty of Engineering, The University of New South Wales, Sydney 2052, Australia;
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Gaurav Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Nidarshana Chaturvedi Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Samruddhi Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Vileparle-West, Mumbai 400056, India; (S.K.); (G.K.)
| | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Vileparle-West, Mumbai 400056, India; (S.K.); (G.K.)
| | | | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur 133001, India;
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| |
Collapse
|
10
|
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
|
11
|
Schobert R, Biersack B. Chemical and Biological Aspects of Garcinol and Isogarcinol: Recent Developments. Chem Biodivers 2019; 16:e1900366. [PMID: 31386266 DOI: 10.1002/cbdv.201900366] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/06/2019] [Indexed: 11/08/2022]
Abstract
The natural polyisoprenylated benzophenone derivatives garcinol and isogarcinol are secondary plant metabolites isolated from various Garcinia species including Garcinia indica. This review takes stock of the recent chemical and biological research into these interesting natural compounds over the last five years. New biological sources and chemical syntheses are discussed followed by new insights into the activity of garcinol and isogarcinol against cancer, pathogenic bacteria, parasite infections and various inflammatory diseases.
Collapse
Affiliation(s)
- Rainer Schobert
- Organic Chemistry Laboratory, University of Bayreuth, 95447, Bayreuth, Germany
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, 95447, Bayreuth, Germany
| |
Collapse
|
12
|
Targeting TRAIL. Bioorg Med Chem Lett 2019; 29:2527-2534. [PMID: 31383590 DOI: 10.1016/j.bmcl.2019.07.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/24/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), also known as Apo2L, has been investigated in the past decade for its promising anticancer activity due to its ability to selectively induce apoptosis in tumoral cells by binding to TRAIL receptors (TRAIL-R). Macromolecules such as agonistic monoclonal antibodies and recombinant TRAIL have not proven efficacious in clinical studies, therefore several small molecules acting as TRAIL-R agonists are emerging in the scientific literature. In this work we focus on systemizing these drug molecules described in the past years, in order to better understand and predict the requirements for a novel anti-tumoral therapy based on the TRAIL-R-induced apoptotic mechanism.
Collapse
|
13
|
Benzophenones from Anemarrhena asphodeloides Bge. Exhibit Anticancer Activity in HepG2 Cells via the NF-κB Signaling Pathway. Molecules 2019; 24:molecules24122246. [PMID: 31208101 PMCID: PMC6630431 DOI: 10.3390/molecules24122246] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 01/15/2023] Open
Abstract
A chemical investigation of the fibrous roots of Anemarrhena asphodeloides Bge. led to the isolation of four benzophenones, including one new compound (1) and three known ones (2–4). Comprehensive 1D, 2D NMR and HRESIMS data established the structures of the isolated compounds. The absolute configurations were determined by comparison of the calculated optical rotation (OR) with experimental data. All the isolates were evaluated for their cytotoxicities on hepatocellular carcinoma cell lines (HepG2 and Hep3B). Compound 1 showed strong cytotoxicity against HepG2 and Hep3B cells, with IC50 values at 153.1 and 180.6 nM. Through MTT assay, flow cytometry and Western blot analysis, compound 1 demonstrated the ability to stimulate apoptosis via the NF-κB signaling pathway in HepG2 cells. These benzophenones are potential lead compounds for the development of better treatments for hepatocellular carcinoma.
Collapse
|
14
|
WP1130 Enhances TRAIL-Induced Apoptosis through USP9X-Dependent miR-708-Mediated Downregulation of c-FLIP. Cancers (Basel) 2019; 11:cancers11030344. [PMID: 30862047 PMCID: PMC6469024 DOI: 10.3390/cancers11030344] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/06/2019] [Accepted: 03/08/2019] [Indexed: 12/20/2022] Open
Abstract
WP1130, a partially selective deubiquitinases (DUB) inhibitor, inhibits the deubiquitinating activities of USP5, USP9X, USP14, USP37, and UCHL1. In this study, we investigate whether WP1130 exerts sensitizing effect on TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in human renal carcinoma cells. Combinations of WP1130 and TRAIL significantly induced apoptosis in renal carcinoma, lung carcinoma and hepatocellular carcinoma cells, but not in normal cells (human mesangial cells (MC) and normal mouse kidney cells (TCMK-1)). The downregulation of c-FLIP protein expression was involved in combined treatment-induced apoptosis. WP1130-induced c-FLIP downregulation was regulated by microRNA (miR)-708 upregulation via inhibition of USP9X. Interestingly, knockdown of USP9X markedly induced c-FLIP downregulation, upregulation of miR-708 expression and sensitivity to TRAIL. Furthermore, ectopic expression of USP9X prevented c-FLIP downregulation and apoptosis upon combined treatment. In sum, WP1130 sensitized TRAIL-induced apoptosis through miR-708-mediated downregulation of c-FLIP by inhibition of USP9X.
Collapse
|
15
|
Farhan M, Malik A, Ullah MF, Afaq S, Faisal M, Farooqi AA, Biersack B, Schobert R, Ahmad A. Garcinol Sensitizes NSCLC Cells to Standard Therapies by Regulating EMT-Modulating miRNAs. Int J Mol Sci 2019; 20:ijms20040800. [PMID: 30781783 PMCID: PMC6413107 DOI: 10.3390/ijms20040800] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 12/24/2022] Open
Abstract
Garcinol, a dietary factor obtained from Garcinia indica, modulates several key cellular signaling pathways as well as the expression of miRNAs. Acquired resistance to standard therapies, such as erlotinib and cisplatin, is a hallmark of non-small cell lung cancer (NSCLC) cells that often involves miRNA-regulated epithelial-to-mesenchymal transition (EMT). We used A549 cells that were exposed to transforming growth factor beta 1 (TGF-β1), resulting in A549M cells with mesenchymal and drug resistant phenotype, and report that garcinol sensitized resistant cells with mesenchymal phenotype to erlotinib as well as cisplatin with significant decrease in their IC50 values. It also potentiated the apoptosis-inducing activity of erlotinib in A549M and the endogenously mesenchymal H1299 NSCLC cells. Further, garcinol significantly upregulated several key EMT-regulating miRNAs, such as miR-200b, miR-205, miR-218, and let-7c. Antagonizing miRNAs, through anti-miRNA transfections, attenuated the EMT-modulating activity of garcinol, as determined by mRNA expression of EMT markers, E-cadherin, vimentin, and Zinc Finger E-Box Binding Homeobox 1 (ZEB1). This further led to repression of erlotinib as well as cisplatin sensitization, thus establishing the mechanistic role of miRNAs, particularly miR-200c and let-7c, in garcinol-mediated reversal of EMT and the resulting sensitization of NSCLC cells to standard therapies.
Collapse
Affiliation(s)
- Mohd Farhan
- College of Basic Sciences, King Faisal University, Hofuf, 400, Al Ahsa 31982, Saudi Arabia.
| | - Arshi Malik
- Clinical Biochemistry, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia.
| | - Mohammad Fahad Ullah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Science, University of Tabuk, Tabuk 71491, Saudi Arabia.
| | - Sarah Afaq
- Clinical Biochemistry, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia.
| | - Mohd Faisal
- Department of Psychiatry, University Hospital Limerick, Limerick V94 T9PX, Ireland.
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan.
| | - Bernhard Biersack
- Organic Chemistry Laboratory, Universitätsstr. 30, 95447 Bayreuth, Germany.
| | - Rainer Schobert
- Organic Chemistry Laboratory, Universitätsstr. 30, 95447 Bayreuth, Germany.
| | - Aamir Ahmad
- Department of Pathology, Wayne State University and Karmanos Cancer Institute, Detroit, MI 48201, USA.
| |
Collapse
|
16
|
Fayyaz S, Javed Z, Attar R, Farooqi AA, Yaylim I, Ahmad A. MicroRNA regulation of TRAIL mediated signaling in different cancers: Control of micro steering wheels during the journey from bench-top to the bedside. Semin Cancer Biol 2019; 58:56-64. [PMID: 30716480 DOI: 10.1016/j.semcancer.2019.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 12/14/2022]
Abstract
Large-scale sequencing methodologies have helped us identify numerous genomic alterations and we have started to scratch the surface of many new targets for treatment of cancer and the associated predictive biomarkers. TRAIL (TNF-related apoptosis-inducing ligand) is a highly appreciated anti-cancer molecule because of its ability to selectively target cancer cells. However, confluence of information suggests that cancer cells develop resistance against TRAIL-based therapeutics. It is being realized that overexpression of anti-apoptotic proteins and inactivation of pro-apoptotic proteins significantly impairs TRAIL triggered apoptosis, particularly in clinical settings. Re-balancing of pro-and anti-apoptotic proteins and upregulation of death receptors with functionally active extrinsic and intrinsic apoptotic pathways are necessary to sensitize cancer cells to TRAIL based therapeutics. microRNAs (miRNAs) are involved in regulation of myriad of molecular processes and characterized into oncogenic and tumor suppressor miRNAs. Accumulating data has identified miRNAs which positively or negatively regulate TRAIL mediated signaling in cancer cells, helping us understand different steps at which TRAIL-mediated apoptotic signaling can be targeted. Here, we assess the status of our understanding of the mechanisms related to miRNA regulation of TRAIL mediated signaling, as well as the existing gaps therein, and discuss the challenges and opportunities that will help us get closer to personalized medicine.
Collapse
Affiliation(s)
- Sundas Fayyaz
- Department of Biochemistry, Rashid Latif Medical College (RLMC), Pakistan
| | - Zeeshan Javed
- Department of Biochemistry, Rashid Latif Medical College (RLMC), Pakistan
| | - Rukset Attar
- Department of Obstetrics and Gynecology, Yeditepe University Hospital, Istanbul, Turkey
| | | | - Ilhan Yaylim
- Department of Molecular Medicine, Aziz Sancar İnstitute of Experimental Medicine, İstanbul University, İstanbul, Turkey
| | - Aamir Ahmad
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA.
| |
Collapse
|