1
|
Yip AJW, Lee YZ, Kow ASF, Wong CSA, Lee MT, Tham CL, Tan JW. Current utilization trend of immortalized mast cell lines in allergy research: a systematic review. Immunol Res 2025; 73:41. [PMID: 39838115 PMCID: PMC11750950 DOI: 10.1007/s12026-024-09562-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 11/07/2024] [Indexed: 01/23/2025]
Abstract
Today, in the modern world, allergic diseases, also described as atopic allergies, are classified as a type of multifactorial disorder due to the complex interplay between genetics, environment, and socioeconomic factors that influence the disease's manifestation, severity, and one's predisposition to allergic diseases. It is undeniable that many reported studies have pointed out that the mast cell is one of the leading key players involved in triggering an allergic reaction. To improve our understanding of the molecular and cellular mechanisms underlying allergy, various mast cell lines have been employed in vitro to study the pathogenesis of allergic diseases for the past decades. However, there is no consensus on many fundamental aspects associated with their use, such as the effects of culture media composition and the type of inducer used for cell degranulation. As the standardization of research protocols and disease models is crucial, we present the outcome of a systematic review of scientific articles using three major immortalized in vitro mast cell lines (HMC-1, LAD2, and RBL-2H3) to study allergy. This systematic review described the cell source, culture conditions, inducers used for degranulation, and mediators released for examination. We hope that the present systematic review may help to standardize the use of immortalized in vitro mast cell lines in allergy research and serve as a user's guide to understand the fundamental aspects of allergy as well to develop an effective allergy therapy in the future for the betterment of human good health and wellbeing.
Collapse
Affiliation(s)
- Ashley Jia Wen Yip
- School of Science, Monash University Malaysia, Jalan Lagoon SelatanSubang Jaya, 47500, Bandar Sunway, Selangor, Malaysia
| | - Yu Zhao Lee
- Faculty of Medicine and Health Sciences, UCSI University, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Audrey Siew Foong Kow
- Faculty of Pharmaceutical Sciences, UCSI University, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Carisa Su-Ann Wong
- School of Science, Monash University Malaysia, Jalan Lagoon SelatanSubang Jaya, 47500, Bandar Sunway, Selangor, Malaysia
| | - Ming-Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Cheras, 56000, Kuala Lumpur, Malaysia
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Ji Wei Tan
- School of Science, Monash University Malaysia, Jalan Lagoon SelatanSubang Jaya, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
2
|
Trigo G, Coelho M, Ferreira CB, Melosini M, Lehmann IS, Reis CP, Gaspar MM, Santos S. Exploring the Biological Activity of Phytocannabinoid Formulations for Skin Health Care: A Special Focus on Molecular Pathways. Int J Mol Sci 2024; 25:13142. [PMID: 39684852 DOI: 10.3390/ijms252313142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Recent advancements have highlighted the potential of cannabis and its phytocannabinoids (pCBs) in skin health applications. These compounds, through their interaction with the endocannabinoid system (ECS), show promise for skin health products. Their ability to regulate inflammation, oxidative stress and cell proliferation makes them useful in addressing skin problems such as inflammation, scarring, healing, acne and aging, positioning them as valuable tools for innovative skincare solutions. In the present work, the cellular and molecular effects of proprietary pCB-based formulations on ECS modulation, inflammation and skin regeneration were investigated. Using human dermal fibroblasts (HDF) and keratinocytes (HaCaT), the effect of formulations in both pre-treatment and treatment scenarios following exposure to stress-inducing agents was assessed. Key molecular markers were analyzed to tackle their efficacy in mitigating inflammation and promoting structural integrity and regeneration. In vitro results showed that these formulations significantly reduced inflammation, promoted skin regeneration and improved structural functions. In vivo studies confirmed that the formulations were well-tolerated and led to noticeable improvements in skin health, including enhanced barrier function. This study demonstrates the safety and efficacy of pCB-based formulations for cosmeceutical applications. By combining molecular analysis with in vivo testing, this research provides new insights into the therapeutic potential of pCBs for managing various skin conditions.
Collapse
Affiliation(s)
- Guilherme Trigo
- R&D&I Department, EXMceuticals Portugal Lda, 1749-016 Lisboa, Portugal
| | - Mariana Coelho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | | | - Matteo Melosini
- R&D&I Department, EXMceuticals Portugal Lda, 1749-016 Lisboa, Portugal
| | | | - Catarina P Reis
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
- Institute of Biophysics and Biomedical Engineering (IBEB), Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
- Institute of Biophysics and Biomedical Engineering (IBEB), Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Susana Santos
- R&D&I Department, EXMceuticals Portugal Lda, 1749-016 Lisboa, Portugal
| |
Collapse
|
3
|
Kim G, Lee SY, Oh S, Jang JW, Lee J, Kim HS, Son KH, Byun K. Anti-Inflammatory Effects of Extracellular Vesicles from Ecklonia cava on 12-O-Tetradecanoylphorbol-13-Acetate-Induced Skin Inflammation in Mice. Int J Mol Sci 2024; 25:12522. [PMID: 39684233 DOI: 10.3390/ijms252312522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Steroids, which are often used to treat the inflammation associated with various skin diseases, have several negative side effects. As Ecklonia cava extract has anti-inflammatory effects in various diseases, we evaluated the efficacy of Ecklonia cava-derived extracellular vesicles (EVEs) in decreasing 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced inflammation. We determined the effect of the EVEs on the TLR4/NF-κB/NLRP3 inflammasome in human keratinocytes and mouse ear skin. TPA-treated human keratinocytes showed an increased expression of TLR4 and its ligands HMGB1 and S100A8. TPA also increased the expression of (1) NF-κB; (2) the NLRP3 inflammasome components NLRP3, ASC, and caspase 1; and (3) the pyroptosis-related factors GSDMD-NT, IL-18, and IL-1β. However, the expression of these molecules decreased in the TPA-treated human keratinocytes after EVE treatment. Similar to the in vitro results, TPA increased the expression of these molecules in mouse ear skin, and EVE treatment decreased their expression. The TPA treatment of skin increased edema, redness, neutrophil infiltration, and epidermal thickness, and EVE reduced these symptoms of inflammation. In conclusion, the EVEs decreased TPA-induced skin inflammation, which was associated with a decrease in the TLR4/NF-κB/NLRP3 inflammasome.
Collapse
Affiliation(s)
- Geebum Kim
- Misogain Dermatology Clinic, Gimpo 10108, Republic of Korea
| | - So Young Lee
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Jong-Won Jang
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| | - Jehyuk Lee
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Doctorbom Clinic, Seoul 06614, Republic of Korea
| | - Hyun-Seok Kim
- Kim Hyun Seok Plastic Surgery Clinic, Seoul 06030, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kyunghee Byun
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| |
Collapse
|
4
|
Bukhari QUA, Della Pelle F, Alvarez-Diduk R, Scroccarello A, Nogués C, Careta O, Compagnone D, Merkoci A. Laser-assembled conductive 3D nanozyme film-based nitrocellulose sensor for real-time detection of H 2O 2 released from cancer cells. Biosens Bioelectron 2024; 262:116544. [PMID: 38963952 DOI: 10.1016/j.bios.2024.116544] [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: 04/17/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024]
Abstract
In this work, a nanostructured conductive film possessing nanozyme features was straightforwardly produced via laser-assembling and integrated into complete nitrocellulose sensors; the cellulosic substrate allows to host live cells, while the nanostructured film nanozyme activity ensures the enzyme-free real-time detection of hydrogen peroxide (H2O2) released by the sames. In detail, a highly exfoliated reduced graphene oxide 3D film decorated with naked platinum nanocubes was produced using a CO2-laser plotter via the simultaneous reduction and patterning of graphene oxide and platinum cations; the nanostructured film was integrated into a nitrocellulose substrate and the complete sensor was manufactured using an affordable semi-automatic printing approach. The linear range for the direct H2O2 determination was 0.5-80 μM (R2 = 0.9943), with a limit of detection of 0.2 μM. Live cell measurements were achieved by placing the sensor in the culture medium, ensuring their adhesion on the sensors' surface; two cell lines were used as non-tumorigenic (Vero cells) and tumorigenic (SKBR3 cells) models, respectively. Real-time detection of H2O2 released by cells upon stimulation with phorbol ester was carried out; the nitrocellulose sensor returned on-site and real-time quantitative information on the H2O2 released proving useful sensitivity and selectivity, allowing to distinguish tumorigenic cells. The proposed strategy allows low-cost in-series semi-automatic production of paper-based point-of-care devices using simple benchtop instrumentation, paving the way for the easy and affordable monitoring of the cytopathology state of cancer cells.
Collapse
Affiliation(s)
- Qurat U A Bukhari
- Nanobioelectronics & Biosensors Group, Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona, Spain; Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Campus "Aurelio Saliceti" Via R. Balzarini 1, 64100, Teramo, Italy
| | - Flavio Della Pelle
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Campus "Aurelio Saliceti" Via R. Balzarini 1, 64100, Teramo, Italy
| | - Ruslan Alvarez-Diduk
- Nanobioelectronics & Biosensors Group, Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona, Spain.
| | - Annalisa Scroccarello
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Campus "Aurelio Saliceti" Via R. Balzarini 1, 64100, Teramo, Italy
| | - Carme Nogués
- Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biociencies, Universitat Autonoma de Barcelona (UAB), 08193 Bellaterra, Barcelona, Spain
| | - Oriol Careta
- Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biociencies, Universitat Autonoma de Barcelona (UAB), 08193 Bellaterra, Barcelona, Spain
| | - Dario Compagnone
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Campus "Aurelio Saliceti" Via R. Balzarini 1, 64100, Teramo, Italy.
| | - Arben Merkoci
- Nanobioelectronics & Biosensors Group, Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona, Spain; ICREA Institució Catalana de Recerca i Estudis Avançats, Passeig de Lluís Companys, 23, 08010, Barcelona, Spain.
| |
Collapse
|
5
|
Wang D, Wang M, Sun S, Zhang C, Song Y, Li J, Song B, Lv H, Wang S, Jiang W. Hypoxia-induced NLRP3 inflammasome activation via the HIF-1α/NF-κB signaling pathway in human dental pulp fibroblasts. BMC Oral Health 2024; 24:1156. [PMID: 39343901 PMCID: PMC11441079 DOI: 10.1186/s12903-024-04936-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Previous studies have reported the link between hypoxic conditions and NLRP3 inflammasome-mediated pulpal inflammation in the progression of pulpitis. However, the underlying mechanism has not been fully elucidated. This study aimed to investigate the role of HIF-1α in the regulation of NLRP3 inflammasome pathway via NF-κB signaling under hypoxic conditions with or without LPS in human dental pulp fibroblasts (HDPFs) during the progression of pulpitis. METHODS HIF-1α plasmids or siRNAs were used to upregulate or downregulate HIF-1α in HDPFs, respectively. The effect of hypoxia with or without LPS on the NF-κB signaling and NLRP3 inflammasome pathway was analyzed by immunofluorescence staining, qRT-PCR, western blotting and ELISA. RESULTS The hypoxic conditions alone induced ASC oligomerization and NLRP3/CASP1 inflammasome pathway activation via NF-κB signaling in a time-dependent manner in HDPFs. The upregulation of HIF-1α further promoted hypoxia-induced ASC oligomerization and NLRP3/CASP1 inflammasome pathway activation via NF-κB signaling compared to the hypoxia-induced group. In comparison, downregulation of HIF-1α inhibited ASC oligomerization and NLRP3/CASP1 inflammasome pathway activation via NF-κB signaling compared to the hypoxia-induced group. Additionally, LPS plus hypoxia further promoted HIF-1α expression and NLRP3/ASC/CASP1 inflammasome pathway activation via NF-κB signaling compared to the hypoxia-induced group. CONCLUSIONS HIF-1α served as a positive regulator of NLRP3/ASC/CASP1 inflammasome pathway activation via NF-κB signaling in HDPFs in the sterile pulpal inflammation and caries-related pulpitis microenvironment. The finding of a novel functional HIF-1α-NF-κB-NLRP3 axis provides insight into the link between the hypoxic microenvironment and pulpal inflammation, thus supporting a promising therapeutic strategy for the control of pulpal inflammation.
Collapse
Affiliation(s)
- Diya Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Minghao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Shukai Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chongyang Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Ya Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Jianing Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Bing Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Haipeng Lv
- Department of Stomatology, Xi'an Daxing Hospital, Xi'an, Shaanxi, China.
| | - Shengchao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China.
| | - Wenkai Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China.
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK.
| |
Collapse
|
6
|
Ebrahimi A, Mehrabi M, Miraghaee SS, Mohammadi P, Fatehi Kafash F, Delfani M, Khodarahmi R. Flavonoid compounds and their synergistic effects: Promising approaches for the prevention and treatment of psoriasis with emphasis on keratinocytes - A systematic and mechanistic review. Int Immunopharmacol 2024; 138:112561. [PMID: 38941673 DOI: 10.1016/j.intimp.2024.112561] [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: 05/09/2024] [Revised: 06/16/2024] [Accepted: 06/23/2024] [Indexed: 06/30/2024]
Abstract
Psoriasis, a chronic autoimmune skin disorder, causes rapid and excessive skin cell growth due to immune system dysfunction. Numerous studies have shown that flavonoids have anti-psoriatic effects by modulating various molecular mechanisms involved in inflammation, cytokine production, keratinocyte proliferation, and more. This study reviewed experimental data reported in scientific literature and used network analysis to identify the potential biological roles of flavonoids' targets in treating psoriasis. 947 records from Web of Sciences, ScienceDirect database, Scopus, PubMed, and Cochrane library were reviewed without limitations until June 26, 2023. 66 articles were included in the systematic review. The ten genes with the highest scores, including interleukin (IL)-10, IL-12A, IL-1β, IL-6, Tumor necrosis factor-α (TNF-α), Janus kinase 2 (JAK 2), Jun N-terminal kinase (JUN), Proto-oncogene tyrosine-protein kinase Src (SRC), Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), and Signal transducer and activator of transcription 3 (STAT3), were identified as the hub genes. KEGG pathway analysis identified connections related to inflammation and autoimmune responses, which are key characteristics of psoriasis. IL-6, STAT3, and JUN's presence in both hub and enrichment genes suggests their important role in flavonoid's effect on psoriasis. This comprehensive study highlights how flavonoids can target biological processes in psoriasis, especially when combined for enhanced effectiveness.
Collapse
Affiliation(s)
- Ali Ebrahimi
- Department of Dermatology, Hajdaie Dermatology Clinic, School of Medicine, Kermanshah University of Medical Sciences (KUMS), Kermanshah, Iran
| | - Masomeh Mehrabi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Seyyed Shahram Miraghaee
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Fatehi Kafash
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohana Delfani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
7
|
Bakhsh T, Alyami NM. Inducing breast cancer cell death: The impact of taxodone on proliferation through apoptosis. Heliyon 2024; 10:e34044. [PMID: 39055854 PMCID: PMC11269907 DOI: 10.1016/j.heliyon.2024.e34044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/19/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer is the most prevalent form of cancer in women and a major contributor to cancer-related fatalities worldwide. Several factors play a role in the development of breast cancer, encompassing age, hormone levels, etc. Taxodone has shown significant anti-tumor properties in both laboratory experiments and living organisms. However, its impact on the human MCF-7 breast cancer cell line has not been researched. This investigation explores the chemo-preventive potential of taxodone in the MCF-7 breast cancer cells. The anticancer potential of taxodone against MCF-7 cells was determined by MTT assay. Further, the induction of apoptosis in MCF-7 breast cancer cells was confirmed via ELISA, which indicated the increased incidences of chromatin condensation and ssDNA breakage in the MCF-7 apoptotic cells upon 24 h of taxodone treatment. The intracellular reactive oxygen species (ROS) level was evaluated using H2DCFDA fluorescent dye to elucidate the mechanism of action triggered upon taxodone treatment. The increasing intercellular ROS level sequentially activated the caspase-mediated apoptosis pathway. Consequently, the outcomes revealed that taxodone decreased the cell viability of MCF-7 dose-dependently. Taxodone triggers apoptosis in MCF-7 cells by increasing intracellular ROS levels and activating the caspase cascade through the mitochondrial apoptosis-induced channel, an early marker of apoptosis onset. Our results indicate that taxodone exhibits anti-proliferative and apoptotic properties against human MCF-7 breast cancer cells, suggesting it to be a natural anticancer agent.
Collapse
Affiliation(s)
- Tahani Bakhsh
- Department of Biology, College of Science, University of Jeddah, Jeddah, 21589, Saudi Arabia
| | - Nouf M. Alyami
- Department of Zoology, College of Science, King Saud University, PO Box -2455, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
8
|
Xu F, Xie L, He J, Huang Q, Shen Y, Chen L, Zeng X. Detection of common pathogenesis of rheumatoid arthritis and atherosclerosis via microarray data analysis. Heliyon 2024; 10:e28029. [PMID: 38628735 PMCID: PMC11019104 DOI: 10.1016/j.heliyon.2024.e28029] [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: 09/20/2023] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 04/19/2024] Open
Abstract
Despite extensive research reveal rheumatoid arthritis (RA) is related to atherosclerosis (AS), common pathogenesis between these two diseases still needs to be explored. In current study, we explored the common pathogenesis between rheumatoid arthritis (RA) and atherosclerosis (AS) by identifying 297 Differentially Expressed Genes (DEGs) associated with both diseases. Through KEGG and GO functional analysis, we highlighted the correlation of these DEGs with crucial biological processes such as the vesicle transport, immune system process, signaling receptor binding, chemokine signaling and many others. Employing Protein-Protein Interaction (PPI) network analysis, we elucidated the associations between DEGs, revealing three gene modules enriched in immune system process, vesicle, signaling receptor binding, Pertussis, and among others. Additionally, through CytoHubba analysis, we pinpointed 11 hub genes integral to intergrin-mediated signaling pathway, plasma membrane, phosphotyrosine binding, chemokine signaling pathway and so on. Further investigation via the TRRUST database identified two key Transcription Factors (TFs), SPI1 and RELA, closely linked with these hub genes, shedding light on their regulatory roles. Finally, leveraging the collective insights from hub genes and TFs, we proposed 10 potential drug candidates targeting the molecular mechanisms underlying RA and AS pathogenesis. Further investigation on xCell revealed that 14 types of cells were all different in both AS and RA. This study underscores the shared pathogenic mechanisms, pivotal genes, and potential therapeutic interventions bridging RA and AS, offering valuable insights for future research and clinical management strategies.
Collapse
Affiliation(s)
- Fan Xu
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, Fujian Province, China
| | - Linfeng Xie
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jian He
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Medical University, Fuzhou, Fujian Province, China
| | - Qiuyu Huang
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, Fujian Province, China
| | - Yanming Shen
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Medical University, Fuzhou, Fujian Province, China
| | - Liangwan Chen
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, Fujian Province, China
| | - Xiaohong Zeng
- Department of Rheumatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
9
|
Dulloo I, Tellier M, Levet C, Chikh A, Zhang B, Blaydon DC, Webb CM, Kelsell DP, Freeman M. Cleavage of the pseudoprotease iRhom2 by the signal peptidase complex reveals an ER-to-nucleus signaling pathway. Mol Cell 2024; 84:277-292.e9. [PMID: 38183983 DOI: 10.1016/j.molcel.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 09/18/2023] [Accepted: 12/08/2023] [Indexed: 01/08/2024]
Abstract
iRhoms are pseudoprotease members of the rhomboid-like superfamily and are cardinal regulators of inflammatory and growth factor signaling; they function primarily by recognizing transmembrane domains of their clients. Here, we report a mechanistically distinct nuclear function of iRhoms, showing that both human and mouse iRhom2 are non-canonical substrates of signal peptidase complex (SPC), the protease that removes signal peptides from secreted proteins. Cleavage of iRhom2 generates an N-terminal fragment that enters the nucleus and modifies the transcriptome, in part by binding C-terminal binding proteins (CtBPs). The biological significance of nuclear iRhom2 is indicated by elevated levels in skin biopsies of patients with psoriasis, tylosis with oesophageal cancer (TOC), and non-epidermolytic palmoplantar keratoderma (NEPPK); increased iRhom2 cleavage in a keratinocyte model of psoriasis; and nuclear iRhom2 promoting proliferation of keratinocytes. Overall, this work identifies an unexpected SPC-dependent ER-to-nucleus signaling pathway and demonstrates that iRhoms can mediate nuclear signaling.
Collapse
Affiliation(s)
- Iqbal Dulloo
- Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK.
| | - Michael Tellier
- Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Clémence Levet
- Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Anissa Chikh
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London E1 2AT, UK
| | - Boyan Zhang
- Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Diana C Blaydon
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London E1 2AT, UK
| | - Catherine M Webb
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London E1 2AT, UK
| | - David P Kelsell
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, Newark Street, London E1 2AT, UK
| | - Matthew Freeman
- Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK.
| |
Collapse
|
10
|
Xia A, Wan J, Li X, Quan J, Chen X, Xu Z, Jiao X. M. tb Rv0927c suppresses the activation of HIF-1α pathway through VHL-mediated ubiquitination and NF-κB/COX-2 pathway to enhance mycobacteria survival. Microbiol Res 2024; 278:127529. [PMID: 37922696 DOI: 10.1016/j.micres.2023.127529] [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: 07/18/2023] [Revised: 09/24/2023] [Accepted: 10/15/2023] [Indexed: 11/07/2023]
Abstract
Mycobacterium tuberculosis (M. tuberculosis), the causative agent of tuberculosis, employs various effector proteins to target and modulate host defenses. Our previous study showed that M. tuberculosis protein Rv0927c can promote the survival of intracellular mycobacteria, but the underlying mechanisms remain poorly understood. Here, we found that Rv0927c inhibited Mycobacterium smegmatis (M. smegmatis) induced hypoxia-inducible factor-1α (HIF-1α) activation in macrophages, and HIF-1α is required for Rv0927c to promote mycobacteria survival. Western blot analysis showed that Rv0927c promoted the proteasomal degradation of HIF-1α via Von Hippel-Lindau (VHL)-mediated ubiquitination and inhibited the nuclear localization of HIF-1α through the NF-κB/COX-2 pathway, thereby suppressing HIF-1α pathway activation. Furthermore, Rv0927c suppressed the host glycolytic metabolism, which is known to be regulated by HIF-1α and depended on the glycolysis process to promote mycobacterial survival. Our findings provide evidence that Rv0927c inhibits the activation of HIF-1α pathway, allowing pathogens to evade host immune responses, suggesting that targeting Rv0927c or HIF-1α might be a potential anti-tuberculosis therapy.
Collapse
Affiliation(s)
- Aihong Xia
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Jiaxu Wan
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry Of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
| | - Xin Li
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry Of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
| | - Juanjuan Quan
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xiang Chen
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry Of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
| | - Zhengzhong Xu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry Of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China.
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry Of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
11
|
Zhang Z, Zhu Z, Zuo X, Wang X, Ju C, Liang Z, Li K, Zhang J, Luo L, Ma Y, Song Z, Li X, Li P, Quan H, Huang P, Yao Z, Yang N, Zhou J, Kou Z, Chen B, Ding T, Wang Z, Hu X. Photobiomodulation reduces neuropathic pain after spinal cord injury by downregulating CXCL10 expression. CNS Neurosci Ther 2023; 29:3995-4017. [PMID: 37475184 PMCID: PMC10651991 DOI: 10.1111/cns.14325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/07/2023] [Accepted: 06/10/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Many studies have recently highlighted the role of photobiomodulation (PBM) in neuropathic pain (NP) relief after spinal cord injury (SCI), suggesting that it may be an effective way to relieve NP after SCI. However, the underlying mechanisms remain unclear. This study aimed to determine the potential mechanisms of PBM in NP relief after SCI. METHODS We performed systematic observations and investigated the mechanism of PBM intervention in NP in rats after SCI. Using transcriptome sequencing, we screened CXCL10 as a possible target molecule for PBM intervention and validated the results in rat tissues using reverse transcription-polymerase chain reaction and western blotting. Using immunofluorescence co-labeling, astrocytes and microglia were identified as the cells responsible for CXCL10 expression. The involvement of the NF-κB pathway in CXCL10 expression was verified using inhibitor pyrrolidine dithiocarbamate (PDTC) and agonist phorbol-12-myristate-13-acetate (PMA), which were further validated by an in vivo injection experiment. RESULTS Here, we demonstrated that PBM therapy led to an improvement in NP relative behaviors post-SCI, inhibited the activation of microglia and astrocytes, and decreased the expression level of CXCL10 in glial cells, which was accompanied by mediation of the NF-κB signaling pathway. Photobiomodulation inhibit the activation of the NF-κB pathway and reduce downstream CXCL10 expression. The NF-κB pathway inhibitor PDTC had the same effect as PBM on improving pain in animals with SCI, and the NF-κB pathway promoter PMA could reverse the beneficial effect of PBM. CONCLUSIONS Our results provide new insights into the mechanisms by which PBM alleviates NP after SCI. We demonstrated that PBM significantly inhibited the activation of microglia and astrocytes and decreased the expression level of CXCL10. These effects appear to be related to the NF-κB signaling pathway. Taken together, our study provides evidence that PBM could be a potentially effective therapy for NP after SCI, CXCL10 and NF-kB signaling pathways might be critical factors in pain relief mediated by PBM after SCI.
Collapse
Affiliation(s)
- Zhihao Zhang
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Zhijie Zhu
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Xiaoshuang Zuo
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Xuankang Wang
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Cheng Ju
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Zhuowen Liang
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Kun Li
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Jiawei Zhang
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Liang Luo
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Yangguang Ma
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Zhiwen Song
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Xin Li
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
- 967 Hospital of People's Liberation Army Joint Logistic Support ForceDalianLiaoningChina
| | - Penghui Li
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Huilin Quan
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Peipei Huang
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Zhou Yao
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Ning Yang
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Jie Zhou
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Zhenzhen Kou
- Department of Anatomy, Histology and Embryology, School of Basic MedicineAir Force Military Medical UniversityXi'anShaanxiChina
| | - Beiyu Chen
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Tan Ding
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Zhe Wang
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| | - Xueyu Hu
- Department of OrthopedicsXijing Hospital, Air Force Military Medical UniversityXi'anShaanxiChina
| |
Collapse
|
12
|
Dey DK, Gahlot H, Chang SN, Kang SC. CopA3 treatment suppressed multidrug resistivity in HCT-116 cell line by p53-induced degradation of hypoxia-inducible factor 1α. Life Sci 2023; 329:121933. [PMID: 37451396 DOI: 10.1016/j.lfs.2023.121933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/08/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
The major reason for multidrug resistance is the failure of chemotherapy in many tumors, including colon cancer. Hypoxia-inducible factor (HIF)-1α is a crucial transcription factor that simulates multiple cellular response to hypoxia. HIF-1α has been known to play a vital role towards tumor resistance; however, its mechanism of action is still not fully elucidated. N this study, we found that HIF-1α remarkably modulated drug resistance-associated proteins upon CopA3 peptide treatment against colon cancer cells. Abnormal rates of tumor growth along with high metastatic potential lacks the susceptibility towards cellular signals is a key characteristic in many tumor types. Moreover, in growing tumors, cells are exposed to insufficient nutrient supply and low oxygen availability. These stress force them to switch into adaptable and aggressive phenotypes. Our study investigated the interaction of HIF-1α and MDR gene association upon CopA3 treatment in the tumor microenvironment. We demonstrate that the multidrug resistance gene is associated with tumor resistance to chemotherapeutics, which upon CopA3 treatment promotes p53 activation and proteasomal degradation of HIF-1α, effecting the angiogenesis response to hypoxia. p53 downregulation augments HIF-1-dependent transcriptional activation of VEGF in response to oxygen deprivation.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea
| | - Himanshi Gahlot
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea
| | - Sukkum Ngullie Chang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| |
Collapse
|
13
|
Chen R, Wang H, Zheng C, Zhang X, Li L, Wang S, Chen H, Duan J, Zhou X, Peng H, Guo J, Zhang A, Li F, Wang W, Zhang Y, Wang J, Wang C, Meng Y, Du X, Zhang H. Polo-like kinase 1 promotes pulmonary hypertension. Respir Res 2023; 24:204. [PMID: 37598171 PMCID: PMC10440037 DOI: 10.1186/s12931-023-02498-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/22/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is a lethal vascular disease with limited therapeutic options. The mechanistic connections between alveolar hypoxia and PH are not well understood. The aim of this study was to investigate the role of mitotic regulator Polo-like kinase 1 (PLK1) in PH development. METHODS Mouse lungs along with human pulmonary arterial smooth muscle cells and endothelial cells were used to investigate the effects of hypoxia on PLK1. Hypoxia- or Sugen5416/hypoxia was applied to induce PH in mice. Plk1 heterozygous knockout mice and PLK1 inhibitors (BI 2536 and BI 6727)-treated mice were checked for the significance of PLK1 in the development of PH. RESULTS Hypoxia stimulated PLK1 expression through induction of HIF1α and RELA. Mice with heterozygous deletion of Plk1 were partially resistant to hypoxia-induced PH. PLK1 inhibitors ameliorated PH in mice. CONCLUSIONS Augmented PLK1 is essential for the development of PH and is a druggable target for PH.
Collapse
Affiliation(s)
- Rongrong Chen
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hongfei Wang
- Department of Cardiac Surgery, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cuiting Zheng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Department of Pathology, Beijing Lab for Cardiovascular Precision Medicine, Key Laboratory of Medical Engineering for Cardiovascular Disease, Capital Medical University, Beijing, China
| | - Xiyu Zhang
- Department of Pathology, Beijing Lab for Cardiovascular Precision Medicine, Key Laboratory of Medical Engineering for Cardiovascular Disease, Capital Medical University, Beijing, China
| | - Li Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shengwei Wang
- Department of Cardiac Surgery, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyu Chen
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Duan
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xian Zhou
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Haiyong Peng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Guo
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Anchen Zhang
- Department of Cardiac Surgery, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feifei Li
- Department of Cardiac Surgery, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wang Wang
- Department of Physiology, Capital Medical University, Beijing, China
| | - Yu Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Wang
- Department of Physiology, Capital Medical University, Beijing, China
| | - Chen Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Meng
- Department of Pathology, Beijing Lab for Cardiovascular Precision Medicine, Key Laboratory of Medical Engineering for Cardiovascular Disease, Capital Medical University, Beijing, China.
| | - Xinling Du
- Department of Cardiac Surgery, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Hongbing Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
14
|
Restivo I, Basilicata MG, Giardina IC, Massaro A, Pepe G, Salviati E, Pecoraro C, Carbone D, Cascioferro S, Parrino B, Diana P, Ostacolo C, Campiglia P, Attanzio A, D’Anneo A, Pojero F, Allegra M, Tesoriere L. A Combination of Polymethoxyflavones from Citrus sinensis and Prenylflavonoids from Humulus lupulus Counteracts IL-1β-Induced Differentiated Caco-2 Cells Dysfunction via a Modulation of NF-κB/Nrf2 Activation. Antioxidants (Basel) 2023; 12:1621. [PMID: 37627616 PMCID: PMC10451557 DOI: 10.3390/antiox12081621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
We here investigated the anti-inflammatory activity of a polymethoxylated flavone-containing fraction (PMFF) from Citrus sinensis and of a prenylflavonoid-containing one (PFF) from Humulus lupulus, either alone or in combination (MIX). To this end, an in vitro model of inflammatory bowel disease (IBD), consisting of differentiated, interleukin (IL)-1β-stimulated Caco-2 cells, was employed. We demonstrated that non-cytotoxic concentrations of either PMFF or PFF or MIX reduced nitric oxide (NO) production while PFF and MIX, but not PMFF, also inhibited prostaglandin E2 release. Coherently, MIX suppressed both inducible NO synthase and cyclooxygenase-2 over-expression besides NF-κB activation. Moreover, MIX increased nuclear factor erythroid 2-related factor 2 (Nrf2) activation, heme oxygenase-1 expression, restoring GSH and reactive oxygen and nitrogen species (RONs) levels. Remarkably, these effects with MIX were stronger than those produced by PMFF or PFF alone. Noteworthy, nobiletin (NOB) and xanthohumol (XTM), two of the most represented phytochemicals in PMFF and PFF, respectively, synergistically inhibited RONs production. Overall, our results demonstrate that MIX enhances the anti-inflammatory and anti-oxidative effects of the individual fractions in a model of IBD, via a mechanism involving modulation of NF-κB and Nrf2 signalling. Synergistic interactions between NOB and XTM emerge as a relevant aspect underlying this evidence.
Collapse
Affiliation(s)
- Ignazio Restivo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | | | - Ilenia Concetta Giardina
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Alessandro Massaro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (M.G.B.); (E.S.); (C.O.); (P.C.)
| | - Emanuela Salviati
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (M.G.B.); (E.S.); (C.O.); (P.C.)
| | - Camilla Pecoraro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (C.P.); (D.C.); (S.C.); (B.P.); (P.D.)
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (C.P.); (D.C.); (S.C.); (B.P.); (P.D.)
| | - Stella Cascioferro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (C.P.); (D.C.); (S.C.); (B.P.); (P.D.)
| | - Barbara Parrino
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (C.P.); (D.C.); (S.C.); (B.P.); (P.D.)
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (C.P.); (D.C.); (S.C.); (B.P.); (P.D.)
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (M.G.B.); (E.S.); (C.O.); (P.C.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (M.G.B.); (E.S.); (C.O.); (P.C.)
| | - Alessandro Attanzio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Antonella D’Anneo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Fanny Pojero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Mario Allegra
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| | - Luisa Tesoriere
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (I.C.G.); (A.M.); (A.A.); (A.D.); (F.P.); (L.T.)
| |
Collapse
|
15
|
Chang SN, Kang SC. Decursinol Angelate Inhibits Glutamate Dehydrogenase 1 Activity and Induces Intrinsic Apoptosis in MDR-CRC Cells. Cancers (Basel) 2023; 15:3541. [PMID: 37509203 PMCID: PMC10377166 DOI: 10.3390/cancers15143541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Colorectal cancer (CRC) was the second most commonly diagnosed cancer worldwide and the second most common cause of cancer-related deaths in Europe in 2020. After CRC patients' recovery, in many cases a patient's tumor returns and develops chemoresistance, which has remained a major challenge worldwide. We previously published our novel findings on the role of DA in inhibiting the activity of GDH1 using in silico and enzymatic assays. No studies have been conducted so far to explain the inhibitory role of DA against glutamate dehydrogenase in MDR-CRC cells. We developed a multidrug-resistant colorectal cancer cell line, HCT-116MDR, after treatment with cisplatin and 5-fluorouracil. We confirmed the MDR phenotype by evaluating the expression of MDR1, ABCB5, extracellular vesicles, polyploidy, DNA damage response markers and GDH1 in comparison with parental HCT-116WT (HCT-116 wild type). Following confirmation, we determined the IC50 and performed clonogenic assay for the efficacy of decursinol angelate (DA) against HCT-116MDR (HCT-116 multidrug resistant). Subsequently, we evaluated the novel interactions of DA with GDH1 and the expression of important markers regulating redox homeostasis and cell death. DA treatment markedly downregulated the expression of GDH1 at 50 and 75 μM after 36 h, which directly correlated with reduced expression of the Krebs cycle metabolites α-ketoglutarate and fumarate. We also observed a systematic dose-dependent downregulation of MDR1, ABCB5, TERT, ERCC1 and γH2AX. Similarly, the expression of important antioxidant markers was also downregulated. The markers for intrinsic apoptosis were notably upregulated in a dose-dependent manner. The results were further validated by flow cytometry and TUNEL assay. Additionally, GDH1 knockdown on both HCT-116WT and HCT-116MDR corresponded to a decreased expression of γH2AX, catalase, SOD1 and Gpx-1, and an eventual increase in apoptosis markers. In conclusion, inhibition of GDH1 increased ROS production, decreased cell proliferation and increased cell death.
Collapse
Affiliation(s)
- Sukkum Ngullie Chang
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Republic of Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Republic of Korea
| |
Collapse
|
16
|
Gharib E, Veilleux V, Boudreau LH, Pichaud N, Robichaud GA. Platelet-derived microparticles provoke chronic lymphocytic leukemia malignancy through metabolic reprogramming. Front Immunol 2023; 14:1207631. [PMID: 37441073 PMCID: PMC10333545 DOI: 10.3389/fimmu.2023.1207631] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/30/2023] [Indexed: 07/15/2023] Open
Abstract
Background It is well established that inflammation and platelets promote multiple processes of cancer malignancy. Recently, platelets have received attention for their role in carcinogenesis through the production of microvesicles or platelet-derived microparticles (PMPs), which transfer their biological content to cancer cells. We have previously characterized a new subpopulation of these microparticles (termed mito-microparticles), which package functional mitochondria. The potential of mitochondria transfer to cancer cells is particularly impactful as many aspects of mitochondrial biology (i.e., cell growth, apoptosis inhibition, and drug resistance) coincide with cancer hallmarks and disease progression. These metabolic aspects are particularly notable in chronic lymphocytic leukemia (CLL), which is characterized by a relentless accumulation of proliferating, immunologically dysfunctional, mature B-lymphocytes that fail to undergo apoptosis. The present study aimed to investigate the role of PMPs on CLL metabolic plasticity leading to cancer cell phenotypic changes. Methods CLL cell lines were co-incubated with different concentrations of human PMPs, and their impact on cell proliferation, mitochondrial DNA copy number, OCR level, ATP production, and ROS content was evaluated. Essential genes involved in metabolic-reprogramming were identified using the bioinformatics tools, examined between patients with early and advanced CLL stages, and then validated in PMP-recipient CLLs. Finally, the impact of the induced metabolic reprogramming on CLLs' growth, survival, mobility, and invasiveness was tested against anti-cancer drugs Cytarabine, Venetoclax, and Plumbagin. Results The data demonstrated the potency of PMPs in inducing tumoral growth and invasiveness in CLLs through mitochondrial internalization and OXPHOS stimulation which was in line with metabolic shift reported in CLL patients from early to advanced stages. This metabolic rewiring also improved CLL cells' resistance to Cytarabine, Venetoclax, and Plumbagin chemo drugs. Conclusion Altogether, these findings depict a new platelet-mediated pathway of cancer pathogenesis. We also highlight the impact of PMPs in CLL metabolic reprogramming and disease progression.
Collapse
Affiliation(s)
- Ehsan Gharib
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Vanessa Veilleux
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Luc H Boudreau
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Nicolas Pichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Gilles A Robichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| |
Collapse
|
17
|
Jiang Q, Wei B, You M, Zhou X. d-mannose blocks the interaction between keratinocytes and Th17 cells to alleviate psoriasis by inhibiting HIF-1α/CCL20 in mice. Int Immunopharmacol 2023; 118:110087. [PMID: 37001381 DOI: 10.1016/j.intimp.2023.110087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Psoriasis is an autoimmune chronic inflammatory skin disease with an unclear pathogenesis that is difficult to cure, causing serious physical and mental burdens for patients. Previous research showed that a mutually reinforcing vicious cycle caused by keratinocytes (KC) and a variety of immune cells plays an important role in psoriatic inflammation. d-Mannose, a widely distributed metabolite in the body, has been found to treat several metabolic diseases, but its impact on psoriasis remains unknown. Our study aims to investigate the effects of d-mannose on psoriasis and its specific mechanism. Here, we found that d-mannose alleviates psoriasis in mice both as oral and topical agents. Specifically, d-mannose down-regulated the expression of hypoxia-inducible factor 1A(HIF-1α) and inhibited the expression of chemokine CCL20 in keratinocytes, thereby inhibiting the local infiltration of Th17 cells and breaking the cycle of keratinocytes-Th17 cells. Overall, our study indicates that d-mannose alleviates cutaneous inflammation in psoriasis by inhibiting the HIF-1α/CCL20/Th17 cells axis, and d-mannose has the potential to be used as an oral and topical agent in the treatment of psoriasis.
Collapse
|
18
|
Dey DK, Sharma C, Vadlamudi Y, Kang SC. CopA3 peptide inhibits MDM2-p53 complex stability in colorectal cancers and activates p53 mediated cell death machinery. Life Sci 2023; 318:121476. [PMID: 36758667 DOI: 10.1016/j.lfs.2023.121476] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
The diverse expression patterns of the tumor suppressor p53 in cancer cells reflect the regulatory efficiency of multiple cellular pathways. By contrast, many human tumors are reported to develop in the presence of wild-type p53. Recently, several oncogene inhibitors have been used clinically to suppress tumor development by functionally reactivating other oncoproteins. On the other hand, p53 reactivation therapies have not been well established, as few of the p53-MDM2 complex inhibitors such as Nutlin-3 induces mutation in p53 gene upon prolonged usage. Therefore, in this study CopA3, a 9-mer dimeric D-type peptide with anticancer activity against the human colorectal cancer cells, was used to explore the efficacy of p53 reactivation in-vitro and in-vivo. The anticancer activity of CopA3 was more selective towards the wild-type p53 expressing cells than the p53 deficient or mutant colorectal cancer cells. In response to this, this study investigated the signaling pathway in vitro and validated its anti-tumor activity in-vivo. The protein-peptide interaction and molecular docking efficiently provided insight into the specific binding affinity of CopA3 to the p53-binding pocket of the MDM2 protein, which efficiently blocked the p53 and MDM2 interaction. CopA3 plays a crucial role in the binding with MDM2 and enhanced the nuclear translocation of the p53 protein, which sequentially activated the downstream targets to trigger the autophagic mediated cell death machinery through the JNK/Beclin-1 mediated pathway. Collectively, CopA3 affected the MDM2-p53 interaction, which suppressed tumor development. This study may provide a novel inhibitor candidate for the MDM2-p53 complex, which could ultimately suppress the growth of colorectal cancer cells without being cytotoxic to the healthy neighboring cells present around the tumor microenvironment.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea
| | - Chanchal Sharma
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea
| | - Yellamandayya Vadlamudi
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| |
Collapse
|
19
|
Wang C, Yang Y, Shao S, Zhang H, Li N, Zhang Z, Liu B. Accurate detection of reactive oxygen species by tuning an elastic motif (GPGGA) 4 in nanopores. Chem Commun (Camb) 2023; 59:4368-4371. [PMID: 36946304 DOI: 10.1039/d3cc00563a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
We have developed a reactive oxygen species (ROS) sensor based on nanopores modified with GGGCEG(GPGGA)4CEG. The formation of an intramolecular disulfide bond oxidized by ROS leads to conformation changes in GGGCEG(GPGGA)4CEG, which then induces an obvious change in the size of the nanopores and a corresponding ionic current change. This work allows the accurate and dynamic monitoring of ROS through the combination of (GPGGA)4 and nanopores.
Collapse
Affiliation(s)
- Cunli Wang
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Lingshui Road, Dalian, 116024, P. R. China.
| | - Yiming Yang
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Lingshui Road, Dalian, 116024, P. R. China.
| | - Shuai Shao
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Lingshui Road, Dalian, 116024, P. R. China.
| | - Hangyu Zhang
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Lingshui Road, Dalian, 116024, P. R. China.
| | - Na Li
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Lingshui Road, Dalian, 116024, P. R. China.
| | - Zhengyao Zhang
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Lingshui Road, Dalian, 116024, P. R. China.
| | - Bo Liu
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Lingshui Road, Dalian, 116024, P. R. China.
| |
Collapse
|
20
|
Park SY, Kim MW, Kang JH, Jung HJ, Hwang JH, Yang SJ, Woo JK, Jeon Y, Lee H, Yoon YS, Seong JK, Oh SH. Novel NF-κB reporter mouse for the non-invasive monitoring of inflammatory diseases. Sci Rep 2023; 13:3556. [PMID: 36864088 PMCID: PMC9981691 DOI: 10.1038/s41598-023-29689-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/08/2023] [Indexed: 03/04/2023] Open
Abstract
Bioluminescence imaging is useful for non-invasively monitoring inflammatory reactions associated with disease progression, and since NF-κB is a pivotal transcription factor that alters expressions of inflammatory genes, we generated novel NF-κB luciferase reporter (NF-κB-Luc) mice to understand the dynamics of inflammatory responses in whole body, and also in various type of cells by crossing NF-κB-Luc mice with cell-type specific Cre expressing mice (NF-κB-Luc:[Cre]). Bioluminescence intensity was significantly increased in NF-κB-Luc (NKL) mice exposed to inflammatory stimuli (PMA or LPS). Crossing NF-κB-Luc mice with Alb-cre mice or Lyz-cre mice generated NF-κB-Luc:Alb (NKLA) and NF-κB-Luc:Lyz2 (NKLL) mice, respectively. NKLA and NKLL mice showed enhanced bioluminescence in liver and macrophages, respectively. To confirm that our reporter mice could be utilized for the non-invasive monitoring of inflammation in preclinical models, we conducted a DSS-induced colitis model and a CDAHFD-induced NASH model in our reporter mice. In both models, our reporter mice reflected the development of these diseases over time. In conclusion, we believe that our novel reporter mouse can be utilized as a non-invasive monitoring platform for inflammatory diseases.
Collapse
Affiliation(s)
- Se Yong Park
- College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Min Woo Kim
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Ju-Hee Kang
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Hyun Jin Jung
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Jung Ho Hwang
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Soo Jung Yang
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Jong Kyu Woo
- Korea Mouse Phenotyping Center (KMPC), College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yoon Jeon
- Graduate School of Cancer Science and Policy, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Yeo Sung Yoon
- College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center (KMPC), College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| | - Seung Hyun Oh
- College of Pharmacy, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
21
|
Chang SN, Park JG, Kang SC. Therapeutic propensity of ginsenosides Rg1 and Rg3 in rhabdomyolysis-induced acute kidney injury and renohepatic crosstalk in rats. Int Immunopharmacol 2023; 115:109602. [PMID: 36580761 DOI: 10.1016/j.intimp.2022.109602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Ginseng is a traditional herbal medicine used for thousands of years in Southeast Asian countries because of its medicinal properties. Ginsenosides Rg1 and Rg3 have demonstrated therapeutic properties against a broad spectrum of diseases. PURPOSE Here in this study, we investigated the therapeutic efficacy of Rg1 and Rg3 in alleviating glycerol-induced acute kidney injury, also known as rhabdomyolysis-induced acute kidney injury (RAKI). METHODS AKI was induced in male Wistar rats through intramuscular injection of 10 mL/kg glycerol and simultaneous oral treatment of ginsenosides Rg1 and Rg3 for 3 days. We also evaluated the therapeutic potential of Rg1 and Rg3 on human embryonic kidney epithelial (HEK-293). Cell viability and LDH assay were performed on HEK-293 cells to evaluate the toxicity of Rg1 and Rg3. Evaluation of important kidney damage markers such as creatinine and blood urea nitrogen (BUN) was carried out at different time points from the rat serum. Histopathological analysis was performed on kidney tissues. We also performed experiments such as ELISA assay, immunohistochemistry, immunofluorescence staining, COMET assay, western blotting, TUNEL assay, and flow cytometry to obtain results. RESULTS Rg1 and Rg3 significantly downregulated the expression of kidney damage markers such as creatinine and BUN in a dose-dependent manner. Histopathological analysis revealed damage across the glomerulus, tubules, and collecting duct rendering the kidney dysfunctional in glycerol treatment groups. However, Rg1 and Rg3 treated groups showed a significant reduction in tubular necrosis at both 10 and 20 mg/kg. There was also a sharp downregulation of oxidative and ER stress markers. Additionally, we observed nuclear translocation of Nrf2 which were more prominent in kidney tissues. Rg1 and Rg3 were also able to mitigate apoptotic cell death in vitro and in vivo evaluated through immunofluorescence staining for p53, TUNEL assay, flow cytometry, and immunoblotting for intrinsic apoptosis markers. CONCLUSION In summary, we conclude that Rg1 and Rg3 exhibited natural therapeutic remedy against AKI.
Collapse
Affiliation(s)
- Sukkum Ngullie Chang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| | - Jae Gyu Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| |
Collapse
|
22
|
Medicinal Chemistry of Anti-HIV-1 Latency Chemotherapeutics: Biotargets, Binding Modes and Structure-Activity Relationship Investigation. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010003. [PMID: 36615199 PMCID: PMC9822059 DOI: 10.3390/molecules28010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
The existence of latent viral reservoirs (LVRs), also called latent cells, has long been an acknowledged stubborn hurdle for effective treatment of HIV-1/AIDS. This stable and heterogeneous reservoir, which mainly exists in resting memory CD4+ T cells, is not only resistant to highly active antiretroviral therapy (HAART) but cannot be detected by the immune system, leading to rapid drug resistance and viral rebound once antiviral treatment is interrupted. Accordingly, various functional cure strategies have been proposed to combat this barrier, among which one of the widely accepted and utilized protocols is the so-called 'shock-and-kill' regimen. The protocol begins with latency-reversing agents (LRAs), either alone or in combination, to reactivate the latent HIV-1 proviruses, then eliminates them by viral cytopathic mechanisms (e.g., currently available antiviral drugs) or by the immune killing function of the immune system (e.g., NK and CD8+ T cells). In this review, we focuse on the currently explored small molecular LRAs, with emphasis on their mechanism-directed drug targets, binding modes and structure-relationship activity (SAR) profiles, aiming to provide safer and more effective remedies for treating HIV-1 infection.
Collapse
|
23
|
Rhabdomyolysis-induced acute kidney injury and concomitant apoptosis induction via ROS-mediated ER stress is efficaciously counteracted by epigallocatechin gallate. J Nutr Biochem 2022; 110:109134. [PMID: 36028100 DOI: 10.1016/j.jnutbio.2022.109134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 01/13/2023]
Abstract
Rhabdomyolysis induced acute kidney injury (RIAKI) is a life-threatening condition responsible for approximately 19-58% of AKI cases worldwide. We performed an intramuscular injection of glycerol (10 mL/kg) in male wistar rats to induce AKI. Epigallocatechin gallate (EGCG) was administered for 3 consecutive days to evaluate its protective effects. We observed significant downregulation in serum creatinine, blood urea nitrogen (BUN) and LDH at different time points on EGCG treatment groups in a dose-dependent manner. Similarly, H&E staining also revealed that EGCG was able to reduce the formation of damaged tubules and tubular necrosis which was prominently spread throughout the kidney tissue of glycerol treatment group. Concomitantly, we observed upregulated inflammation, ER stress and elevated oxidative stress in the glycerol treated group only, which was significantly normalized upon EGCG treatment in both in vitro and in vivo studies. The occurrence of apoptosis in kidney tubules was found to be relatively higher in glycerol treated group and H2O2 treated HEK-293 cells. The results obtained after EGCG treatment revealed a significant decrease in apoptotic cell population, which was further validated by immunofluorescence staining against p53 and comet assay in HEK-293 cells and p53 IHC in kidney tissues. Western blotting also revealed a systemic downregulation of intrinsic mitochondrial apoptotic pathway markers such as bax, bcl-2, pro and cleaved caspase 3, caspase 9 and PARP1. Additionally, the results for flow cytometry analysis and TUNEL assay corroborated apoptotic equilibrium. Conclusively, we reckon EGCG as a multi-therapeutic natural product that can be used the for treatment of AKI.
Collapse
|
24
|
Kim CG, Chang SN, Park SM, Hwang BS, Kang SA, Kim KS, Park JG. Moringa oleifera mitigates ethanol-induced oxidative stress, fatty degeneration and hepatic steatosis by promoting Nrf2 in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154037. [PMID: 35358929 DOI: 10.1016/j.phymed.2022.154037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/04/2022] [Accepted: 03/09/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Moringa oleifera (M. oleifera) is cultivated throughout the world and it is known by numerous regional names and is consumed as medication for various diseases such as hypertension, diabetes, HIV and is potential source of nutrients and natural antioxidants making it among the most useful trees. METHODS We evaluated the therapeutic potential of M. oleifera on ethanol-induced fatty liver. The mice were treated with 30% ethanol (EtOH) alone or in combination with different concentration of M. oleifera extracts (100, 200 and 400 mg/kg). We performed biochemical estimation for the serum of important liver damage markers such as aspartate aminotransferase (AST), alanine aminotransferase (ALT) and triglyceride (TG). We performed histopathological analysis from the liver tissues of different mice groups. We also performed ELISA assay, western blotting analysis and SPECT imaging to obtain our results. RESULTS The results for serum (AST, p < 0.0001), (ALT, p < 0.0006) and triglyceride (TG, p < 0.0003) were found to be significantly reduced in all doses of M. oleifera extract treatment groups in comparison with the ethanol group. H&E staining analysis and scoring revealed a significant reduction in lipid droplet accumulation and a significant reduction of liver steatosis (p < 0.0001), lobular inflammation (p < 0.0013), ballooning (p < 0.0004) and immunohistochemistry for TNF-α. M. oleifera also ameliorated ethanol-induced oxidative stress evaluated through MDA (p < 0.0001), H2DCFDA, JC-1 staining and a significant down-regulation of CYP2E1 enzyme (p < 0.0001) in the 200 and 400 mg/kg groups in comparison with EtOH groups. M. oleifera extract also boosted the antioxidant response evaluated through total GSH assay (p < 0.0001) and nuclear translocation of Nrf2. Furthermore, we performed SPECT imaging and evaluated the liver uptake value (LUV) to assess the extent of liver damage. LUV was observed to be lower in the ethanol group, whereas LUV was higher in control and M. olifera treated groups. CONCLUSION In summary, from this experiment we conclude that M. oleifera extract has the potential to ameliorate ethanol-induced liver damage.
Collapse
Affiliation(s)
- Chang Geon Kim
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang, Gyeongbuk, 37668, Republic of Korea; School of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 712749, Republic of Korea
| | - Sukkum Ngullie Chang
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang, Gyeongbuk, 37668, Republic of Korea; Department of Biotechnology, Daegu University, Gyeongsan, 38453, Republic of Korea
| | - Seon Min Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang, Gyeongbuk, 37668, Republic of Korea; Department of Veterinary Toxicology, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Buyng Su Hwang
- Nakdonggang National Institute of Biological Resources, Sangju, 37242, Republic of Korea
| | - Sung-A Kang
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang, Gyeongbuk, 37668, Republic of Korea
| | - Kil Soo Kim
- Department of Veterinary Toxicology, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea.
| | - Jae Gyu Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang, Gyeongbuk, 37668, Republic of Korea.
| |
Collapse
|
25
|
Van den Broek B, Wuyts C, Sisto A, Pintelon I, Timmermans JP, Somers V, Timmerman V, Hellings N, Irobi J. Oligodendroglia-derived extracellular vesicles activate autophagy via LC3B/BAG3 to protect against oxidative stress with an enhanced effect for HSPB8 enriched vesicles. Cell Commun Signal 2022; 20:58. [PMID: 35513867 PMCID: PMC9069805 DOI: 10.1186/s12964-022-00863-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/12/2022] [Indexed: 01/18/2023] Open
Abstract
Background The contribution of native or modified oligodendroglia-derived extracellular vesicles (OL-EVs) in controlling chronic inflammation is poorly understood. In activated microglia, OL-EVs contribute to the removal of cytotoxic proteins following a proteotoxic stress. Intracellular small heat shock protein B8 (HSPB8) sustain this function by facilitating autophagy and protecting cells against oxidative stress mediated cell death. Therefore, secretion of HSPB8 in OL-EVs could be beneficial for neurons during chronic inflammation. However, how secreted HSPB8 contribute to cellular proteostasis remains to be elucidated. Methods We produced oligodendroglia-derived EVs, either native (OL-EVs) or HSPB8 modified (OL-HSPB8-EVs), to investigate their effects in controlling chronic inflammation and cellular homeostasis. We analyzed the impact of both EV subsets on either a resting or activated microglial cell line and on primary mixed neural cell culture cells. Cells were activated by stimulating with either tumor necrosis factor-alpha and interleukin 1-beta or with phorbol-12-myristate-13-acetate. Results We show that OL-EVs and modified OL-HSPB8-EVs are internalized by C20 microglia and by primary mixed neural cells. The cellular uptake of OL-HSPB8-EVs increases the endogenous HSPB8 mRNA expression. Consistently, our results revealed that both EV subsets maintained cellular homeostasis during chronic inflammation with an increase in the formation of autophagic vesicles. Both EV subsets conveyed LC3B-II and BAG3 autophagy markers with an enhanced effect observed for OL-HSPB8-EVs. Moreover, stimulation with either native or modified OL-HSPB8-EVs showed a significant reduction in ubiquitinated protein, reactive oxygen species and mitochondrial depolarization, with OL-HSPB8-EVs exhibiting a more protective effect. Both EV subsets did not induce cell death in the C20 microglia cell line or the primary mixed neural cultures. Conclusion We demonstrate that the functions of oligodendroglia secreted EVs enriched with HSPB8 have a supportive role, comparable to the native OL-EVs. Further development of engineered oligodendroglia derived EVs could be a novel therapeutic strategy in countering chronic inflammation. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00863-x.
Collapse
Affiliation(s)
- Bram Van den Broek
- Department of Immunology and Infections, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Charlotte Wuyts
- Department of Immunology and Infections, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Angela Sisto
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, Institute Born Bunge and University of Antwerp, Antwerp, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology & Histology, Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology & Histology, Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Antwerp, Belgium
| | - Veerle Somers
- Department of Immunology and Infections, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, Institute Born Bunge and University of Antwerp, Antwerp, Belgium
| | - Niels Hellings
- Department of Immunology and Infections, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Joy Irobi
- Department of Immunology and Infections, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.
| |
Collapse
|
26
|
González-Ballesteros N, Diego-González L, Lastra-Valdor M, Grimaldi M, Cavazza A, Bigi F, Rodríguez-Argüelles MC, Simón-Vázquez R. Immunomodulatory and Antitumoral Activity of Gold Nanoparticles Synthesized by Red Algae Aqueous Extracts. Mar Drugs 2022; 20:md20030182. [PMID: 35323481 PMCID: PMC8953345 DOI: 10.3390/md20030182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 12/11/2022] Open
Abstract
This study reports on the green and cost-efficient synthesis of gold nanoparticles from three different red algae extracts. The nanoparticles synthesized were fully characterized by UV-Vis spectroscopy, HRTEM, and Z-potential. Relevant components occurring in the extracts, such as polysaccharides or phenolic content, were assessed by analytical techniques such as spectrophotometric assays and liquid chromatography. Finally, the antioxidant, antitumoral, and anti-inflammatory potential of both the extracts and the gold nanoparticles synthesized were analyzed in order to determine a possible synergistic effect on the nanoparticles. The results obtained confirmed the obtainment of gold nanoparticles with significant potential as immunotherapeutic agents. The therapeutic potential of these nanoparticles could be higher than that of inert gold nanoparticles loaded with bioactive molecules since the former would allow for higher accumulation into the targeted tissue.
Collapse
Affiliation(s)
| | - Lara Diego-González
- CINBIO, Immunology Group, Universidade de Vigo, 36310 Vigo, Spain; (L.D.-G.); (R.S.-V.)
- Instituto de Investigación Sanitaria Galicia Sur, Hospital Alvaro Cunqueiro, 36312 Vigo, Spain
| | | | - Maria Grimaldi
- Dipartimento Scienze Chimiche, della Vita e della Sostenibilità Ambientale, Università di Parma, 43124 Parma, Italy; (M.G.); (A.C.); (F.B.)
| | - Antonella Cavazza
- Dipartimento Scienze Chimiche, della Vita e della Sostenibilità Ambientale, Università di Parma, 43124 Parma, Italy; (M.G.); (A.C.); (F.B.)
| | - Franca Bigi
- Dipartimento Scienze Chimiche, della Vita e della Sostenibilità Ambientale, Università di Parma, 43124 Parma, Italy; (M.G.); (A.C.); (F.B.)
- Institute of Materials for Electronics and Magnetism, National Research Council, 43124 Parma, Italy
| | | | - Rosana Simón-Vázquez
- CINBIO, Immunology Group, Universidade de Vigo, 36310 Vigo, Spain; (L.D.-G.); (R.S.-V.)
- Instituto de Investigación Sanitaria Galicia Sur, Hospital Alvaro Cunqueiro, 36312 Vigo, Spain
| |
Collapse
|
27
|
89Zr Immuno-PET Imaging of Tumor PD-1 Reveals That PMA Upregulates Lymphoma PD-1 through NFκB and JNK Signaling. Mol Imaging 2022; 2022:5916692. [PMID: 35250391 PMCID: PMC8865856 DOI: 10.1155/2022/5916692] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/21/2021] [Accepted: 01/27/2022] [Indexed: 12/15/2022] Open
Abstract
Immune therapy of T-cell lymphoma requires assessment of tumor-expressed programmed cell death protein-1 (PD-1). Herein, we developed an immuno-PET technique that quantitatively images and monitors regulation of PD-1 expression on T-cell lymphomas. Methods. Anti-PD-1 IgG underwent sulfhydryl moiety-specific conjugation with maleimide-deferoxamine and 89Zr labeling. Binding assays and Western blotting were performed in EL4 murine T-cell lymphoma cells. In vivo pharmacokinetics, biodistribution, and PET were performed in mice. Results. 89Zr-PD-1 IgG binding to EL4 cells was completely blocked by cold antibodies, confirming excellent target specificity. Following intravenous injection into mice, 89Zr-PD-1 IgG showed biexponential blood clearance and relatively low normal organ uptake after five days. PET/CT and biodistribution demonstrated high EL4 tumor uptake that was suppressed by cold antibodies. In EL4 cells, phorbol 12-myristate 13-acetate (PMA) increased 89Zr-PD-1 IgG binding (
%) and dose-dependent augmentation of PD-1 expression (
of controls by 200 ng/ml). FACS showed strong PD-1 expression on all EL4 cells and positive but weaker expression on
% of the mouse spleen lymphocytes. PMA stimulation led to
-fold increase in the proportion of the strongest PD-1 expressing EL4 cells but failed to influence that of PD-1+ mouse lymphocytes. In mice, PMA treatment increased 89Zr-PD-1 IgG uptake in EL4 lymphomas from
to
%ID/g (
), and tumor uptake closely correlated with PD-1 level (
,
). On immunohistochemistry of tumor sections, infiltrating CD8α+ T lymphocytes constituted a small fraction of tumor cells. The entire tumor section showed strong PD-1 staining that was even stronger for PMA-treated mice. Investigation of involved signaling revealed that PMA increased EL4 cell and tumor HIF-1α accumulation and NFκB and JNK activation. Conclusion. 89Zr-PD-1 IgG offered high-contrast PET imaging of tumor PD-1 in mice. This was found to mostly represent binding to EL4 tumor cells, although infiltrating T lymphocytes may also have contributed. PD-1 expression on T-cell lymphomas was upregulated by PMA stimulation, and this was reliably monitored by 89Zr-PD-1 IgG PET. This technique may thus be useful for understanding the mechanisms of PD-1 regulation in lymphomas of living subjects.
Collapse
|
28
|
Rivera-Yañez CR, Ruiz-Hurtado PA, Mendoza-Ramos MI, Reyes-Reali J, García-Romo GS, Pozo-Molina G, Reséndiz-Albor AA, Nieto-Yañez O, Méndez-Cruz AR, Méndez-Catalá CF, Rivera-Yañez N. Flavonoids Present in Propolis in the Battle against Photoaging and Psoriasis. Antioxidants (Basel) 2021; 10:antiox10122014. [PMID: 34943117 PMCID: PMC8698766 DOI: 10.3390/antiox10122014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/09/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022] Open
Abstract
The skin is the main external organ. It protects against different types of potentially harmful agents, such as pathogens, or physical factors, such as radiation. Skin disorders are very diverse, and some of them lack adequate and accessible treatment. The photoaging of the skin is a problem of great relevance since it is related to the development of cancer, while psoriasis is a chronic inflammatory disease that causes scaly skin lesions and deterioration of the lifestyle of people affected. These diseases affect the patient's health and quality of life, so alternatives have been sought that improve the treatment for these diseases. This review focuses on describing the properties and benefits of flavonoids from propolis against these diseases. The information collected shows that the antioxidant and anti-inflammatory properties of flavonoids play a crucial role in the control and regulation of the cellular and biochemical alterations caused by these diseases; moreover, flavones, flavonols, flavanones, flavan-3-ols, and isoflavones contained in different worldwide propolis samples are the types of flavonoids usually evaluated in both diseases. Therefore, the research carried out in the area of dermatology with bioactive compounds of different origins is of great relevance to developing preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Claudia Rebeca Rivera-Yañez
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico;
| | - Porfirio Alonso Ruiz-Hurtado
- Laboratorio de Toxicología de Productos Naturales, Departamento de Farmacia, IPN, Escuela Nacional de Ciencias Biológicas, Av. Wilfrido Massieu, Gustavo A. Madero 07738, Mexico;
| | - María Isabel Mendoza-Ramos
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- Laboratorio de Inmunología, Unidad de Morfofisiología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Julia Reyes-Reali
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- Laboratorio de Inmunología, Unidad de Morfofisiología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Gina Stella García-Romo
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- Laboratorio de Inmunología, Unidad de Morfofisiología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Glustein Pozo-Molina
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- Laboratorio de Genética y Oncología Molecular, Laboratorio 5, Edificio A4, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Aldo Arturo Reséndiz-Albor
- Laboratorio de Inmunidad de Mucosas, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Salvador Díaz Mirón y Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Mexico City 11340, Mexico;
| | - Oscar Nieto-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
| | - Adolfo René Méndez-Cruz
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- Laboratorio de Inmunología, Unidad de Morfofisiología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Claudia Fabiola Méndez-Catalá
- Laboratorio de Genética y Oncología Molecular, Laboratorio 5, Edificio A4, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- División de Investigación y Posgrado, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Correspondence: (C.F.M.-C.); (N.R.-Y.); Tel.: +52-5522-476-721 (N.R.-Y.)
| | - Nelly Rivera-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- División de Investigación y Posgrado, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Correspondence: (C.F.M.-C.); (N.R.-Y.); Tel.: +52-5522-476-721 (N.R.-Y.)
| |
Collapse
|
29
|
Ferreira MS, Resende DISP, Lobo JMS, Sousa E, Almeida IF. Marine Ingredients for Sensitive Skin: Market Overview. Mar Drugs 2021; 19:md19080464. [PMID: 34436303 PMCID: PMC8398991 DOI: 10.3390/md19080464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 12/19/2022] Open
Abstract
Marine ingredients are a source of new chemical entities with biological action, which is the reason why they have gained relevance in the cosmetic industry. The facial care category is the most relevant in this industry, and within it, the sensitive skin segment occupies a prominent position. This work analyzed the use of marine ingredients in 88 facial cosmetics for sensitive skin from multinational brands, as well as their composition and the scientific evidence that supports their efficacy. Marine ingredients were used in 27% of the cosmetic products for sensitive skin and included the species Laminaria ochroleuca, Ascophyllum nodosum (brown macroalgae), Asparagopsis armata (red macroalgae), and Chlorella vulgaris (microalgae). Carotenoids, polysaccharides, and lipids are the chemical classes highlighted in these preparations. Two ingredients, namely the Ascophyllum nodosum extract and Asparagopsis armata extracts, present clinical evidence supporting their use for sensitive skin. Overall, marine ingredients used in cosmetics for sensitive skin are proposed to reduce skin inflammation and improve the barrier function. Marine-derived preparations constitute promising active ingredients for sensitive skin cosmetic products. Their in-depth study, focusing on the extracted metabolites, randomized placebo-controlled studies including volunteers with sensitive skin, and the use of extraction methods that are more profitable may provide a great opportunity for the cosmetic industry.
Collapse
Affiliation(s)
- Marta Salvador Ferreira
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.S.F.); (J.M.S.L.)
- UCIBIO–Applied Molecular Biosciences Unit, MedTech, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Diana I. S. P. Resende
- CIIMAR–Centro Interdisciplinar de Investigação Marinha e Ambiental, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; (D.I.S.P.R.); (E.S.)
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - José M. Sousa Lobo
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.S.F.); (J.M.S.L.)
- UCIBIO–Applied Molecular Biosciences Unit, MedTech, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Emília Sousa
- CIIMAR–Centro Interdisciplinar de Investigação Marinha e Ambiental, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; (D.I.S.P.R.); (E.S.)
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - Isabel F. Almeida
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.S.F.); (J.M.S.L.)
- UCIBIO–Applied Molecular Biosciences Unit, MedTech, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Correspondence: ; Tel.: +351-220-428
| |
Collapse
|
30
|
Dey DK, Kang SC. CopA3 peptide induces permanent cell-cycle arrest in colorectal cancer cells. Mech Ageing Dev 2021; 196:111497. [PMID: 33957217 DOI: 10.1016/j.mad.2021.111497] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 12/23/2022]
Abstract
Cell-cycle arrest reflects an accumulation of responses to DNA damage that sequentially affects cell growth and division. Herein, we analyzed the effect of the 9-mer dimer defensin-like peptide, CopA3, against colorectal cancer cell growth and proliferation in a dose-dependent manner upon 96 h of treatment. As observed, CopA3 treatment significantly affected cancer cell growth, reduced colony formation ability, increased the number of SA-β-Gal positive cells, and remarkably reduced Ki67 protein expression. Notably, in HCT-116 cells, CopA3 (5 μM) treatment effectively increased oxidative stress and, as a result, amplified the endogenous ROS, mitochondrial ROS, and NO content in the cells, which further activated the DNA damage response and caused cell-cycle arrest at the G1 phase. The prolonged cell-cycle arrest elevated the release of inflammatory cytokines in the cell supernatant. Nevertheless, mechanistically, NAC treatment effectively reversed the CopA3 effect and significantly reduced the oxidative stress; subsequently rescuing the cells from G1 phase arrest. Overall, CopA3 treatment can inhibit the growth and proliferation of colorectal cancer cells by inducing cell-cycle arrest through the ROS-mediated pathway.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
31
|
Dey DK, Chang SN, Gu JY, Kim KM, Lee JJ, Kim TH, Kang SC. Ultraviolet B-irradiated mushroom supplementation increased the Ca ++ uptake and ameliorated the LPS-induced inflammatory responses in zebrafish larvae. J Food Biochem 2021; 45:e13742. [PMID: 33931887 DOI: 10.1111/jfbc.13742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/05/2021] [Accepted: 04/07/2021] [Indexed: 12/26/2022]
Abstract
The harmful effects of excessive ultraviolet (UV) exposure are well known. However, moderate exposure to UV radiation is beneficial and required for active vitamin D synthesis in our body. People living in the coldest regions on the earth are unable to expose their skin to the solar UV radiation and, therefore, additional supplementation of Vitamin D2 is recommended. Mushrooms are one such consumable macrofungi, which has high vitamin content and therefore used in various traditional medicines. Particularly, UVB-irradiated mushrooms are rich in active vitamin D content and that is why recommended to include in the daily diets for the patients suffering from the problems associated with bone mineralization. In the present study, we evaluated the cytotoxic effect of mushroom extract (UVB-ME) (Lentinus edodes) treatment against MG-63 cells, HepG2 cells, and CCD 841 CoN cells. Furthermore, we elucidated the potential of UVB-ME on Ca++ uptake in osteoblast-like MG-63 cells. Next, we validated the response of Ca++ uptake on the growth and development of zebrafish larvae. In addition, the anti-inflammatory and immunomodulatory potential of UVB-ME treatment against lipopolysaccharide-induced inflammatory response was also analyzed in vivo. Collectively, the study suggested that dietary supplementation of UVB-irradiated mushroom is beneficial for bone calcification and could modulate the host immune system.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Gyeongsan, Republic of Korea
| | | | - Ji Ye Gu
- Department of Pharmaceutical Science and Technology, Kyungsung University, Busan, Republic of Korea
| | - Kang Min Kim
- Department of Pharmaceutical Science and Technology, Kyungsung University, Busan, Republic of Korea
| | | | - Tae Hee Kim
- Naturetech Co. Ltd., Chungbuk, Republic of Korea
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Republic of Korea
| |
Collapse
|
32
|
Chang SN, Khan I, Kim CG, Park SM, Choi DK, Lee H, Hwang BS, Kang SC, Park JG. Decursinol Angelate Arrest Melanoma Cell Proliferation by Initiating Cell Death and Tumor Shrinkage via Induction of Apoptosis. Int J Mol Sci 2021; 22:4096. [PMID: 33921050 PMCID: PMC8071397 DOI: 10.3390/ijms22084096] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Melanoma is known to aggressively metastasize and is one of the prominent causes of skin cancer mortality. This study was designed to assess the molecular mechanism of decursinol angelate (DA) against murine melanoma cell line (B16F10 cells). Treatment of DA resulted in growth inhibition and cell cycle arrest at G0/G1 (p < 0.001) phase, evaluated through immunoblotting. Moreover, autophagy-related proteins such as ATG-5 (p < 0.0001), ATG-7 (p < 0.0001), beclin-1 (p < 0.0001) and transition of LC3-I to LC3-II (p < 0.0001) were markedly decreased, indicating autophagosome inhibition. Additionally, DA treatment triggered apoptotic events which were corroborated by the occurrence of distorted nuclei, elevated reactive oxygen species (ROS) levels and reduction in the mitochondrial membrane potential. Subsequently, there was an increase in the expression of pro-apoptotic protein Bax in a dose-dependent manner, with the corresponding downregulation of Bcl-2 expression and cytochrome C expression following 24 h DA treatment in A375.SM and B16F10 cells. We substantiated our results for apoptotic occurrence through flow cytometry in B16F10 cells. Furthermore, we treated B16F10 cells with N-acetyl-L-cysteine (NAC). NAC treatment upregulated ATG-5 (p < 0.0001), beclin-1 (p < 0.0001) and LC3-I to LC3-II (p < 0.0001) conversion, which was inhibited in the DA treatment group. We also noticed a systematic upregulation of important markers for progression of G1 cell phase such as CDK-2 (p < 0.029), CDK-4 (p < 0.036), cyclin D1 (p < 0.0003) and cyclin E (p < 0.020) upon NAC treatment. In addition, we also observed a significant fold reduction (p < 0.05) in ROS fluorescent intensity and the expression of Bax (p < 0.0001), cytochrome C (p < 0.0001), cleaved caspase-9 (p > 0.010) and cleaved caspase-3 (p < 0.0001). NAC treatment was able to ameliorate DA-induced apoptosis and cell cycle arrest to support our finding. Our in vivo xenograft model also revealed similar findings, such as downregulation of CDK-2 (p < 0.0001) and CDK-4 (p < 0.0142) and upregulation of Bax (p < 0.0001), cytochrome C (p < 0.0001), cleaved caspase 3 (p < 0.0001) and cleaved caspase 9 (p < 0.0001). In summary, our study revealed that DA is an effective treatment against B16F10 melanoma cells and xenograft mice model.
Collapse
Affiliation(s)
- Sukkum Ngullie Chang
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea;
| | - Imran Khan
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea;
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Chang Geon Kim
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
| | - Seon Min Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
| | - Dong Kyu Choi
- New Drug Development Center, DGMIF, 88 Dongnae-ro, Dong-gu, Daegu 41061, Korea; (D.K.C.); (H.L.)
| | - Heejin Lee
- New Drug Development Center, DGMIF, 88 Dongnae-ro, Dong-gu, Daegu 41061, Korea; (D.K.C.); (H.L.)
| | - Buyng Su Hwang
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Korea;
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea;
| | - Jae Gyu Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
| |
Collapse
|
33
|
Chang SN, Kim SH, Dey DK, Park SM, Nasif O, Bajpai VK, Kang SC, Lee J, Park JG. 5-O-Demethylnobiletin Alleviates CCl 4-Induced Acute Liver Injury by Equilibrating ROS-Mediated Apoptosis and Autophagy Induction. Int J Mol Sci 2021; 22:ijms22031083. [PMID: 33499185 PMCID: PMC7865239 DOI: 10.3390/ijms22031083] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 01/08/2023] Open
Abstract
Polymethoxyflavanoids (PMFs) have exhibited a vast array of therapeutic biological properties. 5-O-Demethylnobiletin (5-DN) is one such PMF having anti-inflammatory activity, yet its role in hepatoprotection has not been studied before. Results from in vitro study revealed that 5-DN did not exert a high level of cytotoxicity on HepG2 cells at 40 μM, and it was able to rescue HepG2 cell death induced by carbon tetrachloride (CCl4). Subsequently, we investigated acute liver injury on BALB/c mice induced by CCl4 through the intraperitoneal injection of 1 mL/kg CCl4 and co-administration of 5-DN at (1 and 2 mg/kg) by oral gavage for 15 days. The results illustrated that treatment with 5-DN attenuated CCl4-induced elevated serum aminotransferase (AST)/alanine aminotransferase (ALT) ratio and significantly ameliorated severe hepatic damage such as inflammation and fibrosis evidenced through lesser aberrations in the liver histology of 5-DN dose groups. Additionally, 5-DN efficiently counteracted and equilibrated the production of ROS accelerated by CCl4 and dramatically downregulated the expression of CYP2E1 vitally involved in converting CCl4 to toxic free radicals and also enhanced the antioxidant enzymes. 5-DN treatment also inhibited cell proliferation and inflammatory pathway abnormally regulated by CCl4 treatment. Furthermore, the apoptotic response induced by CCl4 treatment was remarkably reduced by enhanced Bcl-2 expression and noticeable reduction in Bax, Bid, cleaved caspase 3, caspase 9, and apaf-1 expression. 5-DN treatment also induced the conversion of LC3 and promoted the autophagic flux. Conclusively, 5-DN exhibited hepatoprotective effects in vitro and in vivo and prevented liver fibrosis induced by CCl4.
Collapse
Affiliation(s)
- Sukkum Ngullie Chang
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea; (S.N.C.); (D.K.D.); (S.C.K.)
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.H.K.); (S.M.P.)
| | - Se Ho Kim
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.H.K.); (S.M.P.)
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea
| | - Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea; (S.N.C.); (D.K.D.); (S.C.K.)
| | - Seon Min Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.H.K.); (S.M.P.)
| | - Omaima Nasif
- Department of Physiology, College of Medicine, King Saud University (Medical City), King Khalid University Hospital, P.O. Box 2925, Riyadh 11461, Saudi Arabia;
| | - Vivek K. Bajpai
- Department of Energy and Materials Engineering, Dongguk University-Seoul, 30 Pildong-ro 1-gil, Seoul 04620, Korea
- Correspondence: (V.K.B.); (J.T.L.); (J.G.P.); Fax: +82-32-872-4046 (V.K.B.); +82-53-810-4631 (J.L.); +82-54-223-2780 (J.G.P.)
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea; (S.N.C.); (D.K.D.); (S.C.K.)
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea
- Correspondence: (V.K.B.); (J.T.L.); (J.G.P.); Fax: +82-32-872-4046 (V.K.B.); +82-53-810-4631 (J.L.); +82-54-223-2780 (J.G.P.)
| | - Jae Gyu Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.H.K.); (S.M.P.)
- Correspondence: (V.K.B.); (J.T.L.); (J.G.P.); Fax: +82-32-872-4046 (V.K.B.); +82-53-810-4631 (J.L.); +82-54-223-2780 (J.G.P.)
| |
Collapse
|