1
|
Valle-Sánchez SL, Rodríguez-Ramírez R, Ávila-Villa LA, Villa-Lerma AG, Wall-Medrano A, de la Rosa LA, Muñoz-Bernal ÓA, González-Córdova AF, Arellano-Gil M. Phenolic compounds profile in extracts of Smilax spp., antioxidant activity, and inhibition of advanced glycation end products. Food Chem 2025; 463:141389. [PMID: 39332373 DOI: 10.1016/j.foodchem.2024.141389] [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: 04/25/2024] [Revised: 09/05/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024]
Abstract
Smilax genus possesses bioactive properties attributed to phenolic compounds, which may exhibit antioxidant effects and inhibit the advanced glycation end products (AGEs). However, identifying these phenolic compounds and AGEs has become increasingly relevant to understanding such activities. This study aimed to identify phenolic compounds in extracts of Smilax spp. and evaluate their antioxidant and AGEs inhibitory activities. To achieve this, the Smilax genus was identified via PCR, and phenolic compounds including chlorogenic acid, naringenin-6-C-glucoside, quercetin, quercetin-3-O-glucoside, and myricetin were identified using HPLC-MS/MS. Antioxidant activity was assessed by ferric reducing antioxidant power (FRAP), and radicals such as 2,2-diphenyl-1-picrylhydrazyl (DPPH), and 2,2'-azino-bis-[3-ethyl-benzothiazoline]-6-sulfonic acid (ABTS), while AGEs inhibition was evaluated using a model system formed by bovine serum albumin-glucose. The highest antioxidant activity was 3612.18 mM TE/g, and the inhibition of AGEs was 52.44 %. These results demonstrate that Smilax spp. can inhibit AGEs, neutralize free radicals, and reduce compounds associated with antioxidant capacity.
Collapse
Affiliation(s)
- Sergio Luis Valle-Sánchez
- Laboratorio de Biotecnología y Trazabilidad Molecular de los Alimentos, Instituto Tecnológico de Sonora, 5 de febrero, 818 sur, C.P. 85000 Ciudad Obregón, Sonora, Mexico
| | - Roberto Rodríguez-Ramírez
- Laboratorio de Biotecnología y Trazabilidad Molecular de los Alimentos, Instituto Tecnológico de Sonora, 5 de febrero, 818 sur, C.P. 85000 Ciudad Obregón, Sonora, Mexico.
| | - Luz Angélica Ávila-Villa
- Departamento de Ciencias de la Salud, Universidad de Sonora, C.P. 85010 Ciudad Obregón, Sonora, Mexico
| | - Alma Guadalupe Villa-Lerma
- Laboratorio de Biotecnología y Trazabilidad Molecular de los Alimentos, Instituto Tecnológico de Sonora, 5 de febrero, 818 sur, C.P. 85000 Ciudad Obregón, Sonora, Mexico
| | - Abraham Wall-Medrano
- Instituto de Ciencias Biomédicas, Av. Benjamín Franklin 4650, Zona Pronaf Condominio La Plata, C.P. 32310, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, Chihuahua, Mexico
| | - Laura Alejandra de la Rosa
- Instituto de Ciencias Biomédicas, Av. Benjamín Franklin 4650, Zona Pronaf Condominio La Plata, C.P. 32310, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, Chihuahua, Mexico
| | - Óscar Adrián Muñoz-Bernal
- Instituto de Ciencias Biomédicas, Av. Benjamín Franklin 4650, Zona Pronaf Condominio La Plata, C.P. 32310, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, Chihuahua, Mexico
| | - Aarón Fernando González-Córdova
- Coordinación de Tecnología de Alimentos de Origen Animal, Centro de Investigación en Alimentación y Desarrollo, A. C., C.P. 83000 Hermosillo, Sonora, Mexico
| | - Maritza Arellano-Gil
- Laboratorio de Biotecnología y Trazabilidad Molecular de los Alimentos, Instituto Tecnológico de Sonora, 5 de febrero, 818 sur, C.P. 85000 Ciudad Obregón, Sonora, Mexico
| |
Collapse
|
2
|
Ashrafizadeh M, Aref AR, Sethi G, Ertas YN, Wang L. Natural product/diet-based regulation of macrophage polarization: Implications in treatment of inflammatory-related diseases and cancer. J Nutr Biochem 2024; 130:109647. [PMID: 38604457 DOI: 10.1016/j.jnutbio.2024.109647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Macrophages are phagocytic cells with important physiological functions, including the digestion of cellular debris, foreign substances, and microbes, as well as tissue development and homeostasis. The tumor microenvironment (TME) shapes the aggressiveness of cancer, and the biological and cellular interactions in this complicated space can determine carcinogenesis. TME can determine the progression, biological behavior, and therapy resistance of human cancers. The macrophages are among the most abundant cells in the TME, and their functions and secretions can determine tumor progression. The education of macrophages to M2 polarization can accelerate cancer progression, and therefore, the re-education and reprogramming of these cells is promising. Moreover, macrophages can cause inflammation in aggravating pathological events, including cardiovascular diseases, diabetes, and neurological disorders. The natural products are pleiotropic and broad-spectrum functional compounds that have been deployed as ideal alternatives to conventional drugs in the treatment of cancer. The biological and cellular interactions in the TME can be regulated by natural products, and for this purpose, they enhance the M1 polarization of macrophages, and in addition to inhibiting proliferation and invasion, they impair the chemoresistance. Moreover, since macrophages and changes in the molecular pathways in these cells can cause inflammation, the natural products impair the pro-inflammatory function of macrophages to prevent the pathogenesis and progression of diseases. Even a reduction in macrophage-mediated inflammation can prevent organ fibrosis. Therefore, natural product-mediated macrophage targeting can alleviate both cancerous and non-cancerous diseases.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Translational Sciences, Xsphera Biosciences Inc., Boston, Massachusetts, USA
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Türkiye; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Türkiye.
| | - Lu Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
3
|
Ravi S, Duraisamy P, Krishnan M, Martin LC, Manikandan B, Ramar M. Sitosterol-rich Digera muricata against 7-ketocholesterol and lipopolysaccharide-mediated atherogenic responses by modulating NF-ΚB/iNOS signalling pathway in macrophages. 3 Biotech 2023; 13:331. [PMID: 37670802 PMCID: PMC10475456 DOI: 10.1007/s13205-023-03741-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/10/2023] [Indexed: 09/07/2023] Open
Abstract
Digera muricata L., commonly known as Tartara, is an edible herb used as traditional medicine in many countries of Africa and Asia. This study aimed to elucidate the effect of a phytosterol-rich extract of D. muricata on 7-ketocholesterol-mediated atherosclerosis in macrophages. The extract was examined by phytochemical analyses, GC-MS, TLC, DPPH scavenging and hRBC membrane stabilization assays. Macrophage polarization was studied with experimental groups framed based on alamar blue cell viability and griess assays. Regulations of arginase enzyme activity, ROS generation, mitochondrial membrane potential, cell membrane integrity, pinocytosis, lipid uptake and peroxidation, as well as, intracellular calcium deposition were determined. In addition, expressions of atherogenic mediators were analysed using PCR, ELISA and immunocytochemistry techniques. Diverse phytochemicals with higher free radical scavenging activity and anti-inflammatory potential have been detected in the D. muricata. Co-treatment with D. muricata markedly reduced the atherogenic responses induced by 7KCh in the presence of LPS such as ROS, especially, NO and O2- along with lipid peroxidation. Furthermore, D. muricata significantly normalized mitochondrial membrane potential, cell membrane integrity, pinocytic activity, intracellular lipid accumulation and calcium deposition. These results provided us with the potentiality of D. muricata in ameliorating atherogenesis. Additionally, it decreased the expression of pro-atherogenic mediators (iNOS, COX-2, MMP9, IL-6, IL-1β, CD36, CD163 and TGFβ1) and increased anti-atherogenic mediators (MRC1 and PPARγ) with high cellular expressions of NF-κB and iNOS. Results showed the potential of sitosterol-rich D. muricata as a versatile biomedical therapeutic agent against abnormal macrophage polarization and its associated pathologies.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| | | | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni’s College for Women, Chennai, 600 015 India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| |
Collapse
|
4
|
Lekshmi VS, Asha K, Sanicas M, Asi A, Arya UM, Kumar B. PI3K/Akt/Nrf2 mediated cellular signaling and virus-host interactions: latest updates on the potential therapeutic management of SARS-CoV-2 infection. Front Mol Biosci 2023; 10:1158133. [PMID: 37325475 PMCID: PMC10267462 DOI: 10.3389/fmolb.2023.1158133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
The emergence and re-emergence of viral diseases, which cause significant global mortality and morbidity, are the major concerns of this decade. Of these, current research is focused majorly on the etiological agent of the COVID-19 pandemic, SARS-CoV-2. Understanding the host response and metabolic changes during viral infection may provide better therapeutic targets for the proper management of pathophysiological conditions associated with SARS-CoV-2 infection. We have achieved control over most emerging viral diseases; however, a lack of understanding of the underlying molecular events prevents us from exploring novel therapeutic targets, leaving us forced to witness re-emerging viral infections. SARS-CoV-2 infection is usually accompanied by oxidative stress, which leads to an overactive immune response, the release of inflammatory cytokines, increasing lipid production, and also alterations in the endothelial and mitochondrial functions. PI3K/Akt signaling pathway confers protection against oxidative injury by various cell survival mechanisms including Nrf2-ARE mediated antioxidant transcriptional response. SARS-CoV-2 is also reported to hijack this pathway for its survival within host and few studies have suggested the role of antioxidants in modulating the Nrf2 pathway to manage disease severity. This review highlights the interrelated pathophysiological conditions associated with SARS-CoV-2 infection and the host survival mechanisms mediated by PI3K/Akt/Nrf2 signaling pathways that can help ameliorate the severity of the disease and provide effective antiviral targets against SARS-CoV-2.
Collapse
Affiliation(s)
- V. S. Lekshmi
- Department of Antiviral Research, Institute of Advanced Virology, Thiruvananthapuram, Kerala, India
| | - Kumari Asha
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | | | - Abhila Asi
- Department of Antiviral Research, Institute of Advanced Virology, Thiruvananthapuram, Kerala, India
| | - U. M. Arya
- Department of Antiviral Research, Institute of Advanced Virology, Thiruvananthapuram, Kerala, India
| | - Binod Kumar
- Department of Antiviral Research, Institute of Advanced Virology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
5
|
El-Zahar H, Menze ET, Handoussa H, Osman AK, El-Shazly M, Mostafa NM, Swilam N. UPLC-PDA-MS/MS Profiling and Healing Activity of Polyphenol-Rich Fraction of Alhagi maurorum against Oral Ulcer in Rats. PLANTS (BASEL, SWITZERLAND) 2022; 11:455. [PMID: 35161436 PMCID: PMC8838639 DOI: 10.3390/plants11030455] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 05/04/2023]
Abstract
Camelthorn, Alhagi maurorum Boiss, family Fabaceae has long been used in African folk medicine owing to its richness in pharmacologically active metabolites. The crude extract (CEAM), ethyl acetate fraction (EFAM) and n-butanol (BFAM) fraction of A. maurorum aerial parts were investigated for their total polyphenols and oral antiulcer activity using in-vitro and in-vivo models. The major phenolic compound was isolated from the polyphenol-rich EFAM fraction and identified by conventional and spectroscopic methods of analysis as isorhamnetin-3-O-rutinoside. Furthermore, standardization of EAFM using UPLC-PDA-UV quantified isorhamnetin-3-O-rutinoside as 262.91 0.57 g/mg of the fraction. Analysis of EFAM using UPLC-PDA-MS/MS revealed tentative identification of 25 polyphenolic compounds. EFAM exhibited the most potent free radical scavenging activity against DPPH, with an IC50 (27.73 ± 1.85 µg/mL) and an FRAP value of (176.60 ± 5.21 μM Trolox equivalent (TE)/mg fraction) in comparison with CEAM and BFAM. Acetic acid-induced oral ulcers in a rat model were used to evaluate the healing properties of A. maurorum aerial parts. EFAM significantly decreased tumor necrosis factor-alpha (TNF-α) and interleukin-2 (IL-2) by 36.4% and 50.8%, respectively, in the ulcer tissues while, CEAM and BFAM exhibited lower activity at the same dose. In addition, EFAM led to a significant (p < 0.0001) rise in the expression of proliferating cell nuclear antigen (PCNA), a cell proliferation marker. A. maurorum exhibited a potent healing effect in acetic acid-induced oral ulcers in rats by mitigating the release of pro-inflammatory cytokines and improving PCNA expression.
Collapse
Affiliation(s)
- Hala El-Zahar
- Department of Pharmaceutical Sciences, Faculty of Dentistry, British University in Egypt (BUE), Cairo 11837, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Heba Handoussa
- Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
| | - Ahmed K Osman
- Department of Botany and Microbiology, Faculty of Science, South Valley University, Qena 83523, Egypt
| | - Mohamed El-Shazly
- Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Nada M Mostafa
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Noha Swilam
- Department of Pharmacognosy, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo 11837, Egypt
| |
Collapse
|
6
|
Liskova A, Koklesova L, Samec M, Abdellatif B, Zhai K, Siddiqui M, Šudomová M, Hassan ST, Kudela E, Biringer K, Giordano FA, Büsselberg D, Golubnitschaja O, Kubatka P. Targeting phytoprotection in the COVID-19-induced lung damage and associated systemic effects-the evidence-based 3PM proposition to mitigate individual risks. EPMA J 2021; 12:325-347. [PMID: 34367380 PMCID: PMC8329620 DOI: 10.1007/s13167-021-00249-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/03/2021] [Indexed: 02/06/2023]
Abstract
The risks related to the COVID-19 are multi-faceted including but by far not restricted to the following: direct health risks by poorly understood effects of COVID-19 infection, overloaded capacities of healthcare units, restricted and slowed down care of patients with non-communicable disorders such as cancer, neurologic and cardiovascular pathologies, among others; social risks-restricted and broken social contacts, isolation, professional disruption, explosion of aggression in the society, violence in the familial environment; mental risks-loneliness, helplessness, defenceless, depressions; and economic risks-slowed down industrial productivity, broken delivery chains, unemployment, bankrupted SMEs, inflation, decreased capacity of the state to perform socially important programs and to support socio-economically weak subgroups in the population. Directly or indirectly, the above listed risks will get reflected in a healthcare occupation and workload which is a tremendous long-term challenge for the healthcare capacity and robustness. The article does not pretend to provide solutions for all kind of health risks. However, it aims to present the scientific evidence of great clinical utility for primary, secondary, and tertiary care to protect affected individuals in a cost-effective manner. To this end, due to pronounced antimicrobial, antioxidant, anti-inflammatory, and antiviral properties, naturally occurring plant substances are capable to protect affected individuals against COVID-19-associated life-threatening complications such as lung damage. Furthermore, they can be highly effective, if being applied to secondary and tertiary care of noncommunicable diseases under pandemic condition. Thus, the stratification of patients evaluating specific health conditions such as sleep quality, periodontitis, smoking, chronic inflammation and diseases, metabolic disorders and obesity, vascular dysfunction, and cancers would enable effective managemenet of COVID-19-associated complications in primary, secondary, and tertiary care in the context of predictive, preventive, and personalized medicine (3PM).
Collapse
Affiliation(s)
- Alena Liskova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Lenka Koklesova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Marek Samec
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Basma Abdellatif
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, 24144 Qatar
| | - Kevin Zhai
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, 24144 Qatar
| | - Manaal Siddiqui
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, 24144 Qatar
| | - Miroslava Šudomová
- Museum of Literature in Moravia, Klášter 1, 66461, Rajhrad, Czech Republic
| | - Sherif T.S. Hassan
- Department of Applied Ecology, Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 16500 Prague, Czech Republic
| | - Erik Kudela
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Kamil Biringer
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Frank A. Giordano
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität, Bonn, Germany
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, 24144 Qatar
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
7
|
Mucha P, Skoczyńska A, Małecka M, Hikisz P, Budzisz E. Overview of the Antioxidant and Anti-Inflammatory Activities of Selected Plant Compounds and Their Metal Ions Complexes. Molecules 2021; 26:4886. [PMID: 34443474 PMCID: PMC8398118 DOI: 10.3390/molecules26164886] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous plant compounds and their metal-ion complexes exert antioxidative, anti-inflammatory, anticancer, and other beneficial effects. This review highlights the different bioactivities of flavonoids, chromones, and coumarins and their metal-ions complexes due to different structural characteristics. In addition to insight into the most studied antioxidative properties of these compounds, the first part of the review provides a comprehensive overview of exogenous and endogenous sources of reactive oxygen and nitrogen species, oxidative stress-mediated damages of lipids and proteins, and on protective roles of antioxidant defense systems, including plant-derived antioxidants. Additionally, the review covers the anti-inflammatory and antimicrobial activities of flavonoids, chromones, coumarins and their metal-ion complexes which support its application in medicine, pharmacy, and cosmetology.
Collapse
Affiliation(s)
- Paulina Mucha
- Department of the Chemistry of Cosmetic Raw Materials, Faculty of Pharmacy, Medical University of Łódź, Muszyńskiego 1, 90-151 Łódź, Poland
| | - Anna Skoczyńska
- Department of Pharmacology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Poniatowskiego 15, 41-200 Sosnowiec, Poland;
| | - Magdalena Małecka
- Department of Physical Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163/165, 90-236 Łódź, Poland;
| | - Paweł Hikisz
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Łódź, Poland;
| | - Elzbieta Budzisz
- Department of the Chemistry of Cosmetic Raw Materials, Faculty of Pharmacy, Medical University of Łódź, Muszyńskiego 1, 90-151 Łódź, Poland
| |
Collapse
|
8
|
Liskova A, Samec M, Koklesova L, Samuel SM, Zhai K, Al-Ishaq RK, Abotaleb M, Nosal V, Kajo K, Ashrafizadeh M, Zarrabi A, Brockmueller A, Shakibaei M, Sabaka P, Mozos I, Ullrich D, Prosecky R, La Rocca G, Caprnda M, Büsselberg D, Rodrigo L, Kruzliak P, Kubatka P. Flavonoids against the SARS-CoV-2 induced inflammatory storm. Biomed Pharmacother 2021; 138:111430. [PMID: 33662680 PMCID: PMC7906511 DOI: 10.1016/j.biopha.2021.111430] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/15/2021] [Accepted: 02/21/2021] [Indexed: 12/11/2022] Open
Abstract
The disease severity of COVID-19, especially in the elderly and patients with co-morbidities, is characterized by hypercytokinemia, an exaggerated immune response associated with an uncontrolled and excessive release of proinflammatory cytokine mediators (cytokine storm). Flavonoids, important secondary metabolites of plants, have long been studied as therapeutic interventions in inflammatory diseases due to their cytokine-modulatory effects. In this review, we discuss the potential role of flavonoids in the modulation of signaling pathways that are crucial for COVID-19 disease, particularly those related to inflammation and immunity. The immunomodulatory ability of flavonoids, carried out by the regulation of inflammatory mediators, the inhibition of endothelial activation, NLRP3 inflammasome, toll-like receptors (TLRs) or bromodomain containing protein 4 (BRD4), and the activation of the nuclear factor erythroid-derived 2-related factor 2 (Nrf2), might be beneficial in regulating the cytokine storm during SARS-CoV-2 infection. Moreover, the ability of flavonoids to inhibit dipeptidyl peptidase 4 (DPP4), neutralize 3-chymotrypsin-like protease (3CLpro) or to affect gut microbiota to maintain immune response, and the dual action of angiotensin-converting enzyme 2 (ACE-2) may potentially also be applied to the exaggerated inflammatory responses induced by SARS-CoV-2. Based on the previously proven effects of flavonoids in other diseases or on the basis of newly published studies associated with COVID-19 (bioinformatics, molecular docking), it is reasonable to assume positive effects of flavonoids on inflammatory changes associated with COVID-19. This review highlights the current state of knowledge of the utility of flavonoids in the management of COVID-19 and also points to the multiple biological effects of flavonoids on signaling pathways associated with the inflammation processes that are deregulated in the pathology induced by SARS-CoV-2. The identification of agents, including naturally occurring substances such as flavonoids, represents great approach potentially utilizable in the management of COVID-19. Although not clinically investigated yet, the applicability of flavonoids against COVID-19 could be a promising strategy due to a broad spectrum of their biological activities.
Collapse
Affiliation(s)
- Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Samson M Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Raghad Khalid Al-Ishaq
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Mariam Abotaleb
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Vladimir Nosal
- Department of Neurology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Karol Kajo
- Department of Pathology, St. Elizabeth Cancer Institute Hospital, Bratislava, Slovakia; Biomedical Research Centre, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Tuzla, Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul, Turkey
| | - Aranka Brockmueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Peter Sabaka
- Department of Infectiology and Geographical Medicine, Faculty Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | - Ioana Mozos
- Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania; Center for Translational Research and Systems Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - David Ullrich
- Department of Leadership, Faculty of Military Leadership, University of Defence, Brno, Czech Republic
| | - Robert Prosecky
- 2nd Department of Internal Medicine, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic
| | - Giampiero La Rocca
- Human Anatomy Section, Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo and Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Luis Rodrigo
- Faculty of Medicine, University of Oviedo and Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Peter Kruzliak
- 2nd Department of Surgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic.
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia.
| |
Collapse
|
9
|
Yuan HL, Zhao YL, Ding CF, Zhu PF, Jin Q, Liu YP, Ding ZT, Luo XD. Anti-inflammatory and antinociceptive effects of Curcuma kwangsiensis and its bioactive terpenoids in vivo and in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2020; 259:112935. [PMID: 32387235 DOI: 10.1016/j.jep.2020.112935] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE "Curcumae Radix", the dried rhizomes of Curcuma kwangsiensis documented in Chinese pharmacopoeia, has been traditionally used for the treatment of inflammatory and pain diseases, such as jaundice and red urine, cleaning the heart-fire and depression, arthralgia, and dysmenorrhea. However, according to literature surveys, anti-inflammatory and antinociceptive studies of C. kwangsiensis have been seldom reported so far. AIM OF THE STUDY The current study focuses on the anti-inflammatory and antinociceptive effects of C. kwangsiensis and discovering the bioactive compounds for its traditional usages both in vivo and in vitro, which could provide scientific justification about its traditional use. MATERIAL AND METHODS The anti-inflammatory and antinociceptive assays of various layers (ME, EA, AQS) from C. kwangsiensis were achieved by carrageenan-induced paw edema and acetic acid-induced writhing animal models, respectively. The most bioactive part, EA layer was further phytochemically investigated by multiple step chromatography techniques. The structures of these isolates were unambiguously elucidated by means of extensive spectroscopic and chemical methods, and comparison with corresponding data of the reported literature. Four major sesquiterpenoids (4, 6, 14, and 15) were achieved for their anti-inflammatory and antinociceptive assays by the two aforementioned animal models in vivo. All the isolated compounds were evaluated for their anti-inflammatory effects via detecting inflammatory mediator releases (COX-2, IL-1β, and TNF-α) in RAW 264.7 macrophage cells induced by LPS. RESULTS The ME and EA layers significantly alleviated the paw edema caused by carrageenan and decreased the number of writhes induced by acetic acid at the dose of 200 and/or 100 mg/kg in comparison to the control group (p < 0.01/0.05), and the EA layer exhibited better activity than that of ME layer. Subsequent phytochemical investigation on EA layer of C. kwangsiensis exhibited that three new terpenoid compounds (1-3), identified as (12Z,14R)-7β-hydroxylabda-8(17),12-diene-14,15,16-triol (1), (12Z,14S)- 7β-hydroxlabda-8(17),12-diene-14,15,16-triol (2), and (4S)-hydroxy-(8)-methoxy-(5S)-(H)-guaia1(10),7(11)-dien-12,8-olide (3), together with twenty-two known analogs were isolated. Furthermore, four major sesquiterpenoids (4, 6, 14, and 15) significantly relieved the paw edema and number of writhes at 100 and/or 50 mg/kg (p < 0.05/0.01). Likewise, the majority of sesqui- and diterpenoids isolated could remarkably inhibited the secretion of inflammatory mediators (COX-2, IL-1β, and TNF-α) in LPS-stimulated RAW 264.7 macrophages cells at the concentration of 20 μg/mL, comparable to DXM used as the positive control. All the results suggested that EA layer from C. kwangsiensis possessed the anti-inflammatory and antinociceptive activities, and these sesqui- and diterpenoids could be the effective constituents responsible for relieving inflammation. CONCLUSION The present studies undoubtedly determined the anti-inflammatory and antinociceptive material basis of C. kwangsiensis, including the EA layer and its precise components, which presented equivalent or better anti-inflammatory effects than that of positive control (ASP/DXM) in vivo and in vitro. These results not only would account for scientific knowledge for traditional use of C. kwangsiensis, but also provide credible theoretical foundation for the further development of anti-inflammatory and antinociceptive agents.
Collapse
Affiliation(s)
- Hai-Lian Yuan
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, People's Republic of China; State Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China
| | - Yun-Li Zhao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, People's Republic of China; State Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China
| | - Cai-Feng Ding
- State Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China
| | - Pei-Feng Zhu
- State Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China; University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Qiong Jin
- State Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China; University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Ya-Ping Liu
- State Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China.
| | - Zhong-Tao Ding
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, People's Republic of China.
| | - Xiao-Dong Luo
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, People's Republic of China; State Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China.
| |
Collapse
|
10
|
Wu A, Yang Z, Huang Y, Yuan H, Lin C, Wang T, Zhao Z, Zhou Y, Zhu C. Natural phenylethanoid glycosides isolated from Callicarpa kwangtungensis suppressed lipopolysaccharide-mediated inflammatory response via activating Keap1/Nrf2/HO-1 pathway in RAW 264.7 macrophages cell. JOURNAL OF ETHNOPHARMACOLOGY 2020; 258:112857. [PMID: 32298752 DOI: 10.1016/j.jep.2020.112857] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/25/2020] [Accepted: 04/06/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Callicarpa kwangtungensis, as a characteristic traditional herb in China, has been widely used as indigenous medicine for thousands of years in the treatment of upper respiratory tract infection, tonsillitis, pneumonia and traumatic bleeding in China. Phenylethanoid glycosides (PhGs), as natural polyphenols, are especially abundant in this herb and can be regarded as the representative active ingredients in C. kwangtungensis. AIM OF THIS STUDY This study was performed to investigate the anti-inflammatory pharmacodynamic basis of six PhGs (acteoside, forsythoside B, poliumoside, alyssonoside, parvifloroside A, and syringalide A 3'-α-L-rhanmnopyranoside) isolated from C. kwangtungensis from the perspective of antioxidation. MATERIALS AND METHODS Six PhGs were isolated from the anti-inflammatory extracts of C. kwangtungensis by various chromatographic techniques and their anti-inflammatory activity on RAW 264.7 murine macrophages induced by LPS was investigated by measuring the release of tumor necrosis factor (TNF-α), the colonic interleukin-6 (IL-6), nitric oxide (NO) and reactive oxygen species (ROS). Further, the underlying anti-inflammatory mechanism of two PhGs (forsythoside B and alyssonoside) was explored by determining the expression of Kelch-like ECH-association protein 1 (Keap1), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (OH-1) and quinone oxidoreductase 1 (NQO1). Besides, molecular simulation was also employed to evaluate the binding capacity of two PhGs with Keap1. RESULTS Compared with the model group, six PhGs revealed obviously inhibitory effects on TNF-α, IL-6, NO and the generation of ROS in RAW 264.7 macrophages. Moreover, forsythoside B and alyssonoside could act as the inhibitors of Keap1-Nrf2 interaction, then activated the nuclear translocation of Nrf2 and promoted the upregulated protein expression of HO-1 and NQO1, finally suppressed LPS-induced inflammatory response in RAW 264.7 macrophages. Molecular modeling exhibited hydrogen bonds played a crucial role for the binding of PhGs with the Nrf2 binding site in Keap1 protein. CONCLUSIONS Natural PhGs-induced protection against LPS-induced inflammatory response via activating Keap1/Nrf2/HO-1 signaling pathway in RAW 264.7 macrophages were confirmed, which provided experimental and theoretical basis for the deeper use of C. Kwangtungensis in the treatment and prevention of diseases related to inflammation and oxidative stress.
Collapse
Affiliation(s)
- Aizhi Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Zhiying Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Yimin Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Huan Yuan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Chaozhan Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Tao Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Zhongxiang Zhao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Yuan Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Chenchen Zhu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| |
Collapse
|