1
|
Nicholls LA, Zeile KA, Scotto LD, Ryznar RJ. Timing of dietary effects on the epigenome and their potential protective effects against toxins. Epigenetics 2025; 20:2451495. [PMID: 39825851 DOI: 10.1080/15592294.2025.2451495] [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: 04/15/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/20/2025] Open
Abstract
Exposure to toxins causes lasting damaging effects on the body. Numerous studies in humans and animals suggest that diet has the potential to modify the epigenome and these modifications can be inherited transgenerationally, but few studies investigate how diet can protect against negative effects of toxins. Potential evidence in the primary literature supports that caloric restriction, high-fat diets, high protein-to-carbohydrate ratios, and dietary supplementation protect against environmental toxins and strengthen these effects on their offspring's epigenome. Most notably, the timing when dietary interventions are given - during a parent's early development, pregnancy, and/or lifetime - result in similar transgenerational epigenetic durations. This implies the existence of multiple opportunities to strategically fortify the epigenome. This narrative review explores how to best utilize dietary modifications to modify the epigenome to protect future generations against negative health effects of persistent environmental toxins. Furthermore, by suggesting an ideal diet with specific micronutrients, macronutrients, and food groups, epigenetics can play a key role in the field of preventive medicine. Based on these findings, longitudinal research should be conducted to determine if a high protein, high-fat, and low-carbohydrate diet during a mother's puberty or pregnancy can epigenetically protect against alcohol, tobacco smoke, and air pollution across multiple generations.
Collapse
Affiliation(s)
- Lynnea A Nicholls
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| | - Kendall A Zeile
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| | - London D Scotto
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| | - Rebecca J Ryznar
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
- Department of Biomedical Sciences, Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| |
Collapse
|
2
|
Ioannidis M, Tjepkema J, Uitbeijerse MRP, van den Bogaart G. Immunomodulatory effects of 4-hydroxynonenal. Redox Biol 2025; 85:103719. [PMID: 40489926 DOI: 10.1016/j.redox.2025.103719] [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: 05/26/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025] Open
Abstract
The reactive aldehyde 4-hydroxy-2-nonenal (4-HNE) is a byproduct of lipid peroxidation driven by reactive oxygen species (ROS). 4-HNE covalently binds to macromolecules such as proteins, altering their functions. While 4-HNE is implicated in various ROS-related pathologies, its impact on the immune system remains incompletely understood. This review explores how 4-HNE influences molecular mechanisms involved in inflammation and immune cell functions. 4-HNE modulates inflammation through the interaction with several signaling pathways, including nuclear factor kappa-light-chain enhancement of activated B cells (NF-κB), nuclear factor erythroid 2-related factor (Nrf2), mitogen-activated protein kinases (MAPK), toll-like receptor (TLR) 4, and stimulator of interferon genes (STING), thereby affecting immune responses and modulating cytokine production and inflammasome activation. However, its effects are complex, exhibiting both pro- and anti-inflammatory properties depending on dose and cell type. This review highlights the multiple mechanisms by which 4-HNE modulates the immune cells' responses.
Collapse
Affiliation(s)
- Melina Ioannidis
- Department of Molecular Immunology, Groningen Biomolecular Science and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747AG, Groningen, the Netherlands
| | - Johanna Tjepkema
- Department of Molecular Immunology, Groningen Biomolecular Science and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747AG, Groningen, the Netherlands
| | - Michael R P Uitbeijerse
- Department of Molecular Immunology, Groningen Biomolecular Science and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747AG, Groningen, the Netherlands
| | - Geert van den Bogaart
- Department of Molecular Immunology, Groningen Biomolecular Science and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747AG, Groningen, the Netherlands; Department of Medical Biology and Pathology, University Medical Centre Groningen, Groningen, the Netherlands.
| |
Collapse
|
3
|
Georges HM, Fischer AC, Abrahams VM. Intermediate Signaling Mechanisms Regulating Human Fetal Membrane Responses to Gram-Positive Bacterial Peptidoglycan. Am J Reprod Immunol 2025; 93:e70090. [PMID: 40435036 PMCID: PMC12124414 DOI: 10.1111/aji.70090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/19/2025] [Accepted: 04/29/2025] [Indexed: 06/01/2025] Open
Abstract
PROBLEM Chorioamnionitis and preterm birth are leading causes of neonatal morbidity and mortality. Despite ongoing research, the signaling pathways involved in the pathogenesis of chorioamnionitis-inflammation of the fetal membranes (FM)-are not well understood. Previously, we reported that FMs utilize miR-146a-3p as an endogenously produced danger signal to sequentially activate Toll-like receptor (TLR) 8 and subsequent inflammation following lipopolysaccharide stimulation of TLR4. In this current study, following stimulation of fetal membrane explants by the TLR2 agonist peptidoglycan (PDG), we investigated sequential microRNA-activation of TLR8, intermediate signaling pathways NFκB and MAPK (p38, ERK), and their effects on inflammation and mediators of membrane weakening. METHOD OF STUDY Human FMs explants were treated with or without PDG in the presence or absence of inhibitors to TLR7, TLR8, p65 NFκB, p38 MAPK, or ERK. Culture supernatants were measured for secreted factors by ELISA, tissue RNA was measured for TLR7/8-activating miRs by RT-qPCR, and tissue protein was measured for phosphorylated proteins by Western blot. RESULTS PDG-treated FMs produced elevated levels of TLR8-activating miR-146a-3p in a p65 NFκB-dependent manner. PDG-treated FMs produced elevated levels of the pro-inflammatory cytokine IL-1β, the neutrophil recruiting chemokine IL-8, and membrane weakening MMP1, MMP9, and PGE2 in a TLR8-dependent manner. Except for MMP9, this inflammatory and membrane weakening response to PDG was dependent upon p65 NFκB, p38 MAPK, and ERK signaling. CONCLUSIONS This study gives new insight into the molecular mechanisms involved in FM responses to Gram-positive bacteria and into the pathogenesis of chorioamnionitis.
Collapse
Affiliation(s)
- Hanah M Georges
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Abigail C Fischer
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Vikki M Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
4
|
Li X, Yao L, Cho YC, Lee DY, Cho N, Yoo G, Choi SY, Yoon S, Lim JS. Alleviation of LPS-induced oxidative stress and inflammation by lesbicoumestan (7) via the increase of Nrf2 expression in mouse Kupffer cells. Toxicol Appl Pharmacol 2025; 501:117405. [PMID: 40414571 DOI: 10.1016/j.taap.2025.117405] [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: 12/20/2024] [Revised: 05/18/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Lesbicoumestans are a class of bioactive compounds isolated from the roots of Lespedeza bicolor (L. bicolor). These compounds have been reported to exhibit anticancer and antiproliferative activities. In this study, our primary focus was to examine the antioxidant capabilities of lesbicoumestan (7) (LC-7), a newly isolated coumestan from roots of L. bicolor, using lipopolysaccharide (LPS)-stimulated immortalized mouse Kupffer cells (ImKCs) as the experimental model. The investigation revealed that LC-7 played a pivotal role in inhibiting the production of reactive oxygen species (ROS). Additionally, LC-7 effectively restored the imbalanced glutathione(GSH)/glutathione disulfide ratio and enhanced the activity of glutathione peroxidase (GPx) following cellular exposure to LPS. Moreover, our investigation revealed that LC-7 exhibited the capacity to enhance the expression of heme oxygenase-1 (HO-1), leading to inhibition of nitric oxide (NO) and proinflammatory cytokines production induced by LPS. Notably, LC-7 did not significantly impact the nuclear factor kappa B cells (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. Intriguingly, LC-7 modulated the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway by direct interaction with Kelch-like-ECH-associated protein 1 (Keap1). These findings suggest that LC-7 possesses antioxidant and anti-inflammatory properties in LPS-stimulated ImKCs by upregulating Nrf2 expression.
Collapse
Affiliation(s)
- Xiangying Li
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea
| | - Lulu Yao
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea
| | - Young-Chang Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea
| | - Da Young Lee
- R&D Center, CUOME BIO Co., Ltd., Sandan-gil, Hwasun-eup, Hwasun-gun, Jeollanam-do 58141, Republic of Korea
| | - Namki Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea
| | - Guijae Yoo
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-Gun, Jeollabuk-do, Republic of Korea
| | - Sang Yoon Choi
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-Gun, Jeollabuk-do, Republic of Korea
| | - Somy Yoon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea.
| | - Jae Sung Lim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea.
| |
Collapse
|
5
|
Kim JH, Che DN, Park JH, Shin JY, Jang SI, Cho BO. Anti-inflammatory effects of Elsholtzia ciliata extract on Poly I:C-treated RAW264.7 cells. JOURNAL OF ETHNOPHARMACOLOGY 2025:120026. [PMID: 40412777 DOI: 10.1016/j.jep.2025.120026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 05/20/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Inflammation is a vital biological response to noxious stimuli, including physical injury and pathogenic infection, and involves immune cells and various inflammatory mediators, limiting cell damage and eliminating pathogens. Although essential for healing, inflammation can cause symptoms, such as fever, swelling, pain, and itching, potentially reducing quality of life. Elsholtzia ciliata used in traditional medicine has numerous medicinal characteristics such as antiviral, antibacterial, antipyretic, diaphoretic, carminative, astringent, and diuretic effects. AIM OF THE STUDY This study aimed to evaluate the anti-inflammatory properties of E. ciliata extract (ECE) in RAW264.7 cells treated with polyinosinic polycytidylic acid (Poly I:C). MATERIALS AND METHODS PGE2, IL-1β, TNF-α, IFN-β, and IL-6 levels were quantified by ELISA and/or real-time PCR. COX-2 and iNOS expression was analyzed using western blotting and real-time PCR. Phosphorylation and expression levels of signaling proteins, including AKT, IRF3, TBK1, STAT1, MAPKs, IκB, and IκK were analyzed using western blotting. The active substance of ECE was determined using high-performance liquid chromatography-mass spectrometry (HPLC-MS). RESULTS Our detections revealed that ECE inhibited the levels of nitric oxide and central inflammatory mediators, such as iNOS and COX-2. Furthermore, ECE downregulated the expression of pro-inflammatory cytokines, including PGE2, IL-1β, TNF-α, IFN-β, and IL-6. Additionally, ECE inhibited the phosphorylation of several cell signaling pathways, including AKT, TBK1/IRF3, MAPK, and NF-κB, in Poly I:C-treated RAW264.7 cells. CONCLUSIONS These results highlight E. ciliata as a candidate for mitigating virus-induced inflammation, providing valuable insights into its use in the development of new anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Jang Hoon Kim
- Department of Herbal Crop Research, National Institute of Horticultural & Herbal Science, RDA, 92, Bisan-ro, Soi-myeon, Eumseong-gun, Chungbuk 27709, Republic of Korea.
| | - Denis Nchang Che
- Immunology Laboratory, Seoul Songdo Colorectal Hospital, 78, Dasan-ro, Jung-gu, Seoul 04597, Republic of Korea.
| | - Ji Hyeon Park
- Institute of Health Science, Jeonju University, 303, Cheonjam-ro, Wansan-gu, Jeonju-si, Jeonbuk-do 55069, Republic of Korea.
| | - Jae Young Shin
- Institute of Health Science, Jeonju University, 303, Cheonjam-ro, Wansan-gu, Jeonju-si, Jeonbuk-do 55069, Republic of Korea.
| | - Seon Il Jang
- Institute of Health Science, Jeonju University, 303, Cheonjam-ro, Wansan-gu, Jeonju-si, Jeonbuk-do 55069, Republic of Korea; Department of Health Management, Jeonju University, 303, Cheonjam-ro, Wansan-gu, Jeonju-si, Jeonbuk-do 55069, Republic of Korea.
| | - Byoung Ok Cho
- Institute of Health Science, Jeonju University, 303, Cheonjam-ro, Wansan-gu, Jeonju-si, Jeonbuk-do 55069, Republic of Korea.
| |
Collapse
|
6
|
Mohiti S, Alizadeh E, Bisgaard LS, Ebrahimi-Mameghani M, Christoffersen C. The AhR/P38 MAPK pathway mediates kynurenine-induced cardiomyocyte damage: The dual role of resveratrol in apoptosis and autophagy. Biomed Pharmacother 2025; 186:118015. [PMID: 40153993 DOI: 10.1016/j.biopha.2025.118015] [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: 01/20/2025] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025] Open
Abstract
Chronic kidney disease increases the risk of cardiovascular disease, partly due to uremic toxins, such as Kynurenine (KYN). While KYN contributes to tissue damage, its role in cardiomyocyte apoptosis and autophagy remains unclear. Resveratrol (RSV) can protect against oxidative stress and inflammation, whereas its specific effects on KYN-induced cardiomyopathy are less understood. This study aimed to investigate the role of KYN in cardiomyocyte apoptosis and autophagy and examine the protective effects of RSV against KYN-induced damage. H9C2 cardiomyocytes were cultured and treated with KYN in presence or absence of RSV or inhibitors of the AhR/Src/MAPKs pathway. Cell viability, apoptosis, mitochondrial membrane potential, and autophagy were assessed using MTT, TUNEL, JC-1, and autophagy detection assays. KYN induced apoptosis, and autophagy in H9C2 cells. RSV pretreatment reduced apoptosis but enhanced autophagy in KYN-treated cells. Inhibiting autophagy or blocking apoptosis, increased KYN-induced apoptosis and autophagy, respectively. Additionally, KYN treatment enhanced AhR activation and the phosphorylation of Src and MAPKs proteins, whereas RSV pretreatment decreased AhR activation and ERK phosphorylation. Inhibitors of p38 MAPK and JNK reduced expression of apoptotic proteins. AhR inhibition also reduced the phosphorylation of p38 MAPK and expression of apoptotic proteins while it enhanced autophagy-related protein expression in KYN treated H9C2 cells. In conclusion, our findings suggest that KYN induces cardiomyocyte apoptosis via the AhR/p38 MAPK pathway whereas RSV can protect against the KYN-induced apoptosis while promoting autophagy. Given the high cardiovascular risk in CKD patients, these findings provide in-sight into potential therapeutic strategies targeting KYN-induced cardiomyopathy.
Collapse
Affiliation(s)
- Sara Mohiti
- Student Research Committee, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Line S Bisgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Denmark
| | - Mehrangiz Ebrahimi-Mameghani
- Nutrition Research Center, Department of Biochemistry and Diet Therapy, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
7
|
Kobayashi H, Shigetomi H, Imanaka S. Reassessing the Role of Tissue Factor Pathway Inhibitor 2 in Neoplastic and Non-Neoplastic Lesions. Cancers (Basel) 2025; 17:1447. [PMID: 40361374 PMCID: PMC12071115 DOI: 10.3390/cancers17091447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/20/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
OBJECTIVES Tissue factor pathway inhibitor 2 (TFPI2) is a serine protease inhibitor that suppresses tumors by preventing extracellular matrix degradation and invasion. In many malignancies, the TFPI2 promoter hypermethylation silences its transcription, increasing tumor aggressiveness. However, TFPI2 paradoxically facilitates tumor progression in certain malignancies. Elevated circulating TFPI2 levels correlate with increased cancer aggressiveness and poor prognosis in ovarian, endometrial, and renal cell carcinoma, though the mechanisms underlying its tumor-promoting effects remain unclear. This review consolidates recent findings on TFPI2 regulation, its downstream targets in cellular homeostasis, and its prognostic significance. Additionally, we reassess TFPI2's role in tumorigenesis, particularly in clear cell carcinoma, as well as in chronic inflammation. METHODS A comprehensive literature search was performed in PubMed and Google Scholar without time restriction. RESULTS TFPI2 expression is tightly regulated by transcription factors, signaling molecules, growth factors, cytokines, and epigenetic modification. TFPI2 regulates cell proliferation, inflammation, and extracellular matrix (ECM) remodeling, preserving tissue homeostasis. TFPI2 also regulates vascular endothelial and smooth muscle cell proliferation, key elements of the tumor microenvironment (TME). In the nucleus, it may modulate transcription factors to influence tumor-associated macrophage (TAM) polarization, facilitating cancer invasion. Its expression may be shaped by interactions between cancer cells and TAM activation. Beyond tumorigenesis, TFPI2 contributes to both inflammatory progression and resolution in diabetes, atherosclerosis, and preeclampsia. CONCLUSIONS TFPI2 may interact with TAMs and inflammatory cells to regulate cell proliferation and inflammation, maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms. Clinic MayOne, 871-1 Shijo-cho, Kashihara 634-0813, Japan;
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan;
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan;
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, 3-3-17 Kitatomigaoka-cho, Nara 634-0001, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms. Clinic MayOne, 871-1 Shijo-cho, Kashihara 634-0813, Japan;
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan;
| |
Collapse
|
8
|
Panchalingam S, Kasivelu G. Harnessing marine bioactive compounds: In silico insights into therapeutics for rheumatoid arthritis and major depressive disorder. Comput Biol Chem 2025; 118:108452. [PMID: 40222053 DOI: 10.1016/j.compbiolchem.2025.108452] [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: 01/22/2025] [Revised: 03/17/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025]
Abstract
The quest for the discovery of novel therapeutic agents' increases day by day owing to the increased incidence of drug-resistant infections, chronic diseases, and a need for discovering novel treatments. Conventionally, the sources for molecules of drugs have remained from terrestrial plants and microorganisms, yet the chemical adaptability of marine organisms presents something very unique in chemical terms and remains an uncharted frontier. Marine bioactive compounds-chemicals produced by marine organisms that have positive health impacts on humans-attract particular interest due to their pharmaceutical potential. Marine organisms range from macroalgae (seaweeds), microalgae, and sponges to molluscs, echinoderms, and fish. Each of these categories generates a variety of bioactive compounds that have unique biochemical properties. Many marine-derived compounds have exhibited strong antimicrobial activity, anticancer activity and neuroprotective effects. Despite the enormous potential of marine bioactive compounds in drug discovery, several challenges like Accessibility and Sustainability, Complexity of Marine Compounds, and Regulation and Approval act as bottlenecks in taking them from the lab to the clinic. It is an imperative task to tackle these challenges for a complete development of marine pharmacopoeia. This review emphasizes on the possible application of chemicals emanating from marine sources as lead molecules for the prevention of major depressive disorder and rheumatoid arthritis.
Collapse
Affiliation(s)
- Santhiya Panchalingam
- Centre for Ocean Research, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamil Nadu 600119, India
| | - Govindaraju Kasivelu
- Centre for Ocean Research, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamil Nadu 600119, India.
| |
Collapse
|
9
|
Giovannetti F, Pontecorvi P, Megiorni F, Armentano M, Alisi L, Romano E, Marchese C, Lambiase A, Bruscolini A. Conjunctival MicroRNA Expression Signature in Primary Sjögren's Syndrome Dry Eye: A NanoString-Based Bioinformatic Analysis. Invest Ophthalmol Vis Sci 2025; 66:80. [PMID: 40298889 PMCID: PMC12045114 DOI: 10.1167/iovs.66.4.80] [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: 12/23/2024] [Accepted: 04/03/2025] [Indexed: 04/30/2025] Open
Abstract
Purpose Primary Sjögren's syndrome (pSS) is a chronic autoimmune disease characterized by inflammation and tissue destruction of the salivary and lacrimal glands, leading to sicca symptoms. Dysregulation of microRNAs (miRNAs), key post-transcriptional regulators, has been implicated in pSS, but their role in conjunctival epithelial cells (CECs) remains unclear. This study aimed to identify altered miRNA expression patterns in CEC from patients with pSS and their potential involvement in pSS pathogenesis. Methods CEC samples were collected from six patients with pSS and six healthy controls (HCs) using nylon-tipped swabs. The miRNA expression was profiled using the NanoString nCounter system with minimal RNA input. Differentially expressed (DE) miRNAs were identified via ROSALIND software, and bioinformatics tools (miRNet and miRTargetLink) were applied to construct miRNA-centric networks, predict target genes, and perform pathway enrichment analysis. Results We identified 11 DE miRNAs in patients with pSS compared with the HCs. Key miRNAs, including hsa-miR-548j-3p and hsa-miR-219b-3p, are central to immune and inflammatory regulation pathways. Pathway enrichment analysis highlighted their involvement in processes such as immune cell regulation, inflammatory signaling, and glandular damage. Dysregulated miRNAs modulate key targets, like TNFAIP3, IL6R, IFNAR1, IL7, and ICOSLG, suggesting their potential role in pSS pathogenesis. Conclusions This study underscores the potential of miRNAs as biomarkers and therapeutic targets in pSS-associated dry eye disease. Despite limitations like small sample size and reliance on in silico predictions, our findings provide valuable insights into miRNA-mediated regulation of immune responses and inflammation, paving the way for future diagnostic and therapeutic advancements.
Collapse
Affiliation(s)
| | - Paola Pontecorvi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Marta Armentano
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Ludovico Alisi
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Enrico Romano
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Alice Bruscolini
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
10
|
Liang Y, Liu T, Wang D, Liu Q. Exploring the antimicrobial, anti-inflammatory, antioxidant, and immunomodulatory properties of Chrysanthemum morifolium and Chrysanthemum indicum: a narrow review. Front Pharmacol 2025; 16:1538311. [PMID: 40176916 PMCID: PMC11963160 DOI: 10.3389/fphar.2025.1538311] [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: 12/02/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Infectious diseases continue to be a major global public health concern, which is exacerbated by the increasing prevalence of antimicrobial resistance. This review investigates the potential of herbal medicine, particularly Chrysanthemum morifolium (CM) and Chrysanthemum indicum (CI), in addressing these challenges. Both herbs, documented in traditional Chinese medicine (TCM) and the Pharmacopoeia of the People's Republic of China (2020 edition), are renowned for their heat-clearing and detoxifying properties. Phytochemical studies reveal that these botanicals contain diverse bioactive compounds, including flavonoids, terpenoids, and phenylpropanoids, which exhibit antimicrobial, anti-inflammatory, and antioxidant properties, among other effects. Comparative analysis reveals that distinct compound profiles and differential concentrations of core phytochemicals between CM and CI may lead to differentiated therapeutic advantages in anti-infective applications. By systematically examining their ethnopharmacological origins, phytochemical fingerprints, and pharmacological mechanisms, this review highlights their synergistic potential with conventional antimicrobial therapies through multi-target mechanisms, proposing novel integrative approaches for global health challenges.
Collapse
Affiliation(s)
- Yuqing Liang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tengwen Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingquan Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Institute of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| |
Collapse
|
11
|
Zhang Y, Wang H, Liu K, Sun R, Wang Y, Guo J, Zhou W, Zheng H, Qi Y. Manganese-Based Nanozyme Alleviates Acute Kidney Injury via Nrf2/HO-1 and PI3K/Akt/NF-κB Signaling Pathways. ACS Biomater Sci Eng 2025; 11:1751-1764. [PMID: 39878300 DOI: 10.1021/acsbiomaterials.4c02093] [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] [Indexed: 01/31/2025]
Abstract
Acute renal injury (AKI) has a high incidence rate and mortality, but current treatment methods are limited. As a kind of nanomaterial with enzyme-like activity, nanozyme has shown outstanding advantages in treating AKI according to recent reports. Herein, we assess the potential of manganese-based nanozymes (MnO2-BSA NPs) with excellent biosafety in effectively alleviating AKI. Our findings in vitro and in vivo reveal that MnO2-BSA NPs exert regulatory effects on oxidative stress, inflammation, and apoptosis. These effects are mediated through activation of the Nrf2/HO-1 and PI3K/Akt/NF-κB pathways. Notably, it was observed that the cytoprotective effect of MnO2-BSA NPs is abrogated upon inhibition of Nrf2 expression, highlighting the important role of this transcription factor in cellular protection. In summary, the study demonstrates the protective effect of MnO2-BSA NPs in AKI and provides the molecular mechanisms involved, which can contribute to the advancement of potential therapeutic interventions for nanozyme-based treatments.
Collapse
Affiliation(s)
- Yang Zhang
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Han Wang
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Ke Liu
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Ruimeng Sun
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Yurou Wang
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Jiayu Guo
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Wenxiang Zhou
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Haoran Zheng
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| | - Yanfei Qi
- School of Public Health, Jilin University, Changchun, Jilin, 130021, P. R. China
| |
Collapse
|
12
|
Zhao ZW, Wang YC, Chen PC, Tzeng SF, Chen PS, Kuo YM. Dopamine D1 receptor agonist alleviates post-weaning isolation-induced neuroinflammation and depression-like behaviors in female mice. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2025; 21:6. [PMID: 40065395 PMCID: PMC11895232 DOI: 10.1186/s12993-025-00269-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Major depressive disorder is a significant global cause of disability, particularly among adolescents. The dopamine system and nearby neuroinflammation, crucial for regulating mood and processing rewards, are central to the frontostriatal circuit, which is linked to depression. This study aimed to investigate the effect of post-weaning isolation (PWI) on depression in adolescent mice, with a focus on exploring the involvement of microglia and dopamine D1 receptor (D1R) in the frontostriatal circuit due to their known links with mood disorders. RESULTS Adolescent mice underwent 8 weeks of PWI before evaluating their depression-like behaviors and the activation status of microglia in the frontostriatal regions. Selective D1-like dopamine receptor agonist SKF-81,297 was administered into the medial prefrontal cortex (mPFC) of PWI mice to assess its antidepressant and anti-microglial activation properties. The effects of SKF-81,297 on inflammatory signaling pathways were examined in BV2 microglial cells. After 8 weeks of PWI, female mice exhibited more severe depression-like behaviors than males, with greater microglial activation in the frontostriatal regions. Microglial activation in mPFC was the most prominent among the three frontostriatal regions examined, and it was positively correlated with the severity of depression-like behaviors. Female PWI mice exhibited increased expression of dopamine D2 receptors (D2R). SKF-81,297 treatment alleviated depression-like behaviors and local microglial activation induced by PWI; however, SKF-81,297 induced these alterations in naïve mice. In vitro, SKF-81,297 decreased pro-inflammatory cytokine release and phosphorylations of JNK and ERK induced by lipopolysaccharide, while in untreated BV2 cells, SKF-81,297 elicited inflammation. CONCLUSIONS This study highlights a sex-specific susceptibility to PWI-induced neuroinflammation and depression. While targeting the D1R shows potential in alleviating PWI-induced changes, further investigation is required to evaluate potential adverse effects under normal conditions.
Collapse
Affiliation(s)
- Zi-Wei Zhao
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yun-Chen Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Pei-Chun Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Shun-Fen Tzeng
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Po-See Chen
- Department of Psychiatry, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, 70101, Taiwan
- Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
13
|
Lin N, Abbas-Aghababazadeh F, Su J, Wu AJ, Lin C, Shi W, Xu W, Haibe-Kains B, Liu FF, Kwan JYY. Development of Machine Learning Models for Predicting Radiation Dermatitis in Breast Cancer Patients Using Clinical Risk Factors, Patient-Reported Outcomes, and Serum Cytokine Biomarkers. Clin Breast Cancer 2025:S1526-8209(25)00048-5. [PMID: 40155248 DOI: 10.1016/j.clbc.2025.03.002] [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: 12/10/2024] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Radiation dermatitis (RD) is a significant side effect of radiotherapy experienced by breast cancer patients. Severe symptoms include desquamation or ulceration of irradiated skin, which impacts quality of life and increases healthcare costs. Early identification of patients at risk for severe RD can facilitate preventive management and reduce severe symptoms. This study evaluated the utility of subjective and objective factors, such as patient-reported outcomes (PROs) and serum cytokines, for predicting RD in breast cancer patients. The performance of machine learning (ML) and logistic regression-based models were compared. PATIENTS AND METHODS Data from 147 breast cancer patients who underwent radiotherapy was analyzed to develop prognostic models. ML algorithms, including neural networks, random forest, XGBoost, and logistic regression, were employed to predict clinically significant Grade 2+ RD. Clinical factors, PROs, and cytokine biomarkers were incorporated into the risk models. Model performance was evaluated using nested cross-validation with separate loops for hyperparameter tuning and calculating performance metrics. RESULTS Feature selection identified 18 predictors of Grade 2+ RD including smoking, radiotherapy boost, reduced motivation, and the cytokines interleukin-4, interleukin-17, interleukin-1RA, interferon-gamma, and stromal cell-derived factor-1a. Incorporating these predictors, the XGBoost model achieved the highest performance with an area under the curve (AUC) of 0.780 (95% CI: 0.701-0.854). This was not significantly improved over the logistic regression model, which demonstrated an AUC of 0.714 (95% CI: 0.629-0.798). CONCLUSION Clinical risk factors, PROs, and serum cytokine levels provide complementary prognostic information for predicting severe RD in breast cancer patients undergoing radiotherapy. ML and logistic regression models demonstrated comparable performance for predicting clinically significant RD with AUC>0.70.
Collapse
Affiliation(s)
- Neil Lin
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Farnoosh Abbas-Aghababazadeh
- Princess Margaret Bioinformatics and Computational Genomics Laboratory, University Health Network, Toronto, Canada
| | - Jie Su
- Biostatistics Division, Princess Margaret Cancer Centre, Toronto, Canada
| | - Alison J Wu
- Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Cherie Lin
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Wei Shi
- Research Institute, Princess Margaret Cancer Centre, Toronto, Canada
| | - Wei Xu
- Biostatistics Division, Princess Margaret Cancer Centre, Toronto, Canada
| | - Benjamin Haibe-Kains
- Princess Margaret Bioinformatics and Computational Genomics Laboratory, University Health Network, Toronto, Canada; Research Institute, Princess Margaret Cancer Centre, Toronto, Canada; Department of Computer Science, University of Toronto, Toronto, Canada; Ontario Institute for Cancer Research, Toronto, Canada; Vector Institute for Artificial Intelligence, Toronto, Canada; Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Fei-Fei Liu
- Research Institute, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Jennifer Y Y Kwan
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Research Institute, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.
| |
Collapse
|
14
|
Mohammadi S, Ashtary-Larky D, Mehrbod M, Kouhi Sough N, Salehi Omran H, Dolatshahi S, Amirani N, Asbaghi O. Impacts of supplementation with milk proteins on inflammation: a systematic review and meta-analysis. Inflammopharmacology 2025; 33:1061-1083. [PMID: 39775243 DOI: 10.1007/s10787-024-01615-8] [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: 10/03/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Impacts of milk proteins (MPs) on inflammation are uncertain. The current systematic review and dose-response meta-analysis of randomized controlled trials (RCTs) evaluated the effects of whey protein (WP), casein protein (CP), or MP supplementation on serum levels of cytokines and adipokines in adults. METHODS A comprehensive search of various online databases was conducted to find appropriate clinical trials published until September 2024. A random-effect statistical model was implemented. RESULTS The meta-analysis included 53 RCTs. It was indicated that MP supplements had no substantial effects on serum values of C-reactive protein (CRP), tumor necrosis factor-alpha (TNF-α), adiponectin, and leptin. However, there were statistically significant decreases in serum levels of interleukin-6 (IL-6) following supplementation with MP (weighted mean difference (WMD): - 0.25 pg/mL, 95% CI - 0.48, - 0.03; P = 0.026) in the intervention group compared with the control group. CONCLUSION This study revealed that MP supplementation may not have any considerable impacts on the levels of cytokines and adipokines.
Collapse
Affiliation(s)
- Shooka Mohammadi
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Damoon Ashtary-Larky
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Milad Mehrbod
- School of Medicine, University of Louisville, Louisville, KY, USA
| | | | - Hossein Salehi Omran
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sina Dolatshahi
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niusha Amirani
- Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Omid Asbaghi
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Oulad Ali H, Belboukhari N, Sekkoum K, Belboukhari M, Seddiki LS. Computational Molecular Docking Analysis of Linalool Enantiomers Interaction With Mitogen-Activated Protein Kinase 1 (MAPK1): Insights Into Potential Binding Mechanisms and Affinity. Chirality 2025; 37:e70030. [PMID: 40047200 DOI: 10.1002/chir.70030] [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: 01/27/2025] [Accepted: 02/23/2025] [Indexed: 05/13/2025]
Abstract
Molecular docking analysis of linalool interaction with mitogen-activated protein kinase 1 (MAPK1) provides valuable insights into the potential binding mechanisms and affinity of this interaction. Linalool, a naturally occurring terpene alcohol, has been the subject of increasing interest due to its diverse pharmacological properties, including anti-inflammatory, antioxidant, and anticancer activities. MAPK1 is a crucial signaling protein involved in various cellular processes, including cell proliferation, differentiation, and survival. Using MOE software, we conducted a stereoisomer analysis of (R)- and (S)-linalool in our study. After docking, the ligand was ranked according to their binding energy and the best lead compound was selected based on the highest binding energy. The results showed that the S-linalool isomer showed superior anticancer activity, while the R-linalool molecule showed less activity. This interaction could provide insights into linalool's potential therapeutic applications, highlighting its diverse pharmacological properties.
Collapse
Affiliation(s)
- Halima Oulad Ali
- Bioactive Molecules and Chiral Separation Laboratory, Faculty of Exact Sciences, Tahri Mohammed University, Bechar, Algeria
| | - Nasser Belboukhari
- Bioactive Molecules and Chiral Separation Laboratory, Faculty of Exact Sciences, Tahri Mohammed University, Bechar, Algeria
| | - Khaled Sekkoum
- Bioactive Molecules and Chiral Separation Laboratory, Faculty of Exact Sciences, Tahri Mohammed University, Bechar, Algeria
| | - Mebarka Belboukhari
- Bioactive Molecules and Chiral Separation Laboratory, Faculty of Exact Sciences, Tahri Mohammed University, Bechar, Algeria
| | - Lamia Salima Seddiki
- Bioactive Molecules and Chiral Separation Laboratory, Faculty of Exact Sciences, Tahri Mohammed University, Bechar, Algeria
| |
Collapse
|
16
|
Karthikeyan A, Kim HH, Anjana S, Elena S, Moniruzzaman M, Kalaiselvi S, Kim GS, Min T. Informatics-assisted proteomics revealing the anti-inflammatory effects of satsuma orange ( Citrus unshiu) peel flavonoid extract in LPS-stimulated RAW 264.7 cells. Food Sci Biotechnol 2025; 34:1207-1218. [PMID: 40093549 PMCID: PMC11904023 DOI: 10.1007/s10068-025-01830-1] [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: 08/25/2024] [Revised: 01/15/2025] [Accepted: 01/26/2025] [Indexed: 03/19/2025] Open
Abstract
Citrus unshiu peel (CUP), rich in flavonoids, has been traditionally used for its health benefits. This study investigated the anti-inflammatory effects of CUP flavonoid extract (CUPFE) in lipopolysaccharide (LPS)-activated RAW 264.7 cells through proteomics analysis. CUPFE significantly reduced the inflammatory mediators and cytokines (nitric oxide, IL-6, and CCL-2) production. Quantitative proteomics analysis using LC-MS/MS identified 140 differentially expressed proteins between the CUPFE and LPS groups, with 86 proteins upregulated and 54 downregulated. Notably, CUPFE negatively regulated 56 proteins induced by LPS. Functional enrichment analysis using gene ontology and Kyoto Encyclopedia of Genes and Genomes revealed that most of these proteins are involved in signal transduction pathways (TNF-α, NF-κB, PI3K-Akt, mTOR, and MAPK) regulating inflammatory processes. Further analysis showed that CUPFE interferes these signaling pathways in a dose-dependent manner, counteracting the LPS-induced effects. Collectively, this study reveals CUPFE's anti-inflammatory effects, laying basis for future research on treating inflammation-related conditions. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-025-01830-1.
Collapse
Affiliation(s)
- Adhimoolam Karthikeyan
- Subtropical Horticulture Research Institute, Jeju National University, Jeju, 63243 South Korea
| | - Hun Hwan Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828 South Korea
| | - Sureshbabu Anjana
- Department of Animal Biotechnology, Bio-Resources Computing Research Center, Sustainable Agriculture Research Institute (SARI), Jeju National University, Jeju, 63243 South Korea
| | - Smirnova Elena
- Department of Animal Biotechnology, Bio-Resources Computing Research Center, Sustainable Agriculture Research Institute (SARI), Jeju National University, Jeju, 63243 South Korea
| | - Mohammad Moniruzzaman
- Department of Animal Biotechnology, Bio-Resources Computing Research Center, Sustainable Agriculture Research Institute (SARI), Jeju National University, Jeju, 63243 South Korea
| | - Senthil Kalaiselvi
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu 641003 India
| | - Gon Sup Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828 South Korea
| | - Taesun Min
- Department of Animal Biotechnology, Bio-Resources Computing Research Center, Sustainable Agriculture Research Institute (SARI), Jeju National University, Jeju, 63243 South Korea
| |
Collapse
|
17
|
Sola-Sevilla N, Garmendia-Berges M, Mera-Delgado MC, Puerta E. Context-dependent role of sirtuin 2 in inflammation. Neural Regen Res 2025; 20:682-694. [PMID: 38886935 PMCID: PMC11433891 DOI: 10.4103/nrr.nrr-d-23-02063] [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: 12/20/2023] [Revised: 03/09/2024] [Accepted: 03/30/2024] [Indexed: 06/20/2024] Open
Abstract
Sirtuin 2 is a member of the sirtuin family nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases, known for its regulatory role in different processes, including inflammation. In this context, sirtuin 2 has been involved in the modulation of key inflammatory signaling pathways and transcription factors by deacetylating specific targets, such as nuclear factor κB and nucleotide-binding oligomerization domain-leucine-rich-repeat and pyrin domain-containing protein 3 (NLRP3). However, whether sirtuin 2-mediated pathways induce a pro- or an anti-inflammatory response remains controversial. Sirtuin 2 has been implicated in promoting inflammation in conditions such as asthma and neurodegenerative diseases, suggesting that its inhibition in these conditions could be a potential therapeutic strategy. Conversely, arthritis and type 2 diabetes mellitus studies suggest that sirtuin 2 is essential at the peripheral level and, thus, its inhibition in these pathologies would not be recommended. Overall, the precise role of sirtuin 2 in inflammation appears to be context-dependent, and further investigation is needed to determine the specific molecular mechanisms and downstream targets through which sirtuin 2 influences inflammatory processes in various tissues and pathological conditions. The present review explores the involvement of sirtuin 2 in the inflammation associated with different pathologies to elucidate whether its pharmacological modulation could serve as an effective strategy for treating this prevalent symptom across various diseases.
Collapse
Affiliation(s)
- Noemí Sola-Sevilla
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Maider Garmendia-Berges
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - MCarmen Mera-Delgado
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Elena Puerta
- Department of Pharmaceutical Sciences, Division of Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| |
Collapse
|
18
|
Lim JS, Li X, Lee DY, Yao L, Yoo G, Kim Y, Eum SM, Cho YC, Yoon S, Park SJ. Antioxidant and Anti-Inflammatory Activities of Methanol Extract of Senna septemtrionalis (Viv.) H.S. Irwin & Barneby Through Nrf2/HO-1-Mediated Inhibition of NF-κB Signaling in LPS-Stimulated Mouse Microglial Cells. Int J Mol Sci 2025; 26:1932. [PMID: 40076558 PMCID: PMC11900505 DOI: 10.3390/ijms26051932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Botanical extracts are recognized in traditional medicine for their therapeutic potential and safety standards. Botanical extracts are viable and sustainable alternatives to synthetic drugs, being essential in drug discovery for various diseases. Senna septemtrionalis (Viv.) H.S. Irwin & Barneby is a medical plant traditionally used to treat inflammation. However, its antioxidant and anti-inflammatory properties and the molecular pathways activated in microglial cells require further investigation. Therefore, this study examines the antioxidant and anti-inflammatory properties of Senna septemtrionalis (Viv.) H.S. Irwin & Barneby methanol extracts (SMEs) in lipopolysaccharide (LPS)-stimulated mouse microglial cells. SMEs significantly inhibit LPS-induced nitric oxide (NO) and proinflammatory cytokine production, which are mediated through the dephosphorylation of mitogen-activated protein kinases and inhibition of nuclear factor kappa B (NF-κB) translocation into the nucleus. Additionally, SME treatment upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase (HO)-1, reducing oxidative stress, indicated by a decrease in reactive oxygen species and restoration of the total glutathione content in LPS-stimulated BV2 cells. The inhibitory effects of SMEs on inflammatory mediator production and NF-κB nuclear translocation were significantly reversed by Sn-protoporphyrin, a specific HO-1 inhibitor. These findings demonstrate that SME protects microglial cells by activating the Nrf2/HO-1 pathway and inhibiting NF-κB translocation.
Collapse
Affiliation(s)
- Jae Sung Lim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Xiangying Li
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Da Young Lee
- R&D Center, CUOME BIO Co., Ltd., Sandan-gil, Hwasun-eup, Hwasun-gun 58141, Jeollanam-do, Republic of Korea;
| | - Lulu Yao
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Guijae Yoo
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Jeollabuk-do, Republic of Korea;
| | - Yunyeong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Sang Mi Eum
- International Biological Material Research Center, Korea Research Institute of Bioscience & Biotechnology, 125 Gwahak-ro, Daejeon 34141, Republic of Korea;
| | - Young-Chang Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Somy Yoon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Su-Jin Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Republic of Korea
| |
Collapse
|
19
|
Xi M, Jiang J, Wang B, Wang Y, Di M, Cong Y, Zhang R. Alterations in Methionine Cycle and Wnt/MAPK Signaling Associated with HMBi-Induced Cashmere Growth in Goats. Int J Mol Sci 2025; 26:1663. [PMID: 40004133 PMCID: PMC11855240 DOI: 10.3390/ijms26041663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Methionine (Met) was the first limiting amino acid identified in cashmere goats, and 2-hydroxy-4-(methylthio) butanoic acid isopropyl ester (HMBi) can effectively provide Met and encourage cashmere growth in goats. However, existing studies have primarily centered on the trait of cashmere growth and have not delved into the underlying molecular and physiological mechanisms by which HMBi promotes cashmere growth in goats. In the present study, we combined metabolomic and transcriptomic approaches to reveal the effects of HMBi supplementation and its impact on the gene expressions and metabolic profiles within the skin tissue of Liaoning cashmere goats. A total of 14 female Liaoning cashmere goats were randomly allocated to the control (CON) and HMBi groups. The CON group received a basal diet, and the HMBi group was fed the basal diet plus 1.27% HMBi. Our results show that HMBi supplementation significantly increased (p < 0.05) the cashmere length and decreased the cashmere diameter in the goats. The metabolomics results show that the HMBi supplementation increased (variable importance in projection >1 and p < 0.05) the concentrations of Met, 2-Hydroxy-4-methylthiobutanoic acid (HMB), proline betaine, and 10-hydroxydecanoic acid in the skin tissue of the goats. For HMB degradation and Met cycle-related genes, compared with the CON diets, the HMBi diets elevated (p < 0.05) LDHD, MAT1A, and AHCY by 86.33%, 154.54%, and 147.89% in the skin tissue, respectively. Regarding genes related to cell proliferation and differentiation, the HMBi supplementation increased (p < 0.05) CCND1, CDK4, IVL, and BMP4 by 113.31%, 107.93%, 291.33%, and 186.21%, respectively. The results of the transcriptome evaluation show that the differential expression genes were mainly enriched (p < 0.05) in the Wnt and MAPK signaling pathways. In summary, these findings indicate that the Met cycle, Wnt, and MAPK play important roles in the process of HMBi, promoting cashmere growth in Liaoning cashmere goats.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ruiyang Zhang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China; (M.X.); (J.J.); (B.W.); (Y.W.); (M.D.); (Y.C.)
| |
Collapse
|
20
|
Yang HR, Zahan MN, Hwang DH, Prakash RLM, Ravi DA, Hong IH, Kim WH, Kim JH, Kim E, Kang C. The Therapeutic Potential of Kiwi Extract as a Source of Cysteine Protease Inhibitors on DNCB-Induced Atopic Dermatitis in Mice and Human Keratinocyte HaCaT Cells. Int J Mol Sci 2025; 26:1534. [PMID: 40004009 PMCID: PMC11855533 DOI: 10.3390/ijms26041534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
The discovery of effective cysteine protease inhibitors with crude protein kiwi extracts (CPKEs) has created novel challenges and prospects for pharmaceutical development. Despite extensive research on CPKEs, limited research has been conducted on treating atopic dermatitis (AD). Therefore, the objective of this work was to investigate the anti-inflammatory effects of CPKEs on TNF-α activation in a HaCaT cell model and in a DNCB (1-chloro-2, 4-dinitrochlorobenzene)-induced atopic dermatitis animal model. The molecular weight of the CPKE was determined using SDS-PAGE under non-reducing (17 kDa and 22 kDa) and reducing conditions (25 kDa, 22 kDa, and 15 kDa), whereas gelatin zymography was performed to examine the CPKE's inhibitory impact on cysteine protease (actinidin and papain) activity. Moreover, the CPKE remains stable at 60 °C, with pH levels varying from 4 to 11, as determined by the azocasein assay. CPKE treatment decreased the phosphorylation of mitogen-activated protein kinase (MAPK) and Akt, along with the activation of nuclear factor-kappa B (NF-κB)-p65 in tumor necrosis factor-α (TNF-α)-stimulated HaCaT cells. Five-week-old BALB/c mice were treated with DNCB to act as an AD-like animal model. The topical application of CPKE to DNCB-treated mice for three weeks substantially decreased clinical dermatitis severity and epidermal thickness and reduced eosinophil infiltration and mast cells into ear and skin tissues. These findings imply that CPKE derived from kiwifruit might be a promising therapy option for inflammatory skin diseases such as AD.
Collapse
Affiliation(s)
- Hye Ryeon Yang
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.R.Y.); (M.N.Z.); (D.H.H.); (R.L.M.P.); (D.A.R.); (I.-H.H.); (W.H.K.); (J.-H.K.); (E.K.)
| | - Most Nusrat Zahan
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.R.Y.); (M.N.Z.); (D.H.H.); (R.L.M.P.); (D.A.R.); (I.-H.H.); (W.H.K.); (J.-H.K.); (E.K.)
| | - Du Hyeon Hwang
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.R.Y.); (M.N.Z.); (D.H.H.); (R.L.M.P.); (D.A.R.); (I.-H.H.); (W.H.K.); (J.-H.K.); (E.K.)
| | - Ramachandran Loganathan Mohan Prakash
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.R.Y.); (M.N.Z.); (D.H.H.); (R.L.M.P.); (D.A.R.); (I.-H.H.); (W.H.K.); (J.-H.K.); (E.K.)
| | - Deva Asirvatham Ravi
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.R.Y.); (M.N.Z.); (D.H.H.); (R.L.M.P.); (D.A.R.); (I.-H.H.); (W.H.K.); (J.-H.K.); (E.K.)
| | - Il-Hwa Hong
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.R.Y.); (M.N.Z.); (D.H.H.); (R.L.M.P.); (D.A.R.); (I.-H.H.); (W.H.K.); (J.-H.K.); (E.K.)
- Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Woo Hyun Kim
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.R.Y.); (M.N.Z.); (D.H.H.); (R.L.M.P.); (D.A.R.); (I.-H.H.); (W.H.K.); (J.-H.K.); (E.K.)
- Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jong-Hyun Kim
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.R.Y.); (M.N.Z.); (D.H.H.); (R.L.M.P.); (D.A.R.); (I.-H.H.); (W.H.K.); (J.-H.K.); (E.K.)
- Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Euikyung Kim
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.R.Y.); (M.N.Z.); (D.H.H.); (R.L.M.P.); (D.A.R.); (I.-H.H.); (W.H.K.); (J.-H.K.); (E.K.)
- Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Changkeun Kang
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.R.Y.); (M.N.Z.); (D.H.H.); (R.L.M.P.); (D.A.R.); (I.-H.H.); (W.H.K.); (J.-H.K.); (E.K.)
- Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
21
|
Patel M, Wahezi S, Mavrocordatos P, Abd-Elsayed A. The Effects and Mechanisms of Phytochemicals on Pain Management and Analgesic. Nutrients 2025; 17:633. [PMID: 40004962 PMCID: PMC11858770 DOI: 10.3390/nu17040633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Phytochemicals can be an essential treatment for chronic pain. This narrative review will summarize and critically analyze the evidence surrounding these substances in pain management. We will introduce phytochemicals, discuss their associated mechanisms, and comment on their viability for potential treatment. There have been decades of research on phytochemical therapies for pain management, but the authors limited the scope of the investigation to the last 25 years. This literature review will serve as a foundation for the pain practitioner to understand where these treatments fit in the paradigm for chronic pain treatment. Assess the integration of phytochemicals within pain management fully.
Collapse
Affiliation(s)
- Milan Patel
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA;
| | - Sayed Wahezi
- Department of Pain Managment, Montefiore Medical Center, Bronx, NY 10461, USA
| | | | - Alaa Abd-Elsayed
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA;
| |
Collapse
|
22
|
Hashim NT, Babiker R, Chaitanya NCSK, Mohammed R, Priya SP, Padmanabhan V, Ahmed A, Dasnadi SP, Islam MS, Gismalla BG, Rahman MM. New Insights in Natural Bioactive Compounds for Periodontal Disease: Advanced Molecular Mechanisms and Therapeutic Potential. Molecules 2025; 30:807. [PMID: 40005119 PMCID: PMC11858609 DOI: 10.3390/molecules30040807] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/18/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Periodontal disease is a chronic inflammatory condition that destroys the tooth-supporting structures due to the host's immune response to microbial biofilms. Traditional periodontal treatments, such as scaling and root planing, pharmacological interventions, and surgical procedures, have significant limitations, including difficulty accessing deep periodontal pockets, biofilm recolonization, and the development of antibiotic resistance. In light of these challenges, natural bioactive compounds derived from plants, herbs, and other natural sources offer a promising alternative due to their anti-inflammatory, antioxidant, antimicrobial, and tissue-regenerative properties. This review focuses on the molecular mechanisms through which bioactive compounds, such as curcumin, resveratrol, epigallocatechin gallate (EGCG), baicalin, carvacrol, berberine, essential oils, and Gum Arabic, exert therapeutic effects in periodontal disease. Bioactive compounds inhibit critical inflammatory pathways like NF-κB, JAK/STAT, and MAPK while activating protective pathways such as Nrf2/ARE, reducing cytokine production and oxidative stress. They also inhibit the activity of matrix metalloproteinases (MMPs), preventing tissue degradation and promoting healing. In addition, these compounds have demonstrated the potential to disrupt bacterial biofilms by interfering with quorum sensing, targeting bacterial cell membranes, and enhancing antibiotic efficacy.Bioactive compounds also modulate the immune system by shifting the balance from pro-inflammatory to anti-inflammatory responses and promoting efferocytosis, which helps resolve inflammation and supports tissue regeneration. However, despite the promising potential of these compounds, challenges related to their poor bioavailability, stability in the oral cavity, and the absence of large-scale clinical trials need to be addressed. Future strategies should prioritize the development of advanced delivery systems like nanoparticles and hydrogels to enhance bioavailability and sustain release, alongside long-term studies to assess the effects of these compounds in human populations. Furthermore, combining bioactive compounds with traditional treatments could provide synergistic benefits in managing periodontal disease. This review aims to explore the therapeutic potential of natural bioactive compounds in managing periodontal disease, emphasizing their molecular mechanisms of action and offering insights into their integration with conventional therapies for a more comprehensive approach to periodontal health.
Collapse
Affiliation(s)
- Nada Tawfig Hashim
- Department of Periodontics, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Rasha Babiker
- Department of Physiology, RAK College of Medical Sciences, RAK Medical & Health Science University, Ras-AlKhaimah 11127, United Arab Emirates;
| | - Nallan C. S. K. Chaitanya
- Department of Oral Medicine and Radiology, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Riham Mohammed
- Department Oral Surgery, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Sivan Padma Priya
- Oral Pathology Department, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Vivek Padmanabhan
- Department of Pediatric and Preventive Dentistry, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Ayman Ahmed
- Department of Periodontology and Implantology, Nile University, Khartoum 1847, Sudan;
| | - Shahista Parveen Dasnadi
- Department of Orthodontics, RAK College of Dental, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Md Sofiqul Islam
- Department of Operative Dentistry, RAK College of Dental Sciences, RAK Medical and Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Bakri Gobara Gismalla
- Department of Oral Rehabilitation, Faculty of Dentistry, University of Khartoum, Khartoum 11115, Sudan;
| | - Muhammed Mustahsen Rahman
- Department of Periodontics, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| |
Collapse
|
23
|
Li L, An G, Li F, Zhang D, Zhu X, Liang C, Zhao Y, Xie K, Zhou P, Zhu H, Jin X, Du L. Shared Genes and Pathways in Ulcerative Colitis and Ankylosing Spondylitis: Functional Validation and Implications for Diagnosis. J Inflamm Res 2025; 18:1657-1678. [PMID: 39925932 PMCID: PMC11806757 DOI: 10.2147/jir.s497201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/24/2025] [Indexed: 02/11/2025] Open
Abstract
Background Associations between ulcerative colitis (UC) and ankylosing spondylitis (AS) have been reported in multiple studies, but the common etiologies of UC and AS remain unknown. Thus, in the current study, we aimed to investigate the shared genes and relevant mechanisms in UC and AS. Methods Using datasets for UC (GSE113079) and AS (GSE1797879), we initially identified differentially expressed genes (DEGs) through differential expression analysis. The DEGs from both datasets were intersected to identify common DEGs, relevant to both UC and AS, which were used in receiver operating characteristic (ROC) curve analysis to confirm key genes in the shared pathway. Gene set enrichment analysis (GSEA) was used to obtain information on key gene pathways and interactions with UC or AS-related diseases, followed by immune infiltration analysis. Finally, peripheral blood samples of AS and UC were used to verify the mRNA expression of the eight key genes using reverse transcription-polymerase chain reaction (RT-PCR). Results Our results revealed that GMFG, GNG11, CLEC4D, CMTM2, VAMP5, S100A8, S100A12 and DGKQ are potential diagnostic biomarkers of AS and UC. Rimegepant, eptinezumab, methotrexate, atogepant, and ubrogepant were identified as potential drugs for S100A12 and S100A8 in patients with UC and AS. GSEA showed that these key genes were associated with antigen processing and presentation, natural killer cell mediated cytotoxicity and the T cell receptor signaling pathway in AS and UC, and were significantly associated with immune cells in various immune-related pathways. Subsequent functional experiments revealed significant increases in the mRNA expressions of S100A12 and VAMP5 in patients with AS and UC. Additionally, CLEC4D mRNA expression was notably higher in patients with UC than in healthy controls. Conclusion Key genes and shared pathways were identified in UC and AS, which may improve understanding of their relationship and guide diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Lin Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| | - Guangqi An
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| | - Fuzhen Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| | - Donghui Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| | - Xinyue Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| | - Chunyu Liang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| | - Yu Zhao
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| | - Kunpeng Xie
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| | - Pengyi Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| | - Haiyan Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| | - Xuemin Jin
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| | - Liping Du
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, Henan, People’s Republic of China
| |
Collapse
|
24
|
Yilmaz E, Yilmaz D, Yildiz CG, Cacan E. Upregulation of the MAP2K4 gene triggers endothelial-mesenchymal transition in COVID-19. Mol Biol Rep 2025; 52:180. [PMID: 39888478 DOI: 10.1007/s11033-025-10289-6] [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: 09/22/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND SARS-CoV-2 infection is marked by an excessive inflammatory response, leading to elevated production of pro-inflammatory cytokines through activation of intracellular pathways like mitogen-activated protein kinase (MAPK). Viruses can use the MAPK signaling pathway to their advantage, but the relationship of this pathway to the severe SARS-CoV-2 period has not been fully elucidated. MAP2K4 is involved in the MAPK signaling pathway and affects cellular processes such as cell-cell junction, cell proliferation, differentiation and apoptosis. METHODS AND RESULTS In this study, we sought to determine the associated biomarkers that are involved in the MAP2K4 pathway and elucidate its possible roles in terms of some clinical features associated with COVID-19. We evaluated the expressions of MAP2K4, SNAI1, SLUG, ZEB1 and E-Cadherin. For this purpose, we prospectively recruited 66 individuals, 39 of whom were women and had a mean age of 65 years. The results revealed that MAP2K4 upregulation increased SNAI1 gene expression level whereas E- Cadherin level was decreased in SARS-CoV-2 positive participants. In addition, negative correlations were determined with PLT, Lymphocyte and CKMB and E- Cadherin levels in positive participants. We also observed a negative correlation between the MAP2K4 and AST, and a positive correlation between SLUG and BUN, ZEB1 and CK. CONCLUSIONS We conclude that SARS-CoV-2 infection triggers fibrosis by increasing MAP2K4 regulation. Additionally, this is the first study to demonstrate the possible contribution of MAP2K4 in influencing COVID-19 clinical features, which may be relevant for identifying COVID-19 positive participants with severe complications.
Collapse
Affiliation(s)
- Esra Yilmaz
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Tokat Gaziosmanpasa University, Tokat, 60200, Türkiye
| | - Dilek Yilmaz
- Department of Infectious Diseases and Clinical Microbiology, Yozgat City Hospital, Tokat, 66100, Türkiye
| | - Can Gokay Yildiz
- Department of Emergency Medicine, Tokat City Hospital, Tokat, 60200, Türkiye
| | - Ercan Cacan
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Tokat Gaziosmanpasa University, Tokat, 60200, Türkiye.
| |
Collapse
|
25
|
Iskandar M, Xiao Barbero M, Jaber M, Chen R, Gomez-Guevara R, Cruz E, Westerheide S. A Review of Telomere Attrition in Cancer and Aging: Current Molecular Insights and Future Therapeutic Approaches. Cancers (Basel) 2025; 17:257. [PMID: 39858038 PMCID: PMC11764024 DOI: 10.3390/cancers17020257] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES As cells divide, telomeres shorten through a phenomenon known as telomere attrition, which leads to unavoidable senescence of cells. Unprotected DNA exponentially increases the odds of mutations, which can evolve into premature aging disorders and tumorigenesis. There has been growing academic and clinical interest in exploring this duality and developing optimal therapeutic strategies to combat telomere attrition in aging and cellular immortality in cancer. The purpose of this review is to provide an updated overview of telomere biology and therapeutic tactics to address aging and cancer. METHODS We used the Rayyan platform to review the PubMed database and examined the ClinicalTrial.gov registry to gain insight into clinical trials and their results. RESULTS Cancer cells activate telomerase or utilize alternative lengthening of telomeres to escape telomere shortening, leading to near immortality. Contrarily, normal cells experience telomeric erosion, contributing to premature aging disorders, such as Werner syndrome and Hutchinson-Gilford Progeria, and (2) aging-related diseases, such as neurodegenerative and cardiovascular diseases. CONCLUSIONS The literature presents several promising therapeutic approaches to potentially balance telomere maintenance in aging and shortening in cancer. This review highlights gaps in knowledge and points to the potential of these optimal interventions in preclinical and clinical studies to inform future research in cancer and aging.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sandy Westerheide
- Department of Molecular Biosciences, University of South Florida, 4202 East Fowler Avenue, ISA2015, Tampa, FL 33620, USA; (M.I.); (M.X.B.); (M.J.); (R.C.); (R.G.-G.); (E.C.)
| |
Collapse
|
26
|
Lee SH, Lee SS, Lee GY, Han SY, Kim DS, Lee BH, Yoo YC. Endarachne binghamiae Extract Ameliorates Inflammatory Responses in Macrophages Through Regulation of MAPK, NF-kB and PI3K/AKT Pathways, and Prevents Acute Lung Injury in Mice. Life (Basel) 2025; 15:88. [PMID: 39860028 PMCID: PMC11766595 DOI: 10.3390/life15010088] [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: 12/26/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
In this study, the anti-inflammatory effect of the hot water extract of Endarachne binghamiae (EB-WE), a type of marine brown algae, was investigated in LPS-stimulated RAW 264.7 cells and an acute lung injury (ALI) mouse model induced by intranasal LPS administration. Treatment with EB-WE significantly inhibited NO and pro-inflammatory cytokine (TNF-a and IL-6) production in LPS-stimulated RAW 264.7 cells. In mRNA analysis, the expression of pro-inflammatory cytokines, COX-2, and iNOS mRNAs, was down-regulated by EB-WE treatment. The phosphorylation of MAPK, IkB, and PI3K/AKT molecules responsible for signal pathways during inflammation in LPS-stimulated macrophages was also significantly inhibited by EB-WE. In an in vivo model for ALI, oral administration of EB-WE significantly reduced the level of pro-inflammatory cytokines (TNF-a, IL-1b, and IL-6) and chemokines (MCP-1, CXC-16, CXCL1, and TARC) in serum or bronchoalveolar lavage fluid (BALF) of mice. Similarly to the results in LPS-stimulated RAW 264.7 cells, treatment with EB-WE significantly inhibited intracellular signal pathways mediated by MAPK, IkB, and PI3K/AKT in lung tissues of mice with ALI, and also decreased the expression of mRNAs of inflammatory mediators such as TNF-a, IL-6, iNOS, and COX-2. Furthermore, the inhibitory effect of EB-WE on ALI was apparently confirmed in histological examination through lung tissue staining. Taken together, it is clear that EB-WE has potential activity to effectively ameliorate the inflammatory responses in macrophages through down-regulation of MAPK, NF-kB, and PI3K/AKT activation, and suppress acute lung injury induced by LPS. These findings strongly suggest that EB-WE is a promising natural product beneficial for developing preventive treatments and cures of inflammation-related diseases.
Collapse
Affiliation(s)
- Sang-Hoon Lee
- Department of Microbiology, College of Medicine, Konyang University, Daejeon 32992, Republic of Korea; (S.-H.L.); (S.-S.L.); (G.-Y.L.)
| | - Sang-Seop Lee
- Department of Microbiology, College of Medicine, Konyang University, Daejeon 32992, Republic of Korea; (S.-H.L.); (S.-S.L.); (G.-Y.L.)
| | - Ga-Young Lee
- Department of Microbiology, College of Medicine, Konyang University, Daejeon 32992, Republic of Korea; (S.-H.L.); (S.-S.L.); (G.-Y.L.)
| | - Seung-Yun Han
- Department of Anatomy, College of Medicine, Konyang University, Daejeon 32992, Republic of Korea;
| | - Dong-Sub Kim
- Division of Natural Product Research, Korea Prime Pharmacy Co., Ltd., Gwangju 58144, Republic of Korea;
| | - Bong-Ho Lee
- Department of Chemical Technology, Hanbat National University, Daejeon 34158, Republic of Korea;
| | - Yung-Choon Yoo
- Department of Microbiology, College of Medicine, Konyang University, Daejeon 32992, Republic of Korea; (S.-H.L.); (S.-S.L.); (G.-Y.L.)
| |
Collapse
|
27
|
Li Y, Wang X, Ren Y, Han BZ, Xue Y. Exploring the health benefits of food bioactive compounds from a perspective of NLRP3 inflammasome activation: an insight review. Crit Rev Food Sci Nutr 2025:1-26. [PMID: 39757837 DOI: 10.1080/10408398.2024.2448768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
The food industry has been focusing on food bioactive compounds with multiple physiological and immunological properties that benefit human health. These bioactive compounds, including polyphenols, flavonoids, and terpenoids, have great potential to limit inflammatory responses especially NLRP3 inflammasome activation, which is a key innate immune platform for inflammation. Current studies have revealed numerous food bioactive compounds with promising activities for unraveling immune metabolic disorders and excessive inflammatory responses by directly and indirectly regulating the NLRP3 inflammasome activation. This review explores the food hazards, including microbial and abiotic factors, that may trigger NLRP3-mediated illnesses and inflammation. It also highlights bioactive compounds in food that can suppress NLRP3 inflammasome activation through various mechanisms, linking its activation and inhibition to different pathways. Especially, this review provided further insight into NLRP3-related targets where food bioactive compounds can interact to block the NLRP3 inflammasome activation process, as well as mechanisms on how these compounds facilitate inactivation processes.
Collapse
Affiliation(s)
- Yabo Li
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xinyi Wang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Ying Ren
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Bei-Zhong Han
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yansong Xue
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
28
|
Di Meo C, Tortolani D, Standoli S, Ciaramellano F, Angelucci BC, Tisi A, Kadhim S, Hsu E, Rapino C, Maccarrone M. Cannabinol modulates the endocannabinoid system and shows TRPV1-mediated anti-inflammatory properties in human keratinocytes. Biofactors 2025; 51:e2122. [PMID: 39275884 DOI: 10.1002/biof.2122] [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: 06/03/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024]
Abstract
Cannabinol (CBN) is a secondary metabolite of cannabis whose beneficial activity on inflammatory diseases of human skin has attracted increasing attention. Here, we sought to investigate the possible modulation by CBN of the major elements of the endocannabinoid system (ECS), in both normal and lipopolysaccharide-inflamed human keratinocytes (HaCaT cells). CBN was found to increase the expression of cannabinoid receptor 1 (CB1) at gene level and that of vanilloid receptor 1 (TRPV1) at protein level, as well as their functional activity. In addition, CBN modulated the metabolism of anandamide (AEA) and 2-arachidonoylglicerol (2-AG), by increasing the activities of N-acyl phosphatidylethanolamines-specific phospholipase D (NAPE-PLD) and fatty acid amide hydrolase (FAAH)-the biosynthetic and degradative enzyme of AEA-and that of monoacylglycerol lipase (MAGL), the hydrolytic enzyme of 2-AG. CBN also affected keratinocyte inflammation by reducing the release of pro-inflammatory interleukin (IL)-8, IL-12, and IL-31 and increasing the release of anti-inflammatory IL-10. Of note, the release of IL-31 was mediated by TRPV1. Finally, the mitogen-activated protein kinases (MAPK) signaling pathway was investigated in inflamed keratinocytes, demonstrating a specific modulation of glycogen synthase kinase 3β (GSK3β) upon treatment with CBN, in the presence or not of distinct ECS-directed drugs. Overall, these results demonstrate that CBN modulates distinct ECS elements and exerts anti-inflammatory effects-remarkably via TRPV1-in human keratinocytes, thus holding potential for both therapeutic and cosmetic purposes.
Collapse
Affiliation(s)
- Camilla Di Meo
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Daniel Tortolani
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Sara Standoli
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | | | | | - Annamaria Tisi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Salam Kadhim
- InMed Pharmaceuticals Inc., Vancouver, BC, Canada
| | - Eric Hsu
- InMed Pharmaceuticals Inc., Vancouver, BC, Canada
| | - Cinzia Rapino
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
- European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, Rome, Italy
| |
Collapse
|
29
|
Dash UC, Bhol NK, Swain SK, Samal RR, Nayak PK, Raina V, Panda SK, Kerry RG, Duttaroy AK, Jena AB. Oxidative stress and inflammation in the pathogenesis of neurological disorders: Mechanisms and implications. Acta Pharm Sin B 2025; 15:15-34. [PMID: 40041912 PMCID: PMC11873663 DOI: 10.1016/j.apsb.2024.10.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 05/17/2025] Open
Abstract
Neuroprotection is a proactive approach to safeguarding the nervous system, including the brain, spinal cord, and peripheral nerves, by preventing or limiting damage to nerve cells and other components. It primarily defends the central nervous system against injury from acute and progressive neurodegenerative disorders. Oxidative stress, an imbalance between the body's natural defense mechanisms and the generation of reactive oxygen species, is crucial in developing neurological disorders. Due to its high metabolic rate and oxygen consumption, the brain is particularly vulnerable to oxidative stress. Excessive ROS damages the essential biomolecules, leading to cellular malfunction and neurodegeneration. Several neurological disorders, including Alzheimer's, Parkinson's, Amyotrophic lateral sclerosis, multiple sclerosis, and ischemic stroke, are associated with oxidative stress. Understanding the impact of oxidative stress in these conditions is crucial for developing new treatment methods. Researchers are exploring using antioxidants and other molecules to mitigate oxidative stress, aiming to prevent or slow down the progression of brain diseases. By understanding the intricate interplay between oxidative stress and neurological disorders, scientists hope to pave the way for innovative therapeutic and preventive approaches, ultimately improving individuals' living standards.
Collapse
Affiliation(s)
- Umesh Chandra Dash
- School of Biotechnology, Campus 11, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar 751024, Odisha, India
| | - Nitish Kumar Bhol
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India
| | - Sandeep Kumar Swain
- ICMR-National Institute of Pathology, Sadarjang Hospital Campus, New Delhi 110029, Delhi, India
| | - Rashmi Rekha Samal
- CSIR-Institute of Minerals & Materials Technology, Bhubaneswar 751013, Odisha, India
| | - Prabhat Kumar Nayak
- Bioanalytical Sciences, Research and Development, Enzene Biosciences Limited, Pune 410501, Maharashtra, India
| | - Vishakha Raina
- School of Biotechnology, Campus 11, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar 751024, Odisha, India
| | - Sandeep Kumar Panda
- School of Biotechnology, Campus 11, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar 751024, Odisha, India
| | - Rout George Kerry
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Oslo 0317, Norway
| | - Atala Bihari Jena
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune 411007, India
| |
Collapse
|
30
|
Lee YL, Lee JY, Park JW, Lee J, Lee HH, Lee DH. Protective Effects of Codium fragile Extract against Acetaminophen-Induced Liver Injury. J Microbiol Biotechnol 2024; 34:2675-2862. [PMID: 39473023 PMCID: PMC11729533 DOI: 10.4014/jmb.2409.09061] [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: 10/01/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 12/31/2024]
Abstract
Acetaminophen (APAP) is a well-known analgesic used globally. Generally, APAP has been proven to be safe and effective at therapeutic doses; however, it can cause serious liver damage when administered at high levels. We prepared Codium fragile extract (CFE) using the seaweed C. fragile and confirmed that the CFE contains a substance called Loliolide with antioxidant activity. We performed the present study to determine whether CFE protects HEPG2 cells and BALB/c mice from oxidative stress-induced liver damage. We confirmed that CFE and Loliolide were non-cytotoxic and protected against liver damage by reducing the activities of ALT and AST, which were increased by APAP treatment, and that CFE reduced the mRNA expression of inflammatory cytokines TNF-α and IL-6 and inhibited the phosphorylation of ERK and p38 in HEPG2 cells as determined by RT-PCR and Western blot analyses. Furthermore, the TNF-α and IL-6 levels, which were increased after APAP treatment in BALB/c mice, decreased after CFE treatment. Therefore, we demonstrated that CFE exerts a protective effect against APAP-induced liver injury by suppressing the inflammatory response through anti-inflammatory activity. Our findings provide new perspectives for developing functional foods that utilize seaweeds to improve liver function.
Collapse
Affiliation(s)
- Yea-Lim Lee
- Nbio, Inc., Gangneung 25457, Republic of Korea
| | - Ji-Yun Lee
- Nbio, Inc., Gangneung 25457, Republic of Korea
| | | | - Jin Lee
- Biostream Co., Ltd., Suwon 10442, Republic of Korea
| | - Hyun-Hoo Lee
- Biostream Co., Ltd., Suwon 10442, Republic of Korea
| | - Dae-Hee Lee
- Nbio, Inc., Gangneung 25457, Republic of Korea
- Department of Marine Bio Food Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
| |
Collapse
|
31
|
Yao C, Tang J, Mo Y, Zhong G, Geng X, Yi X, Zhang Q, Li J, Ma H, Zhao S, Zhang G. Polycyclic aromatics-derived benzene carboxylic acids (BPCAs) as a fast predictor of the genotoxicity of combustion particles. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177632. [PMID: 39571819 DOI: 10.1016/j.scitotenv.2024.177632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/04/2024] [Accepted: 11/16/2024] [Indexed: 11/26/2024]
Abstract
Polycyclic aromatic compounds (PAC) are common toxics in combustion particles. Numerous studies on health effects of PAC mixtures focused on limited compounds. It's still challenging to quantify complex PAC mixtures in combustion particles. Recently, benzene polycarboxylic acids (BPCAs) method, which involves conversion of PAC mixtures into a few BPCAs, has been used to quantify complex PAC mixtures in particles. In this study, in vitro biossays were used to evaluate the toxicity of extractable organic matter (EOM) in combustion particles. Analysis with Fourier transform ion cyclotron resonance mass spectrometry (FT-ICR MS) identified ~1000 molecules, mostly aromatics (84.47 ± 5.32 %), that positively associate with the EOM toxicity (p < 0.05). We further employed BPCAs method to quantify PAC mixtures in the EOM of combustion particles, and observed the toxicity (especially genotoxicity) of EOM linearly increases with the abundance of PAC mixtures (r2: 0.68-0.89, p < 0.05), as it is shown by a data set referring to all source types including biomass burning, coal combustion and vehicle exhaust. The genotoxicity of PAC mixtures in EOM of combustion particles was estimated to be 10-13 times that of benzo[a]pyrene at the same mass concentration. Target analysis of 48 PAC was carried out, but a weaker relationship is found for the toxicity of EOM and the abundance of 48 PAC. Taken together, we suggest PAC-derived BPCAs as a fast predictor of the genotoxicity of combustion particles, which could be promising in routine monitoring of PAC pollution in the air.
Collapse
Affiliation(s)
- Chuxin Yao
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiao Tang
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China
| | - Yangzhi Mo
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China
| | - Guangcai Zhong
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China.
| | - Xiaofei Geng
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China
| | - Xin Yi
- State Key Laboratory of Severe Weather & Key Laboratory of Atmospheric Chemistry of CMA, Chinese Academy of Meteorological Sciences, Beijing 100081, China
| | - Qianyu Zhang
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China
| | - Jun Li
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China
| | - Huimin Ma
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China
| | - Shizhen Zhao
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China
| | - Gan Zhang
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China; Guangdong-Hong Kong-Macao Joint Laboratory for Environmental Pollution and Control and Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China
| |
Collapse
|
32
|
Fomichova O, Oliveira PF, Bernardino RL. Exploring the interplay between inflammation and male fertility. FEBS J 2024. [PMID: 39702986 DOI: 10.1111/febs.17366] [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: 10/02/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Male fertility results from a complex interplay of physiological, environmental, and genetic factors. It is conditioned by the properly developed anatomy of the reproductive system, hormonal regulation balance, and the interplay between different cell populations that sustain an appropriate and functional environment in the testes. Unfortunately, the mechanisms sustaining male fertility are not flawless and their perturbation can lead to infertility. Inflammation is one of the factors that contribute to male infertility. In the testes, it can be brought on by varicocele, obesity, gonadal infections, leukocytospermia, physical obstructions or traumas, and consumption of toxic substances. As a result of prolonged or untreated inflammation, the testicular resident cells that sustain spermatogenesis can suffer DNA damage, lipid and protein oxidation, and mitochondrial dysfunction consequently leading to loss of function in affected Sertoli cells (SCs) and Leydig cells (LCs), and the formation of morphologically abnormal dysfunctional sperm cells that lay in the basis of male infertility and subfertility. This is due mainly to the production and secretion of pro-inflammatory mediators, including cytokines, chemokines, and reactive oxygen species (ROS) by local immune cells (macrophages, lymphocytes T, mast cells) and tissue-specific cells [SCs, LCs, peritubular myoid cells (PMCs) and germ cells (GCs)]. Depending on the location, duration, and intensity of inflammation, these mediators can exert their toxic effect on different elements of the testes. In this review, we discuss the most prevalent inflammatory factors that negatively affect male fertility and describe the different ways inflammation can impair male reproductive function.
Collapse
Affiliation(s)
- Oleksandra Fomichova
- UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, Portugal
| | - Raquel L Bernardino
- UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Portugal
| |
Collapse
|
33
|
Huang SW, Hsu MJ, Chen HC, Meleddu R, Distinto S, Maccioni E, Cottiglia F. Suppression of lipopolysaccharide-induced COX-2 expression via p38MAPK, JNK, and C/EBPβ phosphorylation inhibition by furomagydarin A, a benzofuran glycoside from Magydaris pastinacea. J Enzyme Inhib Med Chem 2024; 39:2287420. [PMID: 38058285 PMCID: PMC11792810 DOI: 10.1080/14756366.2023.2287420] [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: 08/14/2023] [Revised: 10/28/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023] Open
Abstract
The phytochemical investigation of the methanol extract of the seeds of Magydaris pastinacea afforded two undescribed benzofuran glycosides, furomagydarins A-B (1, 2), together with three known coumarins. The structures of the new isolates were elucidated after extensive 1D and 2D NMR experiments as well as HR MS. Compound 1 was able to inhibit the COX-2 expression in RAW264.7 macrophages exposed to lipopolysaccharide, a pro-inflammatory stimulus. RT-qPCR and luciferase reporter assays suggested that compound 1 reduces COX-2 expression at the transcriptional level. Further studies highlighted the capability of compound 1 to suppress the LPS-induced p38MAPK, JNK, and C/EBPβ phosphorylation, leading to COX-2 down-regulation in RAW264.7 macrophages.
Collapse
Affiliation(s)
- Shiu-Wen Huang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming Jen Hsu
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Chen Chen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Rita Meleddu
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, Italy
| | - Simona Distinto
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, Italy
| | - Elias Maccioni
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, Italy
| | - Filippo Cottiglia
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, Italy
| |
Collapse
|
34
|
Kim YS, Jung JH, Kim KT. Sorbus commixta Fruit Extract Suppresses Lipopolysaccharide-Induced Neuroinflammation in BV-2 Microglia Cells via the MAPK and NF- κB Signaling Pathways. Molecules 2024; 29:5592. [PMID: 39683751 DOI: 10.3390/molecules29235592] [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: 10/30/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Sorbus commixta Hedl. is a traditional medicinal plant in Korea, China, and Japan with known antioxidative, anti-inflammatory, anti-atherogenic, and anti-melanin activities. However, its anti-neuroinflammatory effects remain largely unknown. In this study, we investigated the inhibitory effects of S. commixta fruit extracts on lipopolysaccharide-stimulated pro-inflammatory factors in BV-2 microglia. We compared the anti-neuroinflammatory activity of S. commixta fruit water extract (SFW) and 70% ethanol extract using a nitric oxide assay. Our data indicated that the SFW (25-100 μg/mL) treatment significantly inhibited excessive nitric oxide production in lipopolysaccharide-stimulated BV-2 microglia compared to the 70% ethanol extract. It also attenuated the expression of inducible nitric oxide synthase, cyclooxygenase-2, and pro-inflammatory cytokines such as interleukin-6 and tumor necrosis factor α. Moreover, SFW exhibited its anti-inflammatory properties by downregulating the expression of factors involved in the extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase pathways and by suppressing nuclear factor kappa B. Caffeic acid was identified as a primary component of SFW showing anti-neuroinflammatory activity. These findings suggest that SFW may offer substantial therapeutic potential for the treatment of neurodegenerative diseases involving microglia activation.
Collapse
Affiliation(s)
- Yon-Suk Kim
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Diseases, Konkuk University, Chungju 27478, Republic of Korea
| | - Jin-Hwa Jung
- Department of Occupational Therapy, Semyung University, Jecheon 27136, Republic of Korea
| | - Ki-Tae Kim
- Department of Korean Medicine, College of Korean Medicine, Semyung University, Jecheon 27136, Republic of Korea
| |
Collapse
|
35
|
Md Samsuzzaman, Hong SM, Lee JH, Park H, Chang KA, Kim HB, Park MG, Eo H, Oh MS, Kim SY. Depression like-behavior and memory loss induced by methylglyoxal is associated with tryptophan depletion and oxidative stress: a new in vivo model of neurodegeneration. Biol Res 2024; 57:87. [PMID: 39574138 PMCID: PMC11580208 DOI: 10.1186/s40659-024-00572-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Depression and memory loss are prevalent neurodegenerative disorders, with diabetic patients facing an elevated risk of brain dysfunction. Methylglyoxal (MGO) formation, which is heightened in diabetes owing to hyperglycemia and gut dysbiosis, may serve as a critical link between diabetes and brain diseases. Despite the high prevalence of MGO, the precise mechanisms underlying MGO-induced depression and memory loss remain unclear. RESULTS We investigated the effect of MGO stress on depression like-behavior and memory loss to elucidate the potential interplay between MGO-induced tryptophan (Trp) metabolism impairment and oxidative stress in the brain. It demonstrates that MGO induces depression-like behavior in mice, as confirmed by the OFT, TST, FST, SPT, and EPM behavioral tests. MGO led to the depletion of Trp and related neurotransmitters as 5-HT, EPI, and DA in the mouse brain. Additionally, MGO reduced the cell count in the DG, CA1, and CA3 hippocampal regions and modulated TPH2 levels in the brain. Notably, co-treatment with MGO and Trp mirrored the effects observed after Trp-null treatment in neurons, including reduced TPH1 and TPH2 levels and inhibition of neuronal outgrowth. Furthermore, MGO significantly altered the expression of key proteins associated with neurodegeneration, such as p-Tau, p-GSK-3β, APP, oAβ, BDNF, NGF, and p-TrkB. Concurrently, MGO activated MAPKs through ROS induction, triggering a redox imbalance by downregulating Nrf-2, Ho-1, TXNRD1, Trx, Sirt-3, and Sirt-5 expression levels, NAD+, and CAT activity in the mouse brain. This led to an accelerated neuroinflammatory response, as evidenced by increased expression of Iba-1, p-NF-κB, and the secretion of IL-6 and TNF-α. Importantly, Trp treatment ameliorated MGO-induced depression like-behavior and memory loss in mice and markedly mitigated increased expression of p-Tau, APP, p-ERK1/2, p-pJNK, and p-NF-κB in the brain. Likewise, Trp treatment also induced the expression of MGO detoxifying factors GLO-I and GLO-II and CAT activity, suggesting the induction of an antioxidant system and reduced inflammation by inhibiting IL-6 and TNF-α secretion. CONCLUSIONS Our data revealed that MGO-induced depression like-behavior and memory deficits resulted from disturbances in Trp, 5-HT, BDNF, and NGF levels, increased p-Tau and APP expression, neuroinflammation, and impaired redox status (Nrf-2/Ho-1/TXNRD1/Sirt3/5) in the brain.
Collapse
Affiliation(s)
- Md Samsuzzaman
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon, 21936, Republic of Korea
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, 21201, USA
| | - Seong-Min Hong
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon, 21936, Republic of Korea
| | - Jae Hyuk Lee
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon, 21936, Republic of Korea
| | - Hyunjun Park
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon, Republic of Korea
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea
| | - Keun-A Chang
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon, Republic of Korea
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Hyun-Bum Kim
- Department of East-West Medical Science, Graduate School of East-West Medical, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, 446-701, Korea
| | - Myoung Gyu Park
- MetaCen Therapeutics Company, Changnyong-daero 256 Beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Hyeyoon Eo
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Myung Sook Oh
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon, 21936, Republic of Korea.
- Gachon Institute of Pharmaceutical Science, Gachon University, #191, Hambakmoe-ro, Yeonsu-gu, Incheon, 21936, Republic of Korea.
| |
Collapse
|
36
|
Han JY, Kim SK, Lim DW, Kwon O, Choi YR, Kang CH, Lee YJ, Lee YM. Anti-Inflammatory Effect of Ethanol Extract from Hibiscus cannabinus L. Flower in Diesel Particulate Matter-Stimulated HaCaT Cells. Nutrients 2024; 16:3805. [PMID: 39599592 PMCID: PMC11597620 DOI: 10.3390/nu16223805] [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: 10/14/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Diesel Particulate Matter (DPM) is a very small particulate matter originating from cities, factories, and the use of fossil fuels in diesel vehicles. When DPM permeates the skin, it causes inflammation, leading to severe atopic dermatitis. Hibiscus cannabinus L. (Kenaf) seeds and leaves possess various beneficial properties, including anti-coagulation, antioxidant, and anti-inflammation effects. In this study, we investigated the anti-inflammatory effects of an ethanol extract of Hibiscus cannabinus L. flower (HCFE) in HaCaT cells stimulated with 100 μg/mL of DPM. METHODS The anthocyanin content of HCFE was analyzed, and its antioxidant capacity was investigated using the DPPH assay. After inducing inflammation with 100 ug/mL of DPM, the cytotoxicity of HCFE 25, 50, and 100 ug/mL was measured, and the inhibitory effect of HCFE on inflammatory mediators was evaluated. RESULTS Anthocyanin and myricetin-3-O-glucoside were present in HCFE and showed high antioxidant capacity. In addition, HCFE decreased the mRNA expression of inflammatory cytokines and chemokines such as IL-1β, IL-4, IL-6, IL-8, IL-13, and MCP-1, and significantly reduced the gene expression of CXCL10, CCL5, CCL17, and CCL22, which are known to increase in atopic dermatitis lesions. Furthermore, HCFE reduced intracellular reactive oxygen species (ROS) production, and down-regulated the activation of NF-κB, MAPKs. Inhibition of the NLRP-3 inflammasome was observed in DPM-stimulated HaCaT cells. In addition, the restoration of filaggrin and involucrin, skin barrier proteins destroyed by DPM exposure, was confirmed. CONCLUSIONS These data suggest that HCFE could be used to prevent and improve skin inflammation and atopic dermatitis through the regulation of inflammatory mediators and the inhibition of skin water loss.
Collapse
Affiliation(s)
- Ji-Ye Han
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea
- Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan 54538, Republic of Korea
| | - Shin-Kyeom Kim
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea
- Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan 54538, Republic of Korea
| | - Do-Won Lim
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea
- Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan 54538, Republic of Korea
| | - Osoung Kwon
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea
- Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan 54538, Republic of Korea
| | - Yu-Rim Choi
- Division of Crops & Food, Jeonbuk-do Agricultural Research & Extension Services, Iksan 54591, Republic of Korea
| | - Chan-Ho Kang
- Division of Crops & Food, Jeonbuk-do Agricultural Research & Extension Services, Iksan 54591, Republic of Korea
| | - Yun Jung Lee
- Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan 54538, Republic of Korea
| | - Young-Mi Lee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea
- Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan 54538, Republic of Korea
| |
Collapse
|
37
|
Hussain MK, Khatoon S, Khan MF, Akhtar MS, Ahamad S, Saquib M. Coumarins as versatile therapeutic phytomolecules: A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155972. [PMID: 39265442 DOI: 10.1016/j.phymed.2024.155972] [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: 04/28/2024] [Revised: 06/23/2024] [Accepted: 07/11/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Coumarins, abundantly distributed in a plethora of biologically active compounds, serve as a fundamental motif in numerous natural products, drugs, and therapeutic leads. Despite their small size, they exhibit a diverse range of biological activities, intriguing researchers with their immense pharmacological potential. PURPOSE This study consolidates the evidence regarding the essential role of coumarins in modern drug discovery, exploring their broad-spectrum pharmaceutical effects, structural versatility, and mechanisms of action across various domains. METHODS For literature search, we utilized PubMed, Google scholar, and SciFinder databases. Keyword and keyword combinations such as "coumarins", "natural coumarins", "specific natural coumarins for particular diseases", and "therapeutic effects" were employed to retrieve relevant studies. The search encompassed articles published between 2005 and 2023. Selection criteria included studies reporting on the pharmacological activities of natural coumarins against various diseases. RESULTS The results highlight the therapeutic potential of natural coumarins against various diseases, demonstrating anti-cancer, anti-oxidant, and anti-inflammatory activities. They also act as monoamine oxidase inhibitors and phosphodiesterase inhibitors, and as anti-thrombotic, anti-diabetic, and hepatoprotective agents. They also show efficacy against diabetic nephropathy, neurodegenerative diseases, microbial infections and many other diseases. CONCLUSION This review underscores the significant role of natural coumarins in medicinal chemistry and drug discovery. Their diverse biological activities and structural versatility make them promising therapeutic agents. This study serves as a catalyst for further research in the field, aiming to address emerging challenges and opportunities in drug development.
Collapse
Affiliation(s)
- Mohd Kamil Hussain
- Department of Chemistry, Govt. Raza P.G. College, Rampur 244901, M.J.P Rohil Khand University, Bareilly, India.
| | | | - Mohammad Faheem Khan
- Department of Biotechnology, Era's Lucknow Medical College, Era University, Lucknow 226003, India
| | - Mohd Sayeed Akhtar
- Department of Botany, Gandhi Faiz-e-Aam College, Shahjahanpur 242001, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Mohammad Saquib
- Department of Chemistry, University of Allahabad, Prayagraj (Allahabad) 211002, India; Department of Chemistry, G. R. P. B. Degree College, P. R. S. University, Prayagraj (Allahabad) 211010, India.
| |
Collapse
|
38
|
Lei H, Su H, Cao L, Zhou X, Liu Y, Li Y, Song X, Wang Y, Guan Q. Investigating Xiaochaihu Decoction's fever-relieving mechanism via network pharmacology, molecular docking, dynamics simulation, and experiments. Anal Biochem 2024; 694:115629. [PMID: 39069245 DOI: 10.1016/j.ab.2024.115629] [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: 04/20/2024] [Revised: 06/29/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Xiaochaihu Decoction(XCHD)is a classic prescription for the treatment of fever, but the mechanism is not clear. In this study, We elucidated the mechanism of action through network pharmacology and molecular docking. A rat fever model was established to verify the prediction results of network pharmacology. The analysis revealed that 120 intersection targets existed between XCHD and fever. The TP53, STAT3, RELA, MAPK1, AKT1, TNF and MAPK14 as potential core targets of XCHD in fever treatment. GO and KEGG pathway enrichment analyses indicated that XCHD may act through pathways such as the AGE-RAGE signaling pathway in diabetic complications, TNF signaling pathway, IL-17 signaling pathway. Molecular docking results demonstrated that quercetin, kaempferol, β-sitosterol, stigmasterol and baicalein exhibited strong binding activity to key targets. Animal experiments showed that XCHD significantly reduced body temperature and levels of IL-1β, IL-6, TNF-α, NO, PGE2, and cAMP in rats with fever. Importantly, no significant difference was observed between the XCHD self-emulsifying nano phase plus suspension phase and XCHD group. XCHD exerts its therapeutic effects on fever through a multi-ingredient, multi-target, and multi-pathway approach.
Collapse
Affiliation(s)
- Hong Lei
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang, 150066, China
| | - Hongbing Su
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang, 150066, China
| | - Ling Cao
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang, 150066, China
| | - Xiaoying Zhou
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang, 150066, China
| | - Yumeng Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang, 150066, China
| | - Ying Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang, 150066, China
| | - Xiaoxue Song
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang, 150066, China
| | - Yanhong Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang, 150066, China
| | - Qingxia Guan
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang, 150066, China.
| |
Collapse
|
39
|
Doswell F, Haley JD, Kaczocha M. Proteomic Analysis of Signaling Pathways Modulated by Fatty Acid Binding Protein 5 (FABP5) in Macrophages. J Pharmacol Exp Ther 2024; 391:289-300. [PMID: 38849143 PMCID: PMC11493448 DOI: 10.1124/jpet.123.002006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Although acute inflammation serves essential functions in maintaining tissue homeostasis, chronic inflammation is causally linked to many diseases. Macrophages are a major cell type that orchestrates inflammatory processes. During inflammation, macrophages undergo polarization and activation, thereby mobilizing pro-inflammatory and anti-inflammatory transcriptional programs that regulate ensuing macrophage functions. Fatty acid binding protein 5 (FABP5) is a lipid chaperone highly expressed in macrophages. FABP5 deletion is implicated in driving macrophages toward an anti-inflammatory phenotype, yet signaling pathways regulated by macrophage-FABP5 have not been systematically profiled. We leveraged proteomic and phosphoproteomic approaches to characterize pathways modulated by FABP5 in M1 and M2 polarized bone marrow-derived macrophages (BMDMs). Stable isotope labeling by amino acids-based analysis of M1 and M2 polarized wild-type and FABP5 knockout BMDMs revealed numerous differentially regulated proteins and phosphoproteins. FABP5 deletion impacted downstream pathways associated with inflammation, cytokine production, oxidative stress, and kinase activity. Toll-like receptor 2 (TLR2) emerged as a novel target of FABP5 and pharmacological FABP5 inhibition blunted TLR2-mediated activation of downstream pathways, ascribing a novel role for FABP5 in TLR2 signaling. This study represents a comprehensive characterization of the impact of FABP5 deletion on the proteomic and phosphoproteomic landscape of M1 and M2 polarized BMDMs. Loss of FABP5 altered pathways implicated in inflammatory responses, macrophage function, and TLR2 signaling. This work provides a foundation for future studies seeking to investigate the therapeutic potential of FABP5 inhibition in pathophysiological states resulting from dysregulated inflammatory signaling. SIGNIFICANCE STATEMENT: This research offers a comprehensive analysis of fatty acid binding protein 5 (FABP5) in macrophages during inflammatory response. The authors employed quantitative proteomic and phosphoproteomic approaches to investigate this utilizing bone marrow-derived macrophages that were M1 and M2 polarized using lipopolysaccharide with interferon γ and interleukin-4, respectively. This revealed multiple pathways related to inflammation that were differentially regulated due to the absence of FABP5. These findings underscore the potential therapeutic significance of macrophage-FABP5 as a candidate for addressing inflammatory-related diseases.
Collapse
Affiliation(s)
- Faniya Doswell
- Molecular and Cellular Biology Program, Stony Brook University, Stony Brook, New York (F.D.); Departments of Anesthesiology (F.D., M.K.) and Pathology (J.D.H.) and Biological Mass Spectrometry Facility, (J.D.H.), Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - John D Haley
- Molecular and Cellular Biology Program, Stony Brook University, Stony Brook, New York (F.D.); Departments of Anesthesiology (F.D., M.K.) and Pathology (J.D.H.) and Biological Mass Spectrometry Facility, (J.D.H.), Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - Martin Kaczocha
- Molecular and Cellular Biology Program, Stony Brook University, Stony Brook, New York (F.D.); Departments of Anesthesiology (F.D., M.K.) and Pathology (J.D.H.) and Biological Mass Spectrometry Facility, (J.D.H.), Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| |
Collapse
|
40
|
ten Hoeve AL, Rodriguez ME, Säflund M, Michel V, Magimel L, Ripoll A, Yu T, Hakimi MA, Saeij JPJ, Ozata DM, Barragan A. Hypermigration of macrophages through the concerted action of GRA effectors on NF-κB/p38 signaling and host chromatin accessibility potentiates Toxoplasma dissemination. mBio 2024; 15:e0214024. [PMID: 39207098 PMCID: PMC11481493 DOI: 10.1128/mbio.02140-24] [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: 07/22/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Mononuclear phagocytes facilitate the dissemination of the obligate intracellular parasite Toxoplasma gondii. Here, we report how a set of secreted parasite effector proteins from dense granule organelles (GRA) orchestrates dendritic cell-like chemotactic and pro-inflammatory activation of parasitized macrophages. These effects enabled efficient dissemination of the type II T. gondii lineage, a highly prevalent genotype in humans. We identify novel functions for effectors GRA15 and GRA24 in promoting CCR7-mediated macrophage chemotaxis by acting on NF-κB and p38 mitogen-activated protein kinase signaling pathways, respectively, with contributions by GRA16/18 and counter-regulation by effector TEEGR. Furthermore, GRA28 boosted chromatin accessibility and GRA15/24/NF-κB-dependent transcription at the Ccr7 gene locus in primary macrophages. In vivo, adoptively transferred macrophages infected with wild-type T. gondii outcompeted macrophages infected with a GRA15/24 double mutant in migrating to secondary organs in mice. The data show that T. gondii, rather than being passively shuttled, actively promotes its dissemination by inducing a finely regulated pro-migratory state in parasitized human and murine phagocytes via co-operating polymorphic GRA effectors. IMPORTANCE Intracellular pathogens can hijack the cellular functions of infected host cells to their advantage, for example, for intracellular survival and dissemination. However, how microbes orchestrate the hijacking of complex cellular processes, such as host cell migration, remains poorly understood. As such, the common parasite Toxoplasma gondii actively invades the immune cells of humans and other vertebrates and modifies their migratory properties. Here, we show that the concerted action of a number of secreted effector proteins from the parasite, principally GRA15 and GRA24, acts on host cell signaling pathways to activate chemotaxis. Furthermore, the protein effector GRA28 selectively acted on chromatin accessibility in the host cell nucleus to selectively boost host gene expression. The joint activities of GRA effectors culminated in pro-migratory signaling within the infected phagocyte. We provide a molecular framework delineating how T. gondii can orchestrate a complex biological phenotype, such as the migratory activation of phagocytes to boost dissemination.
Collapse
Affiliation(s)
- Arne L. ten Hoeve
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Matias E. Rodriguez
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Martin Säflund
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Valentine Michel
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lucas Magimel
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Albert Ripoll
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Tianxiong Yu
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Mohamed-Ali Hakimi
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble, France
| | - Jeroen P. J. Saeij
- Department of Pathology, Microbiology, and Immunology, University of California Davis, Davis, California, USA
| | - Deniz M. Ozata
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Antonio Barragan
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
41
|
Lee SY, Le DD, Bae CS, Park JW, Lee M, Cho SS, Park DH. Oleic acid attenuates asthma pathogenesis via Th1/Th2 immune cell modulation, TLR3/4-NF-κB-related inflammation suppression, and intrinsic apoptotic pathway induction. Front Immunol 2024; 15:1429591. [PMID: 39421735 PMCID: PMC11484255 DOI: 10.3389/fimmu.2024.1429591] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/19/2024] [Indexed: 10/19/2024] Open
Abstract
WHO reported that asthma was responsible for 455,000 deaths in 2019 and asthma patients was evaluated 262 million in May 2023. The incidence is expected to increase as the average life expectancy increases, highlighting asthma as a significant health challenge in an aging society. The etiology of asthma is linked to an imbalance of Th1 and Th2 cells, respiratory inflammation, and pulmonary cell proliferation. The purpose of this study is to investigate the anti-asthmatic effect and potential mechanism of oleic acid. The anti-inflammatory effect of oleic acid was evaluated in an LPS-induced RAW 264.7 cell model, and immune modulation and the anti-apoptotic effect were measured in an ovalbumin-induced BALB/c mouse model. A variety of analytical procedures, such as MTT, qPCR, ELISA, Western blotting, immunofluorescence, gene transfection, immunohistochemistry, and several staining methods (Diff Quik, H&E, PAS), were used to evaluate the effectiveness and mechanisms of these methods. The results from in vitro experiments showed that oleic acid could reduce the levels of inflammatory cytokines (TNF-α, IL-6, and IL-1β), and molecular docking studies suggested that oleic acid could interact with TLR3 and TLR4 proteins to form ligand-protein complexes, showing good binding affinity. Additionally, oleic acid attenuated the expression of MAPK pathway components (JNK, p38 MAPK) and NF-κB pathway constituents (IκB, NF-κB, COX-2, PGE2). In vivo results indicated that oleic acid reduced the levels of inflammatory cells (WBCs and eosinophils) and IgE activity, reduced the expression of the Th2 cell transcription factor GATA-3, and decreased the levels of Th2/Th17-related cytokines (IL-4, TNF-α, and IL-6). Oleic acid also alleviated OVA-induced pathological changes in the lung, such as epithelial cell proliferation, inflammatory cell infiltration, and mucus hypersecretion. OVA restored apoptosis in lung epithelial cells by modulating the expression of Bcl-2 and Bax. In summary, oleic acid has potential as a novel candidate for asthma treatment through its ability to regulate immune cells, exert anti-inflammatory effects, and promote apoptosis, thereby ameliorating asthma manifestations.
Collapse
Affiliation(s)
- Soon-Young Lee
- College of Oriental Medicine, Dongshin University, Naju, Republic of Korea
| | - Duc Dat Le
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Chun-Sik Bae
- College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Jin Woo Park
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Muan, Republic of Korea
| | - Mina Lee
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, Republic of Korea
| | - Seung-Sik Cho
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Muan, Republic of Korea
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan, Republic of Korea
| | - Dae-Hun Park
- College of Oriental Medicine, Dongshin University, Naju, Republic of Korea
| |
Collapse
|
42
|
Mrozewski L, Tharmalingam S, Michael P, Kumar A, Tai TC. C5a Induces Inflammatory Signaling and Apoptosis in PC12 Cells through C5aR-Dependent Signaling: A Potential Mechanism for Adrenal Damage in Sepsis. Int J Mol Sci 2024; 25:10673. [PMID: 39409001 PMCID: PMC11477224 DOI: 10.3390/ijms251910673] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
The complement system is critically involved in the pathogenesis of sepsis. In particular, complement anaphylatoxin C5a is generated in excess during sepsis, leading to cellular dysfunction. Recent studies have shown that excessive C5a impairs adrenomedullary catecholamine production release and induces apoptosis in adrenomedullary cells. Currently, the mechanisms by which C5a impacts adrenal cell function are poorly understood. The PC12 cell model was used to examine the cellular effects following treatment with recombinant rat C5a. The levels of caspase activation and cell death, protein kinase signaling pathway activation, and changes in inflammatory protein expression were examined following treatment with C5a. There was an increase in apoptosis of PC12 cells following treatment with high-dose C5a. Ten inflammatory proteins, primarily involved in apoptosis, cell survival, and cell proliferation, were upregulated following treatment with high-dose C5a. Five inflammatory proteins, involved primarily in chemotaxis and anti-inflammatory functions, were downregulated. The ERK/MAPK, p38/MAPK, JNK/MAPK, and AKT protein kinase signaling pathways were upregulated in a C5aR-dependent manner. These results demonstrate an apoptotic effect and cellular signaling effect of high-dose C5a. Taken together, the overall data suggest that high levels of C5a may play a role in C5aR-dependent apoptosis of adrenal medullary cells in sepsis.
Collapse
Affiliation(s)
- Lucas Mrozewski
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (L.M.); (S.T.); (P.M.); (A.K.)
| | - Sujeenthar Tharmalingam
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (L.M.); (S.T.); (P.M.); (A.K.)
- Medical Science Division, NOSM University, Sudbury, ON P3E 2C6, Canada
| | - Paul Michael
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (L.M.); (S.T.); (P.M.); (A.K.)
| | - Aseem Kumar
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (L.M.); (S.T.); (P.M.); (A.K.)
- Medical Science Division, NOSM University, Sudbury, ON P3E 2C6, Canada
| | - T. C. Tai
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (L.M.); (S.T.); (P.M.); (A.K.)
- Medical Science Division, NOSM University, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
43
|
Hsu CY, Mustafa MA, Moath Omar T, Taher SG, Ubaid M, Gilmanova NS, Nasrat Abdulraheem M, Saadh MJ, Athab AH, Mirzaei R, Karampoor S. Gut instinct: harnessing the power of probiotics to tame pathogenic signaling pathways in ulcerative colitis. Front Med (Lausanne) 2024; 11:1396789. [PMID: 39323474 PMCID: PMC11422783 DOI: 10.3389/fmed.2024.1396789] [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: 03/06/2024] [Accepted: 08/22/2024] [Indexed: 09/27/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease (IBD) marked by persistent inflammation of the mucosal lining of the large intestine, leading to debilitating symptoms and reduced quality of life. Emerging evidence suggests that an imbalance of the gut microbiota plays a crucial role in UC pathogenesis, and various signaling pathways are implicated in the dysregulated immune response. Probiotics are live microorganisms that confer health benefits to the host, have attracted significant attention for their potential to restore gut microbial balance and ameliorate inflammation in UC. Recent studies have elucidated the mechanisms by which probiotics modulate these signaling pathways, often by producing anti-inflammatory molecules and promoting regulatory immune cell function. For example, probiotics can inhibit the nuclear factor-κB (NF-κB) pathway by stabilizing Inhibitor of kappa B alpha (IκBα), dampening the production of proinflammatory cytokines. Similarly, probiotics can modulate the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway, suppressing the activation of STAT1 and STAT3 and thus reducing the inflammatory response. A better understanding of the underlying mechanisms of probiotics in modulating pathogenic signaling pathways in UC will pave the way for developing more effective probiotic-based therapies. In this review, we explore the mechanistic role of probiotics in the attenuation of pathogenic signaling pathways, including NF-κB, JAK/STAT, mitogen-activated protein kinases (MAPKs), Wnt/β-catenin, the nucleotide-binding domain (NOD)-, leucine-rich repeat (LRR)- and pyrin domain-containing protein 3 (NLRP3) inflammasome, Toll-like receptors (TLRs), interleukin-23 (IL-23)/IL-17 signaling pathway in UC.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ, United States
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, Imam Jaafar AL-Sadiq University, Baghdad, Iraq
- Department of Pathological Analyzes, College of Applied Sciences, University of Samarra, Samarra, Iraq
| | - Thabit Moath Omar
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq
| | - Sada Gh Taher
- Department of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Mohammed Ubaid
- Department of MTL, Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Nataliya S. Gilmanova
- Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | | | - Aya H. Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Massoud G, Parish M, Hazimeh D, Moukarzel P, Singh B, Cayton Vaught KC, Segars J, Islam MS. Unlocking the potential of tranilast: Targeting fibrotic signaling pathways for therapeutic benefit. Int Immunopharmacol 2024; 137:112423. [PMID: 38861914 PMCID: PMC11245748 DOI: 10.1016/j.intimp.2024.112423] [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: 01/03/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024]
Abstract
Fibrosis is the excessive deposition of extracellular matrix in an organ or tissue that results from an impaired tissue repair in response to tissue injury or chronic inflammation. The progressive nature of fibrotic diseases and limited treatment options represent significant healthcare challenges. Despite the substantial progress in understanding the mechanisms of fibrosis, a gap persists translating this knowledge into effective therapeutics. Here, we discuss the critical mediators involved in fibrosis and the role of tranilast as a potential antifibrotic drug to treat fibrotic conditions. Tranilast, an antiallergy drug, is a derivative of tryptophan and has been studied for its role in various fibrotic diseases. These include scleroderma, keloid and hypertrophic scars, liver fibrosis, renal fibrosis, cardiac fibrosis, pulmonary fibrosis, and uterine fibroids. Tranilast exerts antifibrotic effects by suppressing fibrotic pathways, including TGF-β, and MPAK. Because it disrupts fibrotic pathways and has demonstrated beneficial effects against keloid and hypertrophic scars, tranilast could be used to treat other conditions characterized by fibrosis.
Collapse
Affiliation(s)
- Gaelle Massoud
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Maclaine Parish
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Dana Hazimeh
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Pamela Moukarzel
- American University of Beirut Medical Center, Faculty of Medicine, Riad El Solh, Beirut, Lebanon
| | - Bhuchitra Singh
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Kamaria C Cayton Vaught
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - James Segars
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA.
| | - Md Soriful Islam
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
45
|
Guo Y, Xu S, Pan X, Xin W, Cao W, Ma W, Li L, Shen Q, Li Z. Psoralen protects neurons and alleviates neuroinflammation by regulating microglial M1/M2 polarization via inhibition of the Fyn-PKCδ pathway. Int Immunopharmacol 2024; 137:112493. [PMID: 38897126 DOI: 10.1016/j.intimp.2024.112493] [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: 04/03/2024] [Revised: 05/26/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
Microglia-mediated neuroinflammation is closely associated with many neurodegenerative diseases. Psoralen has potential for the treatment of many diseases, however, the anti-neuroinflammatory and neuroprotective effects of psoralen have been unclear. This study investigated the anti-neuroinflammatory and neuroprotective effects of psoralen and its regulation of microglial M1/M2 polarization. The LPS-induced mice model was used to test anti-neuroinflammatory effects, regulatory effects on microglia polarization, and neuroprotective effects of psoralen in vivo. The LPS-induced BV2 model was used to test the anti-neuroinflammatory effects and the regulatory effects and mechanisms on microglial M1/M2 polarization of psoralen in vitro. PC12 cell model induced by conditioned medium of BV2 cells was used to validate the protective effects of psoralen against neuroinflammation-induced neuronal damage. These results showed that psoralen inhibited the expression of iNOS, CD86, and TNF-α, and increased the expression of Arg-1, CD206, and IL-10. These results indicated that psoralen inhibited the M1 microglial phenotype and promoted the M2 microglial phenotype. Further studies showed that psoralen inhibited the phosphorylation of Fyn and PKCδ, thereby inhibiting activation of the MAPKs and NF-κB pathways and suppressing the expression of pro-inflammatory cytokines in microglia. Furthermore, psoralen reduced oxidative stress, neuronal damage, and apoptosis via inhibition of neuroinflammation. For the first time, this study showed that psoralen protected neurons and alleviated neuroinflammation by regulating microglial M1/M2 polarization, which may be mediated by inhibition of the Fyn-PKCδ pathway. Thus, psoralen may be a potential agent in the treatment of neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Yaping Guo
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Sai Xu
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Xiaohong Pan
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Wenyu Xin
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Wenli Cao
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Wenya Ma
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Li Li
- Department of Pharmacy, Zhejiang Hospital, Hangzhou 310013, Zhejiang, China
| | - Qi Shen
- Department of Pharmacy, Zhejiang Hospital, Hangzhou 310013, Zhejiang, China.
| | - Zhipeng Li
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China.
| |
Collapse
|
46
|
Merecz-Sadowska A, Sitarek P, Zajdel K, Sztandera W, Zajdel R. Genus Sambucus: Exploring Its Potential as a Functional Food Ingredient with Neuroprotective Properties Mediated by Antioxidant and Anti-Inflammatory Mechanisms. Int J Mol Sci 2024; 25:7843. [PMID: 39063085 PMCID: PMC11277136 DOI: 10.3390/ijms25147843] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The genus Sambucus, mainly Sambucus nigra, has emerged as a valuable source of bioactive compounds with potential neuroprotective properties. This review explores the antioxidant, anti-inflammatory, and neuroregenerative effects of Sambucus-derived compounds and their implications for brain health and cognitive function. In vitro studies have demonstrated the ability of Sambucus extracts to mitigate oxidative stress, modulate inflammatory responses, and promote neural stem cell proliferation and differentiation. In vivo studies using animal models of neurodegenerative diseases, such as Alzheimer's and Parkinson's, have shown that Sambucus compounds can improve cognitive function, motor performance, and neuronal survival while attenuating neuroinflammation and oxidative damage. The neuroprotective effects of Sambucus are primarily attributed to its rich content of polyphenols, particularly anthocyanins, which exert their benefits through multiple mechanisms, including the modulation of signaling pathways involved in inflammation, apoptosis, mitochondrial function, and oxidative stress. Furthermore, the potential of Sambucus as a functional food ingredient is discussed, highlighting its application in various food products and the challenges associated with the stability and bioavailability of its bioactive compounds. This review provides a comprehensive overview of the current state of research on the neuroprotective potential of Sambucus and its derivatives, offering valuable insights for the development of dietary strategies to promote brain health and prevent age-related cognitive decline.
Collapse
Affiliation(s)
- Anna Merecz-Sadowska
- Department of Economic and Medical Informatics, University of Lodz, 90-214 Lodz, Poland;
- Department of Allergology and Respiratory Rehabilitation, Medical University of Lodz, 90-725 Lodz, Poland
| | - Przemysław Sitarek
- Department of Medical Biology, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland;
| | - Karolina Zajdel
- Department of Medical Informatics and Statistics, Medical University of Lodz, 90-645 Lodz, Poland;
| | - Wiktoria Sztandera
- Department of Internal Medicine, Rehabilitation and Physical Medicine, Medical University of Lodz, 90-647 Lodz, Poland;
| | - Radosław Zajdel
- Department of Economic and Medical Informatics, University of Lodz, 90-214 Lodz, Poland;
- Department of Medical Informatics and Statistics, Medical University of Lodz, 90-645 Lodz, Poland;
| |
Collapse
|
47
|
Li J, Luo X, Shiu PHT, Cheng Y, Nie X, Rangsinth P, Lau BWM, Zheng C, Li X, Li R, Lee SMY, Fu C, Seto SW, Zhang J, Leung GPH. Protective effects of Amauroderma rugosum on dextran sulfate sodium-induced ulcerative colitis through the regulation of macrophage polarization and suppression of oxidative stress. Biomed Pharmacother 2024; 176:116901. [PMID: 38878683 DOI: 10.1016/j.biopha.2024.116901] [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: 02/28/2024] [Revised: 05/30/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Amauroderma rugosum (AR) is a medicinal mushroom commonly used to treat inflammation, gastric disorders, epilepsy, and cancers due to its remarkable anti-inflammatory and anti-oxidative properties. This study was designed to evaluate the pharmacological effects of AR and its underlying mechanism of action against ulcerative colitis (UC) in vitro and in vivo. METHODS A UC mouse model was established by administration of dextran sulfate sodium (DSS). AR extract was administered intragastrically to mice for 7 days. At the end of the experiment, histopathology, macrophage phenotype, oxidative stress, and inflammatory status were examined in vivo. Furthermore, RAW 264.7, THP-1, and Caco-2 cells were used to elucidate the mechanism of action of AR in vitro. RESULTS AR extract (0.5-2 mg/mL) significantly suppressed lipopolysaccharide (LPS) and interferon-gamma (IFN-γ)-induced M1 macrophage (pro-inflammatory) polarization in both RAW 264.7 and THP-1 cells. LPS-induced pro-inflammatory mediators (nitric oxide, TNF-α, IL-1β, MCP-1, and IL-6) were reduced by AR extract in a concentration-dependent manner. Similarly, AR extract downregulated MAPK signaling activity in LPS-stimulated RAW 264.7 cells. AR extract elicited a concentration-dependent increase in the mRNA expression of M2 (anti-inflammatory) phenotype markers (CD206, Arg-1, Fizz-1, and Ym-1) in RAW 264.7 cells. Moreover, AR extract suppressed DSS-induced ROS generation and mitochondrial dysfunction in Caco-2 cells. The in vivo experiment revealed that AR extract (200 mg/kg) increased colon length compared to the DSS-treated group. In addition, disease activity index, spleen ratio, body weight, oxidative stress, and colonic inflammation were markedly improved by AR treatment in DSS-induced UC mice. Finally, AR suppressed M1 and promoted M2 macrophage polarization in UC mice. CONCLUSION The AR extract protected against DSS-induced UC by regulating macrophage polarization and suppressing oxidative stress. These valuable findings suggest that adequate intake of AR can prevent and/or treat UC.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China; The Research Centre for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Xi Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Polly Ho-Ting Shiu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yanfen Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Nie
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Panthakarn Rangsinth
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Benson Wui Man Lau
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Chengwen Zheng
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xuebo Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Renkai Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Simon Ming-Yuen Lee
- Department of Food Science and Nutrition, Faculty of Science, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sai-Wang Seto
- Department of Food Science and Nutrition, Faculty of Science, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China; The Research Centre for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China.
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
48
|
Laveriano-Santos EP, Luque-Corredera C, Trius-Soler M, Lozano-Castellón J, Dominguez-López I, Castro-Barquero S, Vallverdú-Queralt A, Lamuela-Raventós RM, Pérez M. Enterolignans: from natural origins to cardiometabolic significance, including chemistry, dietary sources, bioavailability, and activity. Crit Rev Food Sci Nutr 2024:1-21. [PMID: 38952149 DOI: 10.1080/10408398.2024.2371939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
The enterolignans, enterolactone and enterodiol, the main metabolites produced from plant lignans by the gut microbiota, have enhanced bioavailability and activity compared to their precursors, with beneficial effects on metabolic and cardiovascular health. Although extensively studied, the biosynthesis, cardiometabolic effects, and other therapeutic implications of mammalian lignans are still incompletely understood. The aim of this review is to provide a comprehensive overview of these phytoestrogen metabolites based on up-to-date information reported in studies from a wide range of disciplines. Established and novel synthetic strategies are described, as are the various lignan precursors, their dietary sources, and a proposed metabolic pathway for their conversion to enterolignans. The methodologies used for enterolignan analysis and the available data on pharmacokinetics and bioavailability are summarized and their cardiometabolic bioactivity is explored in detail. The special focus given to research on the health benefits of microbial-derived lignan metabolites underscores the critical role of lignan-rich diets in promoting cardiovascular health.
Collapse
Affiliation(s)
- Emily P Laveriano-Santos
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | | | - Marta Trius-Soler
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Julian Lozano-Castellón
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Inés Dominguez-López
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Sara Castro-Barquero
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, Barcelona, Spain
| | - Anna Vallverdú-Queralt
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Rosa M Lamuela-Raventós
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Maria Pérez
- Department of Nutrition, Food Science and Gastronomy, XIA, Faculty of Pharmacy and Food Sciences, Polyphenol Research Group, University of Barcelona, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Santa Coloma de Gramanet, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
49
|
Lundrigan E, Toudic C, Pennock E, Pezacki JP. SARS-CoV-2 Protein Nsp9 Is Involved in Viral Evasion through Interactions with Innate Immune Pathways. ACS OMEGA 2024; 9:26428-26438. [PMID: 38911767 PMCID: PMC11191075 DOI: 10.1021/acsomega.4c02631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/13/2024] [Accepted: 04/23/2024] [Indexed: 06/25/2024]
Abstract
The suppression of the host's innate antiviral immune response by SARS-CoV-2, a contributing factor to the severity of disease, has been considerably studied in recent years. Many of these studies have focused on the actions of the structural proteins of the virus because of their accessibility to host immunological components. However, less is known about SARS-CoV-2 nonstructural and accessory proteins in relation to viral evasion. Herein, we study SARS-CoV-2 nonstructural proteins Orf3a, Orf6, and Nsp9 in a mimicked virus-infected state using poly(I:C), a synthetic analog of viral dsRNA, that elicits the antiviral immune response. Through genome-wide expression profiling, we determined that Orf3a, Orf6, and Nsp9 all modulate the host antiviral signaling transcriptome to varying extents, uniquely suppressing aspects of innate immune signaling. Our data suggest that SARS-CoV-2 Nsp9 hinders viral detection through suppression of RIG-I expression and antagonizes the interferon antiviral cascade by downregulating NF-kB and TBK1. Our data point to unique molecular mechanisms through which the different SARS-CoV-2 proteins suppress immune signaling and promote viral evasion. Nsp9 in particular acts on major elements of the host antiviral pathways to impair the antiviral immune response.
Collapse
Affiliation(s)
- Eryn Lundrigan
- Department of Chemistry and
Biomolecular Sciences, University of Ottawa, Ottawa K1N 6N5, Canada
| | - Caroline Toudic
- Department of Chemistry and
Biomolecular Sciences, University of Ottawa, Ottawa K1N 6N5, Canada
| | - Emily Pennock
- Department of Chemistry and
Biomolecular Sciences, University of Ottawa, Ottawa K1N 6N5, Canada
| | - John Paul Pezacki
- Department of Chemistry and
Biomolecular Sciences, University of Ottawa, Ottawa K1N 6N5, Canada
| |
Collapse
|
50
|
Pasamba EC, Orda MA, Villanueva BHA, Tsai PW, Tayo LL. Transcriptomic Analysis of Hub Genes Reveals Associated Inflammatory Pathways in Estrogen-Dependent Gynecological Diseases. BIOLOGY 2024; 13:397. [PMID: 38927277 PMCID: PMC11201105 DOI: 10.3390/biology13060397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024]
Abstract
Gynecological diseases are triggered by aberrant molecular pathways that alter gene expression, hormonal balance, and cellular signaling pathways, which may lead to long-term physiological consequences. This study was able to identify highly preserved modules and key hub genes that are mainly associated with gynecological diseases, represented by endometriosis (EM), ovarian cancer (OC), cervical cancer (CC), and endometrial cancer (EC), through the weighted gene co-expression network analysis (WGCNA) of microarray datasets sourced from the Gene Expression Omnibus (GEO) database. Five highly preserved modules were observed across the EM (GSE51981), OC (GSE63885), CC (GSE63514), and EC (GSE17025) datasets. The functional annotation and pathway enrichment analysis revealed that the highly preserved modules were heavily involved in several inflammatory pathways that are associated with transcription dysregulation, such as NF-kB signaling, JAK-STAT signaling, MAPK-ERK signaling, and mTOR signaling pathways. Furthermore, the results also include pathways that are relevant in gynecological disease prognosis through viral infections. Mutations in the ESR1 gene that encodes for ERα, which were shown to also affect signaling pathways involved in inflammation, further indicate its importance in gynecological disease prognosis. Potential drugs were screened through the Drug Repurposing Encyclopedia (DRE) based on the up-and downregulated hub genes, wherein a bacterial ribosomal subunit inhibitor and a benzodiazepine receptor agonist were the top candidates. Other drug candidates include a dihydrofolate reductase inhibitor, glucocorticoid receptor agonists, cholinergic receptor agonists, selective serotonin reuptake inhibitors, sterol demethylase inhibitors, a bacterial antifolate, and serotonin receptor antagonist drugs which have known anti-inflammatory effects, demonstrating that the gene network highlights specific inflammatory pathways as a therapeutic avenue in designing drug candidates for gynecological diseases.
Collapse
Affiliation(s)
- Elaine C. Pasamba
- School of Graduate Studies, Mapúa University, Manila City 1002, Philippines; (E.C.P.); (M.A.O.); (B.H.A.V.)
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines
| | - Marco A. Orda
- School of Graduate Studies, Mapúa University, Manila City 1002, Philippines; (E.C.P.); (M.A.O.); (B.H.A.V.)
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines
| | - Brian Harvey Avanceña Villanueva
- School of Graduate Studies, Mapúa University, Manila City 1002, Philippines; (E.C.P.); (M.A.O.); (B.H.A.V.)
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines
| | - Po-Wei Tsai
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan;
| | - Lemmuel L. Tayo
- School of Graduate Studies, Mapúa University, Manila City 1002, Philippines; (E.C.P.); (M.A.O.); (B.H.A.V.)
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines
- Department of Biology, School of Health Sciences, Mapúa University, Makati City 1203, Philippines
| |
Collapse
|