1
|
Abid A, Mekhadmi N, Mlik R, Bentahar A, Bireche K, Frih B, Boussebaa W, Mouane A, Cherrada N, Sanches Silva A, Dekmouche M, Bechki L, Al-Anazi KM, Farah MA, Ali A. Unveiling the Therapeutic Potential of Atractylis aristata Batt. Aqueous Extract: Anti-inflammatory, Antioxidant, Antibacterial, Sedative Activities & Phytochemical Profiling. ChemistryOpen 2025:e202500056. [PMID: 40244084 DOI: 10.1002/open.202500056] [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: 01/24/2025] [Revised: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Medicinal plants possess the potential to yield bioactive compounds that offer significant health benefits; positioning them as valuable and promising sources for the development of innovative pharmaceutical products. This study aims to comprehensively assess the in vitro and in vivo pharmacological effects of the aqueous extract of the plant Atractylis aristata (AEAA) as well as assessments of its phytochemical composition. UPLC-ESI-MS/MS analysis of AEAA revealed a variety of bioactive compounds, including flavonoids and phenolic acids. In antioxidant assays, AEAA demonstrated considerable activity, with IC50 values of 0.269±0.05 mg/mL for DPPH scavenging and 0.0376±0.003 mg/mL for hydrogen peroxide radical inhibition. AEAA exhibited strong anti-inflammatory activity in vitro, with an IC50 value of 2.563 mg/mL in the BSA denaturation test. In vivo, AEAA reduced carrageenan-induced paw edema by 56.51 %, in comparison to an 83.58 % reduction with Ibuprofen®. Antibacterial testing showed AEAA's broad-spectrum activity, with the highest inhibition against Bacillus subtilis (34 mm zone of inhibition). Additionally, AEAA induced significant sedative effects, reducing locomotor activity by 48.98 %. These findings underscore the diverse pharmacological potential in addressing oxidative stress, inflammation, microbial infections, and anxiety of A. aristata, which can be attributed to its rich phytochemical profile.
Collapse
Affiliation(s)
- Asma Abid
- Laboratory of Valorization and Promotion of Saharan Resources, Faculty of Mathematics and Matter Sciences, University of Ouargla, Road of Ghardaia, 30000, Ouargla, Algeria
| | - Nourelhouda Mekhadmi
- Department of Biology, Faculty of Nature and Life Sciences, University of El Oued, 39000, El Oued, Algeria
- Laboratory of the Development and Technology of Saharan Resources (VTRS), Echahid Hamma Lakhdar El Oued University, Algeria
| | - Randa Mlik
- National Institute of Agronomic Research of Algeria, INRAA, P. O. Box 299, Station of, Adrar, Adrar, Algeria
| | - Assia Bentahar
- Laboratory of Phytotherapy Applied to Chronic Diseases, SNV Faculty, University of Setif 1, 19000, Sétif, Algeria
| | - Kamilia Bireche
- Laboratory of Valorization and Promotion of Saharan Resources, Faculty of Mathematics and Matter Sciences, University of Ouargla, Road of Ghardaia, 30000, Ouargla, Algeria
| | - Bariza Frih
- Department of Biology, Faculty of Nature and Life Sciences, University of El Oued, 39000, El Oued, Algeria
- Laboratory of the Development and Technology of Saharan Resources (VTRS), Echahid Hamma Lakhdar El Oued University, Algeria
| | - Walid Boussebaa
- Scientific and Technical Research Center in Physico-Chemical Analysis (CRAPC), Tipaza, Algeria
| | - Aicha Mouane
- Department of Biology, Faculty of Nature and Life Sciences, University of El Oued, 39000, El Oued, Algeria
| | - Nezar Cherrada
- Department of Biology, Faculty of Nature and Life Sciences, University of El Oued, 39000, El Oued, Algeria
- Laboratory of Biodiversity and Application of Biotechnology in Agriculture, University of El Oued, El-Oued, Algeria
| | - Ana Sanches Silva
- University of Coimbra, Faculty of Pharmacy, Coimbra, 3000-548 Coimbra, Portugal
- Centre for Animal Science Studies (CECA), ICETA, 4099-002, Porto, Portugal
- Associate Laboratory for Animal and Veterinary Sciences, 1300-477, Lisbon, Portugal
| | - Messaouda Dekmouche
- Laboratory of Valorization and Promotion of Saharan Resources, Faculty of Mathematics and Matter Sciences, University of Ouargla, Road of Ghardaia, 30000, Ouargla, Algeria
| | - Lazhar Bechki
- Laboratory of Valorization and Promotion of Saharan Resources, Faculty of Mathematics and Matter Sciences, University of Ouargla, Road of Ghardaia, 30000, Ouargla, Algeria
| | - Khalid Mashay Al-Anazi
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohammad Abul Farah
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Ahmad Ali
- Department of Life Sciences, University of Mumbai, Vidyanagari, Mumbai, 400098
| |
Collapse
|
2
|
Li S, Wang S, Zhang L, Ka Y, Zhou M, Wang Y, Tang Z, Zhang J, Wang W, Liu W. Research progress on pharmacokinetics, anti-inflammatory and immunomodulatory effects of kaempferol. Int Immunopharmacol 2025; 152:114387. [PMID: 40054326 DOI: 10.1016/j.intimp.2025.114387] [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: 12/13/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/24/2025]
Abstract
Chronic inflammation (an abnormal state) and autoimmune disease (AD) can both cause multiple organ damage. AD is a heterogeneous group of diseases due to immune dysfunction. Chronic inflammation is closely related to AD and is an important part of AD. With the increasing prevalence of AD, researchers are constantly exploring new drugs with small side effects, considerable curative effects, and lower costs. Kaempferol, a flavonoid, possesses a range of biological functions, including antioxidant, anti-inflammatory, anti-neoplastic, and immunomodulatory capabilities. This compound is prevalent in a variety of plant sources, such as vegetables, fruits, and medicinal herbs traditionally used in Chinese medicine. A plethora of empirical evidence from animal-based research supports the assertion that this particular substance exhibits both anti-inflammatory and immunomodulatory effects, with the curative effect being significant and application prospects. This article mainly summarizes and discusses the pharmacokinetics, drug delivery system, and the mechanism of kaempferol on immune cells, cytokines, signaling pathways, and other aspects. This paper summarizes the existing kaempferol drug delivery system, analyzes the possibility and limitations of kaempferol as a new anti-inflammatory and immunomodulatory drug, and discusses how to apply it in clinical practice. Therefore, kaempferol can more effectively exert its anti-inflammatory and immune-modulating effects, thereby demonstrating therapeutic potential in clinical settings, while reducing patient burden.
Collapse
Affiliation(s)
- Suiran Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Siwei Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Lei Zhang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, China
| | - Yuxiu Ka
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Meijiao Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yiwen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Zhuo Tang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Jiamin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
3
|
Luo J, Chen K, Nong X. Potential regulation of artesunate on bone metabolism through suppressing inflammatory infiltration in type 2 diabetes mellitus. Immunopharmacol Immunotoxicol 2025; 47:147-158. [PMID: 39762719 DOI: 10.1080/08923973.2024.2444953] [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: 01/24/2024] [Accepted: 12/15/2024] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Osteoimmunology is an emerging field that explores the interplay between bone and the immune system. The immune system plays a critical role in the pathogenesis of diabetes and significantly affects bone homeostasis. Artesunate, a first-line treatment for malaria, is known for its low toxicity and multifunctional properties. Increasing evidence suggests that artesunate possesses anti-inflammatory, immunoregulatory, and osteogenic effects. This review aims to explore the relationship between immune regulation and bone metabolism in type 2 diabetes (T2DM) and to investigate the potential therapeutic application of artesunate. METHODS This review systematically examines literature from PubMed/Medline, Elsevier, Web of Science, Embase, the International Diabetes Federation, and other relevant databases. RESULTS This review synthesizes evidence from multiple sources to delineate the relationship between T lymphocytes and T2DM, the regulation of T lymphocyte subsets in bone metabolism, and the effects of artesunate on both T lymphocytes and bone metabolism. Recent studies suggest a bidirectional regulatory relationship between T2DM and T lymphocytes (CD4+ T and CD8+ T) during the onset and progression of the disease, with inflammatory and anti-inflammatory cytokines serving as key mediators. T lymphocyte subsets and their cytokines play a pivotal role in regulating osteogenesis and osteoclastogenesis in pathological conditions. Furthermore, artesunate has shown promise in modulating inflammatory infiltration and bone metabolism. CONCLUSION The accumulated evidence indicates that artesunate exerts regulatory effects on bone metabolism in T2DM by influencing T lymphocyte differentiation.
Collapse
Affiliation(s)
- Jinghong Luo
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Kun Chen
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaolin Nong
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
4
|
Nazakat L, Ali S, Summer M, Nazakat F, Noor S, Riaz A. Pharmacological modes of plant-derived compounds for targeting inflammation in rheumatoid arthritis: A comprehensive review on immunomodulatory perspective. Inflammopharmacology 2025; 33:1537-1581. [PMID: 40074996 DOI: 10.1007/s10787-025-01664-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/28/2024] [Indexed: 03/14/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is one of the most prevalent autoimmune, chronic, inflammatory disease characterized by joint inflammation, synovial swelling, loss of articular structures, swelling, and pain. RA is a major cause of discomfort and disability worldwide, associated with infectious agents, genetic determinants, epigenetic factors, advancing age, obesity, and smoking. Although conventional therapies for RA alleviate the symptoms, but their long-term use is associated with significant side effects. This necessitates the urge to discover complementary and alternative medicine from natural products with minimum side effects. PURPOSE In this review, natural product's potential mechanism of action against RA has been documented in the setting of in-vivo, in-vitro and pre-clinical trials, which provides new treatment opportunities for RA patients. The bioefficacy of these natural product's bioactive compounds must be further studied to discover novel natural medications for RA with high selectivity, improved effectiveness, and economic replacement with minimum side effects. STUDY DESIGN AND METHODS The current review article was designed systematically in chronological order. Plants and their phytochemicals are discussed in an order concerning their mode of action. All the mechanisms of action are depicted in diagrams which are thoroughly generated by the Chembiodraw to maintain the integrity of the work. Moreover, by incorporating the recent data with simple language which is not incorporated previously, we tried to provide a molecular insight to the readers of every level and ethnicity. Moreover, Google Scholar, PubMed, ResearchGate, and Science Direct databases were used to collect the data. SOLUTION Traditionally, various plant extracts and bioactive compounds are effectively used against RA, but their comprehensive pharmacological mechanistic actions are rarely discussed. Therefore, the objective of this study is to systematically review the efficacy and proposed mechanisms of action of different plants and their bioactive compounds including Tripterygium wilfordii Hook F (celastrol and triptolide), Nigella sativa (thymoquinone), Zingiber officinale (shogaols, zingerone), Boswellia serrata (boswellic acids), Curcuma longa (curcumin), and Syzygium aromaticum (eugenol) against rheumatoid arthritis. CONCLUSION These plants have strong anti-inflammatory, anti-oxidant, and anti-arthritic effects in different study designs of rheumatoid arthritis with negligible side effects. Phytomedicines could revolutionize pharmacology as they act through alternative pathways hence seeming biocompatible.
Collapse
Affiliation(s)
- Laiba Nazakat
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan.
| | - Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Fakiha Nazakat
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Shehzeen Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Anfah Riaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| |
Collapse
|
5
|
Wei J, Chen XY, Wang ZJ, Li XY, Zhang MM, Sun T, Zhang XR, Wang DG, Hou C, Meng XM. Galloflavin mitigates acute kidney injury by suppressing LDHA-dependent macrophage glycolysis. Int Immunopharmacol 2025; 150:114265. [PMID: 39955920 DOI: 10.1016/j.intimp.2025.114265] [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: 01/05/2025] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
Macrophage-mediated inflammation is closely linked to the pathogenesis of acute kidney injury (AKI) and the shift of macrophages to a pro-inflammatory phenotype being reliant on glycolytic metabolism. Galloflavin, a polyphenol derived from tea, functions as a lactate dehydrogenase A (LDHA) inhibitor, effectively obstructing glycolytic metabolic pathways. However, the specific immunometabolic regulatory functions of galloflavin in macrophages remain unclear. Here, we observed that galloflavin drives alleviation of glycolytic metabolism levels in lipopolysaccharide (LPS)-induced macrophages (RAW264.7 cells and human peripheral blood mononuclear cells-derived macrophages) through downregulation of LDHA expression, thereby inhibiting macrophage conversion to a pro-inflammatory phenotype and reducing the release of inflammatory cytokines. However, the overexpression of LDHA counteracts the effects of galloflavin in macrophages. In addition, in vivo experiments observed a protective effect of galloflavin against cecal ligation and puncture (CLP) and cisplatin-induced renal injury. The ability of galloflavin to inhibit glycolysis in renal macrophages, thereby regulating their phenotypic transition during AKI was further validated through the isolation of renal primary macrophages. This intervention ultimately ameliorated the inflammatory response and decelerated the progression of AKI. Collectively, galloflavin confers protection against AKI by suppressing glycolysis in macrophages through a LDHA-dependent mechanism, thereby positioning it as a potential therapeutic option for AKI in the future.
Collapse
Affiliation(s)
- Jie Wei
- Department of Nephrology, the Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Xin-Yu Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zhi-Juan Wang
- Department of Nephrology, the Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Xiang-Yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Meng-Meng Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Tao Sun
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xin-Ru Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - De-Guang Wang
- Department of Nephrology, the Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China.
| | - Chao Hou
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
6
|
Mohseni S, Tavakoli A, Ghazipoor H, Pouralimohamadi N, Zare R, Rampp T, Shayesteh M, Pasalar M. Curcumin for the clinical treatment of inflammatory bowel diseases: a systematic review and meta-analysis of placebo-controlled randomized clinical trials. Front Nutr 2025; 12:1494351. [PMID: 40196017 PMCID: PMC11973083 DOI: 10.3389/fnut.2025.1494351] [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: 09/10/2024] [Accepted: 03/11/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Inflammatory Bowel Disease (IBD), encompassing Crohn disease (CD) and ulcerative colitis (UC), presents complex challenges in management due to dysregulated immune responses and genetic predispositions. This study explored the potential of curcumin as an adjunctive therapy in IBD, assessing its efficacy and safety through a systematic review of clinical trials to enhance treatment strategies and outcomes. Methods To identify placebo-controlled randomized clinical trials on curcumin treatment in IBD, databases such as Medline/PubMed, Scopus, Embase, Web of Knowledge, and Google Scholar were searched till May 2024. Inclusion criteria focused on RCTs comparing curcumin with placebo in IBD patients, with data extraction and analysis conducted using established methodologies and tools for comprehensive synthesis and assessment of study findings. Results In this meta-analysis, 13 placebo-controlled RCTs on curcumin treatment in IBD were included after screening 362 records and conducting a full-text review. Most trials focused on UC patients and were published post-2010, utilizing oral curcumin with varying dosages and durations. The analysis showed curcumin's significant efficacy in achieving clinical remission and response in UC patients, with heterogeneity observed. Adverse events and withdrawal rates did not significantly differ between curcumin and placebo groups. In CD patients, curcumin did not show superiority over placebo for clinical and endoscopic remission. Conclusion The findings highlight curcumin's potential as a treatment for UC but indicate inconclusive results for CD, emphasizing the need for further research. The multifaceted mechanisms of curcumin's efficacy in IBD involve anti-inflammatory, antioxidant, microbiota modulatory, and immune-regulating properties. Further research is warranted to enhance understanding and treatment efficacy. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/view/CRD42024567247.
Collapse
Affiliation(s)
- Saeid Mohseni
- Department of Persian Medicine, School of Persian Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Ali Tavakoli
- Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Ghazipoor
- Department of Family Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Pouralimohamadi
- Department of Family Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roghayeh Zare
- Department of Persian Medicine, School of Persian Medicine, Shahid Sadoughi University of Medical Sciences, Ardakan, Yazd, Iran
| | - Thomas Rampp
- Center for Integrative Medicine and Planetary Health, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Maryam Shayesteh
- Department of Traditional Pharmacy and Persian Medicine, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehdi Pasalar
- Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Almeida JE, de Oliveira AC, de Castro Alves CE, Filho SMC, de Oliveira ECP, Zuliani JP, Pontes GS. Diterpenes: Nature's Hidden Gems of Immunomodulation. Int J Mol Sci 2025; 26:2250. [PMID: 40076871 PMCID: PMC11900544 DOI: 10.3390/ijms26052250] [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: 01/14/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Natural products, especially specific metabolites found in many medicinal plants, exhibit extensive therapeutic potential due to their diverse biological characteristics. Among these compounds, diterpenes stand out for their active principles described in phytochemical studies. Diterpenes exhibit immunomodulatory effects by influencing the production of cytokines and other signaling molecules involved in the immune response. These actions contribute to achieving a more balanced immune profile. The ability to selectively and harmoniously modulate the immune response positions compounds derived from natural products is a promising research field in the development of immunomodulatory therapies. Due to the broad biological activities of diterpenes, the use of molecular docking emerges as a relevant tool for the quantitative screening of a large number of these substances. This review comprehensively examines the pharmacological potential of diterpenes in modulating the immune system. It highlights the existing experimental evidence supporting the efficacy and safety of these compounds as potential treatment for immune dysfunctions. Ultimately, this review aims to contribute to the development of new therapeutic strategies in this field.
Collapse
Affiliation(s)
- Josiane Elizabeth Almeida
- Graduate Program in Basic and Applied Immunology, Federal University of Amazonas (UFAM), Manaus 69080-900, AM, Brazil
| | - André Correa de Oliveira
- Analytical Multidisciplinary Support Center, Federal University of Amazonas (UFAM), Manaus 69080-900, AM, Brazil;
| | - Carlos Eduardo de Castro Alves
- Laboratory of Virology and Immunology, Society, Environment and Health Coordination, National Institute of Amazonian Research (INPA), Manaus 69080-001, AM, Brazil;
| | - Selino Monteiro Costa Filho
- Biotechnology and Medicinal Plants Laboratory, Federal University of Western Pará (UFOPA), Santarém 68040-255, PA, Brazil; (S.M.C.F.); (E.C.P.d.O.)
| | - Elaine Cristina Pacheco de Oliveira
- Biotechnology and Medicinal Plants Laboratory, Federal University of Western Pará (UFOPA), Santarém 68040-255, PA, Brazil; (S.M.C.F.); (E.C.P.d.O.)
| | - Juliana Pavan Zuliani
- Laboratory of Cellular Immunology Applied to Health, Oswaldo Cruz Foundation (FIOCRUZ), Porto Velho 21040-900, RO, Brazil;
| | - Gemilson Soares Pontes
- Laboratory of Virology and Immunology, Society, Environment and Health Coordination, National Institute of Amazonian Research (INPA), Manaus 69080-001, AM, Brazil;
- Graduate Program in Hematological Sciences, University of Amazonas State (UEA), Manaus 69080-010, AM, Brazil
| |
Collapse
|
8
|
Chu TW, Ho CC, Chiu HP, Hsu YJ, Hung CT, Sung CH, Chang DC, Chang HH, Hung CF. d-Limonene inhibits cytokines and chemokines expression by regulating NF-kappaB and STAT in HaCat cells and DNCB-induced atopic dermatitis in BALB/c mice. Int Immunopharmacol 2025; 148:114082. [PMID: 39818092 DOI: 10.1016/j.intimp.2025.114082] [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: 12/17/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin disorder characterized by itching and redness, affecting individuals of all ages and significantly impairing their quality of life. The prevalence of AD is rising, posing serious health concern. Relief of itching is a primary treatment objective; however, steroid treatments can lead to adverse effects, including skin barrier thinning. Therefore, there is a pressing need for safer therapeutic alternatives. Limonene, a naturally occurring monocyclic monoterpene found in citrus peel oil, is widely utilized in food, cosmetics, and pharmaceuticals. Research has identified various biological activities of limonene, including antioxidative, anti-inflammatory, immunomodulatory, and antifibrotic properties. This study aims to investigate the therapeutic effects of limonene on atopic dermatitis, focusing on its anti-inflammatory potential. METHODS In this study, we investigated the expression levels of pro-inflammatory cytokines and chemokines, conducting histopathological analyses, and collecting physiological data from BALB/c mice with atopic-like dermatitis induced by 2,4-dinitrochlorobenzene (DNCB) and TNF-α/ IFN-γ-stimulated HaCaT cells. RESULTS In vitro studies indicated that limonene inhibited cytokine and chemokine expression in human keratinocytes and reduced phosphorylation in the MAPK, NF-κB, and JAK/STAT signaling pathways. In vivo, limonene mitigated DNCB-induced skin barrier damage and itching, improving physiological parameters such as trans-epidermal water loss, erythema, and ear thickness. Furthermore, it decreased the mRNA expression of pro-inflammatory cytokines. CONCLUSION Limonene exhibits significant anti-inflammatory effects, highlighting its therapeutic potential for treating atopic dermatitis.
Collapse
Affiliation(s)
- Thomas W Chu
- Department of Dermatology, Far Eastern Memorial Hospital, New Taipei City 22060 Taiwan; Pariser Dermatology Specialists, Newport News, Virginia 23606, USA.
| | - Ching-Chih Ho
- Department of Anesthesiology, Taoyuan Armed Forces General Hospital, Longtan, Taoyuan 325, Taiwan.
| | - Hsin-Pei Chiu
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan.
| | - Yu-Jou Hsu
- PhD Program in Pharmaceutical Biotechnology, Fu Jen Catholic University, New Taipei City 24205, Taiwan.
| | - Chen-Ting Hung
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Chao-Hsien Sung
- Division of Anesthesiology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan.
| | - Der-Chen Chang
- Department of Mathematics and Statistics and Department of Computer Science, Georgetown University, Washington, DC 20057, USA.
| | - Hsun-Hao Chang
- Department of Cardiology, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation), Tainan, Taiwan.
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan; PhD Program in Pharmaceutical Biotechnology, Fu Jen Catholic University, New Taipei City 24205, Taiwan; School of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
9
|
Xie W, Li H, Yu T, Zhu Y, Gao J, Yang X, Cheng H, Qiao H, Zhang X, Gao X, Zhang L, Fang X, Zhang L, Bi Y. Design and Synthesis of Hederagenin Derivatives for the Treatment of Sepsis by Targeting TAK1 and Regulating the TAK1-NF-κB/MAPK Signaling. J Med Chem 2025; 68:2694-2719. [PMID: 39817810 DOI: 10.1021/acs.jmedchem.4c02032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Sepsis is a systemic inflammatory response caused by infection and is a leading cause of death worldwide. We designed and synthesized a series of hederagenin analogues with anti-inflammatory activity. The most effective compound, 14, reduced the release of TNF-α and IL-6 in RAW264.7 cells induced by lipopolysaccharide by affecting NF-κB/MAPK signaling. It demonstrated significant protection against sepsis in vivo and ameliorated histopathological changes in the liver, lungs, and kidneys. It exhibited good safety in subacute toxicity assays. Western blotting results indicated that it reduced the generation of p-p65, p-IκB, p-p38, p-JNK, and p-ERK. Immunofluorescence assay results suggested that the compound inhibited nuclear translocation of p65 and c-Fos. It was found to target TAK1 with a novel molecular backbone, distinct from the few TAK1 inhibitors previously reported. This work provides a new lead structure for the study of TAK1 inhibitors and a potential target for TAK1 in sepsis therapy.
Collapse
Affiliation(s)
- Wenbin Xie
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Haixia Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Tao Yu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Yatong Zhu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Jing Gao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Xiaoli Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Haoran Cheng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Hui Qiao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Xin Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Xiaojin Gao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Lei Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Xianhe Fang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Leiming Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Yi Bi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| |
Collapse
|
10
|
Jiang L, Ma X, Yan Q, Pu D, Fu X, Zhang D. Dihydromyricetin/montmorillonite intercalation compounds ameliorates DSS-induced colitis: Role of intestinal epithelial barrier, NLRP3 inflammasome pathway and gut microbiota. Int J Pharm 2025; 670:125155. [PMID: 39746581 DOI: 10.1016/j.ijpharm.2024.125155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Dihydromyricetin (DHM), the primary active compound in vine tea possesses various pharmacological effects such as anti-inflammatory and antioxidant properties, along with high biosafety. However, its oral delivery remains a significant challenge. Montmorillonite (MMT), the primary component of bentonite, is a commonly used drug in the clinical treatment of gastrointestinal diseases and serves as an excellent drug carrier due to its intercalation capability. In this study, we intercalated DHM into the interlayer spaces of MMT via solution intercalation method combined with rotary evaporation and used it to treat ulcerative colitis in mice. SEM, XRD, and FTIR analyses confirmed the successful synthesis of the DHM/MMT intercalation compound. In vitro studies shown that DHM/MMT eliminated intracellular ROS and suppressed inflammatory genes IL-1β, IL-6, and TNF-α. Moreover, DHM/MMT demonstrated notable therapeutic effects in ulcerative colitis (UC) mice, significantly restoring the intestinal mucosa. Importantly, the therapeutic mechanism of DHM/MMT is closely linked to the inhibition of the NLRP3 signaling pathway. Additionally, this strategy modulated gut microbiota by increasing probiotics and suppressing harmful bacteria, thereby maintaining intestinal homeostasis. In conclusion, DHM/MMT presents a promising strategy for UC treatment.
Collapse
Affiliation(s)
- Luxia Jiang
- Department of Cardiac Surgery ICU, Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China; Key Laboratory of Digestive Diseases, Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China; Cuiying Biomedical Research Center, Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xueni Ma
- Department of Gastroenterology, Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China; Key Laboratory of Digestive Diseases, Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Qi Yan
- Department of Neurology Department, Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Dan Pu
- Department of Gastroenterology, Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China; Key Laboratory of Digestive Diseases, Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xu Fu
- Key Laboratory of Emergency Medicine, Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| | - Dekui Zhang
- Department of Gastroenterology, Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China; Key Laboratory of Digestive Diseases, Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| |
Collapse
|
11
|
Jiang Y, Cao J, Li R, Yu J, Peng Y, Huang Q, Zuo W, Chen J. Tetrahydropalmatine ameliorates peripheral nerve regeneration by enhancing macrophage anti-inflammatory response. Int Immunopharmacol 2025; 147:114000. [PMID: 39765002 DOI: 10.1016/j.intimp.2024.114000] [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: 08/18/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/29/2025]
Abstract
BACKGROUND Peripheral nerve injury (PNI) is a common clinical problem that can result in partial or complete loss of sensory, motor, and autonomic functions. Tetrahydropalmatine (THP), a Corydalis yanhusuo-derived phytochemical alkaloid, possesses hypnotic, soothing, analgesic, and other effects, but little is known about the effect of THP on moderating peripheral nerve regeneration and its possible underlying mechanism of action. PURPOSE In this study, we aim to elucidate the protective function of THP on PNI and further reveal the underlying pharmacological mechanisms. METHODS PNI rats were in suit injection of THP solution at doses of 40 mg/kg for consecutive 3, 7, or 28 days, followed by harvesting the sciatic nerve tissues. The protective effect of THP on PNI was evaluated by electrophysiological test, transmission electron microscopy, immunofluorescence (IF), and western blotting (WB). Macrophage polarization, the expression of inflammatory-related genes and cytokines, and its upstream signaling pathways were detected by IF, WB, enzyme-linked immunosorbent assay (ELISA), mRNA-seq, and WB. In vitro, the Raw 264.7 cells were treated with lipopolysaccharide containing with/without THP. The degree of inflammatory activation and its potential pharmacological mechanism were measured by ELISA, qRT-PCR, IF staining, flow cytometry, and WB. Additionally, a pharmacological agonist or inhibitor was added to the cell medium to further identify the role of THP's potential pharmacological mechanism in regulating inflammatory response via IF and ELISA technology. RESULTS Using the sciatic nerve crush model, we found that THP significantly enhanced the rate of axonal growth and functional recovery, and altered macrophage subtype transformation from the M1/M0 phenotype into the M2 phenotype, inducing the secretion of large amounts of anti-inflammatory factors. Moreover, THP significantly increased the phosphorylation level of PI3K, AKT, GSK3β, and IκBa, and decreased the expression of TLR4 protein and NF-κB phosphorylation. Similarly, in vitro, THP also facilitated Raw 264.7 cell polarization to the M2 subtype under the condition of LPS stimulation. Meanwhile, the change of PI3K/AKT/GSK3β and TLR4/NF-κB signaling-related proteins in vitro was consistent with the results in vivo. Additionally, the THP-medicated anti-inflammatory effect on Raw 264.7 cells was partly eliminated when pharmacological intervention of these two signaling pathways. CONCLUSIONS THP has anti-inflammatory effects on facilitating M2-subtype macrophage polarization, which produces abundant anti-inflammatory cytokines to ameliorate peripheral nerve regeneration. Moreover, the potential mechanism of THP action may be intimately associated with activating the PI3K/AKT/GSK3β axis and inhibiting the TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Yongsheng Jiang
- Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, P.R. China
| | - Jianye Cao
- Wenzhou Medical University, Wenzhou 325035, China
| | - Rui Li
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional KeyTechnology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China.
| | - Jia Yu
- Hangzhou Institute for Food and Drug Control, China
| | - Yan Peng
- Hangzhou Institute for Food and Drug Control, China
| | - Qiong Huang
- Xiangshan Maternal and Child Health Care Family Planning Service Center, China
| | - Wei Zuo
- Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, P.R. China.
| | - Junyue Chen
- Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, P.R. China.
| |
Collapse
|
12
|
Muhtar E, Ylham G, Tiemuer A, Edirs S. Unraveling the Dual Anti-Inflammatory and Antioxidant Mechanisms of Acteoside: Computational Insights and Experimental Validation. Chem Biodivers 2025; 22:e202401564. [PMID: 39365024 DOI: 10.1002/cbdv.202401564] [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: 06/27/2024] [Revised: 09/29/2024] [Accepted: 10/03/2024] [Indexed: 10/05/2024]
Abstract
Acteoside (ACT) is one of the primary bioactive ingredients in Cistanche tubulosa (Schenk). Its remarkable efficacy in treating immune-related and inflammatory disorders has garnered significant interest among scientific circles. However, the anti-inflammatory and antioxidant effects of ACT and its underlying molecular mechanisms require further investigation. In this study, pharmacophore-based reverse docking and molecular dynamics simulations identified potential anti-inflammatory targets in silico. Studies conducted in vitro with lipopolysaccharide (LPS)-induced RAW264.7 cells validated the anti-inflammatory properties of ACT. Methyl thiazolyl tetrazolium (MTT) and lactate dehydrogenase (LDH) assays indicated ACT's non-toxic and growth-promoting effects on cells. ACT significantly reduced nitric oxide (NO) and reactive oxygen species (ROS) production and restored levels of antioxidant enzymes. It also decreased pro-inflammatory cytokines. Western blotting assays indicated that ACT inhibited p38, TNF-α, PI3 K/AKT, and NF-κB signaling pathways. These findings underscore ACT's ability to mitigate acute inflammation in RAW264.7 cells by modulating key signaling pathways and provide the scientific basis for enhancing the medicinal value of ACT and future drug development.
Collapse
Affiliation(s)
- Eldar Muhtar
- Institute of Agro-products Storage and Processing, Xinjiang Key Laboratory of Processing and Preservation of Agricultural Products, Xinjiang Academy of Agricultural Science, Urumqi, Xinjiang, 830091, China
| | - Gulfira Ylham
- Institute of Agro-products Storage and Processing, Xinjiang Key Laboratory of Processing and Preservation of Agricultural Products, Xinjiang Academy of Agricultural Science, Urumqi, Xinjiang, 830091, China
| | - Atawulla Tiemuer
- Institute of Agro-products Storage and Processing, Xinjiang Key Laboratory of Processing and Preservation of Agricultural Products, Xinjiang Academy of Agricultural Science, Urumqi, Xinjiang, 830091, China
| | - Salamet Edirs
- Institute of Agro-products Storage and Processing, Xinjiang Key Laboratory of Processing and Preservation of Agricultural Products, Xinjiang Academy of Agricultural Science, Urumqi, Xinjiang, 830091, China
| |
Collapse
|
13
|
Edo GI, Mafe AN, Ali ABM, Akpoghelie PO, Yousif E, Apameio JI, Isoje EF, Igbuku UA, Garba Y, Essaghah AEA, Ahmed DS, Umar H, Ozsahin DU. Chitosan and its derivatives: A novel approach to gut microbiota modulation and immune system enhancement. Int J Biol Macromol 2025; 289:138633. [PMID: 39675606 DOI: 10.1016/j.ijbiomac.2024.138633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
Chitosan, a biopolymer derived from the deacetylation of chitin found in crustacean shells and certain fungi, has attracted considerable attention for its promising health benefits, particularly in gut microbiota maintenance and immune system modulation. This review critically examines chitosan's multifaceted role in supporting gut health and enhancing immunity, beginning with a comprehensive overview of its sources, chemical structure, and its dual function as a dietary supplement and biomaterial. Chitosan's prebiotic effects are highlighted, with a focus on its ability to selectively stimulate beneficial gut bacteria, such as Bifidobacteria and Lactobacillus, while enhancing gut barrier integrity and inhibiting the growth of pathogenic microorganisms. The review delves deeply into chitosan's immunomodulatory mechanisms, including its impact on antigen-presenting cells, cytokine profiles, and systemic immune responses. A detailed comparative analysis assesses chitosan's efficacy relative to other prebiotics and immunomodulatory agents, examining challenges related to bioavailability and metabolic activity. Beyond its role in gut health, this review explores chitosan's potential as a dual-action agent that not only supports gut microbiota but also fortifies immune resilience. It introduces emerging research on novel chitosan derivatives, such as chitooligosaccharides, and evaluates their enhanced bioactivity for functional food applications. Special attention is given to sustainability, with an exploration of alternative, plant-based sources of chitosan and their implications for both health and environmental stewardship. Also, the review identifies new research avenues, such as the growing interest in chitosan's role in the gut-brain axis and its potential mental health benefits through microbial interactions. By addressing these innovative areas, the review aims to shift the focus from basic health effects to chitosan's broader impact on public health. The findings encourage further exploration, particularly through human trials, and emphasize chitosan's untapped potential in revolutionizing health and disease management.
Collapse
Affiliation(s)
- Great Iruoghene Edo
- Department of Chemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria; Department of Chemistry, College of Sciences, Al-Nahrain University, Baghdad, Iraq.
| | - Alice Njolke Mafe
- Department of Biological Sciences, Faculty of Science, Taraba State University Jalingo, Taraba State, Nigeria
| | - Ali B M Ali
- Department of Air Conditioning Engineering, Faculty of Engineering, Warith Al-Anbiyaa University, Karbala, Iraq
| | - Patrick Othuke Akpoghelie
- Department of Food Science and Technology, Faculty of Science, Delta State University of Science and Technology, Ozoro, Delta State, Nigeria
| | - Emad Yousif
- Department of Chemistry, College of Sciences, Al-Nahrain University, Baghdad, Iraq
| | - Jesse Innocent Apameio
- Department of Biological Sciences, Faculty of Science, Taraba State University Jalingo, Taraba State, Nigeria
| | - Endurance Fegor Isoje
- Department of Science Laboratory Technology (Biochemistry Option), Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Ufuoma Augustina Igbuku
- Department of Chemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Yasal Garba
- Department of Information Engineering, College of Information Engineering, Al-Nahrain University, Baghdad, Iraq
| | - Arthur Efeoghene Athan Essaghah
- Department of Urban and Regional Planning, Faculty of Environmental Sciences, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Dina S Ahmed
- Department of Chemical Industries, Institute of Technology-Baghdad, Middle Technical University, Baghdad, Iraq
| | - Huzaifa Umar
- Operational Research Centre in Healthcare, Near East University, Nicosia, Cyprus
| | - Dilber Uzun Ozsahin
- Operational Research Centre in Healthcare, Near East University, Nicosia, Cyprus; Department of Medical Diagnostic Imaging, College of Health Sciences, University of Sharjah, P.O. Box 27272, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, United Arab Emirates
| |
Collapse
|
14
|
Maiolini TCS, Nicácio KDJ, Rosa W, Miranda DDO, Santos MFC, Bueno PCP, Lago JHG, Sartorelli P, Dias DF, Chagas de Paula DA, Soares MG. Potential anti-inflammatory biomarkers from Myrtaceae essential oils revealed by untargeted metabolomics. Nat Prod Res 2025; 39:985-992. [PMID: 38006221 DOI: 10.1080/14786419.2023.2283758] [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: 05/03/2023] [Revised: 10/21/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023]
Abstract
Many species from Myrtaceae have traditionally been used in traditional medicine as anti-inflammatory, antimicrobial, antidiarrheal, antioxidant and antirheumatic, besides in blood cholesterol reduction. In the present work, the anti-inflammatory activity of essential oils from eighteen Myrtaceae spp. were evaluated according to their ex-vivo anti-inflammatory activity in human blood, and the corresponding biomarkers were determined using untargeted metabolomics data and multivariate data analysis. From these studied species, six displayed anti-inflammatory activity with percentage rates of inhibition of PGE2 release above 70%. Caryophyllene oxide (1), humulene epoxide II (2), β-selinene (3), α-amorphene (4), α-selinene (5), germacrene A (6), β-bisabolene (7), α-muurolene (8), α-humulene (9), β-gurjunene (10), myrcene (11), β-elemene (12), α-cadinol (13), α-copaene (14), E-nerolidol (15) and ledol (16) were annotated as potential anti-inflammatory biomarkers. The results obtained in this study point to essential oils from species of the Myrtaceae family as a rich source of anti-inflammatory agents.
Collapse
Affiliation(s)
| | | | - Welton Rosa
- Chemistry Institute, Federal University of Alfenas, Alfenas, Brazil
| | | | - Mario Ferreira Conceição Santos
- Department of Chemistry and Physics, Center for Exact, Natural and Health Sciences, Federal University of Espírito Santo, Alegre, Brazil
| | - Paula Carolina Pires Bueno
- Leibniz Institute of Vegetable and Ornamental Crops, Department of Plant Biotic Interactions, Grossbeeren, Germany
| | | | - Patricia Sartorelli
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil
| | | | | | | |
Collapse
|
15
|
Wu Z, Song B, Peng F, Zhang Q, Wu S. Zangsiwei prevents particulate matter-induced lung inflammation and fibrosis by inhibiting the TGF-β/SMAD pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118752. [PMID: 39232997 DOI: 10.1016/j.jep.2024.118752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/11/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zangsiwei(ZSW) is a traditional Tibetan medicine from China consisting of extracts of Rhododendron anthopogonoides Maxim, Gentiana Tourn, Corydalis hendersonii Hemsl and Berberis kansuensis C.K.Schneid. Traditionally, ZSW has been used by Tibetan physicians to treat chronic respiratory diseases. The role of ZSW in particulate matter-induced lung inflammation and fibrosis remains unclear. AIM OF THE STUDY Combining non-targeted metabolomics, network pharmacology, and molecular docking to explore the mechanism of ZSW in the treatment of particulate matter-induced lung inflammation and fibrosis, and validated by in vivo and in vitro experiments. MATERIALS AND METHODS The serum metabolite profile post-ZSW administration was first identified utilizing non-targeted metabolomics. Network pharmacology and molecular docking were employed to predict potential bioactive components and their corresponding targets. The in silico predictions were subsequently validated through in vivo studies in mice exposed to PM2.5 and silica dust, as well as in vitro studies utilizing human lung epithelial cells (A549) and lung fibroblasts (MRC5). RESULTS Metabolomic analysis identified specific serum metabolites that were associated with ZSW treatment. Network pharmacology and molecular docking identified key targets involved in the Transforming growth factor-β (TGF-β)/SMAD pathway, which were subsequently validated through in vivo experiments demonstrating a reduction in lung inflammation and fibrosis in ZSW-treated mice. In vitro studies demonstrated that ZSW exerts protective effects against PM2.5-induced cytotoxicity and modulates fibrotic markers in a dose-dependent manner. This is consistent with the inhibition of the TGF-β/SMAD pathway. CONCLUSION Our integrated approach, which combines non-targeted metabolomics, network pharmacology, and molecular docking, followed by rigorous in vivo and in vitro validation, establishes ZSW as a potential therapeutic agent for particulate matter-induced lung inflammation and fibrosis.
Collapse
Affiliation(s)
- Zhijian Wu
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Centre for Evidence-based Medicine, Changsha, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, 410011, China
| | - Boyang Song
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Centre for Evidence-based Medicine, Changsha, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, 410011, China
| | - Fei Peng
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Centre for Evidence-based Medicine, Changsha, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, 410011, China
| | - Quan Zhang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Centre for Evidence-based Medicine, Changsha, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, 410011, China
| | - Shangjie Wu
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Centre for Evidence-based Medicine, Changsha, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, 410011, China.
| |
Collapse
|
16
|
Borgonetti V, Governa P, Morozzi M, Sasia C, Videtta G, Biagi M, Galeotti N. A Standardized Extract of Zingiber officinale Roscoe Regulates Clinical and Biological Outcomes in Two Different EAE Mouse Models. Biomedicines 2025; 13:278. [PMID: 40002693 PMCID: PMC11852164 DOI: 10.3390/biomedicines13020278] [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: 12/30/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system characterized by demyelination and neuronal damage. Current MS therapies are unsatisfactory, and new therapies are encouraged. A correlation between nutritional intake and MS has been speculated. Supplementation of approved immunomodulatory therapy with herbal medicines possessing antioxidant and anti-inflammatory activities could provide benefits to MS patients. Ginger is one of the most widely consumed dietary supplements in the world, commonly used in traditional medicine. Studies demonstrated that ginger may also be beneficial in the management of neurodegenerative diseases. The aim of this study is to investigate the MS therapeutic potential of ginger. Methods: A standardized Zingiber officinale Roscoe extract (ZOE) was orally administered for 14 days. Two experimental autoimmune encephalomyelitis (EAE) models in mice were used. The PLP139-151-EAE relapsing-remitting model and MOG35-55-EAE chronic model. Clinical score, von Frey, hot plate, and rotarod tests were used for behavioral tests. ELISA and Western blotting were used to measure cytokines levels. Evans Blue content was determined spectrophotometrically. Results: ZOE attenuated motor disability and pain hypersensitivity in both models had no effect on body weight loss. ZOE reduced the blood-brain barrier (BBB) permeability in the PLP-EAE models and reduced levels of circulating cytokines (Il-6, IL-17) in the MOG-EAE model. ZOE attenuated spinal cytokines overexpression in both models. Conclusions: ZOE improves EAE symptoms and attenuates the proinflammatory response in both models, representing a promising nutraceutical support to the conventional therapeutic approach to MS.
Collapse
Affiliation(s)
- Vittoria Borgonetti
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy; (V.B.); (M.M.); (C.S.); (G.V.)
| | - Paolo Governa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, 53100 Siena, Italy;
| | - Martina Morozzi
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy; (V.B.); (M.M.); (C.S.); (G.V.)
| | - Chiara Sasia
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy; (V.B.); (M.M.); (C.S.); (G.V.)
| | - Giacomina Videtta
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy; (V.B.); (M.M.); (C.S.); (G.V.)
| | - Marco Biagi
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy;
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy; (V.B.); (M.M.); (C.S.); (G.V.)
| |
Collapse
|
17
|
Samy BA, Raman K, Velayutham S, Senthilkumar N, Thirumalaivasan N, Kanagaraj K, Pothu R, Boddula R, Radwan AB, Al-Qahtani N. Natural product extract fractions as potential arthritis treatments: A detailed analysis using in-silico, in-vivo, and in-vitro methods. Int Immunopharmacol 2025; 144:113595. [PMID: 39580856 DOI: 10.1016/j.intimp.2024.113595] [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: 05/27/2024] [Revised: 10/13/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024]
Abstract
Two characteristics of the systemic autoimmune disease rheumatoid arthritis (RA) include extra-articular involvement and inflammatory arthritis. It is a long-term inflammatory condition that mostly affects the synovial joints and is often triggered by a confluence of environmental factors, including tobacco use, and genetics. The review investigates natural products' role in arthritis through three key approaches. In-silico analysis identifies molecular mechanisms and targets of these products, revealing their potential for therapeutic use. In-vivo studies evaluate how well these products work and their safety in reducing joint inflammation. In-vitro studies focus on how these compounds interact at the cellular level and their effects on signaling pathways. Together, these approaches offer a comprehensive understanding of how natural products could benefit arthritis management. This review focuses on translational studies and highlights the possible role of natural compounds as adjunctive therapies to conventional arthritis treatments. In conclusion, this study indicates that natural products have potential advantages in treating osteoarthritis and rheumatoid arthritis based on in-silico analysis which shows anti-inflammatory effects, in-vivo studies that reduce joint inflammation, and in-vitro studies that amplify arthritis management. To improve the therapeutic advantages of natural products utilized for treating arthritis, an all-inclusive examination has been done to give direction for the following research efforts.
Collapse
Affiliation(s)
- Bharathiraja Anthony Samy
- Department of Pharmacology, JKKMRFs Annai JKK Sampoorani Ammal College of Pharmacy (Affiliated by The Tamil Nadu Dr. M.G.R. Medical University, Chennai, Tamil Nadu, India), B. Komarapalayam, Namakkal, Tamil Nadu, India
| | - Kannan Raman
- Department of Pharmacology, JKKMRFs Annai JKK Sampoorani Ammal College of Pharmacy (Affiliated by The Tamil Nadu Dr. M.G.R. Medical University, Chennai, Tamil Nadu, India), B. Komarapalayam, Namakkal, Tamil Nadu, India
| | - Suresh Velayutham
- Department of Pharmacology, JKKMRFs Annai JKK Sampoorani Ammal College of Pharmacy (Affiliated by The Tamil Nadu Dr. M.G.R. Medical University, Chennai, Tamil Nadu, India), B. Komarapalayam, Namakkal, Tamil Nadu, India
| | - Nangan Senthilkumar
- Department of Chemistry, Allied Sciences, Graphic Era Hill University, Dehradun 248002, India
| | - Natesan Thirumalaivasan
- Department of Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Kuppusamy Kanagaraj
- Center for Supramolecular Chemistry & Catalysis and Department of Chemistry, College of Science, Shanghai University, Shanghai 200444, China.
| | - Ramyakrishna Pothu
- School of Physics and Electronics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Rajender Boddula
- Department of Chemistry, Allied Sciences, Graphic Era Hill University, Dehradun 248002, India; Allied Sciences, Department of Chemistry, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India; Center for Advanced Materials (CAM), Qatar University, 2713 Doha, Qatar.
| | | | - Noora Al-Qahtani
- Center for Advanced Materials (CAM), Qatar University, 2713 Doha, Qatar; Central Laboratories Unit (CLU), Qatar University, Doha 2713, Qatar.
| |
Collapse
|
18
|
Zhang W, Sun M, Liu N, Li X, Sun J, Wang M. Curcumin ameliorates astrocyte inflammation through AXL in cuprizone-induced mice. Toxicol Appl Pharmacol 2025; 494:117170. [PMID: 39586379 DOI: 10.1016/j.taap.2024.117170] [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: 08/20/2024] [Revised: 10/30/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Curcumin has gained global attention owning to its anti-inflammatory, antioxidant, anticancer, and antimicrobial activities. Curcumin has recently been shown to have well-documented effects on neuroinflammation in multiple sclerosis (MS). Astrocytes, the most widely distributed glial cells in the brain, have a significant influence on the regulation of neuroinflammation in MS. However, it is unknown how curcumin exerts neuroprotective effects in astrocytes. To elucidate the mechanism underlying the effects of curcumin on astrocytes, we explored the effect of curcumin on cuprizone (CPZ)-induced mice in vivo and on primary astrocytes in vitro. In this study, we observed that curcumin significantly ameliorated myelin loss and reduced astrocyte activation in the corpus callosum (CC) region in mice induced with CPZ, and in primary astrocytes stimulated with lipopolysaccharide (LPS). Meanwhile, our research indicated that curcumin may exert neuroprotective effects in CPZ-induced mice by downregulating astrocyte-mediated inflammation by AXL. This study provides new insights into possible targeted therapies for MS.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Mengjiao Sun
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Ning Liu
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Xiaoling Li
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Jing Sun
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China.
| |
Collapse
|
19
|
Ranaweera BVLR, Edward D, Harasgama JC, Abeysekera AM, Weerasena OVDSJ, Handunnetti SM. Anti-inflammatory activity and selective inhibition of iNOS gene expression by a polyherbal formulation. J Ayurveda Integr Med 2025; 16:101049. [PMID: 39827650 PMCID: PMC11787581 DOI: 10.1016/j.jaim.2024.101049] [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: 08/03/2023] [Revised: 07/16/2024] [Accepted: 08/08/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Link Samahan® (LS), a product of Link Natural Products (Pvt) Limited, Sri Lanka contains extracts of 14 medicinal plants. It is used as a prophylactic against cold and cold related symptoms. It has immunomodulatory activity, specifically enhancing the humoral immune response. OBJECTIVE To investigate the anti-inflammatory activity (AIA) of LS and related mechanisms. MATERIALS AND METHODS In vivo AIA was assessed by the inhibition of carrageenan-induced rat paw-edema. Mechanisms of AIA were assessed in vitro by the inhibition of reactive oxygen species (ROS) and reactive nitrogen species (RNS) production and expression of inducible nitric oxide synthase (iNOS) gene using rat peritoneal cells. RESULTS LS showed an increasing inhibition in rat paw-edema up to 5 h compared to the biphasic pattern exhibited by the reference drug, Indomethacin. Inhibition by LS at the 1st and 3rd-5th hours (44.7% and 73.0-74.6%) was comparable to indomethacin (47.4% and 83.6-76.5%; p > 0.05) whereas at the 2nd hour, LS exhibited a significantly higher inhibition compared to indomethacin (66.7% vs 28.6%; p < 0.05). LS treatment significantly inhibited the ROS (superoxide; 47.2 ± 0.86%; p < 0.05) and RNS (nitrite; 54.0 ± 0.40%; p < 0.05) production in rat peritoneal cells. Further, specific inhibition of iNOS gene expression in rat peritoneal cells resulted in a significant reduction in RNS production whereas LS had lesser or no inhibitory effect on endothelial NOS and neuronal NOS gene expression. CONCLUSION LS has potent anti-inflammatory activity and selective inhibition of iNOS activity. AIA of LS shown here supports its use as a prophylactic against cold and cold related symptoms.
Collapse
Affiliation(s)
| | - Daniya Edward
- Institute of Biochemistry, Molecular Biology and Biotechnology (IBMBB), University of Colombo, Sri Lanka
| | | | | | | | | |
Collapse
|
20
|
Abhale K, Addepalli V, Desai S, Sanap A, Bhonde R. Effects of Mesenchymal Stem Cell-conditioned Media with Natural Immunomodulatory Agent Resveratrol on Type 1 Diabetes. Curr Drug Discov Technol 2025; 22:e080324227818. [PMID: 38468534 DOI: 10.2174/0115701638276524240305054259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/18/2024] [Accepted: 01/29/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is a condition marked by elevated blood sugar levels and primarily recognized by the destruction of beta cells caused by an autoimmune attack, which is a significant characteristic of T1DM. Recent studies have demonstrated the regenerative potential of conditioned medium therapy. In light of this, the current research sought to assess the impact of Mesenchymal Stem Cell conditioned media (CM) and CM with resveratrol (CM+ Resveratrol) on the management of T1DM in Swiss albino mice. By leveraging and modifying existing conditioned medium therapy, this study aims to evaluate its effectiveness in treating T1DM. MATERIALS & METHODS Diabetes was induced in animals using the diabetes-inducing agent streptozotocin (STZ). The animals were then divided into five groups: Normal control, Disease Control, Resveratrol, Condition Media, and CM + Resveratrol. Treatments were given to the animals accordingly. The study period was 28 days. During this time, the animals were monitored for foodwater intake twice a week, blood glucose levels, and body weight. At the conclusion of the 28-day study period, biochemical estimations were performed for serum insulin levels, C-peptide levels, anti-inflammatory cytokines levels and pro-inflammatory cytokines levels. Additionally, histopathology of the pancreas was performed. RESULTS The test groups showed a significant decrease in blood glucose levels, an increase in Cpeptide levels, and a decrease in pro-inflammatory cytokine levels compared to the disease group. However, no statistically significant change within groups was observed in terms of serum insulin and anti-inflammatory cytokine levels. The improvement in diabetic symptoms, such as polyphagia, polydipsia, and weight loss, was observed in the treatment group, along with pancreatic regeneration, which indicated improved insulin secretion. CONCLUSION In the current investigation, we concluded that CM and CM+ Resveratrol, as natural immunomodulators, have the capacity to regenerate injured pancreatic beta cells and have antidiabetic action, together with immunomodulating impact. Nonetheless, future studies on this therapy appear to be promising.
Collapse
Affiliation(s)
- Krushna Abhale
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
| | | | - Shivani Desai
- Clinical Research and Pharmacovigilance, Serum Institute of India Pvt. Ltd., Hadapsar, Pune
| | - Avinash Sanap
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Pimpri, Pune, India
| | - Ramesh Bhonde
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Pimpri, Pune, India
| |
Collapse
|
21
|
Kacemi R, Campos MG. Bee Pollen as a Source of Biopharmaceuticals for Neurodegeneration and Cancer Research: A Scoping Review and Translational Prospects. Molecules 2024; 29:5893. [PMID: 39769981 PMCID: PMC11677910 DOI: 10.3390/molecules29245893] [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: 09/24/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025] Open
Abstract
Bee Pollen (BP) has many advantageous properties relying on its multitargeting potential, a new tendency in managing many challenging illnesses. In cancer and neurodegeneration, the multiple effects of BP could be of unequaled importance and need further investigation. Although still limited, available data interestingly spotlights some floral sources with promising activities in line with this investigation. Adopting scoping review methodology, we have identified many crucial bioactivities that are widely recognized to individual BP compounds but remain completely untapped in this valuable bee cocktail. A wide range of these compounds have been recently found to be endowed with great potential in modulating pivotal processes in neurodegeneration and cancer pathophysiology. In addition, some ubiquitous BP compounds have only been recently isolated, while the number of studied BPs remains extremely limited compared to the endless pool of plant species worldwide. We have also elucidated that clinical profits from these promising perspectives are still impeded by challenging hurdles such as limited bioavailability of the studied phytocompounds, diversity and lack of phytochemical standardization of BP, and the difficulty of selective targeting in some pathophysiological mechanisms. We finally present interesting insights to guide future research and pave the way for urgently needed and simplified clinical investigations.
Collapse
Affiliation(s)
- Rachid Kacemi
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
| | - Maria G. Campos
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- Coimbra Chemistry Centre (CQC, FCT Unit 313) (FCTUC), University of Coimbra, Rua Larga, 3004-531 Coimbra, Portugal
| |
Collapse
|
22
|
Zheng XQ, Zhang XH, Gao HQ, Huang LY, Ye JJ, Ye JH, Lu JL, Ma SC, Liang YR. Green Tea Catechins and Skin Health. Antioxidants (Basel) 2024; 13:1506. [PMID: 39765834 PMCID: PMC11673495 DOI: 10.3390/antiox13121506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Green tea catechins (GTCs) are a group of bioactive polyphenolic compounds found in fresh tea leaves (Camellia sinensis (L.) O. Kuntze). They have garnered significant attention due to their diverse health benefits and potential therapeutic applications, including as antioxidant and sunscreen agents. Human skin serves as the primary barrier against various external aggressors, including pathogens, pollutants, and harmful ultraviolet radiation (UVR). Skin aging is a complex biological process influenced by intrinsic factors such as genetics and hormonal changes, as well as extrinsic factors like environmental stressors, among which UVR plays a pivotal role in accelerating skin aging and contributing to various dermatological conditions. Research has demonstrated that GTCs possess potent antioxidant properties that help neutralize free radicals generated by oxidative stress. This action not only mitigates cellular damage but also supports the repair mechanisms inherent in human skin. Furthermore, GTCs exhibit anti-carcinogenic effects by inhibiting pathways involved in tumor promotion and progression. GTCs have been shown to exert anti-inflammatory effects through modulation of inflammatory signaling pathways. Chronic inflammation is known to contribute significantly to both premature aging and various dermatological diseases such as psoriasis or eczema. By regulating these pathways effectively, GTCs may alleviate symptoms associated with inflammatory conditions. GTCs can enhance wound healing processes by stimulating angiogenesis. They also facilitate DNA repair mechanisms within dermal fibroblasts exposed to damaging agents. The photoprotective properties attributed to GTCs further underscore their relevance in skincare formulations aimed at preventing sun-induced damage. Their ability to screen UV light helps shield underlying tissues from harmful rays. This review paper aims to comprehensively examine the beneficial effects of GTCs on skin health through an analysis encompassing in vivo and in vitro studies alongside insights into molecular mechanisms underpinning these effects. Such knowledge could pave the way for the development of innovative strategies focused on harnessing natural compounds like GTCs for improved skincare solutions tailored to combat environmental stresses faced by the human epidermis.
Collapse
Affiliation(s)
- Xin-Qiang Zheng
- Tea Research Institute, Zhejiang University, #866, Yuhangtang Road, Hangzhou 310058, China; (X.-Q.Z.); (X.-H.Z.); (H.-Q.G.); (L.-Y.H.); (J.-J.Y.); (J.-H.Y.); (J.-L.L.)
| | - Xue-Han Zhang
- Tea Research Institute, Zhejiang University, #866, Yuhangtang Road, Hangzhou 310058, China; (X.-Q.Z.); (X.-H.Z.); (H.-Q.G.); (L.-Y.H.); (J.-J.Y.); (J.-H.Y.); (J.-L.L.)
| | - Han-Qing Gao
- Tea Research Institute, Zhejiang University, #866, Yuhangtang Road, Hangzhou 310058, China; (X.-Q.Z.); (X.-H.Z.); (H.-Q.G.); (L.-Y.H.); (J.-J.Y.); (J.-H.Y.); (J.-L.L.)
| | - Lan-Ying Huang
- Tea Research Institute, Zhejiang University, #866, Yuhangtang Road, Hangzhou 310058, China; (X.-Q.Z.); (X.-H.Z.); (H.-Q.G.); (L.-Y.H.); (J.-J.Y.); (J.-H.Y.); (J.-L.L.)
| | - Jing-Jing Ye
- Tea Research Institute, Zhejiang University, #866, Yuhangtang Road, Hangzhou 310058, China; (X.-Q.Z.); (X.-H.Z.); (H.-Q.G.); (L.-Y.H.); (J.-J.Y.); (J.-H.Y.); (J.-L.L.)
| | - Jian-Hui Ye
- Tea Research Institute, Zhejiang University, #866, Yuhangtang Road, Hangzhou 310058, China; (X.-Q.Z.); (X.-H.Z.); (H.-Q.G.); (L.-Y.H.); (J.-J.Y.); (J.-H.Y.); (J.-L.L.)
| | - Jian-Liang Lu
- Tea Research Institute, Zhejiang University, #866, Yuhangtang Road, Hangzhou 310058, China; (X.-Q.Z.); (X.-H.Z.); (H.-Q.G.); (L.-Y.H.); (J.-J.Y.); (J.-H.Y.); (J.-L.L.)
| | - Shi-Cheng Ma
- Wuzhou Liubao Tea Research Association, #18, Sanlong Avenue, Changzhou District, Wuzhou 543001, China;
| | - Yue-Rong Liang
- Tea Research Institute, Zhejiang University, #866, Yuhangtang Road, Hangzhou 310058, China; (X.-Q.Z.); (X.-H.Z.); (H.-Q.G.); (L.-Y.H.); (J.-J.Y.); (J.-H.Y.); (J.-L.L.)
| |
Collapse
|
23
|
Khamar F, Atabaki M, Samadi M, Reisi M, Sandoughi M. Immunomodulatory effects of novel nano micelle based curcumin in rheumatoid arthritis patients: A double blind randomized clinical trial. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2024; 5:227-234. [PMID: 39802548 PMCID: PMC11720463 DOI: 10.1515/rir-2024-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/17/2024] [Indexed: 01/16/2025]
Abstract
Background and Objectives Rheumatoid arthritis (RA) is a well-known systemic autoimmune inflammatory disease. This investigation aimed to assess the effects of Sina-curcumin, a novel nano micelle-based curcumin, on immune system responses of RA patients. Methods This pilot study is a randomized double blinded, controlled trial. Patients who fulfilled the European League against Rheumatism-American College of Rheumatology (EULAR-ACR) criteria for RA were assigned to receive curcumin or placebo for 12 weeks. The outcomes of this study were comparison of changes in mean value of Disease Activity Score of 28 joints erythrocyte sedimentation rate (DAS28-ESR), erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), frequency of T helper 1 and T helper 2 cells population. Results From 150 RA patients who were assessed for eligibility, data from 30 patients (15 patients in each group) were analyzed. There was no significant difference between the two groups regarding age (P = 0.6441) and body mass index (BMI, P = 0.6016). Our measurement showed a statistically significant reduction in ESR (P < 0.0001), CRP (P < 0.0001) and a non-significant decrease in DAS28-ESR (P = 0.5125) in the curcumin group. Also, the Th1/Th2 ratio favorably decreased in the curcumin group. This finding was due to a significant increase in Th2 cells (P < 0.0001) and a nonsignificant decrease in Th1 cells (P = 0.1532). Conclusion Our trial findings revealed the immunomodulatory effects of curcumin. It could be used and recommended as adjunctive treatment for RA patients.
Collapse
Affiliation(s)
- Faezeh Khamar
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mahdi Atabaki
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Morteza Samadi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Reisi
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mahnaz Sandoughi
- Rheumatologis, Department of Internal Medicine, Ali Ebn Abitaleb Hospital, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
24
|
An H, Lin B, Huang F, Wang N. Advances in the study of polysaccharides from Anemarrhena asphodeloides Bge.: A review. Int J Biol Macromol 2024; 282:136999. [PMID: 39476924 DOI: 10.1016/j.ijbiomac.2024.136999] [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: 06/06/2024] [Revised: 10/09/2024] [Accepted: 10/26/2024] [Indexed: 11/05/2024]
Abstract
Anemarrhena asphodeloides Bge. (AA), a traditional Chinese medicine, is used clinically to treat inflammation, diabetes, osteoporosis, and tumors. Polysaccharides are the most abundant components in AA, and have antioxidant, immunomodulatory, anti-inflammatory, hypoglycemic, anti-osteoporosis, and laxative effects. It is necessary to conduct a comprehensive analysis on the structure and pharmacological activity of the polysaccharides from AA (PAAs). This review systematically summarizes the structural characteristics of PAAs, including the monosaccharide compositions, molecular weights, and backbone structures. We discuss the relationship between the structure and pharmacological activities of PAAs. The chemical modification methods of PAAs, including zinc chelation, carboxymethylation, and sulfation, are then reviewed. This review may offer new insights for research on the PAAs and polysaccharides with similar structures.
Collapse
Affiliation(s)
- Huan An
- Department of TCM literature, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, China
| | - Bingfeng Lin
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, China
| | - Feihua Huang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, China; Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, China
| | - Nani Wang
- Department of TCM literature, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, China; Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, China; Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, China.
| |
Collapse
|
25
|
Zeng X, Li T, Yang K, Jiang Y, Chen S, Yang S, Zou S, Liu J, Duan P. Natural compound phloretin restores periodontal immune homeostasis via HIF-1α-regulated PI3K/Akt and glycolysis in macrophages. Int Immunopharmacol 2024; 141:112933. [PMID: 39186834 DOI: 10.1016/j.intimp.2024.112933] [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: 06/20/2024] [Revised: 07/27/2024] [Accepted: 08/11/2024] [Indexed: 08/28/2024]
Abstract
Periodontitis is a chronic inflammatory disease that affects about 45 %-50 % of adults worldwide, but the efficacy of current clinical therapies is unsatisfactory due to the complicated periodontal immune microenvironment. Thus, developing drugs that can regulate innate immune cells (e.g., macrophages) is a potent strategy to treat periodontitis. Here, we report that phloretin, a food plant-derived natural compound, is sufficient to alleviate periodontitis through immune regulation. In vivo, phloretin treatment could significantly reduce alveolar bone resorption and periodontal inflammation in mouse periodontitis models. In vitro, phloretin could suppress proinflammatory (M1-like) polarization and cytokine release in macrophages induced by LPS. Mechanistically, the immune regulatory role of phloretin in macrophages may be due to its metabolic regulation effect. Phloretin might restore the balance of M1/M2 macrophage transition in periodontitis by inhibiting HIF-1α-mediated glycolysis and PI3k/Akt pathways, thereby reducing the proinflammatory effect and immune disorder caused by over-activated M1 macrophages. Together, this study highlights that natural compound, such as phloretin, can restore periodontal immune homeostasis by metabolic regulation of macrophages, which may provide novel insight into the treatment of periodontitis.
Collapse
Affiliation(s)
- Xinyi Zeng
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Tiancheng Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200001, China
| | - Kuan Yang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yukun Jiang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shuo Chen
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shuxian Yang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jingping Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China
| | - Peipei Duan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
26
|
Nanjaiah H, Moudgil KD. Targeted Therapy of Antibody-Induced Autoimmune Arthritis Using Peptide-Guided Nanoparticles. Int J Mol Sci 2024; 25:12019. [PMID: 39596089 PMCID: PMC11593680 DOI: 10.3390/ijms252212019] [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: 10/13/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation of the joints and it affects over 18 million people worldwide. Despite the availability of a variety of potent drugs for RA, over 30-40 percent of patients fail to achieve adequate remission, and many patients suffer from systemic adverse effects. Thus, there is an urgent need for a joint-targeted drug delivery system. Nanotechnology-based drug delivery methods offer a promising resource that is largely untapped for RA. Using the T cell-driven rat adjuvant-induced arthritis (AA) model of human RA, we developed a peptide-targeted liposomal drug delivery system for arthritis therapy. It was based on a novel joint-homing peptide ART-2 to guide liposomes entrapping dexamethasone (Dex) to arthritic joints of rats, and this approach was more effective in suppressing arthritis than the unpackaged (free) drug. To de-risk the translation of our innovative drug delivery technology to RA patients, we undertook the validation of ART-2-liposomal delivery in a genetically and mechanistically distinct arthritis model in mice, the collagen antibody-induced arthritis (CAIA) model. Using live imaging for tissue distribution of liposomes in vivo, immunohistochemistry of paws for cellular binding of ART-2, and liposomal Dex delivery, our results fully validated the key findings of the rat model, namely, preferential homing of peptide-functionalized liposomes to arthritic joints compared to healthy joints, and higher efficacy of liposomal Dex than free Dex. These results offer a proof-of-concept for the benefits of targeted drug delivery to the joints and its potential translation to RA patients.
Collapse
Affiliation(s)
- Hemalatha Nanjaiah
- Research and Development, VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kamal D. Moudgil
- Research and Development, VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Rheumatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
27
|
Cho EC, Kwon HS, Lee NY, Oh HJ, Choi YJ. Blood circulation effect of fermented citrus bioconversion product (FCBP) in EA.hy926 endothelial cells and high-fat diet-fed mouse model. Food Nutr Res 2024; 68:10682. [PMID: 39534464 PMCID: PMC11556378 DOI: 10.29219/fnr.v68.10682] [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: 03/24/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background The escalating global burden of cardiovascular diseases, largely driven by unhealthy lifestyle choices and dietary patterns, has intensified the search for effective and safe interventions. With current treatments often marred by significant side effects, the exploration of natural compounds such as flavonoids presents a compelling alternative. Objective This study investigated the effects of fermented citrus bioconversion product (FCBP), a fermented citrus bioflavonoid, on various markers of cardiovascular health in the context of a high-fat diet. Design In vivo, a high-fat diet-induced mouse model was used to assess the effects of FCBP on body weight, serum nitric oxide (NO) levels, activated partial thromboplastin time (aPTT), phosphatidylserine (PS) exposure on red blood cells, and the expression of inflammatory markers Intercellular Adhesion Molecule (ICAM)-1 and Vascular Cell Adhesion Molecule (VCAM)-1 in the thoracic aorta. In vitro, EA.hy926 endothelial cells were used to evaluate the compound's effects on cell viability, NO production, endothelial nitric oxide synthase (eNOS) expression, and cell adhesion molecule (CAM) levels to further understand the mechanisms behind the in vivo findings. Results In vivo, FCBP supplementation led to a dose-dependent reduction in weight gain, a significant decrease in serum NO levels at 10 mg/kg, and reduced ICAM-1 and VCAM-1 expressions in the thoracic aorta, indicating anti-inflammatory properties. PS exposure on red blood cells was also reduced, suggesting decreased procoagulant activity, while aPTT remained unchanged. In vitro, FCBP was non-cytotoxic to endothelial cells, showed a trend toward increased NO production and eNOS expression, and reduced the expression of ICAM-1 and VCAM-1, supporting its potential anti-inflammatory effects. Conclusions FCBP demonstrates potential as a bioactive compound for managing cardiovascular health by reducing inflammation, mitigating weight gain, and influencing blood circulation-related parameters under high-fat diet conditions. Further studies, including diverse models and human trials, are warranted to elucidate its mechanisms and compare its efficacy with established cardiovascular therapeutics.
Collapse
Affiliation(s)
- Eun-Chae Cho
- Department of Convergence Science, Sahmyook University, Seoul, Republic of Korea
| | - Hyuck Se Kwon
- R&D Team, Food & Supplement Health Claims, Vitech Co., Ltd., Wanju, Republic of Korea
| | - Na Young Lee
- R&D Team, Food & Supplement Health Claims, Vitech Co., Ltd., Wanju, Republic of Korea
| | - Hyun Jeong Oh
- R&D Team, Food & Supplement Health Claims, Vitech Co., Ltd., Wanju, Republic of Korea
| | - Yean-Jung Choi
- Department of Food and Nutrition, Sahmyook University, Seoul, Republic of Korea
| |
Collapse
|
28
|
Tenchov R, Sasso JM, Zhou QA. Alzheimer's Disease: Exploring the Landscape of Cognitive Decline. ACS Chem Neurosci 2024; 15:3800-3827. [PMID: 39392435 PMCID: PMC11587518 DOI: 10.1021/acschemneuro.4c00339] [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: 06/03/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. The pathology of AD is marked by the accumulation of amyloid beta plaques and tau protein tangles in the brain, along with neuroinflammation and synaptic dysfunction. Genetic factors, such as mutations in APP, PSEN1, and PSEN2 genes, as well as the APOE ε4 allele, contribute to increased risk of acquiring AD. Currently available treatments provide symptomatic relief but do not halt disease progression. Research efforts are focused on developing disease-modifying therapies that target the underlying pathological mechanisms of AD. Advances in identification and validation of reliable biomarkers for AD hold great promise for enhancing early diagnosis, monitoring disease progression, and assessing treatment response in clinical practice in effort to alleviate the burden of this devastating disease. In this paper, we analyze data from the CAS Content Collection to summarize the research progress in Alzheimer's disease. We examine the publication landscape in effort to provide insights into current knowledge advances and developments. We also review the most discussed and emerging concepts and assess the strategies to combat the disease. We explore the genetic risk factors, pharmacological targets, and comorbid diseases. Finally, we inspect clinical applications of products against AD with their development pipelines and efforts for drug repurposing. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding AD, to outline challenges, and to evaluate growth opportunities to further efforts in combating the disease.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | - Janet M. Sasso
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | | |
Collapse
|
29
|
Zhao X, Zhang Y, Wang P, Liu K, Zheng Y, Wen J, Wang K, Wen X. Layer by layer self-assembled hyaluronic acid nanoarmor for the treatment of ulcerative colitis. J Nanobiotechnology 2024; 22:633. [PMID: 39420343 PMCID: PMC11488142 DOI: 10.1186/s12951-024-02933-3] [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: 05/27/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024] Open
Abstract
Natural compound-based treatments provide innovative ways for ulcerative colitis therapy. However, poor targeting and rapid degradation curtail its application, which needs to be addressed. Inspired by biomacromolecule-based materials, we have developed an orally administrated nanoparticle (GBP@HA NPs) using bovine serum albumin as a carrier for polyphenol delivery. The system synergizes galactosylated bovine serum albumin with two polyphenols, epigallocatechin gallate and tannic acid, which is then encased in "nanoarmor" of ε-Polylysine and hyaluronic acid to boost its stability and targeting. Remarkably, the nanoarmor demonstrated profound therapeutic effects in both acute and chronic mouse models of ulcerative colitis, mitigating disease symptoms via multiple mechanisms, regulating inflammation related factors and exerting a modulatory impact on gut microbiota. Further mechanistic investigations indicate that GBP@HA NPs may act through several pathways, including modulation of Keap1-Nrf2 and NF-κB signaling, as well as Caspase-1-dependent pyroptosis. Consequently, this novel armored nanotherapy promotes the way for enhanced polyphenol utilization in ulcerative colitis treatment research.
Collapse
Affiliation(s)
- Xinxin Zhao
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yuchen Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Pengchong Wang
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an Shaanxi, 710068, China
| | - Kailai Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yunhe Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jinpeng Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ke Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Xiaopeng Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
30
|
Andrade MLDO, Marinho PAF, de Oliveira AM, de Souza TA, Cibulski SP, Alves HDS. Apodanthera glaziovii (Cucurbitaceae) Shows Strong Anti-Inflammatory Activity in Murine Models of Acute Inflammation. Pharmaceutics 2024; 16:1298. [PMID: 39458627 PMCID: PMC11510368 DOI: 10.3390/pharmaceutics16101298] [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: 09/02/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Background/objectives:Apodanthera glaziovii is an endemic species from the semi-arid Brazilian, which has limited toxicological and pharmacological studies. This species belongs to a well-studied family known for its bioactive compounds used in treating inflammatory. This study aimed to identify secondary metabolites in the stems from A. glaziovii, evaluate toxicity, and investigate the anti-inflammatory potential of the stem hydroalcoholic extract (SHE-Ag). Methods: qualitative and quantitative assays were employed to identify secondary metabolites, along with chromatographic analyses and 1H and 13C NMR. Toxicity was assessed through in vitro hemolytic toxicity, in vivo genotoxicity, and oral acute toxicity tests before the pharmacological assays were conducted. Results: phytochemical screening, HPLC and NMR analyses suggested the presence of saponins of the norcucurbitacin class. The SHE-Ag exhibited no hemolytic activity and no mutagenic potential. However, in vivo toxicity at a dose of 2000 mg/kg revealed hematological and biochemical alterations, while the 500 mg/kg dose was safe. In the anti-inflammatory assays, SHE-Ag at 100 mg/kg reduced paw edema by 55.8%, and leukocyte and neutrophil migration by 62% and 68% in the peritonitis model, respectively; inflammatory cell migration by 70% in the air pouch model, outperforming indomethacin, which showed a 54% reduction. Conclusions: these findings indicate that SHE-Ag is rich in saponins, confirmed through HPLC and 1H and 13C NMR analyses. The SHE-Ag also demonstrated low toxicity. The inflammation models used showed a reduction in inflammation, pro-inflammatory cells, and edema, highlighting the significant anti-inflammatory activity of hydroethanolic extract A. glaziovii stems.
Collapse
Affiliation(s)
- Maria Lorena de Oliveira Andrade
- Center of Biological and Health Sciences, Phytochemistry Laboratory, Post-Graduation Program in Pharmaceuticals Sciences, State University of Paraíba, Campina Grande 58429-500, Brazil; (M.L.d.O.A.); (S.P.C.)
| | - Pedro Artur Ferreira Marinho
- Center of Biological and Health Sciences, Pharmacological Analysis Laboratory, Post-Graduation Program in Pharmaceuticals Sciences, State University of Paraíba, Campina Grande 58429-500, Brazil; (P.A.F.M.); (A.M.d.O.)
| | - Alisson Macário de Oliveira
- Center of Biological and Health Sciences, Pharmacological Analysis Laboratory, Post-Graduation Program in Pharmaceuticals Sciences, State University of Paraíba, Campina Grande 58429-500, Brazil; (P.A.F.M.); (A.M.d.O.)
| | - Thalisson Amorim de Souza
- Multi-User Characterization and Analysis Laboratory, Research Institute for Drugs and Medicines (IpeFarM), Federal University of Paraíba, João Pessoa 58059-900, Brazil;
| | - Samuel Paulo Cibulski
- Center of Biological and Health Sciences, Phytochemistry Laboratory, Post-Graduation Program in Pharmaceuticals Sciences, State University of Paraíba, Campina Grande 58429-500, Brazil; (M.L.d.O.A.); (S.P.C.)
| | - Harley da Silva Alves
- Center of Biological and Health Sciences, Phytochemistry Laboratory, Post-Graduation Program in Pharmaceuticals Sciences, State University of Paraíba, Campina Grande 58429-500, Brazil; (M.L.d.O.A.); (S.P.C.)
| |
Collapse
|
31
|
Wang Y, Su L, Hu Z, Peng S, Li N, Fu H, Wang B, Wu H. Resveratrol suppresses liver cancer progression by downregulating AKR1C3: targeting HCC with HSA nanomaterial as a carrier to enhance therapeutic efficacy. Apoptosis 2024; 29:1429-1453. [PMID: 39023830 DOI: 10.1007/s10495-024-01995-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] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
The enzyme AKR1C3 plays a crucial role in hormone and drug metabolism and is associated with abnormal expression in liver cancer, leading to tumor progression and poor prognosis. Nanoparticles modified with HSA can modulate the tumor microenvironment by enhancing photodynamic therapy to induce apoptosis in tumor cells and alleviate hypoxia. Therefore, exploring the potential regulatory mechanisms of resveratrol on AKR1C3 through the construction of HSA-RSV NPs carriers holds significant theoretical and clinical implications for the treatment of liver cancer. The aim of this study is to investigate the targeted regulation of AKR1C3 expression through the loading of resveratrol (RSV) on nanomaterials HSA-RSV NPs (Nanoparticles) in order to alleviate tumor hypoxia and inhibit the progression of hepatocellular carcinoma (HCC), and to explore its molecular mechanism. PubChem database and PharmMapper server were used to screen the target genes of RSV. HCC-related differentially expressed genes (DEGs) were analyzed through the GEO dataset, and relevant genes were retrieved from the GeneCards database, resulting in the intersection of the three to obtain candidate DEGs. GO and KEGG enrichment analyses were performed on the candidate DEGs to analyze the potential cellular functions and molecular signaling pathways affected by the main target genes. The cytohubba plugin was used to screen the top 10 target genes ranked by Degree and further intersected the results of LASSO and Random Forest (RF) to obtain hub genes. The expression analysis of hub genes and the prediction of malignant tumor prognosis were conducted. Furthermore, a pharmacophore model was constructed using PharmMapper. Molecular docking simulations were performed using AutoDockTools 1.5.6 software, and ROC curve analysis was performed to determine the core target. In vitro cell experiments were carried out by selecting appropriate HCC cell lines, treating HCC cells with different concentrations of RSV, or silencing or overexpressing AKR1C3 using lentivirus. CCK-8, clone formation, flow cytometry, scratch experiment, and Transwell were used to measure cancer cell viability, proliferation, migration, invasion, and apoptosis, respectively. Cellular oxygen consumption rate was analyzed using the Seahorse XF24 analyzer. HSA-RSV NPs were prepared, and their characterization and cytotoxicity were evaluated. The biological functional changes of HCC cells after treatment were detected. An HCC subcutaneous xenograft model was established in mice using HepG2 cell lines. HSA-RSV NPs were injected via the tail vein, with a control group set, to observe changes in tumor growth, tumor targeting of NPs, and biological safety. TUNEL, Ki67, and APC-hypoxia probe staining were performed on excised tumor tissue to detect tumor cell proliferation, apoptosis, and hypoxia. Lentivirus was used to silence or overexpress AKR1C3 simultaneously with the injection of HSA-RSV NPs via the tail vein to assess the impact of AKR1C3 on the regulation of HSA-RSV NPs in HCC progression. Bioinformatics analysis revealed that AKR1C3 is an important target gene involved in the regulation of HCC by RSV, which is associated with the prognosis of HCC patients and upregulated in expression. In vitro cell experiments showed that RSV significantly inhibits the respiratory metabolism of HCC cells, suppressing their proliferation, migration, and invasion and promoting apoptosis. Silencing AKR1C3 further enhances the toxicity of RSV towards HCC cells. The characterization and cytotoxicity experiments of nanomaterials demonstrated the successful construction of HSA-RSV NPs, which exhibited stronger inhibitory effects on HCC cells. In vivo, animal experiments further confirmed that targeted downregulation of AKR1C3 by HSA-RSV NPs suppresses the progression of HCC and tumor hypoxia while exhibiting tumor targeting and biological safety. Targeted downregulation of AKR1C3 by HSA-RSV NPs can alleviate HCC tumor hypoxia and inhibit the progression of HCC.
Collapse
Affiliation(s)
- Ying Wang
- Operating Room, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Longxiang Su
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Zhansheng Hu
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Shuang Peng
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Na Li
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Haiyan Fu
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Baoquan Wang
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Huiping Wu
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China.
| |
Collapse
|
32
|
Vadakkan K, Sathishkumar K, Mapranathukaran VO, Ngangbam AK, Nongmaithem BD, Hemapriya J, Nair JB. Critical review on plant-derived quorum sensing signaling inhibitors in pseudomonas aeruginosa. Bioorg Chem 2024; 151:107649. [PMID: 39029321 DOI: 10.1016/j.bioorg.2024.107649] [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: 05/05/2024] [Revised: 06/21/2024] [Accepted: 07/14/2024] [Indexed: 07/21/2024]
Abstract
Pseudomonas aeruginosa, a biofilm-forming organism with complex quorum mechanisms (Las, Rhl, PQS, and IQS), poses an imminent danger to the healthcare sector and renders current treatment options for chemotherapy ineffectual. The pathogen's diverse pathogenicity, antibiotic resistance, and biofilms make it difficult to eradicate it effectively. Quorum sensing, a complex system reliant on cell density, controls P. aeruginosa's pathogenesis. Quorum-sensing genes are key components of P. aeruginosa's pathogenic arsenal, and their expression determines how severe the spread of infection becomes. Over the past ten years, there has been a noticeable increase in the quest for and development of new antimicrobial medications. Quorum sensing may be an effective treatment for infections triggered by bacteria. Introducing quorum-sensing inhibitors as an anti-virulent strategy might be an intriguing therapeutic method that can be effectively employed along with current medications. Amongst the several speculated processes, a unique anti-virulence strategy using anti-quorum sensing and antibiofilm medications for targeting pseudomonal infestations seems to be at the forefront. Due to their noteworthy quorum quenching capabilities, biologically active phytochemicals have become more well-known in the realm of science in this context. Recent research showed how different phytochemical quorum quenching actions affect P. aeruginosa's QS-dependent pathogenicity. This review focuses on the most current data supporting the implementation of plant bio-actives to treat P.aeruginosa-associated diseases, as well as the benefits and future recommendationsof employing them in anti-virulence therapies as a supplementary drug development approach towards conventional antibiotic approaches.
Collapse
Affiliation(s)
- Kayeen Vadakkan
- Department of Biotechnology, St. Mary's College (Autonomous), Thrissur, Kerala 680020, India; Manipur International University, Imphal, Manipur 795140, India.
| | - Kuppusamy Sathishkumar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Thandalam, Chennai, Tamil Nadu 602105, India
| | | | | | | | - Janarthanam Hemapriya
- Department of Microbiology, DKM College for Women, Vellore, Tamil Nadu 632001, India
| | - Jyotsna B Nair
- Department of Biotechnology, JDT Islam College of Arts and Science, Vellimadukunnu, Kozhikode, Kerala 673012, India
| |
Collapse
|
33
|
Saadati F, Modarresi Chahardehi A, Jamshidi N, Jamshidi N, Ghasemi D. Coumarin: A natural solution for alleviating inflammatory disorders. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 7:100202. [PMID: 39398983 PMCID: PMC11470182 DOI: 10.1016/j.crphar.2024.100202] [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: 04/28/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
Coumarin, a naturally occurring compound found in various plants, has a rich history of use in traditional medicine. Recent research has highlighted its anti-inflammatory properties, positioning it as a promising candidate for treating inflammatory disorders such as rheumatoid arthritis, asthma, and inflammatory bowel disease. This narrative review aims to comprehensively summarize the current knowledge regarding coumarin's pharmacological effects in alleviating inflammatory conditions by analyzing preclinical and clinical studies. The review focuses on elucidating the mechanisms through which coumarin exerts its anti-inflammatory effects, including its antioxidant activity, inhibiting pro-inflammatory cytokine production, and modulation of immune cell functions. Additionally, the paper addresses potential limitations of using coumarin, such as concerns about toxicity at high doses or with prolonged use. Before widespread clinical application, further investigation is needed to fully understand coumarin's potential benefits and risks.
Collapse
Affiliation(s)
- Farnoosh Saadati
- Department of Cellular and Molecular Biology, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | | | - Negar Jamshidi
- Kimia Andisheh Teb Medical and Molecular Research Laboratory Co., Tehran, Iran
| | - Nazanin Jamshidi
- Kimia Andisheh Teb Medical and Molecular Research Laboratory Co., Tehran, Iran
| | - Darioush Ghasemi
- Kimia Andisheh Teb Medical and Molecular Research Laboratory Co., Tehran, Iran
| |
Collapse
|
34
|
Yue Y, Han J, Shen X, Zhu F, Liu Y, Zhang W, Xia W, Wu M. Structural characteristics, immune-activating mechanisms in vitro, and immunomodulatory effects in vivo of the exopolysaccharide EPS53 from Streptococcus thermophilus XJ53. Carbohydr Polym 2024; 340:122259. [PMID: 38858019 DOI: 10.1016/j.carbpol.2024.122259] [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: 01/21/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 06/12/2024]
Abstract
Our previous investigations have successfully identified the repeating structural units of EPS53, an exopolysaccharide derived from Streptococcus thermophilus XJ53 fermented milk, and substantiated its potential immunomodulatory properties. The present study further elucidated the structural characteristics of EPS53 and investigated the underlying mechanisms governing its in vitro immunoreactivity as well as its in vivo immunoreactivity. The results obtained from multi-detector high performance gel filtration chromatography revealed that EPS53 adopted a rigid rod conformation in aqueous solution, with the weight-average molecular weight of 1464 kDa, the number-average molecular weight of 694 kDa, and the polydispersity index of 2.11. Congo red experiment confirmed the absence of a triple helix conformation. Scanning electron microscopy showed that EPS53 displayed a three-dimensional fibrous structure covered with flakes. The in vitro findings indicated that EPS53 enhanced phagocytosis ability, reactive oxygen species (ROS) production, and cytokine levels of macrophages via the TLR4-mediated NF-κB/MAPK signaling pathways as confirmed by immunofluorescence staining experiments, inhibition blocking experiments, and Western blot assay. Additionally, the in vivo experiments demonstrated that EPS53 significantly increased macrophage and neutrophil number while enhancing NO and ROS levels in zebrafish larvae; thus, providing further evidence for the immunomodulatory efficacy of EPS53.
Collapse
Affiliation(s)
- Yun Yue
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Jin Han
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Centre of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai 200436, PR China
| | - Xinyan Shen
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Fei Zhu
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Yikang Liu
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Wenqing Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Wei Xia
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China.
| | - Mengqi Wu
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China; Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
35
|
Lv Y, Yang Z, Hai L, Chen X, Wang J, Hu S, Zhao Y, Yuan H, Hu Z, Cui D, Xie J. Differential alterations of CXCR3, CXCR5 and CX3CR1 in patients with immune thrombocytopenia. Cytokine 2024; 181:156684. [PMID: 38936205 DOI: 10.1016/j.cyto.2024.156684] [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: 04/12/2024] [Revised: 06/16/2024] [Accepted: 06/22/2024] [Indexed: 06/29/2024]
Abstract
As a versatile element for maintaining homeostasis, the chemokine system has been reported to be implicated in the pathogenesis of immune thrombocytopenia (ITP). However, research pertaining to chemokine receptors and related ligands in adult ITP is still limited. The states of several typical chemokine receptors and cognate ligands in the circulation were comparatively assessed through various methodologies. Multiple variable analyses of correlation matrixes were conducted to characterize the correlation signatures of various chemokine receptors or candidate ligands with platelet counts. Our data illustrated a significant decrease in relative CXCR3 expression and elevated plasma levels of CXCL4, 9-11, 13, and CCL3 chemokines in ITP patients with varied platelet counts. Flow cytometry assays revealed eminently diminished CXCR3 levels on T and B lymphocytes and increased CXCR5 on cytotoxic T cell (Tc) subsets in ITP patients with certain platelet counts. Meanwhile, circulating CX3CR1 levels were markedly higher on T cells with a concomitant increase in plasma CX3CL1 level in ITP patients, highlighting the importance of aberrant alterations of the CX3CR1-CX3CL1 axis in ITP pathogenesis. Spearman's correlation analyses revealed a strong positive association of peripheral CXCL4 mRNA level, and negative correlations of plasma CXCL4 concentration and certain chemokine receptors with platelet counts, which might serve as a potential biomarker of platelet destruction in ITP development. Overall, these results indicate that the differential expression patterns and distinct activation states of peripheral chemokine network, and the subsequent expansion of circulating CXCR5+ Tc cells and CX3CR1+ T cells, may be a hallmark during ITP progression, which ultimately contributes to thrombocytopenia in ITP patients.
Collapse
Affiliation(s)
- Yan Lv
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ziyin Yang
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lei Hai
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaoyu Chen
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiayuan Wang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shaohua Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuhong Zhao
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Huiming Yuan
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhengjun Hu
- Department of Laboratory Medicine, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou 310060, China.
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Jue Xie
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
36
|
Feng Y, Pan M, Li R, He W, Chen Y, Xu S, Chen H, Xu H, Lin Y. Recent developments and new directions in the use of natural products for the treatment of inflammatory bowel disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155812. [PMID: 38905845 DOI: 10.1016/j.phymed.2024.155812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/13/2024] [Accepted: 06/06/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) represents a significant global health challenge, and there is an urgent need to explore novel therapeutic interventions. Natural products have demonstrated highly promising effectiveness in the treatment of IBD. PURPOSE This study systematically reviews the latest research advancements in leveraging natural products for IBD treatment. METHODS This manuscript strictly adheres to the PRISMA guidelines. Relevant literature on the effects of natural products on IBD was retrieved from the PubMed, Web of Science and Cochrane Library databases using the search terms "natural product," "inflammatory bowel disease," "colitis," "metagenomics", "target identification", "drug delivery systems", "polyphenols," "alkaloids," "terpenoids," and so on. The retrieved data were then systematically summarized and reviewed. RESULTS This review assessed the different effects of various natural products, such as polyphenols, alkaloids, terpenoids, quinones, and others, in the treatment of IBD. While these natural products offer promising avenues for IBD management, they also face challenges in terms of clinical translation and drug discovery. The advent of metagenomics, single-cell sequencing, target identification techniques, drug delivery systems, and other cutting-edge technologies heralds a new era in overcoming these challenges. CONCLUSION This paper provides an overview of current research progress in utilizing natural products for the treatment of IBD, exploring how contemporary technological innovations can aid in discovering and harnessing bioactive natural products for the treatment of IBD.
Collapse
Affiliation(s)
- Yaqian Feng
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Mengting Pan
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Ruiqiong Li
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Weishen He
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yangyang Chen
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Shaohua Xu
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| | - Hui Chen
- Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, China.
| | - Huilong Xu
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| | - Yao Lin
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| |
Collapse
|
37
|
Subudhi RN, Poonia N, Singh D, Arora V. Natural approaches for the management of ulcerative colitis: evidence of preclinical and clinical investigations. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:42. [PMID: 39078427 PMCID: PMC11289194 DOI: 10.1007/s13659-024-00463-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 07/07/2024] [Indexed: 07/31/2024]
Abstract
Ulcerative colitis (UC) is a recurring autoimmune disorder characterized by persistent inflammation in the mucosal lining of the lower part of the large intestine. Conventional treatment options such as salicylates, corticosteroids, and immunosuppressants often come with severe side effects, limited bioavailability, and the development of drug resistance, which hampers their therapeutic effectiveness. Therefore, it is imperative to explore natural strategies as safe and alternative treatments for UC. Currently, around 40% of UC patients find relief through natural constituents, which can help reduce toxic side effects and maintain clinical remission. This review aims to provide a summary of both preclinical and clinical evidence supporting the efficacy of various natural substances in the prophylaxis of UC. These natural options include plant extracts, essential oils, nutraceuticals, and phytochemicals. Furthermore, we will delve into the potential mechanisms that underlie the protective and curative actions of these novel herbal agents. In summary, this review will explore the effectiveness of natural remedies for UC, shedding light on their preclinical and clinical findings and the mechanisms behind their therapeutic actions. These alternatives offer hope for improved treatment outcomes and reduced side effects for individuals suffering from this challenging autoimmune condition.
Collapse
Affiliation(s)
- Rudra Narayan Subudhi
- Department of Pharmaceutics, University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Neelam Poonia
- Department of Pharmaceutics, University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India.
| | - Dilpreet Singh
- Department of Pharmaceutics, University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Vimal Arora
- Department of Pharmaceutics, University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| |
Collapse
|
38
|
Burlec AF, Hăncianu M, Ivănescu B, Macovei I, Corciovă A. Exploring the Therapeutic Potential of Natural Compounds in Psoriasis and Their Inclusion in Nanotechnological Systems. Antioxidants (Basel) 2024; 13:912. [PMID: 39199158 PMCID: PMC11352172 DOI: 10.3390/antiox13080912] [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: 07/02/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
Psoriasis is a chronic inflammatory disease that affects around 2-3% of the world's population. The treatment for this autoimmune disease still remains centered around conventional methods using synthetic substances, even though more recent advancements focus on biological therapies. Given the numerous side effects of such treatments, current research involves plant extracts and constituents that could prove useful in treating psoriasis. The aim of this narrative review is to highlight the most known representatives belonging to classes of natural compounds such as polyphenols (e.g., astilbin, curcumin, hesperidin, luteolin, proanthocyanidins, and resveratrol), alkaloids (e.g., berberine, capsaicin, and colchicine), coumarins (psoralen and 8-methoxypsoralen), and terpenoids (e.g., celastrol, centelloids, and ursolic acid), along with plants used in traditional medicine that could present therapeutic potential in psoriasis. The paper also provides an overview of these compounds' mechanisms of action and current inclusion in clinical studies, as well as an investigation into their potential incorporation in various nanotechnological systems, such as lipid-based nanocarriers or polymeric nanomaterials, that may optimize their efficacy during treatment.
Collapse
Affiliation(s)
- Ana Flavia Burlec
- Department of Drug Analysis, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.F.B.); (A.C.)
| | - Monica Hăncianu
- Department of Pharmacognosy, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania;
| | - Bianca Ivănescu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Irina Macovei
- Department of Drug Analysis, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.F.B.); (A.C.)
| | - Andreia Corciovă
- Department of Drug Analysis, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.F.B.); (A.C.)
| |
Collapse
|
39
|
Laura M, Marzia V, Donatella D, Lorenzo DCM, Carla G, Antiga E, Alice V, Marzia C, Emanuela B. Posidonia oceanica (L.) Delile Is a Promising Marine Source Able to Alleviate Imiquimod-Induced Psoriatic Skin Inflammation. Mar Drugs 2024; 22:300. [PMID: 39057409 PMCID: PMC11277884 DOI: 10.3390/md22070300] [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: 05/23/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Psoriasis is a chronic immune-mediated inflammatory cutaneous disease characterized by elevated levels of inflammatory cytokines and adipokine Lipocalin-2 (LCN-2). Recently, natural plant-based products have been studied as new antipsoriatic compounds. We investigate the ability of a leaf extract of the marine plant Posidonia oceanica (POE) to inhibit psoriatic dermatitis in C57BL/6 mice treated with Imiquimod (IMQ). One group of mice was topically treated with IMQ (IMQ mice) for 5 days, and a second group received POE orally before each topical IMQ treatment (IMQ-POE mice). Psoriasis Area Severity Index (PASI) score, thickness, and temperature of the skin area treated with IMQ were measured in both groups. Upon sacrifice, the organs were weighed, and skin biopsies and blood samples were collected. Plasma and lesional skin protein expression of IL-17, IL-23, IFN-γ, IL-2, and TNF-α and plasma LCN-2 concentration were evaluated by ELISA. PASI score, thickness, and temperature of lesional skin were reduced in IMQ-POE mice, as were histological features of psoriatic dermatitis and expression of inflammatory cytokines and LCN-2 levels. This preliminary study aims to propose P. oceanica as a promising naturopathic anti-inflammatory treatment that could be introduced in Complementary Medicine for psoriasis.
Collapse
Affiliation(s)
- Micheli Laura
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Firenze, Italy; (M.L.); (D.C.M.L.); (G.C.)
| | - Vasarri Marzia
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Biochemistry Section, University of Florence, 50134 Firenze, Italy; (V.M.); (D.D.)
| | - Degl’Innocenti Donatella
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Biochemistry Section, University of Florence, 50134 Firenze, Italy; (V.M.); (D.D.)
- CIBM, Applied Ecology and Marine Biology Interuniversity Centre “G. Bacci”, 57128 Livorno, Italy
| | - Di Cesare Mannelli Lorenzo
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Firenze, Italy; (M.L.); (D.C.M.L.); (G.C.)
| | - Ghelardini Carla
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Firenze, Italy; (M.L.); (D.C.M.L.); (G.C.)
| | - Emiliano Antiga
- Department of Health Sciences, Dermatology Section, University of Florence, 50125 Firenze, Italy; (E.A.); (C.M.)
| | - Verdelli Alice
- Central Tuscany Local Health Authority, Department of Multidimensional Medicine, Immuno-Rheumatology and Infectious Diseases Area, Dermatology SOC, Dermatological Rare Diseases SOS, 50125 Firenze, Italy;
| | - Caproni Marzia
- Department of Health Sciences, Dermatology Section, University of Florence, 50125 Firenze, Italy; (E.A.); (C.M.)
| | - Barletta Emanuela
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Experimental Pathology and Oncology Section, University of Florence, 50134 Firenze, Italy
| |
Collapse
|
40
|
Ushasree MV, Jia Q, Do SG, Lee EY. New opportunities and perspectives on biosynthesis and bioactivities of secondary metabolites from Aloe vera. Biotechnol Adv 2024; 72:108325. [PMID: 38395206 DOI: 10.1016/j.biotechadv.2024.108325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/10/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024]
Abstract
Historically, the genus Aloe has been an indispensable part of both traditional and modern medicine. Decades of intensive research have unveiled the major bioactive secondary metabolites of this plant. Recent pandemic outbreaks have revitalized curiosity in aloe metabolites, as they have proven pharmacokinetic profiles and repurposable chemical space. However, the structural complexity of these metabolites has hindered scientific advances in the chemical synthesis of these compounds. Multi-omics research interventions have transformed aloe research by providing insights into the biosynthesis of many of these compounds, for example, aloesone, aloenin, noreugenin, aloin, saponins, and carotenoids. Here, we summarize the biological activities of major aloe secondary metabolites with a focus on their mechanism of action. We also highlight the recent advances in decoding the aloe metabolite biosynthetic pathways and enzymatic machinery linked with these pathways. Proof-of-concept studies on in vitro, whole-cell, and microbial synthesis of aloe compounds have also been briefed. Research initiatives on the structural modification of various aloe metabolites to expand their chemical space and activity are detailed. Further, the technological limitations, patent status, and prospects of aloe secondary metabolites in biomedicine have been discussed.
Collapse
Affiliation(s)
- Mrudulakumari Vasudevan Ushasree
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Qi Jia
- Unigen, Inc., 2121 South street suite 400 Tacoma, Washington 98405, USA
| | - Seon Gil Do
- Naturetech, Inc., 29-8, Yongjeong-gil, Chopyeong-myeon, Jincheon-gun, Chungcheongbuk-do 27858, Republic of Korea
| | - Eun Yeol Lee
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.
| |
Collapse
|
41
|
Bullard BM, McDonald SJ, Cardaci TD, VanderVeen BN, Mohammed AD, Kubinak JL, Pierre JF, Chatzistamou I, Fan D, Hofseth LJ, Murphy EA. Panaxynol improves crypt and mucosal architecture, suppresses colitis-enriched microbes, and alters the immune response to mitigate colitis. Am J Physiol Gastrointest Liver Physiol 2024; 326:G591-G606. [PMID: 38469632 PMCID: PMC11376977 DOI: 10.1152/ajpgi.00004.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/21/2024] [Accepted: 02/28/2024] [Indexed: 03/13/2024]
Abstract
Ulcerative colitis (UC) is an idiopathic inflammatory disease of the large intestine, which impacts millions worldwide. Current interventions aimed at treating UC symptoms can have off-target effects, invoking the need for alternatives that may provide similar benefits with less unintended consequences. This study builds on our initial data, which showed that panaxynol-a novel, potent, bioavailable compound found in American ginseng-can suppress disease severity in murine colitis. Here we explore the underlying mechanisms by which panaxynol improves both chronic and acute murine colitis. Fourteen-week-old C57BL/6 female mice were either given three rounds of dextran sulfate sodium (DSS) in drinking water to induce chronic colitis or one round to induce acute colitis. Vehicle or panaxynol (2.5 mg/kg) was administered via oral gavage three times per week for the study duration. Consistent with our previous findings, panaxynol significantly (P < 0.05) improved the disease activity index and endoscopic scores in both models. Using the acute model to examine potential mechanisms, we show that panaxynol significantly (P < 0.05) reduced DSS-induced crypt distortion, goblet cell loss, and mucus loss in the colon. 16S Sequencing revealed panaxynol altered microbial composition to suppress colitis-enriched genera (i.e., Enterococcus, Eubacterium, and Ruminococcus). In addition, panaxynol significantly (P < 0.05) suppressed macrophages and induced regulatory T-cells in the colonic lamina propria. The beneficial effects of panaxynol on mucosal and crypt architecture, combined with its microbial and immune-mediated effects, provide insight into the mechanisms by which panaxynol suppresses murine colitis. Overall, this data is promising for the use of panaxynol to improve colitis in the clinic.NEW & NOTEWORTHY In the current study, we report that panaxynol ameliorates chemically induced murine colitis by improving colonic crypt and mucosal architecture, suppressing colitis-enriched microbes, reducing macrophages, and promoting the differentiation of regulatory T-cells in the colonic lamina propria. This study suggests that this novel natural compound may serve as a safe and effective treatment option for colitis patients.
Collapse
Affiliation(s)
- Brooke M Bullard
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Sierra J McDonald
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Thomas D Cardaci
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Brandon N VanderVeen
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Ahmed D Mohammed
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Jason L Kubinak
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Joseph F Pierre
- Department of Nutritional Sciences, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Lorne J Hofseth
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States
| | - E Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| |
Collapse
|
42
|
Vieira SF, Reis RL, Ferreira H, Neves NM. Plant-derived bioactive compounds as key players in the modulation of immune-related conditions. PHYTOCHEMISTRY REVIEWS 2024. [DOI: 10.1007/s11101-024-09955-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 03/07/2024] [Indexed: 01/03/2025]
Abstract
AbstractThe immune system is a complex and fundamental network for organism protection. A minimal unbalance in the host defense system homeostasis can originate severe repercussions in human health. Fundamentally, immune-related diseases can arise from its compromise (immunodeficiency diseases), overactivation against itself (autoimmune diseases) or harmless substances (allergies), and failure of eliminating the harmful agent (chronic inflammation). The notable advances and achievements in the immune system diseases pathophysiology have been allowing for a dramatic improvement of the available treatments. Nevertheless, they present some drawbacks, including the inappropriate benefit/risk ratio. Therefore, there is a strong and urgent need to develop effective therapeutic strategies. Nature is a valuable source of bioactive compounds that can be explored for the development of new drugs. Particularly, plants produce a broad spectrum of secondary metabolites that can be potential prototypes for innovative therapeutic agents. This review describes the immune system and the inflammatory response and examines the current knowledge of eight plants traditionally used as immunomodulatory medicines (Boswellia serrata, Echinacea purpurea, Laurus nobilis, Lavandula angustifolia, Olea europaea, Salvia officinalis, Salvia rosmarinus, and Taraxacum officinale). Moreover, the issues responsible for possible biologic readout inconsistencies (plant species, age, selected organ, developmental stage, growth conditions, geographical location, drying methods, storage conditions, solvent of extraction, and extraction method) will also be discussed. Furthermore, a detailed list of the chemical composition and the immunomodulatory mechanism of action of the bioactive compounds of the selected plant extracts are presented. This review also includes future perspectives and proposes potential new avenues for further investigation.
Collapse
|
43
|
Dan YL, Yang YQ, Zhu DC, Bo L, Lei SF. Accelerated biological aging as a potential risk factor for rheumatoid arthritis. Int J Rheum Dis 2024; 27:e15156. [PMID: 38665050 DOI: 10.1111/1756-185x.15156] [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: 02/08/2024] [Revised: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 05/31/2024]
Abstract
OBJECTS Previous studies have suggested a potential correlation between rheumatoid arthritis (RA) and biological aging, but the intricate connections and mechanisms remain elusive. METHODS In our study, we focused on two specific measures of biological age (PhenoAge and BioAge), which are derived from clinical biomarkers. The residuals of these measures, when compared to chronological age, are defined as biological age accelerations (BAAs). Utilizing the extensive UK Biobank dataset along with various genetic datasets, we conducted a thorough assessment of the relationship between BAAs and RA at both the individual and aggregate levels. RESULTS Our observational studies revealed positive correlations between the two BAAs and the risk of developing both RA and seropositive RA. Furthermore, the genetic risk score (GRS) for PhenoAgeAccel was associated with an increased risk of RA and seropositive RA. Linkage disequilibrium score regression (LDSC) analysis further supported these findings, revealing a positive genetic correlation between PhenoAgeAccel and RA. PLACO analysis identified 38 lead pleiotropic single nucleotide polymorphisms linked to 301 genes, providing valuable insights into the potential mechanisms connecting PhenoAgeAccel and RA. CONCLUSION In summary, our study has successfully revealed a positive correlation between accelerated biological aging, as measured by BAAs, and the susceptibility to RA.
Collapse
Affiliation(s)
- Yi-Lin Dan
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu, China
| | - Yi-Qun Yang
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu, China
| | - Dong-Cheng Zhu
- Department of Orthopedics, Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Suqian, Jiangsu, China
| | - Lin Bo
- Department of Rheumatology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shu-Feng Lei
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
44
|
Dao TNP, Onikanni SA, Fadaka AO, Sibuyi NRS, Le MH, Chang HH. Phytotherapeutic potential of compounds identified from fractionated extracts of Morus alba L., as an inhibitor of interleukin-6 in the treatment of rheumatoid arthritis: computational approach. J Biomol Struct Dyn 2024:1-14. [PMID: 38525928 DOI: 10.1080/07391102.2024.2330713] [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: 10/03/2023] [Accepted: 03/06/2024] [Indexed: 03/26/2024]
Abstract
The presence of HLA-DRB1 alleles that encode critical points associated with environmental interactions is associated with increased risk of rheumatoid arthritis caused by anti-citrullinated protein antibodies. Therefore, interleukin-6 (IL-6), a multifunctional cytokine that controls both local and systemic acute inflammatory responses through its ability to induce a phase response, plays a serious role. Its overexpression leads to pathological challenges such as rheumatoid arthritis and menopausal osteoporosis. However, targeting the IL-6 receptor and its region could be the major step in controlling the overexpression of this cytokine for therapeutic importance. Therefore, our research explored the computational insight needed to investigate the anti-RFA potential of phytochemicals from fractionated extracts of Morus alba L. against receptors, which have been implicated as druggable targets for the treatment of rheumatoid arthritis. In this study, fifty-nine (59) previously isolated and characterized phytochemicals from M. alba L. were identified from the literature and retrieved from the PubChem database. In silico screening was used to assess the mode of action of these phytochemicals from M. alba L. against receptors that may serve as therapeutic targets for rheumatoid arthritis. Molecular docking studies, toxicity prediction, drug visualization and molecular dynamics simulation (MD) of the ligands together with the receptor-identified target were carried out using the Schrodinger Molecular Drug Discovery Suite. The findings indicated that a selected group of ligands displayed significant binding strength to specific amino acid residues, revealing an important link between the building blocks of proteins (amino acids) and ligands at the inhibitor binding site through traditional chemical interactions, such as interactions between hydrophobic and hydrogen bonds. The binding affinities of the receptors were carefully checked via comparison with those of the approved ligands, and the results suggested structural and functional changes in the lead compounds. Therefore, the bioactive component from M. alba L. could be a lead foot interleukin-6 (IL-6) inhibitor and could be a promising lead compound for the treatment of rheumatoid arthritis and related challenges.
Collapse
Affiliation(s)
- Tran Nhat Phong Dao
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan (ROC)
- Faculty of Traditional Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Sunday Amos Onikanni
- College of Medicine, Graduate Institute of Biomedical Sciences, China Medical University, Taiwan (ROC)
- Department of Chemical Sciences, Biochemistry Unit, Afe-Babalola University, Ado-Ekiti, Nigeria
| | | | - Nicole Remaliah Samantha Sibuyi
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, University of the Western Cape, Bellville, South Africa
| | - Minh Hoang Le
- Faculty of Traditional Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Hen-Hong Chang
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan (ROC)
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan (ROC)
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan (ROC)
| |
Collapse
|
45
|
Liu Z, Nong K, Qin X, Fang X, Zhang B, Chen W, Wang Z, Wu Y, Shi H, Wang X, Liu Y, Guan Q, Zhang H. The antimicrobial peptide Abaecin alleviates colitis in mice by regulating inflammatory signaling pathways and intestinal microbial composition. Peptides 2024; 173:171154. [PMID: 38242174 DOI: 10.1016/j.peptides.2024.171154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/13/2024] [Accepted: 01/14/2024] [Indexed: 01/21/2024]
Abstract
Abaecin is a natural antimicrobial peptide (AMP) rich in proline from bees. It is an important part of the innate humoral immunity of bees and has broad-spectrum antibacterial ability. This study aimed to determine the effect of Abaecin on dextran sulfate sodium (DSS) -induced ulcerative colitis (UC) in mice and to explore its related mechanisms. Twenty-four mice with similar body weight were randomly divided into 4 groups. 2.5% DSS was added to drinking water to induce colitis in mice. Abaecin and PBS were administered rectally on the third, fifth, and seventh days of the experimental period. The results showed that Abaecin significantly alleviated histological damage and intestinal mucosal barrier damage caused by colitis in mice, reduced the concentration of pro-inflammatory cytokines IL-1β, IL-6, TNF-α, IFN-γ, and the phosphorylation of NF-κB / MAPK inflammatory signaling pathway proteins, and improved the composition of intestinal microorganisms. These findings suggest that Abaecin may have potential prospects for the treatment of UC.
Collapse
Affiliation(s)
- Zhineng Liu
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Keyi Nong
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Xinyun Qin
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Xin Fang
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Bin Zhang
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Wanyan Chen
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Zihan Wang
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Yijia Wu
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Huiyu Shi
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Xuemei Wang
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Youming Liu
- Yibin Academy of Agricultural Sciences, Yibin 644600, China
| | - Qingfeng Guan
- College of Life and Health, Hainan University, Haikou 570228, China
| | - Haiwen Zhang
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China.
| |
Collapse
|
46
|
Wang N, Li F, Du J, Hao J, Wang X, Hou Y, Luo Z. Quercetin Protects Against Global Cerebral ischemia‒reperfusion Injury by Inhibiting Microglial Activation and Polarization. J Inflamm Res 2024; 17:1281-1293. [PMID: 38434580 PMCID: PMC10906675 DOI: 10.2147/jir.s448620] [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: 11/10/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
Background This study aims to investigate the protective effect of quercetin against global cerebral ischemia‒reperfusion (GCI/R) injury in rats and elucidate the underlying mechanism. Methods A GCI/R injury rat model was established using a four-vessel occlusion (4-VO) method. An oxygen-glucose deprivation/reoxygenation (OGD/R) injury model was induced in BV2 cells. The extent of injury was assessed by evaluating neurological deficit scores (NDS) and brain water content and conducting behavioral tests. Pathomorphological changes in the prefrontal cortex were examined. Additionally, the study measured the levels of inflammatory cytokines, the degree of microglial activation and polarization, and the protein expression of Toll-like receptor 4 (TLR4) and TIR-domain-containing adaptor inducing interferon-β (TRIF). Results Quercetin pretreatment significantly ameliorated neurological impairment, improved learning and memory abilities, and reduced anxiety in rats subjected to GCI/R injury. Furthermore, quercetin administration effectively mitigated neuronal injury and brain edema. Notably, it suppressed microglial activation and hindered polarization toward the M1 phenotype. Simultaneously, quercetin downregulated the expression of TLR4 and TRIF proteins and attenuated the release of IL-1β and TNF-α. Conclusion This study highlights the novel therapeutic potential of quercetin in alleviating GCI/R injury. Quercetin demonstrates its neuroprotective effects by inhibiting neuroinflammation and microglial activation while impeding their transformation into the M1 phenotype through modulation of the TLR4/TRIF pathway.
Collapse
Affiliation(s)
- Naigeng Wang
- Department of Anesthesiology, Honghui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Fei Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, People’s Republic of China
| | - Jing Du
- Second Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, People’s Republic of China
| | - Jianhong Hao
- Department of Anesthesiology, Honghui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Xin Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, People’s Republic of China
| | - Yueru Hou
- Second Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, People’s Republic of China
| | - Zhenguo Luo
- Department of Anesthesiology, Honghui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
47
|
Salvan da Rosa J, Bramorski Mohr ET, Lubschinski TL, Vieira GN, Rossa TA, Mandolesi Sá M, Dalmarco EM. Interference in Macrophage Balance (M1/M2): The Mechanism of Action Responsible for the Anti-Inflammatory Effect of a Fluorophenyl-Substituted Imidazole. Mediators Inflamm 2024; 2024:9528976. [PMID: 38405621 PMCID: PMC10894048 DOI: 10.1155/2024/9528976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/19/2024] [Accepted: 02/03/2024] [Indexed: 02/27/2024] Open
Abstract
Traditionally, the treatment of inflammatory conditions has focused on the inhibition of inflammatory mediator production; however, many conditions are refractory to this classical approach. Recently, an alternative has been presented by researchers to solve this problem: The immunomodulation of cells closely related to inflammation. Hence, macrophages, a critical key in both innate and acquired immunity, have been presented as an alternative target for the development of new medicines. In this work, we tested the fluorophenyl-imidazole for its anti-inflammatory activity and possible immunomodulatory effect on RAW 264.7 macrophages. We also evaluated the anti-inflammatory effect of the compound, and the macrophage repolarization to M2 was confirmed by the ability of the compound to reduce the M1 markers TNF-α, IL-6, MCP-1, IL-12p70, IFN-γ, and TLR4, the high levels of p65 phosphorylated, iNOS and COX-2 mRNA expression, and the fact that the compound was not able to induce the production of M1 markers when used in macrophages without lipopolysaccharide (LPS) stimulation. Moreover, fluorophenyl-imidazole had the ability to increase the M2 markers IL-4, IL-13, CD206, apoptosis and phagocytosis levels, arginase-1, and FIZZ-1 mRNA expression before LPS stimulation. Similarly, it was also able to induce the production of these same M2 markers in macrophages without being induced with LPS. These results reinforce the affirmation that the fluorophenyl-imidazole has an important anti-inflammatory effect and demonstrates that this effect is due to immunomodulatory activity, having the ability to trigger a repolarization of macrophages from M1 to M2a. These facts suggest that this molecule could be used as an alternative scaffold for the development of a new medicine to treat inflammatory conditions, where the anti-inflammatory and proregenerative properties of M2a macrophages are desired.
Collapse
Affiliation(s)
- Julia Salvan da Rosa
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Eduarda Talita Bramorski Mohr
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Tainá Larissa Lubschinski
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Guilherme Nicácio Vieira
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Thais Andreia Rossa
- Department of Chemistry, Center for Physical and Mathematical Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Marcus Mandolesi Sá
- Department of Chemistry, Center for Physical and Mathematical Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Eduardo Monguilhott Dalmarco
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| |
Collapse
|
48
|
Zhao Q, Wu Y, Wu X, Liu M, Nan L. Single-cell transcriptome analysis reveals keratinocyte subpopulations contributing to psoriasis in corneum and granular layer. Skin Res Technol 2024; 30:e13572. [PMID: 38279596 PMCID: PMC10818132 DOI: 10.1111/srt.13572] [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: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Psoriasis is a chronic, inflammatory skin disease that is common and relapses easily. While the importance of keratinocyte proliferation in psoriasis development is well-documented, the specific functional subpopulations of epidermal keratinocytes associated with this disease remain enigmatic. MATERIALS AND METHODS Therefore, in our analysis of single-cell transcriptome data from both normal and psoriatic skin tissues, we observed significant increases in certain keratinocytes in the stratum corneum (KC) and stratum granulosum (KG) within psoriatic skin. Furthermore, we identified upregulated expression of specific secreted factors known to promote inflammatory responses. Additionally, we conducted a KEGG pathway enrichment analysis on these identified subsets. RESULTS In the stratum corneum, the expression of FTL was upregulated in HIST1H1C+ KC. S100P+ KC displayed a significant increase in the expression of both S100P and S100A10, whereas PRR9+ KC showed upregulated expression of DEFB4B, S100A8, and S100A12. SLURP1+ KC was characterized by elevated expression levels of IL-36G, SLURP1, and S100A12. Meanwhile, in the stratum granulosum, KRT1+ KG highly expressed SLURP1, S100A7, S100A8, and S100A9, while DEFB4B expression was upregulated in PI3+ KG. Our findings indicated that subsets within the stratum corneum primarily participate in pathways related to MAPK, NOD-like receptors, HIF-1, cell senescence, and other crucial processes. In contrast, subsets in the stratum granulosum were predominantly associated with pathways involving MAPK, NOD-like receptors, HIF-1, Hippo, mTOR, and IL-17. CONCLUSION These findings not only uncover the keratinocyte subsets linked to psoriasis but also unveil the molecular mechanisms and related signaling pathways that drive psoriasis development. This knowledge opens new horizons for the development of innovative clinical treatment strategies for psoriasis.
Collapse
Affiliation(s)
- Qianya Zhao
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
- Department of DermatologyGansu Provincial HospitalLanzhouGansuChina
| | - Yan Wu
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
| | - Xianwei Wu
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
- Department of DermatologyGansu Provincial HospitalLanzhouGansuChina
| | - Meng Liu
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
- Department of DermatologyGansu Provincial HospitalLanzhouGansuChina
| | - Lisheng Nan
- First Clinical Medical CollegeGansu University of Chinese MedicineLanzhouGansuChina
- Department of DermatologyGansu Provincial HospitalLanzhouGansuChina
| |
Collapse
|
49
|
Li Z, Li X, Shi P, Li P, Fu Y, Tan G, Zhou J, Zeng J, Huang P. Modulation of Acute Intestinal Inflammation by Dandelion Polysaccharides: An In-Depth Analysis of Antioxidative, Anti-Inflammatory Effects and Gut Microbiota Regulation. Int J Mol Sci 2024; 25:1429. [PMID: 38338707 PMCID: PMC10855136 DOI: 10.3390/ijms25031429] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Acute colitis is a complex disease that can lead to dysregulation of the gut flora, inducing more complex parenteral diseases. Dandelion polysaccharides (DPSs) may have potential preventive and therapeutic effects on enteritis. In this study, LPS was used to induce enteritis and VC was used as a positive drug control to explore the preventive and therapeutic effects of DPS on enteritis. The results showed that DPS could repair the intestinal barrier, down-regulate the expression of TNF-α, IL-6, IL-1β, and other pro-inflammatory factors, up-regulate the expression of IL-22 anti-inflammatory factor, improve the antioxidant capacity of the body, and improve the structure of intestinal flora. It is proved that DPS can effectively prevent and treat LPS-induced acute enteritis and play a positive role in promoting intestinal health.
Collapse
Affiliation(s)
- Zhu Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Xinyao Li
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Panpan Shi
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Pingping Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yue Fu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Guifeng Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Junjuan Zhou
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Jianguo Zeng
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Peng Huang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
50
|
Al-Naemi HA, Alasmar RM, Al-Ghanim K. Alcoholic extracts of Teucrium polium exhibit remarkable anti-inflammatory activity: In vivo study. BIOMOLECULES & BIOMEDICINE 2024; 24:82-88. [PMID: 37289437 PMCID: PMC10787626 DOI: 10.17305/bb.2023.9239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/09/2023]
Abstract
Teucrium polium (germander, Lamiaceae) is a local plant in Qatar that has been used in folk medicine to treat numerous illnesses. It is known for its antioxidant, analgesic, anticancer, and antibacterial activities. This study aimed to evaluate the anti-inflammatory activity of Teucrium polium (TP) extract by α-carrageen-induced paw edema in adult Sprague Dawley rats. The animals were randomly grouped into control, acute inflammation, and plant extract groups. Acute inflammation was induced by a sub-plantar injection of 100 µL of 1% α-carrageenan into the rat's right hind paw. Three different doses of the ethanolic extract of TP were tested at different time periods (1, 3, and 5 hours). All doses of the TP ethanolic extract showed significant inhibition of α-carrageenan-induced rat paw edema in a dose-dependent manner in both early and late phases of edema formation. The size of the α-carrageen induced paw edema was significantly reduced one, three, and five hours after TP extract injection compared to the acute inflammation group. This inhibition was accompanied by high expression of interleukin 10 (IL-10) and low expression of monocyte chemoattractant protein 1 (MCP-1), IL-1β and tumor necrosis factor alpha (TNF-α). The results indicated that the ethanolic extracts of TP possess significant anti-inflammatory and potential pharmaceutical properties.
Collapse
Affiliation(s)
- Hamda A Al-Naemi
- Laboratory Animal Research Centre, Qatar University, Doha, Qatar
- Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar
| | | | - Kaltham Al-Ghanim
- Social and Economic Survey Research Institute (SESRI), Qatar University, Doha, Qatar
| |
Collapse
|