1
|
Luo X, Li J, Cen Z, Feng G, Hong M, Huang L, Long Q. Exploring the therapeutic potential of lupeol: A review of its mechanisms, clinical applications, and advances in bioavailability enhancement. Food Chem Toxicol 2025; 196:115193. [PMID: 39662867 DOI: 10.1016/j.fct.2024.115193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024]
Abstract
Lupeol, a naturally occurring triterpenoid, has garnered significant attention for its diverse range of biological activities and potential therapeutic applications. This comprehensive review delves into the various aspects of lupeol, including its sources, extraction methods, chemical characteristics, pharmacokinetics, safety evaluation, mechanisms of action, and applications in disease treatment. We highlight the compound's unique carbon skeleton and its role in inflammation regulation, antioxidant activity, and broad-spectrum antimicrobial effects. The review also underscores lupeol's potential in cancer therapy, cardiovascular protection, metabolic disease management, and wound healing. Furthermore, we discuss the challenges and future perspectives of lupeol's clinical application, emphasizing the need for further research to improve its bioavailability and explore its full therapeutic potential. The review concludes by recognizing the significance of lupeol in drug development and healthcare, with expectations for future breakthroughs in medical applications.
Collapse
Affiliation(s)
- Xia Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ji Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zhifeng Cen
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Gang Feng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Meiqi Hong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Lizhen Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Qinqiang Long
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
2
|
Sahoo BM, Banik BK, Sharma S, Singh B. Current Insights into Therapeutic Potential of Terpenoids as Anticancer Agents. Anticancer Agents Med Chem 2025; 25:339-356. [PMID: 39440731 DOI: 10.2174/0118715206342920241008062115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Cancer is regarded as one of the main causes of death globally. Future predictions indicate that the death rate from cancer will keep rising, which may reach 11.4 million in 2030. Carcinogenesis refers to the phenomenon of transforming a normal cell into a cancer cell. Cancer is characterized by unregulated and uncontrolled cell division due to alterations at the molecular and genetic levels. Gene mutations can speed up the rate of cell division, which leads to cancer. Metastasis entails the dissemination of cancer cells from the primary site to distant regions of the body via the circulatory or lymphatic systems. OBJECTIVE This review is mainly focusing on the anticancer properties of terpenoids. In the case of human beings, several types of cancers can be treated clinically based on the form and phase of the cancer. So, there are different types of treatment regimens available for the management of cancer, such as immunotherapy, hormonal therapy, radiation therapy, and chemotherapy. METHODS Several problems are associated with cancer therapy, including chemoresistance, severe toxicity, relapse, and metastasis. To minimize these complications, natural products like terpenoids seem to be beneficial for the effective management of cancer. RESULTS Experimental results revealed that the anticancer potential of terpenoids is due to activation of apoptosis and stimulation of cell cycle arrest. Some of the terpenoids exhibit anticancer effects by inhibiting angiogenesis and metastasis via the regulation of several signaling pathways intracellularly. Certain terpenoids have been shown to work in concert with anticancer medications (doxorubicin, cisplatin, paclitaxel, and 5-fluorouracil) to provide synergistic effects. These terpenoids have also been shown to be effective against cancer cells that are resistant to several drug therapies. CONCLUSION The current study will focus on signaling pathways and mode of action of several types of terpenoids as anticancer agents. Further, it will provide insights into the ongoing clinical trials and prospective pathways for the advancement of terpenoids as possible anti-cancer agents.
Collapse
Affiliation(s)
- Biswa Mohan Sahoo
- School of Pharmacy and Life Sciences, Centurion University of Technology & Management, Jatni, Bhubaneswar, Khurda, 752050, Odisha, India
| | - Bimal Krishna Banik
- Department of Mathematics and Natural Sciences, College of Sciences and Human Studies, Prince Mohammad Bin Fahd University, Al Khobar, Kingdom of Saudi Arabia
| | - Shikha Sharma
- Department of Pharmaceutical Science, Lords University, Alwar, 301028, Rajasthan, India
| | - Bhupendra Singh
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248002, India
- Department of Pharmacy, S.N. Medical College, Agra, 282002, India
| |
Collapse
|
3
|
Dalimunthe A, Carensia Gunawan M, Dhiya Utari Z, Dinata MR, Halim P, Estherina S. Pakpahan N, Sitohang AI, Sukarno MA, Yuandani, Harahap Y, Setyowati EP, Park MN, Yusoff SD, Zainalabidin S, Prananda AT, Mahadi MK, Kim B, Harahap U, Syahputra RA. In-depth analysis of lupeol: delving into the diverse pharmacological profile. Front Pharmacol 2024; 15:1461478. [PMID: 39605919 PMCID: PMC11598436 DOI: 10.3389/fphar.2024.1461478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/30/2024] [Indexed: 11/29/2024] Open
Abstract
Lupeol, a naturally occurring lupane-type pentacyclic triterpenoid, is widely distributed in various edible vegetables, fruits, and medicinal plants. Notably, it is found in high concentrations in plants like Tamarindus indica, Allanblackia monticola, and Emblica officinalis, among others. Quantitative studies have highlighted its presence in Elm bark, Olive fruit, Aloe leaf, Ginseng oil, Mango pulp, and Japanese Pear bark. This compound is synthesized from squalene through the mevalonate pathway and can also be synthetically produced in the lab, addressing challenges in natural product synthesis. Over the past four decades, extensive research has demonstrated lupeol's multifaceted pharmacological properties, including anti-inflammatory, antioxidant, anticancer, and antibacterial effects. Despite its significant therapeutic potential, clinical applications of lupeol have been limited by its poor water solubility and bioavailability. Recent advancements have focused on nano-based delivery systems to enhance its bioavailability, and the development of various lupeol derivatives has further amplified its bioactivity. This review provides a comprehensive overview of the latest advancements in understanding the pharmacological benefits of lupeol. It also discusses innovative strategies to improve its bioavailability, thereby enhancing its clinical efficacy. The aim is to consolidate current knowledge and stimulate further research into the therapeutic potential of lupeol and its derivatives.
Collapse
Affiliation(s)
- Aminah Dalimunthe
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Mega Carensia Gunawan
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Zahirah Dhiya Utari
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Muhammad Riza Dinata
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | | | - Alex Insandus Sitohang
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - M. Andriansyah Sukarno
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Yuandani
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | | | | | - Moon Nyeo Park
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Syaratul Dalina Yusoff
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Satirah Zainalabidin
- Biomedical Science, Centre of Toxicology and Health Risk Study, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Arya Tjipta Prananda
- Faculty of Medicine, Universitas Sumatera Utara, Medan, Sumatera Utara, Indonesia
| | - Mohd Kaisan Mahadi
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Bonglee Kim
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Urip Harahap
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
4
|
Fatma H, Jameel M, Siddiqui AJ, Kuddus M, Buali NS, Bahrini I, Siddique HR. Chemotherapeutic potential of lupeol against cancer in pre-clinical model: A systematic review and meta-analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155777. [PMID: 38943695 DOI: 10.1016/j.phymed.2024.155777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/11/2024] [Accepted: 05/22/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND Extensive research on Lupeol's potential in cancer prevention highlights its ability to target various cancer-related factors. It regulates proliferative markers, modulates signaling pathways, including PI3K/AKT/mTOR, and influences inflammatory and apoptotic mechanisms. Additionally, Lupeol demonstrates selectivity in killing cancer cells while sparing normal cells, thus minimizing the risk of toxic effects on healthy tissues. HYPOTHESIS Therefore, we aimed to explore Lupeol's potential roles as a chemotherapeutic agent and as a sensitizer to chemotherapy by reviewing various animal-based studies published on its effects. STUDY DESIGN We conducted a comprehensive search across databases, including PubMed, PMC, Cochrane, EuroPMC, and ctri.gov.in to identify pertinent articles. Our focus was solely on published animal studies examining Lupeol's anti-cancer effects, with reviewers independently assessing bias risk and resolving discrepancies through consensus. RESULT 20 studies were shortlisted. The results demonstrated that Lupeol brings changes in the tumor volume by [Hedges's g: -6.62; 95 % CI: -8.68, -4.56; τ2: 24.36, I2: 96.50 %; p < 0.05] and tumor weight by [Hedges's g: -3.97; 95 % CI: -5.20, -2.49; τ2: 2.70, I2: 79.27 %; p <0.05]. The high I2, negative Egger's value, and asymmetrical funnel plot show the publication bias among the studies. Further, Lupeol in combination with other chemotherapeutic agents showed better outcomes as compared to them alone [Hedges's g: -6.38; 95 % CI: -11.82, -0.94; τ2: 46.91; I2: 98.68 %; p <0.05]. Lupeol also targets various signaling molecules and pathways to exert an anti-cancer effect. CONCLUSION In conclusion, Lupeol significantly reduces tumor volume and weight. Combining Lupeol with other chemotherapy agents shows promise for enhancing anti-cancer effects. However, high variability among studies and evidence of publication bias suggest caution in interpreting results.
Collapse
Affiliation(s)
- Homa Fatma
- Molecular Cancer Genetics & Translational Research Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh-202002, Uttar Pradesh, India
| | - Mohd Jameel
- Molecular Cancer Genetics & Translational Research Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh-202002, Uttar Pradesh, India
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha'il, Hail, PO Box 2440, Kingdom of Saudi Arabia.
| | - Mohammed Kuddus
- Department of Biochemistry, College of Medicine, University of Ha'il, Hail, Kingdom of Saudi Arabia
| | - Nouha Saleh Buali
- Department of Biology, College of Science, University of Ha'il, Hail, PO Box 2440, Kingdom of Saudi Arabia
| | - Insaf Bahrini
- Department of Biology, College of Science, University of Ha'il, Hail, PO Box 2440, Kingdom of Saudi Arabia
| | - Hifzur R Siddique
- Molecular Cancer Genetics & Translational Research Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh-202002, Uttar Pradesh, India.
| |
Collapse
|
5
|
Sherapura A, Siddesh BM, Malojirao VH, Thirusangu P, Avin BRV, Kumari NS, Ramachandra YL, Prabhakar BT. Steroidal alkaloid solanidine impedes hypoxia-driven ATM phosphorylation to switch on anti-angiogenesis in lung adenocarcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154981. [PMID: 37531902 DOI: 10.1016/j.phymed.2023.154981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/14/2023] [Accepted: 07/15/2023] [Indexed: 08/04/2023]
Abstract
PURPOSE The declined oxygen tension in the cancer cell leads to the hypoxic adaptive response and favors establishment of tumor micro environment [TEM]. The complex TME consists of interwoven hypoxic HIF-1α and DNA damage repair ATM signaling. The ATM/HIF-1α phosphorylation switch on angiogenesis and abort apoptosis. Targeting this signaling nexus would be a novel therapeutic strategy for the treatment of cancer. BACKGROUND Steroidal alkaloid solanidine is known for varied pharmacological role but with less molecular evidences. Our earlier findings on solanidine proven its anti-neoplastic activity by inducing apoptosis in lung cancer. In continued research, efforts have been made to establish the underlying molecular signaling in induction of DNA damage in prevailing hypoxic TME. METHODS The solanidine induced DNA damage was assessed trough alkali COMET assay; signaling nexus and gene expression profile analysis through IB, qRT-PCR, Gelatin Zymography, IHC, IF and ELISA. Pathophysiological modulations assessed through tube formation, migration, invasion assays. Anti-angiogenic studies through CAM, rat aorta, matrigel assays and corneal neovascularization assay. Anti-tumor activity through in-vivo DLA ascites tumor model and LLC model. RESULTS The results postulates, inhibition of hypoxia driven DDR proteins pATMser1981/pHIF-1αser696 by solanidine induces anti-angiogenesis. Systematic study of both non-tumorigenic and tumorigenic models in-vitro as well as in-vivo experimental system revealed the angio-regression mediated anticancer effect in lung cancer. These effects are due to the impeded expression of angiogenic mediators such as VEGF, MMP2&9 and inflammatory cytokines IL6 and TNFα to induce pathophysiological changes CONCLUSION: The study establishes new role of solanidine by targeting ATM/HIF-1α signaling to induce anti-angiogenesis for the first time. The study highlights the potentiality of plant based phytomedicine solanidine which can targets the multiple hallmarks of cancer by targeting interwoven signaling crosstalk. Such an approach through solanidine necessary to counteract heterogeneous complexity of cancer which could be nearly translated into drug.
Collapse
Affiliation(s)
- Ankith Sherapura
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577203, Karnataka, India
| | - B M Siddesh
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577203, Karnataka, India
| | - Vikas H Malojirao
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577203, Karnataka, India; Division for DNA Repair Research, Department of Neurosurgery, Centre for Neuroregeneration, Houston Methodist, Fannin Street, Houston, TX, USA
| | - Prabhu Thirusangu
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577203, Karnataka, India; Department of Experidmental Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | - B R Vijay Avin
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577203, Karnataka, India; Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois at Chicago, Chicago, 60612, USA
| | - N Suchetha Kumari
- Department of Biochemistry, K.S. Hegde Medical College, Nitte University, Mangalore, India
| | - Y L Ramachandra
- Postgraduate Department of Studies and Research in Biotechnology, Kuvempu University, Shankaraghatta, 577 451, Karnataka, India
| | - B T Prabhakar
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577203, Karnataka, India.
| |
Collapse
|
6
|
López-Huerta FA, Teresa Ramírez-Apan M, Méndez-Cuesta CA, Nieto-Camacho A, Hernández-Ortega S, Almeida-Aguirre EK, Cerbón MA, Delgado G. Synthesis, Biological Evaluation, Molecular Docking Studies and In-silico ADMET Evaluation of Pyrazines of Pentacyclic Triterpenes. Bioorg Chem 2022; 125:105924. [DOI: 10.1016/j.bioorg.2022.105924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/20/2022] [Accepted: 05/30/2022] [Indexed: 11/02/2022]
|
7
|
Jyothi M, Banumathi, Zabiulla, Sherapura A, Khamees HA, Prabhakar B, Khanum SA. Synthesis, structure analysis, DFT calculations and energy frameworks of new coumarin appended oxadiazoles, to regress ascites malignancy by targeting VEGF mediated angiogenesis. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
8
|
Investigation of Lupeol as Anti-Melanoma Agent: An In Vitro-In Ovo Perspective. Curr Oncol 2021; 28:5054-5066. [PMID: 34940064 PMCID: PMC8700590 DOI: 10.3390/curroncol28060425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/14/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
Malignant melanoma (MM) represents the most life-threatening skin cancer worldwide, with a narrow and inefficient chemotherapeutic arsenal available in advanced disease stages. Lupeol (LUP) is a triterpenoid-type phytochemical possessing a broad spectrum of pharmacological properties, including a potent anticancer effect against several neoplasms (e.g., colorectal, lung, and liver). However, its potential as an anti-melanoma agent has been investigated to a lesser extent. The current study focused on exploring the impact of LUP against two human MM cell lines (A375 and RPMI-7951) in terms of cell viability, confluence, morphology, cytoskeletal distribution, nuclear aspect, and migration. Additionally, the in ovo antiangiogenic effect has been also examined. The in vitro results indicated concentration-dependent and selective cytotoxicity against both MM cell lines, with estimated IC50 values of 66.59 ± 2.20 for A375, and 45.54 ± 1.48 for RPMI-7951, respectively, accompanied by a reduced cell confluence, apoptosis-specific nuclear features, reorganization of cytoskeletal components, and inhibited cell migration. In ovo, LUP interfered with the process of angiogenesis by reducing the formation of neovascularization. Despite the potential anti-melanoma effect illustrated in our in vitro-in ovo study, further investigations are required to elucidate the underlying LUP-induced effects in A375 and RPMI-7951 MM cells.
Collapse
|
9
|
Targeting HIF-1α by newly synthesized Indolephenoxyacetamide (IPA) analogs to induce anti-angiogenesis-mediated solid tumor suppression. Pharmacol Rep 2021; 73:1328-1343. [PMID: 33904146 DOI: 10.1007/s43440-021-00266-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hypoxic microenvironment is a common feature of solid tumors, which leads to the promotion of cancer. The transcription factor, HIF-1α, expressed under hypoxic conditions stimulates tumor angiogenesis, favoring HIF-1α as a promising anticancer agent. On the other hand, synthetic Indolephenoxyacetamide derivatives are known for their pharmacological potentiality. With this background here, we have synthesized, characterized, and validated the new IPA (8a-n) analogs for anti-tumor activity. METHODS The new series of IPA (8a-n) were synthesized through a multi-step reaction sequence and characterized based on the different spectroscopic analysis FT-IR, 1H, 13C NMR, mass spectra, and elemental analyses. Cell-based screening of IPA (8a-n) was assessed by MTT assay. Anti-angiogenic efficacy of IPA (8k) validated through CAM, Rat corneal, tube formation and migration assay. The underlying molecular mechanism is validated through zymogram and IB studies. The in vivo anti-tumor activity was measured in the DLA solid tumor model. RESULTS Screening for anti-proliferative studies inferred, IPA (8k) is a lead molecule with an IC50 value of ˜5 μM. Anti-angiogenic assays revealed the angiopreventive activity through inhibition of HIF-1α and modulation downstream regulatory genes, VEGF, MMPs, and P53. The results are confirmative in an in vivo solid tumor model. CONCLUSION The IPA (8k) is a potent anti-proliferative molecule with anti-angiogenic activity and specifically targets HIF1α, thereby modulates its downstream regulatory genes both in vitro and in vivo. The study provides scope for new target-specific drug development against HIF-1α for the treatment of solid tumors.
Collapse
|
10
|
Liu K, Zhang X, Xie L, Deng M, Chen H, Song J, Long J, Li X, Luo J. Lupeol and its derivatives as anticancer and anti-inflammatory agents: Molecular mechanisms and therapeutic efficacy. Pharmacol Res 2020; 164:105373. [PMID: 33316380 DOI: 10.1016/j.phrs.2020.105373] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/17/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023]
Abstract
Lupeol is a natural triterpenoid that widely exists in edible fruits and vegetables, and medicinal plants. In the last decade, a plethora of studies on the pharmacological activities of lupeol have been conducted and have demonstrated that lupeol possesses an extensive range of pharmacological activities such as anticancer, antioxidant, anti-inflammatory, and antimicrobial activities. Pharmacokinetic studies have indicated that absorption of lupeol by animals was rapid despite its nonpolar characteristics, and lupeol belongs to class II BCS (biopharmaceutics classification system) compounds. Moreover, the bioactivities of some isolated or synthesized lupeol derivatives have been investigated, and these results showed that, with modification to C-3 or C-19, some derivatives exhibit stronger activities, e.g., antiprotozoal or anticancer activity. This review aims to summarize the advances in pharmacological and pharmacokinetic studies of lupeol in the last decade with an emphasis on its anticancer and anti-inflammatory activities, as well as the research progress of lupeol derivatives thus far, to provide researchers with the latest information, point out the limitations of relevant research at the current stage and the aspects that should be strengthened in future research.
Collapse
Affiliation(s)
- Kai Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Xumin Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Long Xie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Mao Deng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Huijuan Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jiawen Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jiaying Long
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| | - Jia Luo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| |
Collapse
|
11
|
Taleb Agha M, Baharetha HM, Al-Mansoub MA, Tabana YM, Kaz Abdul Aziz NH, Yam MF, Abdul Majid AMS. Proapoptotic and Antiangiogenic Activities of Arctium Lappa L. on Breast Cancer Cell Lines. SCIENTIFICA 2020; 2020:7286053. [PMID: 32509375 PMCID: PMC7254072 DOI: 10.1155/2020/7286053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/16/2020] [Accepted: 04/23/2020] [Indexed: 06/10/2023]
Abstract
In this study, the bioactivity-guided fractionation was conducted on the aerial parts of Arctium lappa L. and then the extracts were tested in vitro on breast cancer (MCF-7), colorectal cancer (HCT-116), and normal cells (EA.hy926). The n-hexane fraction (EHX) of the ethanolic extract showed strong activity against both MCF-7 and EA.hy926 cell lines (IC50 values: 14.08 ± 3.64 and 27.25 ± 3.45 μg/mL, respectively). The proapoptotic activity of EHX was assessed using MCF-7. Morphological alterations were visualized using Hoechst staining and a transmission electron microscope. Cancer cell signal transduction pathways were investigated, and EHX significantly upregulated p53, TGF-β, and NF-κB. Furthermore, EHX was found to disrupt the metastatic cascade of breast cancer cells by the inhibition of cell proliferation, migration, invasion, and colonization. The antiangiogenic activity of EHX fraction showed potent inhibition of rat aorta microvessels with IC50 value: 4.34 ± 1.64 μg/mL. This result was supported by the downregulation of VEGF-A expression up to 54%. Over 20 compounds were identified in EHX using GC-MS, of which stigmasterol, ß-sitosterol, and 3-O-acetyllupeol are the major active compounds. Phytochemical analysis of EHX showed higher phenolic and flavonoid contents with a substantial antioxidant activity. In conclusion, this work demonstrated that A. lappa has valuable anticancer activity and antiangiogenic properties that might be useful in breast cancer therapy.
Collapse
Affiliation(s)
- Mohamad Taleb Agha
- EMAN Testing & Research Laboratory, Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Pinang, Malaysia
| | - Hussein M. Baharetha
- EMAN Testing & Research Laboratory, Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Pinang, Malaysia
- Department of Pharmacy, College of Medicine and Health Sciences, Hadhramout University, Al Mukalla, Hadhramout, Yemen
| | - Majed Ahmed Al-Mansoub
- EMAN Testing & Research Laboratory, Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Pinang, Malaysia
| | - Yasser M. Tabana
- EMAN Testing & Research Laboratory, Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Pinang, Malaysia
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Nur Hidayah Kaz Abdul Aziz
- EMAN Testing & Research Laboratory, Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Pinang, Malaysia
| | - Mun Fei Yam
- EMAN Testing & Research Laboratory, Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Pinang, Malaysia
| | - Amin Malik Shah Abdul Majid
- EMAN Testing & Research Laboratory, Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Pinang, Malaysia
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Canberra, Australia
| |
Collapse
|
12
|
Zhang H, Wang J, Guo R. Application Value of Color Doppler Ultrasound and Ultrasound Contrast in the Differential Diagnosis of Ovarian tumor. Pak J Med Sci 2020; 36:80-84. [PMID: 32063936 PMCID: PMC6994900 DOI: 10.12669/pjms.36.2.847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective: To study the value of color Doppler ultrasound and ultrasound contrast in differential diagnosis of ovarian tumors. Methods: Ninety-six patients with ovarian tumors who were treated in our hospital from May 2017 to July 2018 and confirmed by pathological examination were selected as the research subjects. All patients were examined by color Doppler ultrasound and ultrasound contrast. The sensitivity, specificity and accuracy of the two methods were compared, and the parameters of ultrasound contrast in the diagnosis of benign and malignant tumors were observed and compared. Results: The sensitivity, specificity and accuracy of ultrasound contrast in the diagnosis of ovarian tumors were higher than those of color Doppler ultrasound (P<0.05). There were significant differences in the time of initiation enhancement, time to peak and perfusion intensity in the diagnosis of benign and malignant lesions by ultrasound contrast (P<0.05). Conclusion: In the differential diagnosis of ovarian tumors, ultrasound contrast has more advantages than color Doppler ultrasound in displaying the blood perfusion information of tumors. It has high diagnostic accuracy and clinical application value.
Collapse
Affiliation(s)
- Haijing Zhang
- Haijing Zhang, Department of Ultrasonic Medicine, Binzhou People's Hospital, Shandong, 256610, China
| | - Jinming Wang
- Jinming Wang, Department of Ultrasonic Medicine, Binzhou People's Hospital, Shandong, 256610, China
| | - Rui Guo
- Rui Guo, Department of Ultrasonic Medicine, Binzhou People's Hospital, Shandong, 256610, China
| |
Collapse
|
13
|
Biswas T, Dwivedi UN. Plant triterpenoid saponins: biosynthesis, in vitro production, and pharmacological relevance. PROTOPLASMA 2019; 256:1463-1486. [PMID: 31297656 DOI: 10.1007/s00709-019-01411-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/01/2019] [Indexed: 05/26/2023]
Abstract
The saponins are a diverse class of natural products, with a broad scale distribution across different plant species. Chemically characterized as triterpenoid glycosides, they posses a 30C oxidosqualene precursor-based aglycone moiety (sapogenin), to which glycosyl residues are subsequently attached to yield the corresponding saponin. Based on the chemically distinct aglycone moieties, broadly, they are divided into triterpenoid saponins (dammaranes, ursanes, oleananes, lupanes, hopanes, etc.) and the sterol glycosides. This review aims to present in detail the biosynthesis patterns of the different aglycones from a common precursor and their glycosylation patterns to yield the functionally active glycoside. The review also presents recent advances in the pharmacological activities of these saponins, particularly as potent anti-neoplastic pharmacophores, antioxidants, or anti-viral/antibacterial agents. Since alternate production pedestals for these pharmacologically important triterpenes via cell and tissue cultures are an attractive option for their sustainable production, recent trends in the variety and scale of in vitro production of plant triterpenoids have also been discussed.
Collapse
Affiliation(s)
- Tanya Biswas
- Department of Biochemistry, University of Lucknow, Lucknow, 226007, India
| | - Upendra N Dwivedi
- Department of Biochemistry, University of Lucknow, Lucknow, 226007, India.
- Institute for Development of Advanced Computing, ONGC Centre for Advanced Studies, University of Lucknow, Lucknow, 226007, India.
| |
Collapse
|
14
|
Bhattacharyya S, Mitra D, Ray S, Biswas N, Banerjee S, Majumder B, Mustafi SM, Murmu N. Reversing effect of Lupeol on vasculogenic mimicry in murine melanoma progression. Microvasc Res 2018; 121:52-62. [PMID: 30381268 DOI: 10.1016/j.mvr.2018.10.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/16/2018] [Accepted: 10/24/2018] [Indexed: 12/11/2022]
Abstract
Vasculogenic mimicry, an endothelia-independent tumor microcirculation has been found in various cancers and is thought to be achieved by cancer stem like cells. Dacarbazine resistance is one of the most common features of melanoma and recent studies suggest that the mode of resistance is closely related to the formation of vasculogenic mimicry. In our work, we examined the anticancer effect of Lupeol, a novel phytochemical with Dacarbazine in vivo and in vitro. Results demonstrated adequate cytotoxicity followed by down regulation of CD 133 expression in Lupeol treated B16-F10 cell line. In solid tumor model the drug also inhibited vasculogenic mimicry along with angiogenesis by altering both the cancer stem cell as well as the endothelial progenitor cell population. Lupeol hindered the maturation of bone marrow derived endothelial progenitors and thus, retarded the formation of rudimentary tumor microvessels. Notably, Dacarbazine treatment demonstrated unresponsiveness to B16-F10 cells in both in vivo and in vitro model via upregulation of CD 133 expression and increased formation of vasculogenic mimicry tubes. Together, these data indicate that Lupeol alone can become a proficient agent in treating melanoma, inhibiting vasculogenic mimicry and might play a significant role in subduing Dacarbazine induced drug resistance.
Collapse
Affiliation(s)
- Sayantan Bhattacharyya
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Debarpan Mitra
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Sudipta Ray
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Nirjhar Biswas
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Samir Banerjee
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Biswanath Majumder
- Department of Molecular Pathology and Cancer Biology, Mitra Biotech, 202, Narayana Nethralaya, Hosur Main Road, Bangalore 560099, India
| | - Saunak Mitra Mustafi
- Department of Pathology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Nabendu Murmu
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India.
| |
Collapse
|
15
|
Pereira RCG, Soares DCF, Oliveira DCP, de Sousa GF, Vieira-Filho SA, Mercadante-Simões MO, Lula I, Silva-Cunha A, Duarte LP. Triterpenes from leaves of Cheiloclinium cognatum and their in vivo antiangiogenic activity. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2018; 56:360-366. [PMID: 29388257 DOI: 10.1002/mrc.4716] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 06/07/2023]
Affiliation(s)
- Rafael C G Pereira
- Departamento de Química, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Pampulha, CEP, Belo Horizonte, MG, 31270-901, Brazil
| | - Daniel C F Soares
- Universidade Federal de Itajubá, Campus Itabira, Rua Irmã Ivone Drumond, 200, Distrito Industrial II, CEP, Itabira, MG, 35903-087, Brazil
| | - Diogo C P Oliveira
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Pampulha, CEP, Belo Horizonte, MG, 31270-901, Brazil
| | - Grasiely F de Sousa
- Departamento de Química, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Pampulha, CEP, Belo Horizonte, MG, 31270-901, Brazil
| | - Sidney A Vieira-Filho
- Escola de Farmácia, Universidade Federal de Ouro Preto, Campus Morro do Cruzeiro, s/n, CEP, Ouro Preto, MG, 35400-000, Brazil
| | - Maria O Mercadante-Simões
- Departamento de Biologia Geral, Universidade de Montes Claros, Avenida Dr. Ruy Braga, s/n, CEP, Montes Claros, MG, 39401-089, Brazil
| | - Ivana Lula
- Departamento de Química, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Pampulha, CEP, Belo Horizonte, MG, 31270-901, Brazil
| | - Armando Silva-Cunha
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Pampulha, CEP, Belo Horizonte, MG, 31270-901, Brazil
| | - Lucienir P Duarte
- Departamento de Química, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, Pampulha, CEP, Belo Horizonte, MG, 31270-901, Brazil
| |
Collapse
|
16
|
Mohammed YHE, Ara Khanum S. The critical role of novel benzophenone analogs on tumor growth inhibition targeting angiogenesis and apoptosis. MEDCHEMCOMM 2018; 9:639-656. [PMID: 30108955 PMCID: PMC6072443 DOI: 10.1039/c7md00593h] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/14/2018] [Indexed: 11/21/2022]
Abstract
In modern biology, one of the major topics of importance is progress in anti-cancer drugs with specific targets. The angiopreventive and in vitro tumor inhibition activities of novel synthetic benzophenone analogs have been investigated intensively and explored in a very systematic way. Novel benzophenone analogs (9a-d and 10a-d) substituted with methyl, chloro and fluoro groups at different positions on an identical chemical backbone and incorporating variations in the number of substituents have been synthesized in a multistep process and characterized. In this study, we further evaluate the newly synthesized compounds for their cytotoxic and anti-proliferative effects against A549, HeLa and MCF-7 cells. The potent lead compound was further assessed for anti-angiogenic effects. Through the structure-activity relationship, we found that an increase in the number of methyl, chloro and fluoro groups in a benzophenone ring on compound 9d resulted in higher potency compared to other compounds. Tumor inhibition was notably promoted, and this was reflected in effects on neovessel formation in in vivo systems, such as the CAM. Compound 9d interacts with rVEGF through hydrogen bonds in silico, thereby down-regulating the expression of VEGF in angiogenesis. From our investigation, it is suggested on the basis of clonogenesis and cell migration assays that compound 9d has the potency to exhibit prolonged activity against cancer progression, through cell cycle arrest at the G2/M phase. In addition, compound 9d inhibits A549 cells through caspase-activated DNase-mediated apoptosis.
Collapse
Affiliation(s)
- Yasser Hussein Eissa Mohammed
- Department of Chemistry , Yuvaraja's College , University of Mysore , Mysore -570005 , Karnataka , India . ; ; Tel: +91 99018 88755
- Department of Biochemistry , Faculty of Applied Science College , University of Hajjah , Yemen
| | - Shaukath Ara Khanum
- Department of Chemistry , Yuvaraja's College , University of Mysore , Mysore -570005 , Karnataka , India . ; ; Tel: +91 99018 88755
| |
Collapse
|
17
|
Eissa Mohammed YH, Thirusangu P, Zabiulla, V V, B.T P, Khanum SA. The anti-invasive role of novel synthesized pyridazine hydrazide appended phenoxy acetic acid against neoplastic development targeting matrix metallo proteases. Biomed Pharmacother 2017; 95:375-386. [DOI: 10.1016/j.biopha.2017.08.105] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 08/16/2017] [Accepted: 08/23/2017] [Indexed: 01/26/2023] Open
|
18
|
Schmidt C, Loos C, Jin L, Schmiech M, Schmidt CQ, Gaafary ME, Syrovets T, Simmet T. Acetyl-lupeolic acid inhibits Akt signaling and induces apoptosis in chemoresistant prostate cancer cells in vitro and in vivo. Oncotarget 2017; 8:55147-55161. [PMID: 28903409 PMCID: PMC5589648 DOI: 10.18632/oncotarget.19101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 06/27/2017] [Indexed: 11/30/2022] Open
Abstract
The triterpenoid acetyl-lupeolic acid (ac-LA) isolated from the oleogum resin of Boswellia carterii reduced the viability of a panel of cancer cell lines more efficiently than lupeol. There was no detectable intracellular conversion of ac-LA to lupeol and vice versa. In contrast to docetaxel, ac-LA did not induce selection of treatment-resistant cancer cells. By various parameters including DNA fragmentation, ac-LA was shown to induce apoptosis in androgen-independent PC-3 cells, whereas in MDA-MB-231 breast cancer cells, ac-LA led to cell accumulation in the G2/M phase of the cell cycle, but not to apoptosis. In silico docking combined with in vitro kinase assays implied that ac LA potently inhibits Akt mainly by direct binding to the pleckstrin homology domain. Consistently, an Akt1 mutant deficient of the PH domain afforded partial resistance to ac-LA and complete resistance to lupeol and the Akt inhibitor III. Ac-LA inhibited phosphorylation of downstream targets of the Akt signaling pathway, which was followed by inhibition of the mTOR target p70 ribosomal six protein kinase and the nuclear accumulation of p65/NF-κB, β-catenin, and c-myc, as well as loss of the mitochondrial membrane potential. Ac-LA exhibited antiproliferative, proapoptotic, and antitumorigenic effects on PC-3-tumors xenografted either on chick chorioallantoic membranes or in nude mice. Ac-LA exhibited a clearly better safety profile than docetaxel or lupeol during chronic administration in vivo. In contrast to lupeol, ac-LA also inhibited release of vascular endothelial growth factor in vitro and accordingly angiogenesis in vivo. Thus, ac-LA deserves further exploration as a potential new antitumor compound.
Collapse
Affiliation(s)
- Claudia Schmidt
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany.,Present address: Rommelag CMO, Sulzbach-Laufen, Germany
| | - Cornelia Loos
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany.,Present address: Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| | - Lu Jin
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Michael Schmiech
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Christoph Q Schmidt
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Menna El Gaafary
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany.,Present address: Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt
| | - Tatiana Syrovets
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| |
Collapse
|
19
|
Vigneshwaran V, Thirusangu P, Vijay Avin BR, Krishna V, Pramod SN, Prabhakar BT. Immunomodulatory glc/man-directed Dolichos lablab lectin (DLL) evokes anti-tumour response in vivo by counteracting angiogenic gene expressions. Clin Exp Immunol 2017; 189:21-35. [PMID: 28268243 DOI: 10.1111/cei.12959] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2017] [Indexed: 12/31/2022] Open
Abstract
Neovascularization and jeopardized immunity has been critically emphasized for the establishment of malignant progression. Lectins are the diverse class of carbohydrate interacting proteins, having great potential as immunopotentiating and anti-cancer agents. The present investigation sought to demonstrate the anti-proliferative activity of Dolichos lablab lectin (DLL) encompassing immunomodulatory attributes. DLL specific to glucose and mannose carbohydrate moieties has been purified to homogeneity from the common dietary legume D. lablab. Results elucidated that DLL agglutinated blood cells non-specifically and displayed striking mitogenicity to human and murine lymphocytes in vitro with interleukin (IL)-2 production. The DLL-conditioned medium exerted cytotoxicity towards malignant cells and neoangiogenesis in vitro. Similarly, in-vivo anti-tumour investigation of DLL elucidated the regressed proliferation of ascitic and solid tumour cells, which was paralleled with blockade of tumour neovasculature. DLL-treated mice showed an up-regulated immunoregulatory cytokine IL-2 in contrast to severely declined levels in control mice. Mechanistic validation revealed that DLL has abrogated the microvessel formation by weakening the proangiogenic signals, specifically nuclear factor kappa B (NF-κB), hypoxia inducible factor 1α (HIF-1 α), matrix metalloproteinase (MMP)-2 and 9 and vascular endothelial growth factor (VEGF) in malignant cells leading to tumour regression. In summary, it is evident that the dietary lectin DLL potentially dampens the malignant establishment by mitigating neoangiogenesis and immune shutdown. For the first time, to our knowledge, this study illustrates the critical role of DLL as an immunostimulatory and anti-angiogenic molecule in cancer therapeutics.
Collapse
Affiliation(s)
- V Vigneshwaran
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, India
- Laboratory for Immunomodulation and Inflammation Biology, Department of Studies and Research in Biochemistry, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, India
| | - P Thirusangu
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, India
| | - B R Vijay Avin
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, India
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - V Krishna
- Postgraduate Department of Studies and Research in Biotechnology and Bioinformatics, Kuvempu University, Shankaraghatta, Shivamogga, Karnataka, India
| | - S N Pramod
- Laboratory for Immunomodulation and Inflammation Biology, Department of Studies and Research in Biochemistry, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, India
| | - B T Prabhakar
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, India
| |
Collapse
|
20
|
Scutellarein antagonizes the tumorigenesis by modulating cytokine VEGF mediated neoangiogenesis and DFF-40 actuated nucleosomal degradation. Biochem Biophys Res Commun 2017; 484:85-92. [DOI: 10.1016/j.bbrc.2017.01.067] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 01/14/2017] [Indexed: 12/30/2022]
|
21
|
A tumoural angiogenic gateway blocker, Benzophenone-1B represses the HIF-1α nuclear translocation and its target gene activation against neoplastic progression. Biochem Pharmacol 2016; 125:26-40. [PMID: 27838496 DOI: 10.1016/j.bcp.2016.11.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/07/2016] [Indexed: 11/21/2022]
Abstract
Hypoxia is an important module in all solid tumours to promote angiogenesis, invasion and metastasis. Stabilization and subsequent nuclear localization of HIF-1α subunits result in the activation of tumour promoting target genes such as VEGF, MMPs, Flt-1, Ang-1 etc. which plays a pivotal role in adaptation of tumour cells to hypoxia. Increased HIF-α and its nuclear translocation have been correlated with pronounced angiogenesis, aggressive tumour growth and poor patient prognosis leading to current interest in HIF-1α as an anticancer drug target. Benzophenone-1B ([4-(1H-benzimidazol-2-ylmethoxy)-3,5-dimethylphenyl]-(4-methoxyphenyl) methanone, or BP-1B) is a new antineoplastic agent with potential angiopreventive effects. Current investigation reports the cellular biochemical modulation underlying BP-1B cytotoxic/antiangiogenic effects. Experimental evidences postulate that BP-1B exhibits the tumour specific cytotoxic actions against various cancer types with prolonged action. Moreover BP-1B efficiently counteracts endothelial cell capillary formation in in-vitro, in-vivo non-tumour and tumour angiogenic systems. Molecular signaling studies reveal that BP-1B arrests nuclear translocation of HIF-1α devoid of p42/44 pathway under CoCl2 induced hypoxic conditions in various cancer cells thereby leading to abrogated HIF-1α dependent activation of VEGF-A, Flt-1, MMP-2, MMP -9 and Ang-1 angiogenic factors resulting in retarded cell migration and invasions. The in-vitro results were reproducible in the reliable in-vivo solid tumour model. Taken together, we conclude that BP-1B impairs angiogenesis by blocking nuclear localization of HIF-1α which can be translated into a potent HIF-1α inhibitor.
Collapse
|
22
|
Thirusangu P, Vigneshwaran V, Prashanth T, Vijay Avin BR, Malojirao VH, Rakesh H, Khanum SA, Mahmood R, Prabhakar BT. BP-1T, an antiangiogenic benzophenone-thiazole pharmacophore, counteracts HIF-1 signalling through p53/MDM2-mediated HIF-1α proteasomal degradation. Angiogenesis 2016; 20:55-71. [PMID: 27743086 DOI: 10.1007/s10456-016-9528-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 10/07/2016] [Indexed: 12/16/2022]
Abstract
Hypoxia is a feature of all solid tumours, contributing to tumour progression. Activation of HIF-1α plays a critical role in promoting tumour angiogenesis and metastasis. Since its expression is positively correlated with poor prognosis for cancer patients, HIF-1α is one of the most convincing anticancer targets. BP-1T is a novel antiproliferative agent with promising antiangiogenic effects. In the present study, the molecular mechanism underlying cytotoxic/antiangiogenic effects of BP-1T on tumour/non-tumour angiogenesis was evaluated. Evidences show that BP-1T exhibits potent cytotoxicity with prolonged activity and effectively regressed neovessel formation both in reliable non-tumour and tumour angiogenic models. The expression of CoCl2-induced HIF-1α was inhibited by BP-1T in various p53 (WT)-expressing cancer cells, including A549, MCF-7 and DLA, but not in mutant p53-expressing SCC-9 cells. Mechanistically, BP-1T mediates the HIF-1α proteasomal degradation by activating p53/MDM2 pathway and thereby downregulated HIF-1α-dependent angiogenic genes such as VEGF-A, Flt-1, MMP-2 and MMP-9 under hypoxic condition of in vitro and in vivo solid tumour, eventually leading to abolition of migration and invasion. Based on these observations, we conclude that BP-1T acts on HIF-1α degradation through p53/MDM2 proteasome pathway.
Collapse
Affiliation(s)
- Prabhu Thirusangu
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, 577203, India
| | - V Vigneshwaran
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, 577203, India
| | - T Prashanth
- Department of Chemistry, Yuvaraja's College (Autonomous), University of Mysore, Mysore, Karnataka, 570 005, India
| | - B R Vijay Avin
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, 577203, India
- Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Vikas H Malojirao
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, 577203, India
| | - H Rakesh
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, 577203, India
| | - Shaukath Ara Khanum
- Department of Chemistry, Yuvaraja's College (Autonomous), University of Mysore, Mysore, Karnataka, 570 005, India
| | - Riaz Mahmood
- Postgraduate Department of Studies and Research in Biotechnology and Bioinformatics, Kuvempu University, Shankaraghatta, Shivamogga, Karnataka, 577203, India
| | - B T Prabhakar
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga, Karnataka, 577203, India.
| |
Collapse
|
23
|
Vigneshwaran V, Thirusangu P, Madhusudana S, Krishna V, Pramod SN, Prabhakar B. The latex sap of the ‘Old World Plant’ Lagenaria siceraria with potent lectin activity mitigates neoplastic malignancy targeting neovasculature and cell death. Int Immunopharmacol 2016; 39:158-171. [DOI: 10.1016/j.intimp.2016.07.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 07/18/2016] [Accepted: 07/22/2016] [Indexed: 01/13/2023]
|
24
|
Ambasta RK, Jha SK, Kumar D, Sharma R, Jha NK, Kumar P. Comparative study of anti-angiogenic activities of luteolin, lectin and lupeol biomolecules. J Transl Med 2015; 13:307. [PMID: 26385094 PMCID: PMC4575424 DOI: 10.1186/s12967-015-0665-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 09/08/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Angiogenesis is a hallmark feature in the initiation, progression and growth of tumour. There are various factors for promotion of angiogenesis on one hand and on the other hand, biomolecules have been reported to inhibit cancer through anti-angiogenesis mechanism. Biomolecules, for instance, luteolin, lectin and lupeol are known to suppress cancer. This study aims to compare and evaluate the biomolecule(s) like luteolin, lupeol and lectin on CAM assay and HT-29 cell culture to understand the efficacy of these drugs. METHOD The biomolecules have been administered on CAM assay, HT-29 cell culture, cell migration assay. Furthermore, bioinformatics analysis of the identified targets of these biomolecules have been performed. RESULT Luteolin has been found to be better in inhibiting angiogenesis on CAM assay in comparison to lupeol and lectin. In line with this study when biomolecules was administered on cell migration assay via scratch assay method. We provided evidence that Luteolin was again found to be better in inhibiting HT-29 cell migration. In order to identify the target sites of luteolin for inhibition, we used software analysis for identifying the best molecular targets of luteolin. Using software analysis best target protein molecule of these biomolecules have been identified. VEGF was found to be one of the target of luteolin. Studies have found several critical point mutation in VEGF A, B and C. Hence docking analysis of all biomolecules with VEGFR have been performed. Multiple allignment result have shown that the receptors are conserved at the docking site. CONCLUSION Therefore, it can be concluded that luteolin is not only comparatively better in inhibiting blood vessel in CAM assay, HT-29 cell proliferation and cell migration assay rather the domain of VEGFR is conserved to be targeted by luteolin, lupeol and lectin.
Collapse
Affiliation(s)
- Rashmi K Ambasta
- Department of Biotechnology, Delhi Technological University (Former Delhi College of Engineering), Delhi, India. .,School of Biosciences and Technology, Vellore Institute of Technology, University (VITU), Vellore, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, Delhi Technological University (Former Delhi College of Engineering), Delhi, India. .,School of Biosciences and Technology, Vellore Institute of Technology, University (VITU), Vellore, India.
| | - Dhiraj Kumar
- Department of Biotechnology, Delhi Technological University (Former Delhi College of Engineering), Delhi, India.
| | - Renu Sharma
- Department of Biotechnology, Delhi Technological University (Former Delhi College of Engineering), Delhi, India.
| | - Niraj Kumar Jha
- Department of Biotechnology, Delhi Technological University (Former Delhi College of Engineering), Delhi, India. .,School of Biosciences and Technology, Vellore Institute of Technology, University (VITU), Vellore, India.
| | - Pravir Kumar
- Department of Biotechnology, Delhi Technological University (Former Delhi College of Engineering), Delhi, India. .,School of Biosciences and Technology, Vellore Institute of Technology, University (VITU), Vellore, India. .,Neurology Department, Adjunct Faculty, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
25
|
Okino Delgado CH, Fleuri LF. Orange and mango by-products: Agro-industrial waste as source of bioactive compounds and botanical versus commercial description—A review. FOOD REVIEWS INTERNATIONAL 2015. [DOI: 10.1080/87559129.2015.1041183] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|