1
|
Dos Santos JM, Marangoni Faoro JA, Fava de Souza M, de Matos Balsalobre N, Leite Kassuya CA, Kappel Trichez VD, Mussury Franco da Silva RM, Formagio ASN. Anti-arthritic potential and antioxidant properties of infusion, fractions and flavonoid glycosides from Dipteryx alata (baru) leaves. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:118973. [PMID: 39454705 DOI: 10.1016/j.jep.2024.118973] [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: 07/17/2024] [Revised: 10/13/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dipteryx alata Vogel., popularly known as "baru", is a native species of Brazilian cerrado used by "Ribeirinhos" in the North Araguaia microregion. In the traditional medicine, maceration of barks or leaves infusion are used to treat back and muscle pain, osteoporosis and rheumatism. However, few studies have demonstrated the pharmacological effects of this species. AIM OF THE STUDY The goal of this study was to perform phytochemicals studies of lyophilized infusion of D. alata leaves (LI-DA), as well as obtaining ethyl acetate fraction (EAF-DA) and hydromethanolic fraction (HMF-DA), and isolated flavonoids. The antioxidant of LI-DA, EAF-DA and HMF-DA, anti-inflammatory effects of LI-DA and quercetin-3-O-β-glucoside-7-O-α-rhamnoside (DA-1) and quercetin-7-O-α-rhamnoside (DA-2) were performed while in silico tests were used for absorption, distribution, metabolism, excretion and toxicity predictions of DA-1 and DA-2. MATERIALS AND METHODS LI-DA, EAF-DA and HMF-DA were evaluated in antioxidant assays (2, 2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid - ABTS; 2,2-diphenyl-1-picrylhydrazyl - DPPH; hydrogen peroxide - H2O2; reducing power and oxidation of β-carotene). The DA-1 and DA-2 were isolated from EAF-DA using chromatographic methods and characterized by Nuclear Magnetic Resonance (NMR) spectrometer. The Programs ProTox 3.0 and ADMETlab 2.0 were used for the prediction studies of DA-1 and DA-2. Mice received a single dose of LI-DA (3, 30, and 100 mg/kg), DA-1 (3 mg/kg) and DA-2 (3 mg/kg) and were subjected to inflammation induced by Complete Freund's Adjuvant (CFA) and in the zymosan-induced articular inflammation model. RESULTS DA-1 and DA-2 have been identified for the first time in the leaves of D. alata. LI-DA, EAF-DA and HMF-DA demonstrated a high level of antioxidant activity as measured by ABTS (IC50 ≤ 5.62 μg/mL) and DPPH (IC50 ≤ 11.45 μg/mL). Oral administration of LI-DA (3, 30 and 100 mg/kg), DA-1 (3 mg/kg) and DA-2 (3 mg/kg) showed significantly reduced edema, cold and mechanical allodynia in the CFA-induced inflammation model (24 h). LI-DA (3, 30, and 100 mg/kg) and DA-1 (3 mg/kg) reduced leukocytes migration into the joint cavity, mechanical allodynia, edema and NO production in mice (24 h) in the zymosan-induced articular inflammation model. Additionally, DA-2 (3 mg/kg) reduced leukocyte migration and LI-DA (30 mg/kg) reduced protein exudation (24 h) in zymosan model. DA-1 and DA-2 showed good oral bioavailability and low toxicity predicted by the ProTox model. CONCLUSION This is the first chemical and biological study performed of D. alata infusion and two quercetin glycoside derivatives. The results indicated promising potential for the treatment of inflammation, pain, and rheumatism, supporting the traditional use of the infusion obtained from the leaves of D. alata.
Collapse
Affiliation(s)
- Jéssica Maurino Dos Santos
- Federal University of Grande Dourados - UFGD, Faculty of Health Sciences, Postgraduate Program in Health Sciences, 79804-970, Dourados, Mato Grosso do Sul, Brazil.
| | - Janaine Alberto Marangoni Faoro
- Federal University of Grande Dourados - UFGD, Faculty of Health Sciences, Postgraduate Program in Health Sciences, 79804-970, Dourados, Mato Grosso do Sul, Brazil.
| | - Maiara Fava de Souza
- Federal University of Grande Dourados - UFGD, Faculty of Health Sciences, Postgraduate Program in Health Sciences, 79804-970, Dourados, Mato Grosso do Sul, Brazil.
| | - Natalia de Matos Balsalobre
- Federal University of Grande Dourados - UFGD, Faculty of Health Sciences, Postgraduate Program in Health Sciences, 79804-970, Dourados, Mato Grosso do Sul, Brazil.
| | - Candida Aparecida Leite Kassuya
- Federal University of Grande Dourados - UFGD, Faculty of Health Sciences, Postgraduate Program in Health Sciences, 79804-970, Dourados, Mato Grosso do Sul, Brazil.
| | - Virginia Demarchi Kappel Trichez
- Federal University of Grande Dourados - UFGD, Faculty of Health Sciences, Postgraduate Program in Health Sciences, 79804-970, Dourados, Mato Grosso do Sul, Brazil.
| | - Rosilda Mara Mussury Franco da Silva
- Federal University of Grande Dourados - UFGD, Faculty of Biological and Environmental Sciences, 79804-970, Dourados, Mato Grosso do Sul, Brazil.
| | - Anelise Samara Nazari Formagio
- Federal University of Grande Dourados - UFGD, Faculty of Health Sciences, Postgraduate Program in Health Sciences, 79804-970, Dourados, Mato Grosso do Sul, Brazil.
| |
Collapse
|
2
|
Feng S, Tang J, Wei X, Lu Z, Xu Y, Zhang T, Han H. Swertia cincta and its main active ingredients regulate the PPAR-α pathway in anti-cholestatic liver injury. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118956. [PMID: 39423946 DOI: 10.1016/j.jep.2024.118956] [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: 07/19/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Swertia cincta is a traditional remedy for cholestasis commonly utilised in Yunnan, China. Despite its widespread use, the specific active components and underlying mechanisms of action remain poorly understood. AIM OF THIS STUDY This study aimed to investigate the therapeutic properties, mechanisms, and active compounds of Swertia cincta in an animal model of cholestasis induced by alpha-naphthylisothiocyanate (ANIT). MATERIALS AND METHODS UHPLC/Q-TOF-MS and high-performance liquid chromatography (HPLC) were utilised to analyse the blood components of Swertia cincta. An ANIT-induced cholestatic liver injury animal model was established, and metabolomics was employed to explore the potential mechanisms of Swertia cincta in treating cholestatic liver injury. Hepatocellular injury induced by taurochenodeoxycholic acid was evaluated in vitro, and key bioactive components of Swertia cincta for cholestatic liver injury treatment were identified and confirmed using the ANIT-induced mouse model. RESULTS The established HPLC method demonstrates good specificity and reproducibility, enabling the simultaneous determination of six components in Swertia cincta. Results from serum biochemical indicators and liver pathology analysis indicated that Swertia cincta exhibits promising anti-cholestasis liver injury effects. Specifically, gentiopicroside, loganic acid, and isoorientin were identified as key active ingredients in treating cholestatic liver injury. Their mechanism of action primarily involves regulating PPAR-α, FXR, CYP3A4, NTCP, CAR, and CPT2. By modulating PPAR-α and bile acid metabolism-related proteins, reducing pro-inflammatory factors, enhancing bile acid transport, and promoting fatty acid oxidation to reduce lipid accumulation, Swertia cincta exerts protective and therapeutic effects against cholestatic liver injury. Notably, gentian bitter glycosides appear to be the most critical components for this effect. CONCLUSION Swertia cincta may improve cholestatic liver injury by activating the peroxisome proliferator-activated receptor alpha pathway, and the key active compounds were gentiopicroside, loganic acid, and isoorientin.
Collapse
Affiliation(s)
- Shuaixia Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Jie Tang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Xia Wei
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Zou Lu
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Ying Xu
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Han Han
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| |
Collapse
|
3
|
Zhu J, Zhang Y, Sun Y, Yu F, Lu Y, Hu Q, Guo J, Zhang H, Chen T, Lian F, Wang J, Li X, Xiao J. Mesoporous Prussian blue nanoparticle neuroconduit for the biological therapy targeting oxidative stress reduction, inflammation inhibition, and nerve regeneration. J Nanobiotechnology 2025; 23:1. [PMID: 39743507 DOI: 10.1186/s12951-024-02937-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/16/2024] [Indexed: 01/04/2025] Open
Abstract
The applications of nanomaterials in regenerative medicine encompass a broad spectrum. The functional nanomaterials, such as Prussian blue and its derivative nanoparticles, exhibit potent anti-inflammatory and antioxidant properties. By combining it with the corresponding scaffold carrier, the fusion of nanomaterials and biotherapy can be achieved, thereby providing a potential avenue for clinical treatment. The present study demonstrates the fabrication of a Mesoporous Prussian blue nanoparticles (MPBN) functionalized Inverse Opal Film (IOF) neuroconduit for peripheral nerve repair through reverse replication and freeze-drying techniques. The binding of MPBN to the neuroconduit can effectively decreasing reactive oxygen species and inflammatory factors in the vicinity of the residual nerve, thereby providing protective effects on the damaged nerve. Furthermore, comprehensive behavioral, electrophysiological, and pathological analyses unequivocally substantiate the efficacy of MPBN in increasing nerve structure regeneration and ameliorating denervation-induced myopathy. Moreover, MPBN enhances the antioxidant capacity of Schwann cells by activating the AMPK/SIRT1/PGC-1 pathway. The findings suggest that MPBN, a biocompatible nanoparticle, can safeguard damaged nerves by optimizing the microenvironment surrounding nerve cells and augmenting the antioxidant capacity of nerve cells, thereby facilitating nerve regeneration and repair. This also establishes a theoretical foundation for exploring the integration and clinical translation between nanomaterials and biotherapy.
Collapse
Affiliation(s)
- Junyi Zhu
- Department of Hand Surgery and Peripheral Neurosurgery, Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yijia Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yinuo Sun
- Department of Hand Surgery and Peripheral Neurosurgery, Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Fangzheng Yu
- Department of Hand Surgery and Peripheral Neurosurgery, Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yang Lu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Qianqian Hu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Jiali Guo
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Haijuan Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Tianling Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Feifei Lian
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Jian Wang
- Department of Hand Surgery and Peripheral Neurosurgery, Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaokun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Jian Xiao
- Department of Hand Surgery and Peripheral Neurosurgery, Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
4
|
Wang F, Zhang M, Yin L, Zhou Z, Peng Z, Li W, Chen H, Yu G, Tang J. The tryptophan metabolite kynurenic acid ameliorates septic colonic injury through activation of the PPARγ signaling pathway. Int Immunopharmacol 2024; 147:113651. [PMID: 39742725 DOI: 10.1016/j.intimp.2024.113651] [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: 09/12/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 01/04/2025]
Abstract
Sepsis is the leading cause of death among critically ill patients in clinical practice, making it urgent to reduce its incidence and mortality rates. In sepsis, macrophage dysfunction often worsens and complicates the condition. M1 and M2 macrophages, two distinct types, contribute to pro-inflammatory and anti-inflammatory effects, respectively. An imbalance between them is a major cause of sepsis. The aim of this study was to explore the potential of a differential metabolite between M1 and M2 macrophages in mitigating septic colonic injury via multiomics in combination with clinical data and animal experiments. Using nontargeted metabolomics analysis, we found that Kynurenic acid (KYNA), a metabolite of tryptophan metabolism, was significantly upregulated in the supernatant of M2 macrophages. Furthermore, we discovered that the level of KYNA was significantly decreased in sepsis in both human and mouse serum and was negatively correlated with inflammatory factor levels. In vivo experiments demonstrated that KYNA can effectively alleviate septic colon injury and reduce inflammatory factor levels in mice, indicating that KYNA plays a very important protective role in sepsis. Mechanistically, KYNA promotes the transition of M1 macrophages to M2 macrophages by inhibiting the NF-κB signaling pathway and alleviates septic colonic injury through the PPARγ/NF-κB axis. This article reveals that KYNA, a differentially abundant metabolite between M1 and M2 macrophages, can become a new strategy for alleviating septic colon injury.
Collapse
Affiliation(s)
- Fei Wang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, China
| | - Meng Zhang
- Department of Pneumology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, China
| | - Liping Yin
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, China
| | - Ziyang Zhou
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, China
| | - Ziyao Peng
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, China
| | - Wenweiran Li
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, China
| | - Hui Chen
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, China.
| | - Guohong Yu
- Department of Emergency Medicine, Baoshan Second People's Hospital, Baoshan College of Traditional Chinese Medicine, 13 Zhengyang South Road, Baoshan, Yunnan 678000, China.
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, China.
| |
Collapse
|
5
|
Meng L, Gu T, Yu P, Zhang Z, Wei Z. The role of microglia in Neuroinflammation associated with cardiopulmonary bypass. Front Cell Neurosci 2024; 18:1496520. [PMID: 39742156 PMCID: PMC11685197 DOI: 10.3389/fncel.2024.1496520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Cardiopulmonary bypass (CPB) and deep hypothermic circulatory arrest (DHCA) are indispensable core techniques in cardiac surgery. Numerous studies have shown that cardiopulmonary bypass and deep hypothermic circulatory arrest are associated with the occurrence of neuroinflammation, accompanied by the activation of microglia. Microglia, as macrophages in the central nervous system, play an irreplaceable role in neuroinflammation. Current research on neuroinflammation induced by microglia activation mainly focuses on neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, neuropathic pain, acquired brain injury, and others. However, there is relatively limited research on microglia and neuroinflammation under conditions of cardiopulmonary bypass and deep hypothermic circulatory arrest. The close relationship between cardiopulmonary bypass, deep hypothermic circulatory arrest, and cardiac surgery underscores the importance of identifying targets for intervening in neuroinflammation through microglia. This could greatly benefit cardiac surgery patients during cardiopulmonary bypass and the perioperative period, significantly improving patient prognosis. This review article provides the first comprehensive discussion on the signaling pathways associated with neuroinflammation triggered by microglia activation, the impact of cardiopulmonary bypass on microglia, as well as the current status and advancements in cardiopulmonary bypass animal models. It provides new insights and methods for the treatment of neuroinflammation related to cardiopulmonary bypass and deep hypothermic circulatory arrest, holding significant importance for clinical treatment by cardiac surgeons, management strategies by cardiopulmonary bypass physicians, and the development of neurologically related medications.
Collapse
Affiliation(s)
- Lingda Meng
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Tianxiang Gu
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Peng Yu
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhiwei Zhang
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhijing Wei
- Department of Trauma Center, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Siddiqui S, Liu F, Kanthasamy AG, McGrail M. Stat3 mediates Fyn kinase-driven dopaminergic neurodegeneration and microglia activation. Dis Model Mech 2024; 17:dmm052011. [PMID: 39641161 PMCID: PMC11646115 DOI: 10.1242/dmm.052011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/17/2024] [Indexed: 12/07/2024] Open
Abstract
The Alzheimer's disease and Parkinson's disease risk locus FYN kinase is implicated in neurodegeneration and inflammatory signaling. To investigate in vivo mechanisms of Fyn-driven neurodegeneration, we built a zebrafish neural-specific Gal4:UAS model of constitutively active FynY531F signaling. Using in vivo live imaging, we demonstrated that neural FynY531F expression leads to dopaminergic neuron loss and mitochondrial aggregation in 5 day larval brain. Dopaminergic loss coincided with microglia activation and induction of tnfa, il1b and il12a inflammatory cytokine expression. Transcriptome analysis revealed Stat3 signaling as a potential Fyn target. Chemical inhibition experiments confirmed Fyn-driven dopaminergic neuron loss, and the inflammatory response was dependent upon activation of Stat3 and NF-κB pathways. Dual chemical inhibition demonstrated that Stat3 acts synergistically with NF-κB in dopaminergic neuron degeneration. These results identify Stat3 as a novel downstream effector of Fyn signaling in neurodegeneration and inflammation.
Collapse
Affiliation(s)
- Sahiba Siddiqui
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, USA
- Interdepartmental Genetics and Genomics Graduate Program (IGG), Iowa State University, Ames, IA 50011, USA
| | - Fang Liu
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, USA
| | - Anumantha G. Kanthasamy
- Center for Brain Science and Neurodegenerative Diseases, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA
| | - Maura McGrail
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, USA
- Interdepartmental Genetics and Genomics Graduate Program (IGG), Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
7
|
Li P, Zhao S, Teng Y, Han S, Yang Y, Wu M, Guo S, Ding B, Xiao L, Yi D. Dietary supplementary with ellagic acid improves the intestinal barrier function and flora structure of broiler chicken challenged with E. coli K88. Poult Sci 2024; 103:104429. [PMID: 39461273 PMCID: PMC11544055 DOI: 10.1016/j.psj.2024.104429] [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: 08/07/2024] [Revised: 09/26/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024] Open
Abstract
Ellagic acid (EA) contributes to the immunity and anti-oxidant function of body, whereas there are few reports about its effect on the intestinal health and growth performance of broiler chickens. Hence, the present study was arranged to investigate the effect of dietary supplementary with EA on the intestinal barrier function and flora structure of broiler chickens challenged with Escherichia coli K88 (E. coli K88). A total of 216 healthy 1-day-old, Ross 308 broilers with uniform weight were randomly assigned into three treatment groups, six replicates in each group and twelve birds in each replicate. Broilers in the control (CTR) group and E. coli K88 infected group (ETEC) were fed with the basic diet, and 200 mg/kg EA was supplemented into the diet of the E. coli K88 infected group treated with EA (EAETEC). The animal trial had lasted for 42 days, and the outcomes showed that the ADG and ADFI during the animal trial, the jejunal villi height (VH) and the ratio of VH to crypt depth (CD) tended to be decreased with E. coli K88 treated (P< 0.05). Additionally, the level of serum diamine oxidase (DAO) and intestinal malondialdehyde (MDA) were elevated, the activity of intestinal total superoxide dismutase (T-SOD) and glutathione peroxidase (GSH-Px), the mRNA levels in jejunal claudin-1 and occludin were down-regulated with E. coli K88 treated as well as the transcription levels of ileal Mucin-2, aquaporin-3 (AQP-3) and Na+/H+ exchanger proteins-3 (NHE-3) (P< 0.05). In addition, E. coli K88 down-regulated the α-diversity index of cecal flora, the ratio of Bacteroidota to Firmicutes and the relative abundance of Barnesiella were up-regulated and it of Alistipes was descended with E. coli K88 treated (P< 0.05). Beyond that, the content of propionic acid in the cecal chyme was decreased and the amino acid metabolic pathways were inhibited with E. coli K88 challenged (P< 0.05). Additionally, there was a significant positive correlation between the relative abundance of Alistipes and the levels of butyric acid in the caecal chyme and the activity of GSH-Px in the intestine (P< 0.05). Interestingly, dietary supplementary with EA could reshape the intestinal flora structure and alleviate the above negative effects of E. coli K88 on broiler chickens. In conclusion, dietary supplementary with ellagic acid improved the intestinal barrier function and flora structure of broiler chickens challenged with E. coli K88.
Collapse
Affiliation(s)
- Peng Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China; Engineering Research Center of Feed Protein Resources of Agricultural By-products, Ministry of Education,Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China
| | - Shengnan Zhao
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China; Engineering Research Center of Feed Protein Resources of Agricultural By-products, Ministry of Education,Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China
| | - Yi Teng
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China; Engineering Research Center of Feed Protein Resources of Agricultural By-products, Ministry of Education,Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China
| | - Shaochen Han
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China; Engineering Research Center of Feed Protein Resources of Agricultural By-products, Ministry of Education,Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China
| | - Yuzhu Yang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China; Engineering Research Center of Feed Protein Resources of Agricultural By-products, Ministry of Education,Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China
| | - Mengjun Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China; Engineering Research Center of Feed Protein Resources of Agricultural By-products, Ministry of Education,Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China
| | - Shuangshuang Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China; Engineering Research Center of Feed Protein Resources of Agricultural By-products, Ministry of Education,Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China
| | - Bingying Ding
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China; Engineering Research Center of Feed Protein Resources of Agricultural By-products, Ministry of Education,Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China
| | - Lei Xiao
- Hubei Lan Good microbial Technology Co., Ltd. Yichang, Hubei 443100, PR China
| | - Dan Yi
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China; Engineering Research Center of Feed Protein Resources of Agricultural By-products, Ministry of Education,Wuhan Polytechnic University, Wuhan, Hubei 430023, PR China.
| |
Collapse
|
8
|
Kou Y, Jing Q, Yan X, Chen J, Shen Y, Ma Y, Xiang Y, Li X, Liu X, Liu Z, Wei Y, Wang Y. 4-Hydroxybenzoic acid restrains Nlrp3 inflammasome priming and activation via disrupting PU.1 DNA binding activity and direct antioxidation. Chem Biol Interact 2024; 404:111262. [PMID: 39389441 DOI: 10.1016/j.cbi.2024.111262] [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: 06/25/2024] [Revised: 08/30/2024] [Accepted: 10/08/2024] [Indexed: 10/12/2024]
Abstract
Reactive oxygen species (ROS) production is considered central to triggering the nucleotide-binding domain-like receptor family pyrin domain containing 3 (Nlrp3) inflammasome activation and the subsequent inflammatory responses. Coenzyme Q10 (CoQ10) plays a critical role in maintaining intracellular ROS homeostasis and inhibiting excessive Nlrp3 inflammasome activation. However, direct supplementation of CoQ10 showed unsatisfactory clinical improvement due to its limited absorption and bioavailability. Therefore, stimulating endogenous CoQ10 biosynthesis by supplementing CoQ10 precursors may provide a more promising therapeutic approach. In this study, we described the role of 4-hydroxybenzoic acid (4-HBA), a precursor of CoQ10, in attenuating excessive inflammatory responses. We found that while supplementation of 4-HBA inhibited the priming and activation of Nlrp3 inflammasome, this effect was independent of its metabolic transformation into CoQ10. 4-HBA itself exhibits antioxidative activities. Furthermore, 4-HBA can disrupt the binding activity of PU.1 on the promoters of Tlr4 and Md2, thereby directly suppressing Nlrp3 inflammasome priming during LPS-induced inflammatory responses. Therefore, strategically utilizing 4-HBA or increasing 4-HBA intake may represent a potential strategy for reducing excessive inflammation.
Collapse
Affiliation(s)
- Yanbo Kou
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Qiyue Jing
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Xiaoqing Yan
- Laboratory of Emergency Medicine, Second Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Junru Chen
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Yusi Shen
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Yulu Ma
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Yaoyao Xiang
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Xiangyang Li
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Xiangye Liu
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Zhuanzhuan Liu
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Yanxia Wei
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China
| | - Yugang Wang
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, China; Jiangsu International Laboratory of Immunity and Metabolism, China.
| |
Collapse
|
9
|
Jindal J, Hill J, Harte J, Dunachie SJ, Kronsteiner B. Starvation and infection: The role of sickness-associated anorexia in metabolic adaptation during acute infection. Metabolism 2024; 161:156035. [PMID: 39326837 DOI: 10.1016/j.metabol.2024.156035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Sickness-associated anorexia, the reduction in appetite seen during infection, is a widely conserved and well-recognized symptom of acute infection, yet there is very little understanding of its functional role in recovery. Anorexic sickness behaviours can be understood as an evolutionary strategy to increase tolerance to pathogen-mediated illness. In this review we explore the evidence for mechanisms and potential metabolic benefits of sickness-associated anorexia. Energy intake can impact on the immune response, control of inflammation and tissue stress, and on pathogen fitness. Fasting mediators including hormone-sensitive lipase, peroxisome proliferator-activated receptor-alpha (PPAR-α) and ketone bodies are potential facilitators of infection recovery through multiple pathways including suppression of inflammation, adaptation to lipid utilising pathways, and resistance to pathogen-induced cellular stress. However, the effect and benefit of calorie restriction is highly heterogeneous depending on both the infection and the metabolic status of the host, which has implications regarding clinical recommendations for feeding during different infections.
Collapse
Affiliation(s)
- Jessy Jindal
- The Medical School, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Jennifer Hill
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Jodie Harte
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Susanna J Dunachie
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.
| | - Barbara Kronsteiner
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
10
|
Fernandes-da-Silva A, Santana-Oliveira DA, Oliveira ASD, Ferreira TAM, Monteiro NC, Silva-Veiga FM, Martins FF, Cummins CL, Romeiro LAS, Souza-Mello V. LDT409 (pan-PPAR partial agonist) mitigates metabolic dysfunction-associated steatotic liver disease in high-fructose-fed mice. Mol Cell Endocrinol 2024; 594:112380. [PMID: 39332468 DOI: 10.1016/j.mce.2024.112380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 09/29/2024]
Abstract
AIM This study sought to evaluate the effects of LDT409, a pan-PPAR partial agonist obtained from the main industrial waste from cashew nut processing, on hepatic remodeling, highlighting energy metabolism and endoplasmic reticulum (ER) stress in high-fructose-fed mice. METHODS Male C57BL/6 mice received a control diet (C) or a high-fructose diet (HFRU) for ten weeks. Then, a five-week treatment started: C, C-LDT409, HFRU, and HFRU-LDT409. The LDT409 (40 mg/kg of body weight) was mixed with the diets. RESULTS The HFRU diet caused insulin resistance and endoplasmic reticulum (ER) stress. High Pparg and decreased Ppara expression increased steatosis and pro-fibrogenic gene expression in livers of HFRU-fed mice. Suppressed lipogenic factors, orchestrated by PPAR-gamma, and mitigated ER stress concomitant with the increase in beta-oxidation driven by PPAR-alpha mediated the LDT409 beneficial effects. CONCLUSIONS LDT409 may represent a potential low-cost approach to treat metabolic dysfunction-associated steatotic liver disease, which does not currently have a specific treatment.
Collapse
Affiliation(s)
- Aline Fernandes-da-Silva
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daiana Araujo Santana-Oliveira
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andressa S de Oliveira
- Graduate Program in Pharmaceutical Sciences, Department of Pharmacy, Health Sciences Faculty, University of Brasília, Brasília, DF, Brazil; Laboratory of Development of Therapeutic Innovations (LDT), Center for Tropical Medicine, Faculty of Medicine, University of Brasília, Brasília, DF, Brazil
| | - Thaís A M Ferreira
- Graduate Program in Pharmaceutical Sciences, Department of Pharmacy, Health Sciences Faculty, University of Brasília, Brasília, DF, Brazil; Laboratory of Development of Therapeutic Innovations (LDT), Center for Tropical Medicine, Faculty of Medicine, University of Brasília, Brasília, DF, Brazil
| | - Natália Cipriano Monteiro
- Graduate Program in Pharmaceutical Sciences, Department of Pharmacy, Health Sciences Faculty, University of Brasília, Brasília, DF, Brazil; Laboratory of Development of Therapeutic Innovations (LDT), Center for Tropical Medicine, Faculty of Medicine, University of Brasília, Brasília, DF, Brazil
| | - Flávia Maria Silva-Veiga
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiane Ferreira Martins
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Luiz Antonio Soares Romeiro
- Graduate Program in Pharmaceutical Sciences, Department of Pharmacy, Health Sciences Faculty, University of Brasília, Brasília, DF, Brazil; Laboratory of Development of Therapeutic Innovations (LDT), Center for Tropical Medicine, Faculty of Medicine, University of Brasília, Brasília, DF, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
11
|
Sattari M, Amri J, Shahaboddin ME, Sattari M, Tabatabaei-Malazy O, Azmon M, Meshkani R, Panahi G. The protective effects of fisetin in metabolic disorders: a focus on oxidative stress and associated events. J Diabetes Metab Disord 2024; 23:1753-1771. [PMID: 39610486 PMCID: PMC11599505 DOI: 10.1007/s40200-024-01502-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/09/2024] [Indexed: 11/30/2024]
Abstract
Abstract Metabolic syndrome is increasingly recognized as a significant precursor to various chronic diseases, contributing to a growing public health concern. Its complex pathogenesis involves multiple interrelated mechanisms, with oxidative stress identified as a cornerstone that exacerbates other pathogenic pathways. This study elucidates the molecular mechanisms by which oxidative stress intensifies metabolic disturbances, particularly insulin resistance. Some recent research has focused on fisetin, a natural product known for its potential benefits in diabetes and its associated microvascular and macrovascular complications. This paper compiles a comprehensive collection of findings by reviewing studies conducted over the past decade, detailing dosages, investigated markers, and their respective outcomes. Notably, a recurrent finding was fisetin's ability to enhance Nrf2, a principal regulator of antioxidant defense, in both metabolic and non-metabolic diseases. Furthermore, intriguing results suggest that the effects of Nrf2 extend beyond oxidative stress modulation, demonstrating favorable impacts on tissue-specific functions in metabolic regulation. This highlights fisetin not only as an antioxidant but also as a potential therapeutic agent for improving metabolic health and mitigating the effects of metabolic syndrome. In conclusion, fisetin can enhance the body's antioxidant defenses by modulating the Nrf2 pathway while also improving metabolic health through its effects on inflammation, cell survival, and energy metabolism, offering a comprehensive approach to managing metabolic disorders. Graphical Abstract
Collapse
Affiliation(s)
- Mahboobe Sattari
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, I.R Iran
| | - Jamal Amri
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, I.R Iran
| | - Mohammad Esmaeil Shahaboddin
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohadese Sattari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ozra Tabatabaei-Malazy
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzyeh Azmon
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghodratollah Panahi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Galgani G, Bray G, Martelli A, Calderone V, Citi V. In Vitro Models of Diabetes: Focus on Diabetic Retinopathy. Cells 2024; 13:1864. [PMID: 39594613 PMCID: PMC11592768 DOI: 10.3390/cells13221864] [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: 10/14/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Diabetic retinopathy is a major eye complication in patients with diabetes mellitus, and it is the leading cause of blindness and visual impairment in the world. Chronic hyperglycemia induces endothelial damage with consequent vascular lesions, resulting in global vasculitis, which affects the small vessels of the retina. These vascular lesions cause ischemic conditions in certain areas of the retina, with a consequent increase in the release of pro-angiogenic mediators. In addition to pharmacological interventions for controlling the blood glycaemic level, the main strategies for treating diabetic retinopathy are the intravitreal injections of drugs, surgical treatments, and vitrectomies. The complexity of diabetic retinopathy is due to its close interactions with different cell types (endothelial cells, astrocytes, and Müller cells). The evaluation of the efficacy of novel pharmacological strategies is mainly performed through in vivo models. However, the use of different animal species leads to heterogenic results and ethical concerns. For these reasons, the development of new and reliable in vitro models, such as cell co-cultures and eye organoids, represents an urgent need in this area of research. This review features an overview of the in vitro models used to date and highlights the advances in technology used to study this pathology.
Collapse
Affiliation(s)
- Giulia Galgani
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
| | - Giorgia Bray
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
- Interuniversity Centre for the Promotion of the 3R Principles in Teaching and Research, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
- Interuniversity Centre for the Promotion of the 3R Principles in Teaching and Research, Italy
| | - Valentina Citi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
- Interuniversity Centre for the Promotion of the 3R Principles in Teaching and Research, Italy
| |
Collapse
|
13
|
Fadaly WAA, Elshewy A, Nemr MTM, Abdou K, Sayed AM, Kahk NM. Discovery of novel thiazole derivatives containing pyrazole scaffold as PPAR-γ Agonists, α-Glucosidase, α-Amylase and COX-2 inhibitors; Design, synthesis and in silico study. Bioorg Chem 2024; 152:107760. [PMID: 39197383 DOI: 10.1016/j.bioorg.2024.107760] [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: 06/19/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
A novel series of thiazole derivatives with pyrazole scaffold 16a-l as hybrid rosiglitazone/celecoxib analogs was designed, synthesized and tested for its PPAR-γ activation, α-glucosidase, α-amylase and COX-2 inhibitory activities. Regarding the anti-diabetic activity, all compounds were assessed in vitro against PPAR-γ activation, α-glucosidase and α-amylase inhibition in addition to in vivo hypoglycemic activity (one day and 15 days studies). Compounds 16b, 16c, 16e and 16 k showed good PPAR-γ activation (activation % ≈ 72-79 %) compared to that of the reference drug rosiglitazone (74 %). In addition, the same derivatives 16b, 16c, 16e and 16 k showed the highest inhibitory activities against α-glucosidase (IC50 = 0.158, 0.314, 0.305, 0.128 μM, respectively) and against α-amylase (IC50 = 32.46, 23.21, 7.74, 35.85 μM, respectively) compared to the reference drug acarbose (IC50 = 0.161 and 31.46 μM for α-glucosidase and α-amylase, respectively). The most active derivatives 16b, 16c, 16e and 16 k also revealed good in vivo hypoglycemic effect comparable to that of rosiglitazone. In addition, compounds 16b and 16c had the best COX-2 selectivity index (S.I. = 18.7, 31.7, respectively) compared to celecoxib (S.I. = 10.3). In vivo anti-inflammatory activity of the target derivatives 16b, 16c, 16e and 16 k supported the results of in vitro screening as the derivatives 16b and 16c (ED50 = 8.2 and 24 mg/kg, respectively) were more potent than celecoxib (ED50 = 30 mg/kg). In silico docking, ADME, toxicity, and molecular dynamic studies were carried out to explain the interactions of the most active anti-diabetic and anti-inflammatory compounds 16b, 16c, 16e and 16 k with the target enzymes in addition to their physiochemical parameters.
Collapse
Affiliation(s)
- Wael A A Fadaly
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Ahmed Elshewy
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini street 11562, Cairo, Egypt; Department of Natural and Applied Sciences, College of Arts and Sciences, The American University of Iraq-Baghdad (AUIB), Baghdad, Iraq
| | - Mohamed T M Nemr
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini street 11562, Cairo, Egypt.
| | - Kareem Abdou
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Ahmed M Sayed
- Department of Pharmacognosy, Collage of Pharmacy, Almaaqal University, 61014 Basrah, Iraq
| | - Nesma M Kahk
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
14
|
Wang XY, Hao M, Li YP, Zhang J, Xu QS, Yang F, Yang ZC, Xiong YR, Gong ES, Luo JH, Zou Q. Structural characteristics of a purified Evodiae fructus polysaccharide and its gastroprotection and relevant mechanism against alcohol-induced gastric lesions in rats. Int J Biol Macromol 2024; 281:136410. [PMID: 39395514 DOI: 10.1016/j.ijbiomac.2024.136410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/12/2024] [Accepted: 10/06/2024] [Indexed: 10/14/2024]
Abstract
Evodiae fructus polysaccharide (EFP) has been previously shown to protect against alcohol-induced gastric lesions. However, which and how active fractions in EFP exert gastroprotection remains unclear. This study aimed to characterize the structure of the purified fraction (EFP-2-1) of EFP, and investigate its gastroprotection and underlying mechanisms. EFP-2-1 was obtained through column chromatography, and was characterized using instrumental analytical techniques. Gastroprotective effect of EFP-2-1 was evaluated using alcohol-induced gastric lesions in rats, and its mechanism was explored through proteomics, metabolomics and diversity sequencing. Results showed that EFP-2-1 had a molecular weight of 7.3 kDa, and consisted mainly of rhamnose, galacturonic acid, galactose and arabinose. Its backbone contained HG and RG-I domains, and branched with →5)-α-l-Araf-(1→, α-l-Araf-(1→ and →4)-β-d-Galp-(1→ residues. EFP-2-1 reduced gastric lesions and the levels of MDA, TNF-α and IL-6, activated PPARγ, primarily altered protein digestion and absorption and bile secretion metabolic pathways, regulated gut microbiota like Faecalibaculum and Lachnoclostridium, and increased short-chain fatty acids production. Correlations were observed among the gut microbiota, metabolites and biochemical indexes influenced by EFP-2-1. These findings suggest that EFP-2-1 is an active fraction of EFP for protecting against alcohol-induced gastric lesions, which may be linked to PPARγ activation, gut microbiota and serum metabolism.
Collapse
Affiliation(s)
- Xiao-Yin Wang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China.
| | - Ming Hao
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China.
| | - Yan-Ping Li
- Scientific Research Center, Gannan Medical University, Ganzhou 341000, China.
| | - Jun Zhang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China.
| | - Quan-Sheng Xu
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China.
| | - Fan Yang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China.
| | - Zi-Chao Yang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China.
| | - Yu-Rou Xiong
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China.
| | - Er-Sheng Gong
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China.
| | - Jiang-Hong Luo
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China.
| | - Qi Zou
- School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
15
|
Ali GF, Hassanein EHM, Mohamed WR. Molecular mechanisms underlying methotrexate-induced intestinal injury and protective strategies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8165-8188. [PMID: 38822868 PMCID: PMC11522073 DOI: 10.1007/s00210-024-03164-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/13/2024] [Indexed: 06/03/2024]
Abstract
Methotrexate (MTX) is a folic acid reductase inhibitor that manages various malignancies as well as immune-mediated inflammatory chronic diseases. Despite being frequently prescribed, MTX's severe multiple toxicities can occasionally limit its therapeutic potential. Intestinal toxicity is a severe adverse effect associated with the administration of MTX, and patients are significantly burdened by MTX-provoked intestinal mucositis. However, the mechanism of such intestinal toxicity is not entirely understood, mechanistic studies demonstrated oxidative stress and inflammatory reactions as key factors that lead to the development of MTX-induced intestinal injury. Besides, MTX causes intestinal cells to express pro-inflammatory cytokines like interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), which activate nuclear factor-kappa B (NF-κB). This is followed by the activation of the Janus kinase/signal transducer and activator of the transcription3 (JAK/STAT3) signaling pathway. Moreover, because of its dual anti-inflammatory and antioxidative properties, nuclear factor erythroid-2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) has been considered a critical signaling pathway that counteracts oxidative stress in MTX-induced intestinal injury. Several agents have potential protective effects in counteracting MTX-provoked intestinal injury such as omega-3 polyunsaturated fatty acids, taurine, umbelliferone, vinpocetine, perindopril, rutin, hesperidin, lycopene, quercetin, apocynin, lactobacillus, berberine, zinc, and nifuroxazide. This review aims to summarize the potential redox molecular mechanisms of MTX-induced intestinal injury and how they can be alleviated. In conclusion, studying these molecular pathways might open the way for early alleviation of the intestinal damage and the development of various agent plans to attenuate MTX-mediated intestinal injury.
Collapse
Affiliation(s)
- Gaber F Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62514, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Assiut Branch, Al-Azhar University, Assiut, 71524, Egypt
| | - Wafaa R Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62514, Egypt.
| |
Collapse
|
16
|
Yu L, Yang Y, Wang J, Bao Z, Zheng M, Wang X, Zhu Y, Wulasihan M. PDCD4 promotes inflammation/fibrosis by activating the PPAR‑γ/NF‑κB pathway in mouse atrial myocytes. Mol Med Rep 2024; 30:209. [PMID: 39301631 PMCID: PMC11425064 DOI: 10.3892/mmr.2024.13333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
Fibrosis is the basis of structural remodeling in atrial fibrillation (AF), during which inflammation is crucial. Programmed cell death factor 4 (PDCD4) is a newly identified inflammatory gene, with unknown mechanisms of action in AF. The present study aimed to elucidate the effects of PDCD4 on the inflammation and structural remodeling of atrial myocytes. For this purpose, a PDCD4 overexpression plasmid (oePDCD4) and PDCD4 small interfering (si)RNA (siPDCD4) were used to modulate PDCD4 expression in mouse atrial myocytes (HL‑1 cells). The expression of PDCD4 was detected using reverse transcription‑quantitative PCR and western blot analysis. The optimal drug concentrations of peroxisome proliferator‑activated receptor γ (PPARγ) agonist (pioglitazone hydrochloride), NF‑κB inhibitor (CBL0137), PPARγ inhibitor (GW9962) and NF‑κB agonist (betulinic acid) were screened using a Cell Counting Kit‑8 assay. The levels of inflammatory factors were detected using enzyme‑linked immunosorbent assays, the expression levels of fibrosis‑related proteins and NF‑κB subunits were detected using western blot analysis, and the expression of phosphorylated (p‑)p65/p65 was detected using immunofluorescence staining. The results revealed that PDCD4 overexpression increased the levels of fibrotic factors (collagen I, collagen III, fibronectin, α‑smooth muscle actin and matrix metalloproteinase 2), pro‑inflammatory cytokines (IFN‑γ, IL‑6, IL‑17A and TNF‑α) and p‑p65, whereas it reduced the levels of anti‑inflammatory cytokines (IL‑4) in HL‑1 cells. Additionally, treatment with the PPARγ agonist and NF‑κB inhibitor reversed the levels of fibrotic‑, pro‑inflammatory and anti‑inflammatory factors in oePDCD4‑HL‑1 cells. By contrast, PDCD4 silencing exerted the opposite effects on fibrotic factors, pro‑inflammatory cytokines, anti‑inflammatory cytokines and p‑p65. In addition, treatment with the PPARγ inhibitor and NF‑κB agonist reversed the levels of fibrotic‑, pro‑inflammatory and anti‑inflammatory factors in siPDCD4‑HL‑1 cells. In conclusion, the present study demonstrated that PDCD4 may induce inflammation and fibrosis by activating the PPARγ/NF‑κB signaling pathway, thereby promoting the structural remodeling of atrial myocytes in AF.
Collapse
Affiliation(s)
- Li Yu
- Department of Integrated Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, P.R. China
| | - Yuchun Yang
- Department of Integrated Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, P.R. China
| | - Jiao Wang
- Department of Integrated Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, P.R. China
| | - Zhen Bao
- Department of Integrated Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, P.R. China
| | - Meijuan Zheng
- Department of Integrated Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, P.R. China
| | - Xi Wang
- Department of Integrated Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, P.R. China
| | - Yu Zhu
- Department of Integrated Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, P.R. China
| | - Muhuyati Wulasihan
- School of Health Management, Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| |
Collapse
|
17
|
Zhou Y, Xiong X, Cheng Z, Chen Z, Wu S, Yu Y, Liu Y, Chen G, Li L. Ginsenoside Rb1 Alleviates DSS-Induced Ulcerative Colitis by Protecting the Intestinal Barrier Through the Signal Network of VDR, PPARγ and NF-κB. Drug Des Devel Ther 2024; 18:4825-4838. [PMID: 39494151 PMCID: PMC11531243 DOI: 10.2147/dddt.s481769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/05/2024] [Indexed: 11/05/2024] Open
Abstract
Purpose Ginseng (Panax ginseng Meyer) is an herbal medicine used in traditional Chinese medicine (TCM), has the effects of treating colitis and other diseases. Ginsenoside Rb1 (GRb1), a major component of ginseng, modulates autoimmunity and metabolism. However, the mechanism underlying GRb1 treatment of ulcerative colitis (UC) has not yet been elucidated. UC is a refractory inflammatory bowel disease (IBD) with a high recurrence rate, and researches on new drugs for UC have been in the spotlight for a long time. Methods Mice with DSS-induced UC were treated with GRb1 or 0.9% saline for 10 days. Colon tissue of UC mice was collected to detect the levels of intestinal inflammatory cytokines and integrity of the intestinal barrier. RNA-seq and network pharmacology were used to predict the therapeutic targets of GRb1 during UC treatment. Results GRb1 treatment alleviated intestinal inflammation and improved intestinal barrier dysfunction in UC mice. Specifically, GRb1 downregulated the levels of pro-inflammatory cytokines such as TNF-α and IL-6, while upregulating the level of the anti-inflammatory cytokine IL-10. Additionally, GRb1 treatment increased the levels of tight junction proteins including ZO-1, Occludin, and E-cadherin, which are crucial for maintaining intestinal barrier integrity. Further analyses using RNA-seq and network pharmacology suggested that these effects might involve the regulation of GRb1 in the signal transduction network of VDR, PPARγ, and NF-κB. Conclusion The study demonstrated that GRb1 effectively alleviated UC by modulating intestinal inflammation and protecting the integrity of the intestinal barrier through the signal transduction network of VDR, PPARγ, and NF-κB.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xinyu Xiong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Zhe Cheng
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Zekai Chen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Shizhen Wu
- College of Acupuncture and Bone Injury, Hubei University of Chinese Medicine, Wuhan, 430061, People’s Republic of China
| | - Yan Yu
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yujin Liu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Guang Chen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Lingli Li
- Department of Traditional Chinese Medicine, Wuhan Fourth Hospital, Wuhan, 430033, People’s Republic of China
| |
Collapse
|
18
|
Zhao X, Zhang Y, Wang P, Liu K, Zheng Y, Wen J, Wang K, Wen X. Layer by layer self-assembled hyaluronic acid nanoarmor for the treatment of ulcerative colitis. J Nanobiotechnology 2024; 22:633. [PMID: 39420343 PMCID: PMC11488142 DOI: 10.1186/s12951-024-02933-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024] Open
Abstract
Natural compound-based treatments provide innovative ways for ulcerative colitis therapy. However, poor targeting and rapid degradation curtail its application, which needs to be addressed. Inspired by biomacromolecule-based materials, we have developed an orally administrated nanoparticle (GBP@HA NPs) using bovine serum albumin as a carrier for polyphenol delivery. The system synergizes galactosylated bovine serum albumin with two polyphenols, epigallocatechin gallate and tannic acid, which is then encased in "nanoarmor" of ε-Polylysine and hyaluronic acid to boost its stability and targeting. Remarkably, the nanoarmor demonstrated profound therapeutic effects in both acute and chronic mouse models of ulcerative colitis, mitigating disease symptoms via multiple mechanisms, regulating inflammation related factors and exerting a modulatory impact on gut microbiota. Further mechanistic investigations indicate that GBP@HA NPs may act through several pathways, including modulation of Keap1-Nrf2 and NF-κB signaling, as well as Caspase-1-dependent pyroptosis. Consequently, this novel armored nanotherapy promotes the way for enhanced polyphenol utilization in ulcerative colitis treatment research.
Collapse
Affiliation(s)
- Xinxin Zhao
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yuchen Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Pengchong Wang
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an Shaanxi, 710068, China
| | - Kailai Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yunhe Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jinpeng Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ke Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Xiaopeng Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
19
|
Su Z, Hu Q, Li X, Wang Z, Xie Y. The Influence of Circadian Rhythms on DNA Damage Repair in Skin Photoaging. Int J Mol Sci 2024; 25:10926. [PMID: 39456709 PMCID: PMC11507642 DOI: 10.3390/ijms252010926] [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: 08/15/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Circadian rhythms, the internal timekeeping systems governing physiological processes, significantly influence skin health, particularly in response to ultraviolet radiation (UVR). Disruptions in circadian rhythms can exacerbate UVR-induced skin damage and increase the risk of skin aging and cancer. This review explores how circadian rhythms affect various aspects of skin physiology and pathology, with a special focus on DNA repair. Circadian regulation ensures optimal DNA repair following UVR-induced damage, reducing mutation accumulation, and enhancing genomic stability. The circadian control over cell proliferation and apoptosis further contributes to skin regeneration and response to UVR. Oxidative stress management is another critical area where circadian rhythms exert influence. Key circadian genes like brain and muscle ARNT-like 1 (BMAL1) and circadian locomotor output cycles kaput (CLOCK) modulate the activity of antioxidant enzymes and signaling pathways to protect cells from oxidative stress. Circadian rhythms also affect inflammatory and immune responses by modulating the inflammatory response and the activity of Langerhans cells and other immune cells in the skin. In summary, circadian rhythms form a complex defense network that manages UVR-induced damage through the precise regulation of DNA damage repair, cell proliferation, apoptosis, inflammatory response, oxidative stress, and hormonal signaling. Understanding these mechanisms provides insights into developing targeted skin protection and improving skin cancer prevention.
Collapse
Affiliation(s)
- Zhi Su
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
| | - Qianhua Hu
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
| | - Xiang Li
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
| | - Zirun Wang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
| | - Ying Xie
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
| |
Collapse
|
20
|
Sun Y, Lu T, Pan J, He H, Xu M, Chen Y, Chen Y, Fang P, Ye X, Li S, Hu H, Yu S. Dual tobramycin and docosahexaenoic acid loaded nanoemulsions combating Pseudomonas aeruginosa-induced pulmonary infection. Colloids Surf B Biointerfaces 2024; 242:114088. [PMID: 39003845 DOI: 10.1016/j.colsurfb.2024.114088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) typically forms biofilms in vivo, which exhibit high resistance and complicate eradication efforts. Additionally, persistent inflammation and excessive oxidative stress can lead to severe lung dysfunction, facilitating bacterial colonization and infection. Herein, we prepared oil-in-water (O/W) nanoemulsions (TD-αT NEs) by using PEG5k-block-PCL5k and α-tocopherol to encapsulate tobramycin (TOB). To enhance TOB's drug load, a hydrophobic ion pair (TDIP) composed of TOB and docosahexaenoic acid (DHA) was pre-prepared. TD-αT NEs was not only easily prepared and aerosolized, but stable in both physics and chemistry. The negatively charged TD-αT NEs facilitated penetration through mucus, reaching infection sites. Subsequently, TD-αT NEs permeated biofilms due to their small size and released drugs via lipase-triggered carrier dissociation, aiding in eradicating internal bacteria within biofilms (with a 16-fold reduction in CFU vs. free TOB group). TD-αT NEs simultaneously exerted superior anti-inflammatory effects, reducing levels of pro-inflammatory cytokines (NO, IL-6, IL-8, and TNF-α) while increasing the level of anti-inflammatory cytokine (IL-10). It was achieved through the upregulation of PPAR-γ and downregulation of NF-κB signaling, thus mitigating the lung damage. In addition, TD-αT NEs demonstrated strong antioxidant activity, alleviating the oxidative stress induced by P. aeruginosa. Notably, when administered via inhalation, TD-αT NEs significantly reduced the lung bacterial burden, lung inflammation, and oxidative stress in vivo compared to TOB solution. TD-αT NEs could prove beneficial in treating chronic pulmonary infections induced by P. aeruginosa through a comprehensive strategy, specifically enhancing biofilm eradication, reducing inflammation, and alleviating oxidative stress.
Collapse
Affiliation(s)
- Yingying Sun
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Tianli Lu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Jieyi Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Haonan He
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Mao Xu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Yujun Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Yan Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Pengchao Fang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Xiaoxing Ye
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Shuxuan Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China
| | - Haiyan Hu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China; State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China.
| | - Shihui Yu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China; State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Sun Yat-Sen University, University Town, Guangzhou 510006, PR China.
| |
Collapse
|
21
|
Byron M, Lection J, Foster RA, Chenier T, Wagner B, Diel de Amorim M. Prostaglandin-related genes are differentially expressed in equine endometrium with different biopsy grade, degrees of inflammation, and fibrosis. Theriogenology 2024; 227:151-156. [PMID: 39089078 DOI: 10.1016/j.theriogenology.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 08/03/2024]
Abstract
Prostaglandins have many roles in the equine reproductive tract, including but not limited to luteolysis, luteal support, ovulation, transport through the uterine tube, uterine contraction, embryonic mobility, inflammation, and fibrosis. Altered secretion of inflammatory proteins are likely to disrupt the balance of endometrial function and could impair fertility. Our overall goal was to measure the expression of several prostaglandin- and inflammation-related genes in mares with different degrees of endometrial histological changes. Our hypothesis was that mares with neutrophilic and lymphocytic plasmocytic inflammation, fibrosis, or different biopsy grades would have altered concentrations of prostaglandin E2 (PGE2) and F2α (PGF2α), as well as altered expression of inflammation- and prostaglandin-related genes, compared to mares with minimal to no histological changes on biopsy evaluation. Forty-five endometrial biopsies from estrous mares were assessed by a reproductive pathologist for the degree of neutrophilic inflammation, lymphocytic and plasmocytic inflammation, and fibrosis, and a biopsy grade was assigned based on the Kenney-Doig system. A low-volume uterine lavage was collected from a subset of twenty-six mares prior to biopsy collection and was used to measure PGE2 and PGF2α concentrations via ELISA. Total RNA was extracted from biopsies and mRNA expression was evaluated for twenty-five genes of interest. A restricted maximum likelihood linear model was used to compare differences of mRNA expression, with a statistical significance set at P < 0.05. There was no difference in the abundance of PGE2 or PGF2α between any of the variables tested. Mares with endometrial biopsy grade I had lower expression of NF-kB, PTGS1 and HPGD compared to grade IIA or IIB (P < 0.05). Mares with neutrophilic inflammation had decreased expression of NF-kB, PTGS1, PTGER4, CBR1, mPGES2 and PTGIS compared to mares without inflammation. Mares with mild or minimal endometrial fibrosis had increased expression of mPGES2 and PTGIS, compared to mares with moderate endometrial fibrosis. In conclusion, several genes were identified to be differentially expressed in mares with histological changes compared to mares with no to minimal histological changes. The presence of inflammation and fibrosis may alter the concentration of prostaglandins in endometrial tissue, which could impair many of the uterine reproductive and immune functions during estrus, affecting early embryo survival.
Collapse
Affiliation(s)
- Michael Byron
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Jennine Lection
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Robert A Foster
- Department of Pathobiology, Ontario Veterinary College, Guelph, ON, Canada
| | - Tracey Chenier
- Department of Population Medicine, Ontario Veterinary College, Guelph, ON, Canada
| | - Bettina Wagner
- Department of Population Medicine and Diagnostic Sciences, Cornell University, Ithaca, NY, USA
| | - Mariana Diel de Amorim
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
22
|
Warren WG, Osborn M, Duffy P, Yates A, O'Sullivan SE. Potential safety implications of fatty acid-binding protein inhibition. Toxicol Appl Pharmacol 2024; 491:117079. [PMID: 39218163 DOI: 10.1016/j.taap.2024.117079] [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: 06/07/2024] [Revised: 08/15/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Fatty acid-binding proteins (FABPs) are small intracellular proteins that regulate fatty acid metabolism, transport, and signalling. There are ten known human isoforms, many of which are upregulated and involved in clinical pathologies. As such, FABP inhibition may be beneficial in disease states such as cancer, and those involving the cardiovascular system, metabolism, immunity, and cognition. Recently, a potent, selective FABP5 inhibitor (ART26.12), with 90-fold selectivity to FABP3 and 20-fold selectivity to FABP7, was found to be remarkably benign, with a no-observed-adverse-effect level of 1000 mg/kg in rats and dogs, showing no genotoxicity, cardiovascular, central, or respiratory toxicity. To understand the potential implication of FABP inhibition more fully, this review systematically assessed literature investigating genetic knockout, knockdown, and pharmacological inhibition of FABP3, FABP4, FABP5, or FABP7. Analysis of the literature revealed that animals bred not to express FABPs showed the most biological effects, suggesting key roles of these proteins during development. FABP ablation sometimes exacerbated symptoms of disease models, particularly those linked to metabolism, inflammatory and immune responses, cardiac contractility, neurogenesis, and cognition. However, FABP inhibition (genetic silencing or pharmacological) had a positive effect in many more disease conditions. Several polymorphisms of each FABP gene have also been linked to pathological conditions, but it was unclear how several polymorphisms affected protein function. Overall, analysis of the literature to date suggests that pharmacological inhibition of FABPs in adults is of low risk.
Collapse
Affiliation(s)
- William G Warren
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom.
| | - Myles Osborn
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom
| | - Paul Duffy
- Apconix Ltd., Alderley Park, Cheshire SK10 4TG, United Kingdom
| | - Andrew Yates
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom
| | | |
Collapse
|
23
|
Ali Z, Al-Ghouti MA, Abou-Saleh H, Rahman MM. Unraveling the Omega-3 Puzzle: Navigating Challenges and Innovations for Bone Health and Healthy Aging. Mar Drugs 2024; 22:446. [PMID: 39452854 PMCID: PMC11509197 DOI: 10.3390/md22100446] [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: 08/27/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Omega-3 polyunsaturated fatty acids (ω-3 PUFAs, n-3 PUFAs), including eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and alpha-linolenic acid (ALA), are essential polyunsaturated fats primarily obtained from fatty fish and plant-based sources. Compelling evidence from preclinical and epidemiological studies consistently suggests beneficial effects of ω-3 PUFAs on bone health and healthy aging processes. However, clinical trials have yielded mixed results, with some failing to replicate these benefits seen in preclinical models. This contraindication is mainly due to challenges such as low bioavailability, potential adverse effects with higher doses, and susceptibility to oxidation of ω-3 fatty acids, hindering their clinical effectiveness. This review comprehensively discusses recent findings from a clinical perspective, along with preclinical and epidemiological studies, emphasizing the role of ω-3 PUFAs in promoting bone health and supporting healthy aging. Additionally, it explores strategies to improve ω-3 PUFA efficacy, including nanoparticle encapsulation and incorporation of specialized pro-resolving mediators (SPM) derived from DHA and EPA, to mitigate oxidation and enhance solubility, thereby improving therapeutic potential. By consolidating evidence from various studies, this review underscores current insights and future directions in leveraging ω-3 PUFAs for therapeutic applications.
Collapse
Affiliation(s)
- Zayana Ali
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Mohammad Ahmed Al-Ghouti
- Environmental Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Haissam Abou-Saleh
- Biomedical Sciences Department, College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Md Mizanur Rahman
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| |
Collapse
|
24
|
Boateng E, Bonilla-Martinez R, Ahlemeyer B, Garikapati V, Alam MR, Trompak O, Oruqaj G, El-Merhie N, Seimetz M, Ruppert C, Günther A, Spengler B, Karnati S, Baumgart-Vogt E. It takes two peroxisome proliferator-activated receptors (PPAR-β/δ and PPAR-γ) to tango idiopathic pulmonary fibrosis. Respir Res 2024; 25:345. [PMID: 39313791 PMCID: PMC11421181 DOI: 10.1186/s12931-024-02935-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/01/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant lung epithelial phenotypes, fibroblast activation, and increased extracellular matrix deposition. Transforming growth factor-beta (TGF-β)1-induced Smad signaling and downregulation of peroxisomal genes are involved in the pathogenesis and can be inhibited by peroxisome proliferator-activated receptor (PPAR)-α activation. However, the three PPARs, that is PPAR-α, PPAR-β/δ, and PPAR-γ, are known to interact in a complex crosstalk. METHODS To mimic the pathogenesis of lung fibrosis, primary lung fibroblasts from control and IPF patients with comparable levels of all three PPARs were treated with TGF-β1 for 24 h, followed by the addition of PPAR ligands either alone or in combination for another 24 h. Fibrosis markers (intra- and extracellular collagen levels, expression and activity of matrix metalloproteinases) and peroxisomal biogenesis and metabolism (gene expression of peroxisomal biogenesis and matrix proteins, protein levels of PEX13 and catalase, targeted and untargeted lipidomic profiles) were analyzed after TGF-β1 treatment and the effects of the PPAR ligands were investigated. RESULTS TGF-β1 induced the expected phenotype; e.g. it increased the intra- and extracellular collagen levels and decreased peroxisomal biogenesis and metabolism. Agonists of different PPARs reversed TGF-β1-induced fibrosis even when given 24 h after TGF-β1. The effects included the reversals of (1) the increase in collagen production by repressing COL1A2 promoter activity (through PPAR-β/δ activation); (2) the reduced activity of matrix metalloproteinases (through PPAR-β/δ activation); (3) the decrease in peroxisomal biogenesis and lipid metabolism (through PPAR-γ activation); and (4) the decrease in catalase protein levels in control (through PPAR-γ activation) and IPF (through a combined activation of PPAR-β/δ and PPAR-γ) fibroblasts. Further experiments to explore the role of catalase showed that an overexpression of catalase protein reduced collagen production. Additionally, the beneficial effect of PPAR-γ but not of PPAR-β/δ activation on collagen synthesis depended on catalase activity and was thus redox-sensitive. CONCLUSION Our data provide evidence that IPF patients may benefit from a combined activation of PPAR-β/δ and PPAR-γ.
Collapse
Affiliation(s)
- Eistine Boateng
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
- Department of Medical Education, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - Rocio Bonilla-Martinez
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
| | - Barbara Ahlemeyer
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
| | - Vannuruswamy Garikapati
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University, 35392, Giessen, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307, Dresden, Germany
| | - Mohammad Rashedul Alam
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
| | - Omelyan Trompak
- Department of Internal Medicine VIII, Eberhard Karls University, 72076, Tübingen, Germany
| | - Gani Oruqaj
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
- Department of Internal Medicine II, Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University, 35392, Giessen, Germany
| | - Natalia El-Merhie
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
- Institute for Lung Health (ILH), Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University, 35392, Giessen, Germany
| | - Michael Seimetz
- Excellence Cluster Cardio-Pulmonary System, German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, 35392, Giessen, Germany
| | - Clemens Ruppert
- Excellence Cluster Cardio-Pulmonary System, German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, 35392, Giessen, Germany
- UGMLC Giessen Biobank, Universities of Giessen and Marburg Lung Center, 35392, Giessen, Germany
| | - Andreas Günther
- Excellence Cluster Cardio-Pulmonary System, German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, 35392, Giessen, Germany
- Center for Interstitial and Rare Lung Diseases, Department of Internal Medicine, German Center for Lung Research, Universities of Giessen and Marburg Lung Center, 35392, Giessen, Germany
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University, 35392, Giessen, Germany
| | - Srikanth Karnati
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
- Institute for Anatomy and Cell Biology, Julius Maximilians University, 97070, Würzburg, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Aulweg 123, 35392, Giessen, Germany.
| |
Collapse
|
25
|
Li D, Li F, Zhou Y, Tang Y, Hu Z, Wu Q, Xie T, Lin Q, Wang H, Luo F. Role and Mechanism of Sialic Acid in Alleviating Acute Lung Injury through In Vivo and In Vitro Models. Foods 2024; 13:2984. [PMID: 39335912 PMCID: PMC11431537 DOI: 10.3390/foods13182984] [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: 08/10/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Excessive inflammatory reactions are the most important pathological injury factor in acute lung injury (ALI). Our recent study found that sialic acid had an anti-colitis effect. In this study, the effect of sialic acid (SA) on acute lung inflammation was investigated. A lipopolysaccharide (LPS)-induced ALI animal model and LPS-stimulated HUVEC cell model were used to evaluate the anti-inflammatory effect of SA and study its molecular mechanisms. Compared with the LPS group, the lung index of the SA group decreased from 0.79 ± 0.05% to 0.58 ± 0.06% (LPS + 50 SA) and 0.62 ± 0.02% (LPS + 100 SA), with p < 0.01, suggesting that SA could improve the pulmonary edema of mice and alleviate LPS-induced lung injury. Transcriptome research identified 26 upregulated genes and 25 downregulated genes involved in the protection of SA against ALI. These genes are mainly related to the MAPK and NF-κB signaling pathways. Our study also proved that SA markedly downregulated the expression of inflammatory factors and blocked the JNK/p38/PPAR-γ/NF-κB pathway. Meanwhile, SA treatment also upregulated the expression of HO-1 and NQO1 in ALI mice. In vitro, SA obviously repressed the expressions of inflammatory cytokines and the JNK/p38-NF-κB/AP-1 pathway. SA also regulated the expression of oxidative stress-related genes through the Nrf2 pathway. Taken together, SA exhibits a protective role by modulating the anti-inflammatory and anti-oxidation pathways in ALI, and it may be a promising candidate for functional foods to prevent ALI.
Collapse
Affiliation(s)
- Dan Li
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
- Hunan Engineering Research Center of Full Life-Cycle Energy-Efficient Buildings and Environmental Health, School of Civil Engineering, Central South University of Forestry and Technology, Changsha 410004, China;
| | - Fangyan Li
- Hunan Engineering Research Center of Full Life-Cycle Energy-Efficient Buildings and Environmental Health, School of Civil Engineering, Central South University of Forestry and Technology, Changsha 410004, China;
| | - Yaping Zhou
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Yiping Tang
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Zuomin Hu
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Qi Wu
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Tiantian Xie
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Qinlu Lin
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| | - Hanqing Wang
- Hunan Engineering Research Center of Full Life-Cycle Energy-Efficient Buildings and Environmental Health, School of Civil Engineering, Central South University of Forestry and Technology, Changsha 410004, China;
| | - Feijun Luo
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Processing and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (D.L.); (Y.Z.); (Y.T.); (Z.H.); (Q.W.); (T.X.); (Q.L.)
| |
Collapse
|
26
|
Kim HJ, Kim H, Song J, Hong JY, Lee EH, Londhe AM, Choi JW, Park SJ, Oh E, Yoon H, Hwang H, Hahn D, Jung K, Kwon S, Kadayat TM, Ma MJ, Joo J, Kim J, Bae JH, Hwang H, Pae AN, Cho SJ, Park JH, Chin J, Kang H, Park KD. Highly potent and selective PPARδ agonist reverses memory deficits in mouse models of Alzheimer's disease. Theranostics 2024; 14:6088-6108. [PMID: 39431021 PMCID: PMC11488110 DOI: 10.7150/thno.96707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/06/2024] [Indexed: 10/22/2024] Open
Abstract
Rationale: Alzheimer's disease (AD) is a progressive neurodegenerative disease accompanied by neurotoxicity, excessive inflammation, and cognitive impairment. The peroxisome proliferator-activated receptor (PPAR) δ is a potential target for AD. However, its regulatory mechanisms and therapeutic potential in AD remain unclear. We aimed to investigate if the activation of PPARδ using a highly selective and potent agonist could provide an effective therapeutic strategy against AD. Methods: We synthesized a novel PPARδ agonist, 5a, containing a selenazole group and determined the X-ray crystal structure of its complex with PPARδ. The drug-like properties of 5a were assessed by analyzing cytochrome P450 (CYP) inhibition, microsomal stability, pharmacokinetics, and mutagenicity. We investigated the anti-inflammatory effects of 5a using lipopolysaccharide (LPS)-stimulated BV-2 microglia and neuroinflammatory mouse model. The therapeutic efficacy of 5a was evaluated in AD mice with scopolamine-induced memory impairment and APP/PS1 by analyzing cognitive function, glial reactivity, and amyloid pathology. Results: Compound 5a, the most potent and selective PPARδ agonist, was confirmed to bind hPPARδ in a complex by X-ray crystallographic analysis. PPARδ activation using 5a showed potent anti-inflammatory effects in activated glial cells and mouse model of neuroinflammation. Administration of 5a inhibited amyloid plaque deposition by suppressing the expression of neuronal beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), and reduced abnormal glial hyperactivation and inflammatory responses, resulting in improved learning and memory in the APP/PS1 mouse model of AD. Conclusion: We identified that specific activation of PPARδ provides therapeutic effects on multiple pathogenic phenotypes of AD, including neuroinflammation and amyloid deposition. Our findings suggest the potential of PPARδ as a promising drug target for treating AD.
Collapse
Affiliation(s)
- Hyeon Jeong Kim
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Haelee Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jaeyoung Song
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jun Young Hong
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, Seoul National University, NS-80 Seoul 08826, Republic of Korea
- Department of Systems Biology, Yonsei University, Seoul 03722, Republic of Korea
| | - Elijah Hwejin Lee
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ashwini M. Londhe
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ji Won Choi
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Sun Jun Park
- Cureverse, lnc., H2 building, KIST, Seoul 02792, Republic of Korea
| | - Eunseok Oh
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, Seoul National University, NS-80 Seoul 08826, Republic of Korea
| | - Heeseok Yoon
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Hoosang Hwang
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, Seoul National University, NS-80 Seoul 08826, Republic of Korea
| | - Dongyup Hahn
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, Seoul National University, NS-80 Seoul 08826, Republic of Korea
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kyungjin Jung
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Sugyeong Kwon
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Tara Man Kadayat
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Min Jung Ma
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jeongmin Joo
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jina Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jae Hyun Bae
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Hayoung Hwang
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Ae Nim Pae
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Sung Jin Cho
- Cureverse, lnc., H2 building, KIST, Seoul 02792, Republic of Korea
| | - Jong-Hyun Park
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Jungwook Chin
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
- Cureverse, lnc., H2 building, KIST, Seoul 02792, Republic of Korea
| | - Heonjoong Kang
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, Seoul National University, NS-80 Seoul 08826, Republic of Korea
- Research Institute of Oceanography, Seoul National University, NS-80, Seoul 08826, Republic of Korea
| | - Ki Duk Park
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| |
Collapse
|
27
|
Olender D, Kujawski J, Skóra B, Baranowska-Wójcik E, Sowa-Kasprzak K, Pawełczyk A, Zaprutko L, Szwajgier D, Szychowski KA. Bis-chalcones obtained via one-pot synthesis as the anti-neurodegenerative agents and their effect on the HT-22 cell line. Heliyon 2024; 10:e37147. [PMID: 39286165 PMCID: PMC11403034 DOI: 10.1016/j.heliyon.2024.e37147] [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: 06/17/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
In the area of research on neurodegenerative diseases, the current challenge is to search for appropriate research methods that would detect these diseases at the earliest possible stage, but also new active structures that would reduce the rate of the disease progression and minimize the intensity of their symptoms experienced by the patient. The chalcones are considered in the context of candidates for new drugs dedicated to the fight against neurodegenerative diseases. The synthesis of bis-chalcone derivatives (3a-3d), as aim molecules was performed. Their structures were established by applying 1H NMR, 13C NMR, MS, FT-IR and UV-Vis spectra. All bis-chalcones were synthesized from terephthalaldehyde and appropriate aromatic ketone as substrates in the Claisen-Schmidt condensation method and evaluated in the biological tests and in silico analysis. Compounds exerted antioxidant activity using the HORAC method (3a-3d) and decreased the activities of GPx, COX-2 (3b-3d), GR (3a-3c) and CAT (3a,3b). The high anti-neurodegenerative potential of all four bis-chalcones was observed by inhibition of acetyl- (AChE) and butyrylcholinesterase (BChE) and a positive effect on the mouse hippocampal neuronal HT-22 cell line (LDH release and PGC-1α, PPARγ and GAPDH protein expression). TD-DFT method (computing a number of descriptors associated with HOMO-LUMO electron transition: electronegativity, chemical hardness and potential, first ionization potential, electron affinity) was employed to study the spectroscopic properties. This method showed that the first excited state of compounds was consistent with their maximum absorption in the computed UV-Vis spectra, which showed good agreement with the experimental spectrum using PBE1PBE functional. Using in silico approach, interactions of bis-chalcones with selected targets (aryl hydrocarbon receptor (AhR) PAS-A Domain, ligand binding domain of human PPAR-γ, soman-aged human BChE-butyrylthiocholine complex, Torpedo californica AChE:N-piperidinopropyl-galanthamine complex and the COX-2-celecoxib complex) were characterized. Results obtained in in silico models were consistent with in vitro experiments.
Collapse
Affiliation(s)
- Dorota Olender
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznań, Poland
| | - Jacek Kujawski
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznań, Poland
| | - Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225, Rzeszów, Poland
| | - Ewa Baranowska-Wójcik
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, Skromna 8, 20-704, Lublin, Poland
| | - Katarzyna Sowa-Kasprzak
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznań, Poland
| | - Anna Pawełczyk
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznań, Poland
| | - Lucjusz Zaprutko
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznań, Poland
| | - Dominik Szwajgier
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, Skromna 8, 20-704, Lublin, Poland
| | - Konrad A Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225, Rzeszów, Poland
| |
Collapse
|
28
|
Lu J, Zhang J, Jiang H, Hu Z, Zhang Y, He L, Yang J, Xie Y, Wu D, Li H, Zeng K, Tan P, Xiao Q, Song Z, Pan C, Bai X, Yu X. Vangl2 suppresses NF-κB signaling and ameliorates sepsis by targeting p65 for NDP52-mediated autophagic degradation. eLife 2024; 12:RP87935. [PMID: 39269442 PMCID: PMC11398866 DOI: 10.7554/elife.87935] [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] [Indexed: 09/15/2024] Open
Abstract
Van Gogh-like 2 (Vangl2), a core planar cell polarity component, plays an important role in polarized cellular and tissue morphology induction, growth development, and cancer. However, its role in regulating inflammatory responses remains elusive. Here, we report that Vangl2 is upregulated in patients with sepsis and identify Vangl2 as a negative regulator of The nuclear factor-kappaB (NF-κB) signaling by regulating the protein stability and activation of the core transcription component p65. Mice with myeloid-specific deletion of Vangl2 (Vangl2ΔM) are hypersusceptible to lipopolysaccharide (LPS)-induced septic shock. Vangl2-deficient myeloid cells exhibit enhanced phosphorylation and expression of p65, therefore, promoting the secretion of proinflammatory cytokines after LPS stimulation. Mechanistically, NF-κB signaling-induced-Vangl2 recruits E3 ubiquitin ligase PDLIM2 to catalyze K63-linked ubiquitination on p65, which serves as a recognition signal for cargo receptor NDP52-mediated selective autophagic degradation. Taken together, these findings demonstrate Vangl2 as a suppressor of NF-κB-mediated inflammation and provide insights into the crosstalk between autophagy and inflammatory diseases.
Collapse
Affiliation(s)
- Jiansen Lu
- Department of Joint Surgery, the Fifth Affiliated Hospital, Southern Medical UniversityGuangzhouChina
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Jiahuan Zhang
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Huaji Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Department of Orthopaedics, Yuebei People's Hospital Affiliated to Medical College of Shantou UniversityShaoguanChina
| | - Zhiqiang Hu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yufen Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Lian He
- Department of Pharmacology, School of Medicine, Southern University of Science and TechnologyShenzhenChina
- Institute of Biosciences and Technology, College of Medicine, Texas A&M UniversityHoustonUnited States
| | - Jianwu Yang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yingchao Xie
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Dan Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Hongyu Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Ke Zeng
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Peng Tan
- Department of Pharmacology, School of Medicine, Southern University of Science and TechnologyShenzhenChina
- Klarman Cell Observatory, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Qingyue Xiao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Zijing Song
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Chenglong Pan
- Department of Joint Surgery, the Fifth Affiliated Hospital, Southern Medical UniversityGuangzhouChina
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Xiao Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Lab of Single Cell Technology and Application, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
29
|
Roy S, Kant S, Das Saha K, Jha T. Chrysin-functionalized gold nanoparticles and paclitaxel exhibit synergistic impact on lung cancer cell lines via regulating the AKT/PPAR-ϒ/β-catenin pathway. Drug Dev Ind Pharm 2024:1-14. [PMID: 39159138 DOI: 10.1080/03639045.2024.2393327] [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: 04/13/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024]
Abstract
Lung cancer has become progressively widespread, posing a challenge to traditional chemotherapeutic drugs such as platinum compounds and paclitaxel (PTX) owing to growing resistance. Along with that, the chemotherapeutic drugs infer major side effects. The usage of natural compounds as chemosensitizers to boost the efficacy of these chemotherapeutic drugs and minimizing their toxicity is a plausible approach. In our investigation, we employed PTX as the standard chemotherapeutic agent and utilized chrysin-functionalized gold nanoparticles (CHR-AuNPs) to augment its cytotoxicity. Gold nanoparticles were chosen for their inherent cytotoxic properties and ability to enhance chrysin's bioavailability and solubility. Characterization of CHR-AuNP revealed spherical nanoparticles within the nano-size range (35-70 nm) with a stable negative zeta potential of -22 mV, confirmed by physicochemical analyses including UV-visible spectroscopy, Fourier transform infrared (FTIR) spectral analysis, and visual observation of the wine-red coloration. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay cytotoxicity studies demonstrated CHR-AuNP's superior efficacy compared to CHR alone, with synergistic effects observed in combination with PTX, validated by Compusyn software. Morphological changes indicative of apoptosis were more pronounced with combined treatment, corroborated by acridine orange/ethidium bromide (AO/EtBr) staining and Annexin V assays. Furthermore, the combination treatment amplified reactive oxygen species (ROS) production and destabilized mitochondrial membrane potential, while altering the expression of pro-apoptotic and anti-apoptotic proteins. Exploring the mechanistic pathways, we found that the drugs upregulated PPAR-γ expression while suppressing Akt and overexpressing PTEN, thereby impeding the Wnt/β-catenin pathway commonly dysregulated in lung cancer. This highlights the potential of low-dose combination therapy with PTX and CHR-AuNP as a promising strategy for addressing lung cancer's challenges.
Collapse
Affiliation(s)
- Saheli Roy
- Department of Pharmaceutical Technology, School of Health and Medical Sciences, Adamas University, Kolkata, India
| | - Shashi Kant
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology (CSIR-IICB), Kolkata, India
| | - Krishna Das Saha
- Cancer Biology & Inflammatory Disorder Division, Indian Institute of Chemical Biology (CSIR-IICB), Kolkata, India
| | - Tarun Jha
- Department of Pharmaceutical Technology, Division of Medicinal and Pharmaceutical Chemistry, Natural Science Laboratory, Jadavpur University, Kolkata, India
| |
Collapse
|
30
|
Tiwari P, Tripathi LP. Long Non-Coding RNAs, Nuclear Receptors and Their Cross-Talks in Cancer-Implications and Perspectives. Cancers (Basel) 2024; 16:2920. [PMID: 39199690 PMCID: PMC11352509 DOI: 10.3390/cancers16162920] [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: 06/05/2024] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) play key roles in various epigenetic and post-transcriptional events in the cell, thereby significantly influencing cellular processes including gene expression, development and diseases such as cancer. Nuclear receptors (NRs) are a family of ligand-regulated transcription factors that typically regulate transcription of genes involved in a broad spectrum of cellular processes, immune responses and in many diseases including cancer. Owing to their many overlapping roles as modulators of gene expression, the paths traversed by lncRNA and NR-mediated signaling often cross each other; these lncRNA-NR cross-talks are being increasingly recognized as important players in many cellular processes and diseases such as cancer. Here, we review the individual roles of lncRNAs and NRs, especially growth factor modulated receptors such as androgen receptors (ARs), in various types of cancers and how the cross-talks between lncRNAs and NRs are involved in cancer progression and metastasis. We discuss the challenges involved in characterizing lncRNA-NR associations and how to overcome them. Furthering our understanding of the mechanisms of lncRNA-NR associations is crucial to realizing their potential as prognostic features, diagnostic biomarkers and therapeutic targets in cancer biology.
Collapse
Affiliation(s)
- Prabha Tiwari
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Lokesh P. Tripathi
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Kanagawa, Japan
- AI Center for Health and Biomedical Research (ArCHER), National Institutes of Biomedical Innovation, Health and Nutrition, Kento Innovation Park NK Building, 3-17 Senrioka Shinmachi, Settsu 566-0002, Osaka, Japan
| |
Collapse
|
31
|
Manusov EG, Diego VP, Almeida M, Ortiz D, Curran JE, Galan J, Leandro AC, Laston S, Blangero J, Williams-Blangero S. Genotype-by-Environment Interactions in Nonalcoholic Fatty Liver Disease and Chronic Illness among Mexican Americans: The Role of Acculturation Stress. Genes (Basel) 2024; 15:1006. [PMID: 39202366 PMCID: PMC11353877 DOI: 10.3390/genes15081006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
This study examines the complex interplay of genetic and environmental interactions that shape chronic illness risk. Evidence is mounting for the role of genetic expression and the immune response in the pathogenesis of chronic disease. In the Rio Grande Valley of south Texas, where 90% of the population is Mexican American, chronic illnesses (including obesity, diabetes, nonalcoholic liver disease, and depression) are reaching epidemic proportions. This study leverages an ongoing family study of the genetic determinants of risk for obesity, diabetes, hypertension, hyperlipidemia, and depression in a Mexican American population. Data collected included blood pressure, BMI, hepatic transaminases, HbA1c, depression (BDI-II), acculturation/marginalization (ARSMA-II), and liver health as assessed by elastography. Heritability and genotype-by-environment (G×E) interactions were analyzed, focusing on the marginalization/separation measure of the ARSMA-II. Significant heritabilities were found for traits such as HbA1c (h2 = 0.52), marginalization (h2 = 0.30), AST (h2 = 0.25), ALT (h2 = 0.41), and BMI (h2 = 0.55). Genotype-by-environment interactions were significant for HbA1c, AST/ALT ratio, BDI-II, and CAP, indicating that genetic factors interact with marginalization to influence these traits. This study found that acculturation stress exacerbates the genetic response to chronic illness. These findings underscore the importance of considering G×E interactions in understanding disease susceptibility and may inform targeted interventions for at-risk populations. Further research is warranted to elucidate the underlying molecular pathways and replicate these findings in diverse populations.
Collapse
Affiliation(s)
- Eron G. Manusov
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Vincent P. Diego
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
| | - Marcio Almeida
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - David Ortiz
- School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA;
| | - Joanne E. Curran
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Jacob Galan
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Ana C. Leandro
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Sandra Laston
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - John Blangero
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Sarah Williams-Blangero
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| |
Collapse
|
32
|
Xiao C, Liu Y, Zhao W, Liang Y, Cui C, Yang S, Fang W, Miao L, Yuan Z, Lin Z, Zhai B, Zhao Z, Zhang L, Ma H, Jin H, Cao Y. The comparison of meat yield, quality, and flavor between small-tailed Han sheep and two crossbred sheep and the verification of related candidate genes. Front Nutr 2024; 11:1399390. [PMID: 39149545 PMCID: PMC11324605 DOI: 10.3389/fnut.2024.1399390] [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: 03/11/2024] [Accepted: 07/12/2024] [Indexed: 08/17/2024] Open
Abstract
Introduction In Northeast China, Dorper and Australian White rams are commonly crossbred with small-tailed Han (STH) ewes to improve the offspring's meat yield and quality. However, the differences in traits and the flavor between the crossbred sheep and STH sheep remain unclear. In addition, the candidate genes potentially influencing the meat quality in the three sheep breeds require further verification. Methods A total of 18 2-month-old healthy rams were raised over a period of 5 months, which included 6 STH, 6 Dorper and small-tailed Han crossbred (Do × STH), and 6 Australian white and small-tailed Han crossbred (Au × STH) offspring. The differences in slaughter, meat quality traits, fatty acid and amino acid composition in the muscular longissimus dorsi (MLD), and volatile compounds in the semitendinosus muscle were compared between the sheep breeds. The candidate genes related to intramuscular fat (IMF) content and fatty acids were validated. Results The results of this study revealed that the crossbred sheep had higher body weight, carcass weight, bone weight, net meat weight, and IMF content than the STH sheep (p < 0.05). The Do × STH offspring had a higher pH value (24 h), moisture content, and cooking percentage; they also had redder and brighter meat color. The content of myristate, palmitic, and margaric acids in the crossbred sheep was higher than that in the STH sheep (p < 0.05). The Do × STH offspring had the highest saturated fatty acid content (p < 0.05). The Au × STH offspring had the highest protein content (p < 0.05). The arachidonic acid and amino acid (Asp, Ala, Ile, Leu, Lys, Thr, and essential amino acid) contents were higher in the STH sheep than in the crossbred sheep (p < 0.05). The odor activity value (OAV) analysis showed that most of the aldehydes in the Au × STH offspring had higher values. The PDK4 gene expression was positively associated with the IMF content and was negatively correlated with the linoleic acid content in the Do × STH sheep (p < 0.05). The TMEM273 gene expression was positively associated with linoleic and arachidonic acid contents and was negatively correlated with oleic and palmitic acid contents in the Do × STH sheep (p < 0.05). Discussion The results showed the differences between the crossbred sheep and STH sheep and provided the candidate genes related to meat quality in sheep.
Collapse
Affiliation(s)
- Cheng Xiao
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
- Research Institute for Farm Animal Biology (FBN), Institute of Muscle Biology and Growth, Dummerstorf, Germany
- Institute of Agricultural and Environmental Sciences, Rostock University, Rostock, Germany
| | - Yu Liu
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Wenjun Zhao
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
- College of Agriculture, YanBian University, Yanji, China
| | - Yingjia Liang
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Chao Cui
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Shaoying Yang
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - WenWen Fang
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Lisheng Miao
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Zhiyu Yuan
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Zihan Lin
- College of Plant Protection, Jilin Agricultural University, Changchun, China
| | - Bo Zhai
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Zhongli Zhao
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Lichun Zhang
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Huihai Ma
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Haiguo Jin
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| | - Yang Cao
- Institute of Animal and Veterinary Sciences, Jilin Academy of Agricultural Sciences, Gongzhuling, China
| |
Collapse
|
33
|
Wang K, Xue Y, Liu Y, Su X, Wei L, Lv C, Zhang X, Zhang L, Jia L, Zheng S, Ma Y, Yan H, Jiang G, Song H, Wang F, Lin Q, Hou Y. The detoxification ability of sex-role reversed seahorses determines the sexual dimorphism in immune responses to benzo[a]pyrene exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 933:173088. [PMID: 38735333 DOI: 10.1016/j.scitotenv.2024.173088] [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: 12/20/2023] [Revised: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
Sexual dimorphism in immune responses is an essential factor in environmental adaptation. However, the mechanisms involved remain obscure owing to the scarcity of data from sex-role-reversed species in stressed conditions. Benzo[a]pyrene (BaP) is one of the most pervasive and carcinogenic organic pollutants in coastal environments. In this study, we evaluated the potential effects on renal immunotoxicity of the sex-role-reversed lined seahorse (Hippocampus erectus) toward environmental concentrations BaP exposure. Our results discovered the presence of different energy-immunity trade-off strategies adopted by female and male seahorses during BaP exposure. BaP induced more severe renal damage in female seahorses in a concentration-dependent manner. BaP biotransformation and detoxification in seahorses resemble those in mammals. Benzo[a]pyrene-7,8-dihydrodiol-9,10-oxide (BPDE) and 9-hydroxybenzo[a]pyrene (9-OH-BaP) formed DNA adducts and disrupted Ca2+ homeostasis may together attribute the renal immunotoxicity. Sexual dimorphisms in detoxification of both BPDE and 9-OH-BaP, and in regulation of Ca2+, autophagy and inflammation, mainly determined the extent of renal damage. Moreover, the mechanism of sex hormones regulated sexual dimorphism in immune responses needs to be further elucidated. Collectively, these findings contribute to the understanding of sexual dimorphism in the immunotoxicity induced by BaP exposure in seahorses, which may attribute to the dramatic decline in the biodiversity of the genus.
Collapse
Affiliation(s)
- Kai Wang
- School of Agriculture, Ludong University, Yantai 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai 264025, China.
| | - Yuanyuan Xue
- School of Agriculture, Ludong University, Yantai 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai 264025, China
| | - Yali Liu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China.
| | - Xiaolei Su
- School of Agriculture, Ludong University, Yantai 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai 264025, China
| | - Lei Wei
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Chunhui Lv
- School of Agriculture, Ludong University, Yantai 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai 264025, China
| | - Xu Zhang
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Lele Zhang
- School of Agriculture, Ludong University, Yantai 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai 264025, China
| | - Longwu Jia
- School of Agriculture, Ludong University, Yantai 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai 264025, China
| | - Shiyi Zheng
- School of Agriculture, Ludong University, Yantai 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai 264025, China
| | - Yicong Ma
- School of Agriculture, Ludong University, Yantai 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai 264025, China
| | - Hansheng Yan
- School of Agriculture, Ludong University, Yantai 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai 264025, China
| | - Guangjun Jiang
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Hongce Song
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Fang Wang
- Department of Pathology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264025, China
| | - Qiang Lin
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Yuping Hou
- School of Life Sciences, Ludong University, Yantai 264025, China
| |
Collapse
|
34
|
Szlachcikowska D, Tabęcka-Łonczyńska A, Holota S, Roman O, Shepeta Y, Lesyk R, Szychowski KA. Role of Ciminalum-4-thiazolidinone Hybrids in Molecular NF-κB Dependent Pathways. Int J Mol Sci 2024; 25:7329. [PMID: 39000436 PMCID: PMC11242080 DOI: 10.3390/ijms25137329] [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: 05/13/2024] [Revised: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
A range of hybrid molecules incorporating the ciminalum moiety in the thiazolidinone ring demonstrate significant anticancer and antimicrobial properties. Therefore, the aim of our study was to evaluate the properties and mechanism of action of two 4-thiazolidinone-based derivatives, i.e., 3-{5-[(Z,2Z)-2-chloro-3-(4-nitrophenyl)-2-propenylidene]-4-oxo-2-thioxothiazolidin-3-yl}propanoic acid (Les-45) and 5-[2-chloro-3-(4-nitrophenyl)-2-propenylidene]-2-(3-hydroxyphenylamino)thiazol-4(5H)-one (Les-247). In our study, we analyzed the impact of Les-45 and Les-247 on metabolic activity, caspase-3 activity, and the expression of genes and proteins related to inflammatory and antioxidant defenses and cytoskeleton rearrangement in healthy human fibroblasts (BJ) and a human lung carcinoma cell line (A549). The cells were exposed to increasing concentrations (1 nM to 100 μM) of the studied compounds for 24 h and 48 h. A decrease in the metabolic activity in the BJ and A549 cell lines was induced by both compounds at a concentration range from 10 to 100 µM. Both compounds decreased the mRNA expression of NRF2 (nuclear factor erythroid 2-related factor 2) and β-actin in the BJ cells. Interestingly, a significant decrease in the level of NF-κB gene and protein expression was detected in the BJ cell line, suggesting a direct impact of the studied compounds on the inhibition of inflammation. However, more studies are needed due to the ability of Les-45 and Les-247 to interfere with the tubulin/actin cytoskeleton, i.e., a critical system existing in eukaryotic cells.
Collapse
Affiliation(s)
- Dominika Szlachcikowska
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (D.S.); (R.L.); (K.A.S.)
| | - Anna Tabęcka-Łonczyńska
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (D.S.); (R.L.); (K.A.S.)
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (S.H.); (O.R.)
- Department of Organic Chemistry and Pharmacy, Lesya Ukrainka Volyn National University, Volya Avenue 13, 43025 Lutsk, Ukraine
| | - Olexandra Roman
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (S.H.); (O.R.)
| | - Yulia Shepeta
- Department of Pharmaceutical Chemistry, National Pirogov Memorial Medical University, Pirogov 56, 21018 Vinnytsia, Ukraine;
| | - Roman Lesyk
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (D.S.); (R.L.); (K.A.S.)
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (S.H.); (O.R.)
| | - Konrad A. Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (D.S.); (R.L.); (K.A.S.)
| |
Collapse
|
35
|
Khoshnavay Foumani M, Amirshahrokhi K, Namjoo Z, Niapour A. Carvedilol attenuates inflammatory reactions of lipopolysaccharide-stimulated BV2 cells and modulates M1/M2 polarization of microglia via regulating NLRP3, Notch, and PPAR-γ signaling pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4727-4736. [PMID: 38133658 DOI: 10.1007/s00210-023-02914-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Microglial cells coordinate immune responses in the central nervous system. Carvedilol (CVL) is a non-selective β-blocker with anti-inflammatory and anti-oxidant effects. This study aims to investigate the anti-inflammatory effects and the underlying mechanisms of CVL on lipopolysaccharide (LPS)-induced inflammation in microglial BV2 cells. BV2 cells were stimulated with LPS, and the protective effects of CVL were investigated via measurement of cell viability, reactive oxygen species (ROS), and interleukin (IL)-1β liberation. The protein levels of some inflammatory cascade, Notch, and peroxisome proliferator-activated receptor (PPAR)-γ pathways and relative markers of M1/M2 microglial phenotypes were assessed. Neuroblastoma SH-SY5Y cells were cultured with a BV2-conditioned medium (CM), and the capacity of CVL to protect cell viability was evaluated. CVL displayed a protective effect against LPS stress through reducing ROS and down-regulating of nuclear factor kappa B (NF-κB) p65, NLR family pyrin domain containing-3 (NLRP3), and IL-1β proteins. LPS treatment significantly increased the levels of the M1 microglial marker inducible nitric oxide synthase (iNOS) and M1-associated cleaved-NOTCH1 and hairy and enhancer of split-1 (HES1) proteins. Conversely, LPS treatment reduced the levels of the M2 marker arginase-1 (Arg-1) and PPAR-γ proteins. CVL pre-treatment reduced the protein levels of iNOS, cleaved-NOTCH1, and HES1, while increased Arg-1 and PPAR-γ. CM of CVL-primed BV2 cells significantly improved SH-SY5Y cell viability as compared with the LPS-induced cells. CVL suppressed ROS production and alleviated the expression of inflammatory markers in LPS-stimulated BV2 cells. Our results demonstrated that targeting Notch and PPAR-γ pathways as well as directing BV2 cell polarization toward the M2 phenotype may provide a therapeutic strategy to suppress neuroinflammation by CVL.
Collapse
Affiliation(s)
- Mohammadjavad Khoshnavay Foumani
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Keyvan Amirshahrokhi
- Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Zeinab Namjoo
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ali Niapour
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
36
|
Pázmándi K, Szöllősi AG, Fekete T. The "root" causes behind the anti-inflammatory actions of ginger compounds in immune cells. Front Immunol 2024; 15:1400956. [PMID: 39007134 PMCID: PMC11239339 DOI: 10.3389/fimmu.2024.1400956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
Ginger (Zingiber officinale) is one of the most well-known spices and medicinal plants worldwide that has been used since ancient times to treat a plethora of diseases including cold, gastrointestinal complaints, nausea, and migraine. Beyond that, a growing body of literature demonstrates that ginger exhibits anti-inflammatory, antioxidant, anti-cancer and neuroprotective actions as well. The beneficial effects of ginger can be attributed to the biologically active compounds of its rhizome such as gingerols, shogaols, zingerone and paradols. Among these compounds, gingerols are the most abundant in fresh roots, and shogaols are the major phenolic compounds of dried ginger. Over the last two decades numerous in vitro and in vivo studies demonstrated that the major ginger phenolics are able to influence the function of various immune cells including macrophages, neutrophils, dendritic cells and T cells. Although the mechanism of action of these compounds is not fully elucidated yet, some studies provide a mechanistic insight into their anti-inflammatory effects by showing that ginger constituents are able to target multiple signaling pathways. In the first part of this review, we summarized the current literature about the immunomodulatory actions of the major ginger compounds, and in the second part, we focused on the possible molecular mechanisms that may underlie their anti-inflammatory effects.
Collapse
Affiliation(s)
| | | | - Tünde Fekete
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
37
|
Elkhamary A, Gerner I, Bileck A, Oreff GL, Gerner C, Jenner F. Comparative proteomic profiling of the ovine and human PBMC inflammatory response. Sci Rep 2024; 14:14939. [PMID: 38942936 PMCID: PMC11213919 DOI: 10.1038/s41598-024-66059-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/26/2024] [Indexed: 06/30/2024] Open
Abstract
Understanding the cellular and molecular mechanisms of inflammation requires robust animal models. Sheep are commonly used in immune-related studies, yet the validity of sheep as animal models for immune and inflammatory diseases remains to be established. This cross-species comparative study analyzed the in vitro inflammatory response of ovine (oPBMCs) and human PBMCs (hPBMCs) using mass spectrometry, profiling the proteome of the secretome and whole cell lysate. Of the entire cell lysate proteome (oPBMCs: 4217, hPBMCs: 4574 proteins) 47.8% and in the secretome proteome (oPBMCs: 1913, hPBMCs: 1375 proteins) 32.8% were orthologous between species, among them 32 orthologous CD antigens, indicating the presence of six immune cell subsets. Following inflammatory stimulation, 71 proteins in oPBMCs and 176 in hPBMCs showed differential abundance, with only 7 overlapping. Network and Gene Ontology analyses identified 16 shared inflammatory-related terms and 17 canonical pathways with similar activation/inhibition patterns in both species, demonstrating significant conservation in specific immune and inflammatory responses. However, ovine PMBCs also contained a unique WC1+γδ T-cell subset, not detected in hPBMCs. Furthermore, differences in the activation/inhibition trends of seven canonical pathways and the sets of DAPs between sheep and humans, emphasize the need to consider interspecies differences in translational studies and inflammation research.
Collapse
Affiliation(s)
- A Elkhamary
- Department for Companion Animals and Horses, Veterm, University Equine Hospital, Vetmeduni Vienna, Vienna, Austria
- Department for Surgery, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - I Gerner
- Department for Companion Animals and Horses, Veterm, University Equine Hospital, Vetmeduni Vienna, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - A Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - G L Oreff
- Department for Companion Animals and Horses, Veterm, University Equine Hospital, Vetmeduni Vienna, Vienna, Austria
| | - C Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - F Jenner
- Department for Companion Animals and Horses, Veterm, University Equine Hospital, Vetmeduni Vienna, Vienna, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| |
Collapse
|
38
|
Artimovič P, Badovská Z, Toporcerová S, Špaková I, Smolko L, Sabolová G, Kriváková E, Rabajdová M. Oxidative Stress and the Nrf2/PPARγ Axis in the Endometrium: Insights into Female Fertility. Cells 2024; 13:1081. [PMID: 38994935 PMCID: PMC11240766 DOI: 10.3390/cells13131081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Successful pregnancy depends on precise molecular regulation of uterine physiology, especially during the menstrual cycle. Deregulated oxidative stress (OS), often influenced by inflammatory changes but also by environmental factors, represents a constant threat to this delicate balance. Oxidative stress induces a reciprocally regulated nuclear factor erythroid 2-related factor 2/peroxisome proliferator-activated receptor-gamma (Nrf2/PPARγ) pathway. However, increased PPARγ activity appears to be a double-edged sword in endometrial physiology. Activated PPARγ attenuates inflammation and attenuates OS to restore redox homeostasis. However, it also interferes with physiological processes during the menstrual cycle, such as hormonal signaling and angiogenesis. This review provides an elucidation of the molecular mechanisms that support the interplay between PPARγ and OS. Additionally, it offers fresh perspectives on the Nrf2/PPARγ pathway concerning endometrial receptivity and its potential implications for infertility.
Collapse
Affiliation(s)
- Peter Artimovič
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Zuzana Badovská
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Silvia Toporcerová
- Department of Gynaecology and Obstetrics, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia;
| | - Ivana Špaková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Lukáš Smolko
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Gabriela Sabolová
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Eva Kriváková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Miroslava Rabajdová
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| |
Collapse
|
39
|
Briganti S, Mosca S, Di Nardo A, Flori E, Ottaviani M. New Insights into the Role of PPARγ in Skin Physiopathology. Biomolecules 2024; 14:728. [PMID: 38927131 PMCID: PMC11201613 DOI: 10.3390/biom14060728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a transcription factor expressed in many tissues, including skin, where it is essential for maintaining skin barrier permeability, regulating cell proliferation/differentiation, and modulating antioxidant and inflammatory responses upon ligand binding. Therefore, PPARγ activation has important implications for skin homeostasis. Over the past 20 years, with increasing interest in the role of PPARs in skin physiopathology, considerable effort has been devoted to the development of PPARγ ligands as a therapeutic option for skin inflammatory disorders. In addition, PPARγ also regulates sebocyte differentiation and lipid production, making it a potential target for inflammatory sebaceous disorders such as acne. A large number of studies suggest that PPARγ also acts as a skin tumor suppressor in both melanoma and non-melanoma skin cancers, but its role in tumorigenesis remains controversial. In this review, we have summarized the current state of research into the role of PPARγ in skin health and disease and how this may provide a starting point for the development of more potent and selective PPARγ ligands with a low toxicity profile, thereby reducing unwanted side effects.
Collapse
Affiliation(s)
| | | | | | - Enrica Flori
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy; (S.B.); (S.M.); (A.D.N.); (M.O.)
| | | |
Collapse
|
40
|
Shan X, Li J, Hong B, Yin H, Lu Z, Wang G, Yu N, Peng D, Wang L, Zhang C, Chen W. Comparative efficacy of sweated and non-sweated Salvia miltiorrhiza Bge. extracts on acute myocardial ischemia via regulating the PPARα/RXRα/NF-κB signaling pathway. Heliyon 2024; 10:e31923. [PMID: 38845919 PMCID: PMC11154627 DOI: 10.1016/j.heliyon.2024.e31923] [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: 01/12/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Salvia miltiorrhiza Bge. (S. miltiorrhiza) is a well-known traditional Chinese medicine for the treatment of cardiovascular diseases. The processing of S. miltiorrhiza requires the raw herbs to sweat first and then dry. The aim of this study was to investigate the anti-acute myocardial ischemia (AMI) of S. miltiorrhiza extracts (including tanshinones and phenolic acids) before and after sweating, and to further explore whether the "sweating" primary processing affected the efficacy of S. miltiorrhiza. The AMI animal model was established by subcutaneous injection of isoprenaline hydrochloride (ISO). After treatment, the cardiac function of rats was evaluated by electrocardiogram (ECG), biochemical, and histochemical analysis. Moreover, the regulation of S. miltiorrhiza extracts on the peroxisome proliferator-activated receptor α (PPARα)/retinoid X receptor α (RXRα)/nuclear transcription factor-kappa B (NF-κB) signaling pathway of rats was assessed by the Western blotting. The results showed that sweated and non-sweated S. miltiorrhiza extracts including tanshinones and phenolic acids significantly reduced ST-segment elevation in ECG and the myocardial infarction area in varying degrees. Meanwhile, sweated and non-sweated S. miltiorrhiza reversed the activities of aspartate transaminase (AST), lactic dehydrogenase (LDH), creatine kinase-MB (CK-MB), and superoxide dismutase (SOD), as well as the levels of interleukin-6 (IL-6), interleukin-10 (IL-10), and tumor necrosis factor-α (TNF-α) in AMI rats. Concurrently, the results of Western blotting revealed that S. miltiorrhiza extracts regulated the PPARα/RXRα/NF-κB signaling pathway to exert an anti-inflammatory effect. Most importantly, sweated S. miltiorrhiza tanshinones extracts are more effective than the non-sweated S. miltiorrhiza, and the anti-inflammatory efficacy of tanshinones extract was also better than that of phenolic acid extract. Although phenolic acid extracts before and after sweating were effective in anti-AMI, there was no significant difference between them. In conclusion, both tanshinones and phenolic acids extracts of sweated and non-sweated S. miltiorrhiza promote anti-oxidative stress and anti-inflammatory against AMI via regulating the PPARα/RXRα/NF-κB signaling pathway. Further, the comparations between sweated and non-sweated S. miltiorrhiza extracts indicate that sweated S. miltiorrhiza tanshinones extracts have better therapeutic effects on AMI.
Collapse
Affiliation(s)
- Xiaoxiao Shan
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Junying Li
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Bangzhen Hong
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Huihui Yin
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Ziyi Lu
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Guokai Wang
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
| | - Nianjun Yu
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Daiyin Peng
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Lei Wang
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Caiyun Zhang
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
| | - Weidong Chen
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| |
Collapse
|
41
|
Abdel-Reheim MA, Ali GF, Hassanein EHM, Mohamed WR. Role of Nrf2/HO-1, PPAR-γ, and cytoglobin signals in the pathogenesis of methotrexate-induced testicular intoxication in rats and the protective effect of diacerein. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4235-4246. [PMID: 38060042 DOI: 10.1007/s00210-023-02876-w] [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: 09/16/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
Methotrexate (MTX) is an inhibitor of folic acid reductase used in managing a variety of malignancies. Testicular injury by MTX is one of its serious adverse effects. The current investigation aims to assess the protective effects of diacerein (DIA) on testicular injury by MTX and clarify the possible underlying mechanisms. Testicular injury in rats was induced by a single injection of 20 mg/kg body weight of MTX. DIA was given in 25 mg/kg body weight/day and 50 mg/kg body weight/day doses for 10 days. Compared to the MTX group, DIA attenuated testicular intoxication as evidenced by improvement of testicular histopathological abnormalities and increased serum testosterone and luteinizing hormone. DIA attenuated testicular oxidative stress changes by lowering testicular MDA and boosting GSH content and SOD activity. Moreover, administration of DIA attenuated MTX-induced testicular inflammation, as proved by decreased TNF-α and IL-6. At the molecular level, DIA induced significant upregulation in Nrf2, HO-1, PPAR-γ, and cytoglobin protein expression. The present results proved that DIA, in a dose-dependent manner, exhibited notable amelioration of testicular toxicity induced by MTX through augmentation of anti-inflammatory and antioxidant effects combined by upregulating Nrf2/HO-1, PPAR-γ, and cytoglobin signaling.
Collapse
Affiliation(s)
- Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, 11961, Shaqra, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Gaber F Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Wafaa R Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| |
Collapse
|
42
|
Rawat S, Singh G, Prasad A. Investigating the Taenia solium Fatty Acid Binding Protein Superfamily for Their Immunological Outlook and Prospect for Therapeutic Targets. ACS OMEGA 2024; 9:22557-22572. [PMID: 38826528 PMCID: PMC11137695 DOI: 10.1021/acsomega.3c09253] [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: 11/20/2023] [Revised: 04/22/2024] [Accepted: 05/07/2024] [Indexed: 06/04/2024]
Abstract
Taenia solium, like other helminthic parasites, lacks key components of cellular machinery required for endogenous lipid biosynthesis. This deficiency compels the parasite to obtain all of its lipid requirements from its host. The passage of lipids across the cell membrane is tightly regulated. To facilitate effective lipid transport, the cestode parasite utilizes certain lipid binding proteins called FABPs. These FABPs bind with the lipid ligands and allow the transport of lipids across the membranes and into the cytosol. Here, by integrating a computational with homology protein prediction tools, we had identified five FABPs in the T. solium proteome. We confirmed their presence by RNA expression analysis of respective genes from the parasite's cysticerci transcript. During the molecular modeling and MD simulation studies, two of them, TsM_000544100 and TsM_001185100, were most stable. Furthermore, they had a robust interaction with the IgG1 molecule, as evidenced by MD simulation. In addition, by employing in silico screening, we had identified potential ligand interacting residues that are present on the probable druggable site. In combination with in vitro cysticidal assays, enalaprilat dihydrate showed efficacy against cysticerci, which suggests that FABPs play a significant role in the cysticercus life cycle. Together, we provided a detailed distribution of all FABPs expressed by T. solium cysticerci and the critical role of TsM_001185100 in cysticercus viability.
Collapse
Affiliation(s)
- Suraj
S. Rawat
- School
of Biosciences and Bioengineering, Indian
Institute of Technology Mandi, Mandi, Himachal Pradesh 175005, India
| | - Gagandeep Singh
- Dayanad
Medical College and Hospital, Ludhiana, Punjab 141001,India
| | - Amit Prasad
- School
of Biosciences and Bioengineering, Indian
Institute of Technology Mandi, Mandi, Himachal Pradesh 175005, India
- Indian
Knowledge System and Mental Health Centre, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh 175005, India
- Centre
for Human-Computer Interaction, Indian Institute
of Technology Mandi, Mandi, Himachal Pradesh 175005, India
| |
Collapse
|
43
|
Tian A, Zheng Y, Li H, Zhang Z, Du L, Huang X, Sun L, Wu H. Eicosapentaenoic acid activates the P62/KEAP1/NRF2 pathway for the prevention of diabetes-associated cognitive dysfunction. Food Funct 2024; 15:5251-5271. [PMID: 38680120 DOI: 10.1039/d4fo00774c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Diabetes-associated cognitive dysfunction (DCD) is a severe complication of diabetes mellitus (DM), threatening the life quality of the diabetic population. However, there is still a lack of effective approaches for its intervention. Eicosapentaenoic acid (EPA) is an omega-3 polyunsaturated fatty acid that was not previously investigated for its effect on DCD. In this study, EPA was found to improve DCD in a mouse model of type 2 DM (T2DM) induced by streptozotocin and a high-fat diet, exhibiting profound protective effects on cognitive dysfunction, neuronal loss, and cerebral oxidative stress and inflammation. While EPA did not attenuate advanced glycation end product-induced neuron injury, we hypothesized that EPA might protect neurons by regulating microglia polarization, the effect of which was confirmed by the co-culture of neurons and lipopolysaccharide-stimulated microglia. RNA sequencing identified nuclear factor-erythroid-2-related factor 2 (NRF2) antioxidant signaling as a major target of EPA in microglia. Mechanistically, EPA increased sequestosome-1 (SQSTM1 or P62) levels that might structurally inhibit Kelch-like ECH associated protein 1 (KEAP1), leading to nuclear translocation of NRF2. P62 and NRF2 predominantly mediated EPA's effect since the knockdown of P62 or NRF2 abolished EPA's protective effect on microglial oxidative stress and inflammation and sequential neuron injuries. Moreover, the regulation of P62/KEPA1/NRF2 axes by EPA was confirmed in the hippocampi of diabetic mice. The present work presents EPA as an effective nutritional approach and microglial P62/KEAP1/NRF2 as molecular targets for the intervention of DCD.
Collapse
Affiliation(s)
- Ao Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| | - Yan Zheng
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Hui Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Zhiyue Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Lei Du
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| | - Xiaoli Huang
- Department of Nutrition, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Rd., Jinan, Shandong 250012, China.
| | - Lei Sun
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Rd., Jinan, Shandong 250012, China.
| | - Hao Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| |
Collapse
|
44
|
Pomianek T, Zagórska-Dziok M, Skóra B, Ziemlewska A, Nizioł-Łukaszewska Z, Wójciak M, Sowa I, Szychowski KA. Comparison of the Antioxidant and Cytoprotective Properties of Extracts from Different Cultivars of Cornus mas L. Int J Mol Sci 2024; 25:5495. [PMID: 38791533 PMCID: PMC11122231 DOI: 10.3390/ijms25105495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Cornus mas L. is a rich source of vitamin C and polyphenols. Due to their health-benefit properties, C. mas L. extracts have been used in, e.g., dermatology and cosmetology, and as a food supplement. Peroxisome proliferator-activated receptor gamma (PPARγ) and its co-activator (PGC-1α) are now suspected to be the main target of active substances from C. mass extracts, especially polyphenols. Moreover, the PPARγ pathway is involved in the development of different diseases, such as type 2 diabetes mellitus (DM2), cancers, skin irritation, and inflammation. Therefore, the aim of the present study was to evaluate the PPARγ pathway activation by the most popular water and ethanol extracts from specific C. mas L. cultivars in an in vitro model of the human normal fibroblast (BJ) cell line. We analyzed the content of biologically active compounds in the extracts using the UPLC-DAD-MS technique and revealed the presence of many polyphenols, including gallic, quinic, protocatechuic, chlorogenic, and ellagic acids as well as iridoids, with loganic acid being the predominant component. In addition, the extracts contained cyanidin 3-O-galactoside, pelargonidin 3-O-glucoside, and quercetin 3-glucuronide. The water-ethanol dark red extract (DRE) showed the strongest antioxidant activity. Cytotoxicity was assessed in a normal skin cell line, and positive effects of all the extracts with concentrations ranging from 10 to 1000 µg/mL on the cells were shown. Our data show that the studied extracts activate the PPARγ/PGC-1α molecular pathway in BJ cells and, through this mechanism, initiate antioxidant response. Moreover, the activation of this molecular pathway may increase insulin sensitivity in DM2 and reduce skin irritation.
Collapse
Affiliation(s)
- Tadeusz Pomianek
- Department of Management, Faculty of Administration and Social Sciences, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland;
| | - Martyna Zagórska-Dziok
- Department of Technology of Cosmetic and Pharmaceutical Products, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (M.Z.-D.); (A.Z.); (Z.N.-Ł.)
| | - Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland;
| | - Aleksandra Ziemlewska
- Department of Technology of Cosmetic and Pharmaceutical Products, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (M.Z.-D.); (A.Z.); (Z.N.-Ł.)
| | - Zofia Nizioł-Łukaszewska
- Department of Technology of Cosmetic and Pharmaceutical Products, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (M.Z.-D.); (A.Z.); (Z.N.-Ł.)
| | - Magdalena Wójciak
- Department of Analytical Chemistry, Medical University of Lublin, Aleje Raclawickie 1, 20-059 Lublin, Poland; (M.W.); (I.S.)
| | - Ireneusz Sowa
- Department of Analytical Chemistry, Medical University of Lublin, Aleje Raclawickie 1, 20-059 Lublin, Poland; (M.W.); (I.S.)
| | - Konrad A. Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland;
| |
Collapse
|
45
|
Liu X, Miao R, Liu K, Xie Q, Zheng P, Zhu J, Zhang Y, Peng F. Panoramic analysis of cell death patterns reveals prognostic and immune profiles of head and neck squamous cell carcinoma. Am J Cancer Res 2024; 14:2584-2607. [PMID: 38859838 PMCID: PMC11162683 DOI: 10.62347/pmda6193] [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: 02/21/2024] [Accepted: 05/15/2024] [Indexed: 06/12/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) has been characterized by a low therapeutic response and poor prognosis. Currently, there are no reliable predictive models for HNSCC progression and therapeutic efficacy. This study explores the role of diverse patterns of cell death in tumor development, positing them as predictive factors of HNSCC prognosis. We utilized bulk transcriptome and single-cell transcriptome, align with clinical information from TCGA and GEO database, to analyze genes associated with 15 types of cell death and construct a cell death index (CDI) signature. The associations of CDI with tumor-infiltrating immune cells and immunotherapy-related biomarkers were also evaluated using various algorithms. The CDI signature emerged as a robust prognosis biomarker that could identify patients who can benefit potentially from immunotherapy, thus improving diagnostic accuracy and optimizing clinical decisions in HNSCC management. Notably, we discovered that CAAP1 deficiency not only induced apoptosis but also enhanced anti-tumor immunity, suggesting its potential as a target for clinical drug development.
Collapse
Affiliation(s)
- Xinyu Liu
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| | - Rui Miao
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South UniversityNo. 87 Xiangya Road, Changsha 410008, Hunan, The People’s Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan ProvinceNo. 87 Xiangya Road, Changsha 410008, Hunan, The People’s Republic of China
| | - Kui Liu
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| | - Qun Xie
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| | - Penghui Zheng
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| | - Junai Zhu
- Department of Gastroenterology, The Chinese University of Hong KongHong Kong SAR, China
| | - Ying Zhang
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| | - Fusen Peng
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| |
Collapse
|
46
|
Lv S, Huang J, Luo Y, Wen Y, Chen B, Qiu H, Chen H, Yue T, He L, Feng B, Yu Z, Zhao M, Yang Q, He M, Xiao W, Zou X, Gu C, Lu R. Gut microbiota is involved in male reproductive function: a review. Front Microbiol 2024; 15:1371667. [PMID: 38765683 PMCID: PMC11099273 DOI: 10.3389/fmicb.2024.1371667] [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: 01/16/2024] [Accepted: 04/08/2024] [Indexed: 05/22/2024] Open
Abstract
Globally, ~8%-12% of couples confront infertility issues, male-related issues being accountable for 50%. This review focuses on the influence of gut microbiota and their metabolites on the male reproductive system from five perspectives: sperm quality, testicular structure, sex hormones, sexual behavior, and probiotic supplementation. To improve sperm quality, gut microbiota can secrete metabolites by themselves or regulate host metabolites. Endotoxemia is a key factor in testicular structure damage that causes orchitis and disrupts the blood-testis barrier (BTB). In addition, the gut microbiota can regulate sex hormone levels by participating in the synthesis of sex hormone-related enzymes directly and participating in the enterohepatic circulation of sex hormones, and affect the hypothalamic-pituitary-testis (HPT) axis. They can also activate areas of the brain that control sexual arousal and behavior through metabolites. Probiotic supplementation can improve male reproductive function. Therefore, the gut microbiota may affect male reproductive function and behavior; however, further research is needed to better understand the mechanisms underlying microbiota-mediated male infertility.
Collapse
Affiliation(s)
- Shuya Lv
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Jingrong Huang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Yadan Luo
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Yuhang Wen
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Baoting Chen
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Hao Qiu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Huanxin Chen
- Gastrointestinal Surgery, Suining First People's Hospital, Suining, China
| | - Tianhao Yue
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Lvqin He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Baochun Feng
- Gastrointestinal Surgery, Suining First People's Hospital, Suining, China
| | - Zehui Yu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Mingde Zhao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Qian Yang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Manli He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Wudian Xiao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Xiaoxia Zou
- Gastrointestinal Surgery, Suining First People's Hospital, Suining, China
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Congwei Gu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ruilin Lu
- Gastrointestinal Surgery, Suining First People's Hospital, Suining, China
| |
Collapse
|
47
|
Purushothaman K, Crawford AD, Rocha SD, Göksu AB, Lange BM, Mydland LT, Vij S, Qingsong L, Øverland M, Press CM. Cyberlindnera jadinii yeast as a functional protein source: Modulation of immunoregulatory pathways in the intestinal proteome of zebrafish ( Danio rerio). Heliyon 2024; 10:e26547. [PMID: 38468924 PMCID: PMC10925985 DOI: 10.1016/j.heliyon.2024.e26547] [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: 11/17/2023] [Revised: 02/09/2024] [Accepted: 02/15/2024] [Indexed: 03/13/2024] Open
Abstract
Yeasts contain bioactive components that can enhance fish immune robustness and disease resistance. Our study focused on analyzing intestinal immunoregulatory pathways in zebrafish (Danio rerio) using iTRAQ and 2D LC-MS/MS to quantify intestinal proteins. Zebrafish were fed either control diet (C) or diet supplemented with autolyzed Cyberlindnera jadinii (ACJ). KEGG analysis revealed that ACJ yeast diet induced increased abundance of proteins related to arginine and proline metabolism, phagosome, C-lectin receptor signaling, ribosome and PPAR signaling pathways, which can modulate and enhance innate immune responses. ACJ yeast diet also showed decreased abundance of proteins associated with inflammatory pathways, including apoptosis, necroptosis and ferroptosis. These findings indicate boosted innate immune response and control of inflammation-related pathways in zebrafish intestine. Our findings in the well annotated proteome of zebrafish enabled a detailed investigation of intestinal responses and provide insight into health-beneficial effects of yeast species C. jadinii, which is relevant for aquaculture species.
Collapse
Affiliation(s)
- Kathiresan Purushothaman
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Alexander D. Crawford
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Sérgio D.C. Rocha
- Department of Animal and Aquaculture Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, P.O. Box 5003, Ås, Norway
| | - Aleksandar B. Göksu
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Byron Morales Lange
- Department of Animal and Aquaculture Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, P.O. Box 5003, Ås, Norway
| | - Liv Torunn Mydland
- Department of Animal and Aquaculture Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, P.O. Box 5003, Ås, Norway
| | - Shubha Vij
- School of Applied Science, Republic Polytechnic, 9 Woodlands Avenue 9, Singapore 738964, Singapore
- Tropical Futures Institute, James Cook University Singapore, 149 Sims Drive, 387380, Singapore
| | - Lin Qingsong
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore
| | - Margareth Øverland
- Department of Animal and Aquaculture Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, P.O. Box 5003, Ås, Norway
| | - Charles McL. Press
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
48
|
Ye B, Wang Q, Ye Q, Wang D, Wang Z, Dong Z, Zou J. Effects of different combinations of koumine and gelsemine on growth performance, intestinal health, and transcriptome of Cyprinus carpio. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133130. [PMID: 38086301 DOI: 10.1016/j.jhazmat.2023.133130] [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: 07/17/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 02/08/2024]
Abstract
Koumine (KM) and gelsemine (GS) have shown significant benefits in livestock production, but their potential in aquaculture remains largely unexplored. This study examined the impact of different KM and GS combinations as feed additives on C. carpio (90 fish per group, initial weight 1.95 ± 0.08 g). KM and GS were introduced in ratios of 2:2 (mg/kg), 2:1 (mg/kg), and 2:0.67 (mg/kg) over a 10-week aquaculture experiment. The results demonstrate that the 2:1 (mg/kg) group increases the villus length, muscular layer thickness, crude protein, and crude fat content. Regarding fatty acid content, KM and GS enhance the levels of various fatty acids, including the total saturated fatty acid and total monounsaturated fatty acid. Additionally, KM and GS improve the composition and function of the intestinal microbiota. The 2:1 (mg/kg) group significantly elevates the enzymatic activities of SOD, MDA, CAT and upregulates the expression of immune-related genes such as toll-like receptor 2, transforming growth factor β, and glutathione S-transferase. Transcriptomic analysis suggests that KM and GS may have potential benefits for nutrient utilization and immune regulation in C. carpio. In summary, this study provides valuable insights into the use of KM and GS as feed additives in aquaculture.
Collapse
Affiliation(s)
- Bin Ye
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Qiujie Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Qiao Ye
- School of Life Sciences, Huizhou University, Huizhou 516007, China
| | - Dongjie Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Zhenlu Wang
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Zaijie Dong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China.
| | - Jixing Zou
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
49
|
Amlerova Z, Chmelova M, Anderova M, Vargova L. Reactive gliosis in traumatic brain injury: a comprehensive review. Front Cell Neurosci 2024; 18:1335849. [PMID: 38481632 PMCID: PMC10933082 DOI: 10.3389/fncel.2024.1335849] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/01/2024] [Indexed: 01/03/2025] Open
Abstract
Traumatic brain injury (TBI) is one of the most common pathological conditions impacting the central nervous system (CNS). A neurological deficit associated with TBI results from a complex of pathogenetic mechanisms including glutamate excitotoxicity, inflammation, demyelination, programmed cell death, or the development of edema. The critical components contributing to CNS response, damage control, and regeneration after TBI are glial cells-in reaction to tissue damage, their activation, hypertrophy, and proliferation occur, followed by the formation of a glial scar. The glial scar creates a barrier in damaged tissue and helps protect the CNS in the acute phase post-injury. However, this process prevents complete tissue recovery in the late/chronic phase by producing permanent scarring, which significantly impacts brain function. Various glial cell types participate in the scar formation, but this process is mostly attributed to reactive astrocytes and microglia, which play important roles in several brain pathologies. Novel technologies including whole-genome transcriptomic and epigenomic analyses, and unbiased proteomics, show that both astrocytes and microglia represent groups of heterogenic cell subpopulations with different genomic and functional characteristics, that are responsible for their role in neurodegeneration, neuroprotection and regeneration. Depending on the representation of distinct glia subpopulations, the tissue damage as well as the regenerative processes or delayed neurodegeneration after TBI may thus differ in nearby or remote areas or in different brain structures. This review summarizes TBI as a complex process, where the resultant effect is severity-, region- and time-dependent and determined by the model of the CNS injury and the distance of the explored area from the lesion site. Here, we also discuss findings concerning intercellular signaling, long-term impacts of TBI and the possibilities of novel therapeutical approaches. We believe that a comprehensive study with an emphasis on glial cells, involved in tissue post-injury processes, may be helpful for further research of TBI and be the decisive factor when choosing a TBI model.
Collapse
Affiliation(s)
- Zuzana Amlerova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Martina Chmelova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Miroslava Anderova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Lydia Vargova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
50
|
Nolsøe JMJ, Underhaug J, Sørskar ÅM, Antonsen SG, Malterud KE, Gani O, Fan Q, Hjorth M, Sæther T, Hansen TV, Stenstrøm YH. Biological Evaluations, NMR Analyses, Molecular Modeling Studies, and Overview of the Synthesis of the Marine Natural Product (-)-Mucosin. Molecules 2024; 29:994. [PMID: 38474506 PMCID: PMC10933799 DOI: 10.3390/molecules29050994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Natural products obtained from marine organisms continue to be a rich source of novel structural architecture and of importance in drug discovery, medicine, and health. However, the success of such endeavors depends on the exact structural elucidation and access to sufficient material, often by stereoselective total synthesis, of the isolated natural product of interest. (-)-Mucosin (1), a fatty acid derivative, previously presumed to contain a rare cis-bicyclo[4.3.0]non-3-ene moiety, has since been shown to be the trans-congener. Analytically, the fused bicyclic ring system in (-)-1 constitutes a particular challenge in order to establish its relative and absolute stereochemistry. Herein, data from biological evaluations, NMR and molecular modeling studies of (-)-1 are presented. An overview of the synthetic strategies enabling the exact structural elucidation of (-)-mucosin (1) is also presented.
Collapse
Affiliation(s)
- Jens M. J. Nolsøe
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, P.O. Box 5003, NO-1433 Ås, Norway; (J.M.J.N.); (T.V.H.)
- Faculty of Biosciences and Aquaculture, Nord University, P.O. Box 1490, NO-8049 Bodø, Norway
| | - Jarl Underhaug
- Department of Chemistry, University of Bergen, Allégaten 41, NO-5007 Bergen, Norway;
| | - Åshild Moi Sørskar
- Department of Pharmacy, Section for Pharmaceutical Chemistry, University of Oslo, P.O. Box 1068, NO-0316 Oslo, Norway; (Å.M.S.); (K.E.M.); (O.G.)
| | - Simen Gjelseth Antonsen
- Department of Mechanical, Electronic and Chemical Engineering, Faculty of Technology, Art and Design, Oslo Metropolitan University, NO-0130 Oslo, Norway;
| | - Karl E. Malterud
- Department of Pharmacy, Section for Pharmaceutical Chemistry, University of Oslo, P.O. Box 1068, NO-0316 Oslo, Norway; (Å.M.S.); (K.E.M.); (O.G.)
| | - Osman Gani
- Department of Pharmacy, Section for Pharmaceutical Chemistry, University of Oslo, P.O. Box 1068, NO-0316 Oslo, Norway; (Å.M.S.); (K.E.M.); (O.G.)
| | - Qiong Fan
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, NO-0317 Oslo, Norway; (Q.F.); (M.H.); (T.S.)
| | - Marit Hjorth
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, NO-0317 Oslo, Norway; (Q.F.); (M.H.); (T.S.)
| | - Thomas Sæther
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, NO-0317 Oslo, Norway; (Q.F.); (M.H.); (T.S.)
| | - Trond V. Hansen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, P.O. Box 5003, NO-1433 Ås, Norway; (J.M.J.N.); (T.V.H.)
- Department of Pharmacy, Section for Pharmaceutical Chemistry, University of Oslo, P.O. Box 1068, NO-0316 Oslo, Norway; (Å.M.S.); (K.E.M.); (O.G.)
| | - Yngve H. Stenstrøm
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, P.O. Box 5003, NO-1433 Ås, Norway; (J.M.J.N.); (T.V.H.)
- Department of Mechanical, Electronic and Chemical Engineering, Faculty of Technology, Art and Design, Oslo Metropolitan University, NO-0130 Oslo, Norway;
| |
Collapse
|