1
|
Wu X, Shao Y, Chen Y, Zhang W, Dai S, Wu Y, Jiang X, Song X, Shen H. Comparative study on the anti-inflammatory and protective effects of different oxygen therapy regimens on lipopolysaccharide-induced acute lung injury in mice. Med Gas Res 2025; 15:171-179. [PMID: 39324894 PMCID: PMC11515059 DOI: 10.4103/mgr.medgasres-d-24-00044] [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: 06/08/2024] [Revised: 06/27/2024] [Accepted: 07/23/2024] [Indexed: 09/27/2024] Open
Abstract
Oxygen therapy after acute lung injury can regulate the inflammatory response and reduce lung tissue injury. However, the optimal exposure pressure, duration, and frequency of oxygen therapy for acute lung injury remain unclear. In the present study, after intraperitoneal injection of lipopolysaccharide in ICR mice, 1.0 atmosphere absolute (ATA) pure oxygen and 2.0 ATA hyperbaric oxygen treatment for 1 hour decreased the levels of proinflammatory factors (interleukin-1beta and interleukin-6) in peripheral blood and lung tissues. However, only 2.0 ATA hyperbaric oxygen increased the mRNA levels of anti-inflammatory factors (interleukin-10 and arginase-1) in lung tissue; 3.0 ATA hyperbaric oxygen treatment had no significant effect. We also observed that at 2.0 ATA, the anti-inflammatory effect of a single exposure to hyperbaric oxygen for 3 hours was greater than that of a single exposure to hyperbaric oxygen for 1 hour. The protective effect of two exposures for 1.5 hours was similar to that of a single exposure for 3 hours. These results suggest that hyperbaric oxygen alleviates lipopolysaccharide-induced acute lung injury by regulating the expression of inflammatory factors in an acute lung injury model and that appropriately increasing the duration and frequency of hyperbaric oxygen exposure has a better tissue-protective effect on lipopolysaccharide-induced acute lung injury. These results could guide the development of more effective oxygen therapy regimens for acute lung injury patients.
Collapse
Affiliation(s)
- Xinhe Wu
- Department of Hyperbaric Oxygen Medicine, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yanan Shao
- Department of Rehabilitation Medicine, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yongmei Chen
- Department of Pathology, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Wei Zhang
- Department of Pathology, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Shirong Dai
- Department of Pathology, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yajun Wu
- Department of Hyperbaric Oxygen Medicine, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xiaoge Jiang
- Department of Rehabilitation Medicine, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xinjian Song
- Department of Rehabilitation Medicine, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Hao Shen
- Department of Rehabilitation Medicine, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
2
|
Ju Z, Li X, Li X, Liang C, Xu Z, Chen H, Xiong D. Stranded heavy fuel oil exposure causes deformities, cardiac dysfunction, and oxidative stress in marine medaka Oryzias melastigma using an oiled-gravel-column system. FISH PHYSIOLOGY AND BIOCHEMISTRY 2025; 51:1-21. [PMID: 39695067 DOI: 10.1007/s10695-024-01437-z] [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: 01/24/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
Heavy fuel oil (HFO) stranded on the coastline poses a potential threat to the health of marine fish after an oil spill. In this study, an oiled-gravel-column (OGC) system was established to investigate the toxic effects of stranded HFO on marine medaka Oryzias melastigma. HFO 380# (sulfur content 2.9%) was chosen as one type of high sulfur fuel oil for acute toxicity tests. The marine medaka larvae were exposed to the OGC system effluents with oil loading rates of 0 (control), 400, 800, 1600, and 3200 µg HFO/g gravel for 144 h, respectively. Results showed that a prevalence of blue sac disease signs presented teratogenic effects, including decreased circulation, ventricular stretch, cardiac hemorrhage, and pericardial edema. Moreover, the treatments (800, 1600, and 3200 µg oil/g gravel) induced severe cardiotoxicity, characterized by significant bradycardia and reduced stroke volume with an overt decrease in cardiac output. Additionally, the antioxidant enzyme activities, including catalase (CAT), peroxidase (POD), and glutathione S-transferase (GST) were significantly upregulated at 800-3200 µg oil/g gravel except for a marked inhibition of CAT activity at 3200 µg oil/g gravel. Furthermore, significantly elevated protein carbonyl (PCO) levels were detected, suggesting that the organisms suffered severe protein oxidative damage subjected to the exposure. Overall, stranded HFO 380# exposure activated the antioxidant defense system (up-regulated POD and GST activities) of marine medaka and disrupted CAT activity, which could result in an oxidative stress state (elevated PCO levels) and might further contribute to cardiac dysfunction, deformities, and mortality.
Collapse
Affiliation(s)
- Zhonglei Ju
- College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, China
| | - Xishan Li
- College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, China.
| | - Xin Li
- College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, China
| | - Cen Liang
- College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, China
| | - Zhu Xu
- College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, China
| | - Huishu Chen
- College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, China
| | - Deqi Xiong
- College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, China.
| |
Collapse
|
3
|
Stinson JA, Sheen A, Lax BM, Yang GN, Duhamel L, Santollani L, Fink E, Palmeri J, Wittrup KD. Tumor Integrin-Targeted Glucose Oxidase Enzyme Promotes ROS-Mediated Cell Death that Combines with Interferon Alpha Therapy for Tumor Control. Mol Cancer Ther 2025; 24:118-130. [PMID: 39382078 PMCID: PMC11695183 DOI: 10.1158/1535-7163.mct-24-0163] [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: 03/06/2024] [Revised: 07/14/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024]
Abstract
Although heightened intratumoral levels of reactive oxygen species (ROS) are typically associated with a suppressive tumor microenvironment, under certain conditions ROS contribute to tumor elimination. Treatment approaches, including some chemotherapy and radiation protocols, increase cancer cell ROS levels that influence their mechanism of cell death and subsequent recognition by the immune system. Furthermore, activated myeloid cells rapidly generate ROS upon encounter with pathogens or infected cells to eliminate disease, and recently, this effector function has been noted in cancer contexts as well. Collectively, ROS-induced cancer cell death may help initiate adaptive antitumor immune responses that could synergize with current approved immunotherapies, for improved control of solid tumors. In this work, we explore the use of glucose oxidase, an enzyme which produces hydrogen peroxide, a type of ROS, to therapeutically mimic the endogenous oxidative burst from myeloid cells to promote antigen generation within the tumor microenvironment. We engineer the enzyme to target pan-tumor-expressed integrins both as a tumor-agnostic therapeutic approach and as a strategy to prolong local enzyme activity following intratumoral administration. We found the targeted enzyme potently induced cancer cell death and enhanced cross-presentation by dendritic cells in vitro and further combined with interferon alpha for long-term tumor control in murine MC38 tumors in vivo. Optimizing the single-dose administration of this enzyme overcomes limitations with immunogenicity noted for other prooxidant enzyme approaches. Overall, our results suggest ROS-induced cell death can be harnessed for tumor control and highlight the potential use of designed enzyme therapies alongside immunotherapy against cancer.
Collapse
Affiliation(s)
- Jordan A. Stinson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Allison Sheen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Brianna M. Lax
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Grace N. Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Lauren Duhamel
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Luciano Santollani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Elizabeth Fink
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Joseph Palmeri
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - K. Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
4
|
Das S, Krishnamoorthy J, Kar RK. Estimating the structural and spatial variables of allantoinase enzyme critical for protein adsorption. Biochem Biophys Res Commun 2025; 743:151161. [PMID: 39693939 DOI: 10.1016/j.bbrc.2024.151161] [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: 09/11/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
Designing enzyme-based sensors necessitates a comprehensive exploration of macromolecular properties. Integrating enzymes with a suitable transducer involves immobilizing them onto a surface, facilitated through adsorption or entrapment techniques. Allantoin, a stable biomarkers metabolite, holds promise for detecting oxidative stress-related complications through its enzyme. In this study, we examined allantoinase from various taxa, with bacterial origin comprising over 70 % of the dataset. Crucial residues such as Asp, His, and Gly in the active binding site and associated hydrophobic area play a critical role in maintaining binding specificity and sensitivity. In this work, we utilized bioinformatics tools to analyze properties such as pI, solubility index, amino acid hydropathy, stability, disordered regions, solvent-accessible surface area, and hydrodynamic parameters. The stability of allantoinase is assessed through surface Cys residues, hydrophobicity, and thermostability. Furthermore, the compactness and spherical geometry of the enzyme, which are crucial for protein adsorption are evaluated through parameters like spatial conformation, asphericity, and hydrodynamic radius distribution. Among the dataset, bacterial allantoinase demonstrates significant adaptability to environmental changes, as indicated by solvent-accessible surface area and instability index. This study highlights the importance of macromolecular properties underscoring their significance in optimizing, calibrating, and ensuring the stability of enzyme-based sensor design.
Collapse
Affiliation(s)
- Sheetal Das
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Assam, 781039, India
| | | | - Rajiv K Kar
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Assam, 781039, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, Assam, 781039, India.
| |
Collapse
|
5
|
Zhang WP, Zhang SY, Zhou Y, Sun WJ, Zhang SF, Lee JS, Wang M, Wang DZ. Divergent responses of an armored and an unarmored dinoflagellate to ocean acidification. HARMFUL ALGAE 2025; 141:102772. [PMID: 39645393 DOI: 10.1016/j.hal.2024.102772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/05/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
Dinoflagellates, both armored and unarmored, with distinct cell wall difference, are being affected by elevated CO2-induced ocean acidification (OA). However, their specific responses to OA are not well understood. In this study, we investigated the physiological and molecular response of the armored species Prorocentrum obtusidens and the unarmored species Karenia mikimotoi to OA over a 28-day period. The results show that the two species responded differently to OA. Cell growth rate, particulate organic carbon (POC) content, and the activities of C4 pathway enzymes decreased in P. obtusidens under future acidified ocean condition (pH 7.8, 1000 μatm pCO2), but the activities of carbonic anhydrase (CA), ribulose-1,5-bisphosphate carboxylase/oxygenase (RubisCO), and superoxide dismutase (SOD) increased. Whereas cell growth rate, contents of Chl a and PON, and SOD activity altered insignificantly in K. mikimotoi, but contents of POC and total carbohydrate, and the activity of RubisCO increased while the activities of CA and C4 pathway enzymes decreased. Transcriptomic analysis indicates that genes associated with antioxidative response, heat shock protein, proteasome, signal transduction, ribosome, and pH regulation were up-regulated in P. obtusidens but down-regulated in K. mikimotoi. Notably, the synthesis of soluble organic matter (i.e., spermidine and trehalose) was enhanced in K. mikimotoi, thereby regulating intracellular pH and improving stress resistance. This study highlights the divergent response of the armored and unarmored dinoflagellates to OA, with the unarmored dinoflagellate exhibiting a higher ability to withstand this stressor. Therefore, caution should be exercised when predicting the behavior and the eventual fate of dinoflagellates in the future acidified ocean.
Collapse
Affiliation(s)
- Wei-Ping Zhang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Shuo-Yu Zhang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Yang Zhou
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Wen-Jing Sun
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Shu-Feng Zhang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - Jae-Seong Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Minghua Wang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China.
| | - Da-Zhi Wang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
6
|
Zhang XJ, Pu YK, Yang PY, Wang MR, Zhang RH, Li XL, Xiao WL. Isolicoflavonol ameliorates acute liver injury via inhibiting NLRP3 inflammasome activation through boosting Nrf2 signaling in vitro and in vivo. Int Immunopharmacol 2024; 143:113233. [PMID: 39366075 DOI: 10.1016/j.intimp.2024.113233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND NOD like receptor pyrin domain containing 3 (NLRP3) inflammasome is involved in innate immunity, and related to liver injury. However, no inflammasome inhibitors are clinically available until now. Our previous research suggests that isolicoflavonol (ILF), isolated from Macaranga indica, is a potent NLRP3 inflammasome inhibitor, but its mechanism is unclear. METHODS Fluorescent imaging and Western blot assay were used to ascertain the effects of ILF on pyroptosis and NLRP3 inflammasome activation in macrophages. Next, Nrf2 signal pathway, its downstream gene transcription and expression were further investigated. ML385, a Nrf2 inhibitor, was used to verify whether ILF targets Nrf2 signaling. A carbon tetrachloride induced liver injury model was introduced to evaluate the liver protection activity of ILF in mice. RESULTS This work revealed that ILF inhibited macrophage LDH release and IL-1β secretion in a dose-dependent manner. ILF had no significant cytotoxic effect on macrophage, it reduced pyroptosis and Gasdermin D N-terminal fragment formation. Moreover, ILF inhibited IL-1β maturation and Caspase-1 cleavage, but did not affect NLRP3, pro-Caspase-1, pro-IL-1β and ASC expression. ILF decreased ASC speck rate and reduced ASC oligomer formation. ILF decreased aggregated JC-1 formation restoring mitochondria membrane potential. In addition, ILF increased Nrf2 expression, extended Nrf2 lifespan and upregulated Nrf2 signaling pathway in macrophages whether the NLRP3 inflammasome was activated or not. Besides, ILF increased Nrf2 nuclear translocation, maintained a high proportion of Nrf2 in the nucleus, and upregulated ARE-related gene transcription and expression. Furthermore, Nrf2 signal inhibition attenuated compound ILF-mediated inhibition of pyroptosis, inflammasome activation and upregulation of Nrf2 signaling. ILF in a liver injury mouse model inhibited NLRP3 inflammasome activation and enhanced Nrf2 signaling. CONCLUSION Our study verified that ILF ameliorates liver injury via inhibiting NLRP3 inflammasome activation through boosting Nrf2 signaling, and highlighted that ILF is a potent anti-inflammatory drug for inflammasome-related liver diseases.
Collapse
Affiliation(s)
- Xing-Jie Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Yu-Kun Pu
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Peng-Yun Yang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Meng-Ru Wang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Rui-Han Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China
| | - Xiao-Li Li
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China.
| | - Wei-Lie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming 650500, Yunnan, China; Southwest United Graduate School, Kunming 650500, Yunnan, China.
| |
Collapse
|
7
|
Stavrovskaya I, Morin BK, Madamba S, Alexander C, Romano A, Alam S, Pavlov L, Mitaishvili E, Peixoto PM. Mitochondrial ROS modulate presynaptic plasticity in the drosophila neuromuscular junction. Redox Biol 2024; 79:103474. [PMID: 39721493 DOI: 10.1016/j.redox.2024.103474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/29/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
The elevated emission of reactive oxygen species (ROS) from presynaptic mitochondria is well-documented in several inflammatory and neurodegenerative diseases. However, the potential role of mitochondrial ROS in presynaptic function and plasticity remains largely understudied beyond the context of disease. Here, we investigated this potential ROS role in presynaptic function and short-term plasticity by combining optogenetics, whole cell electrophysiological recordings, and live confocal imaging using a well-established protocol for induction and measurement of synaptic potentiation in Drosophila melanogaster neuromuscular junctions (NMJ). Optogenetic induction of ROS emission from presynaptic motorneuron mitochondria expressing mitokiller red (mK) resulted in synaptic potentiation, evidenced by an increase in the frequency of spontaneous mini excitatory junction potentials. Notably, this effect was not observed in flies co-expressing catalase, a cytosolic hydrogen peroxide (H2O2) scavenging enzyme. Moreover, the increase in electrical activity did not coincide with synaptic structural changes. The absence of Wnt1/Wg release from synaptic boutons suggested involvement of alternative or non-canonical signaling pathway(s). However, in existing boutons we observed an increase in the active zone (AZ) marker Brp/Erc1, which serves as docking site for the neurotransmitter vesicle release pool. We propose the involvement of putative redox switches in AZ components as the molecular target of mitochondrial H2O2. These findings establish a novel framework for understanding the signaling role of mROS in presynaptic structural and functional plasticity, providing insights into redox-based mechanisms of neuronal communication.
Collapse
Affiliation(s)
- Irina Stavrovskaya
- Baruch College and CUNY Graduate Center, 1 Baruch Way, New York, NY, 10010, USA
| | | | - Stephen Madamba
- Baruch College and CUNY Graduate Center, 1 Baruch Way, New York, NY, 10010, USA
| | | | - Alexis Romano
- Baruch College and CUNY Graduate Center, 1 Baruch Way, New York, NY, 10010, USA
| | - Samia Alam
- Baruch College and CUNY Graduate Center, 1 Baruch Way, New York, NY, 10010, USA
| | - Lucas Pavlov
- Baruch College and CUNY Graduate Center, 1 Baruch Way, New York, NY, 10010, USA
| | - Erna Mitaishvili
- Baruch College and CUNY Graduate Center, 1 Baruch Way, New York, NY, 10010, USA
| | - Pablo M Peixoto
- Baruch College and CUNY Graduate Center, 1 Baruch Way, New York, NY, 10010, USA.
| |
Collapse
|
8
|
Huang Y, Tang Y, Zhang R, Wu X, Yan L, Chen X, Wu Q, Chen Y, Lv Y, Su Y. Role of periodontal ligament fibroblasts in periodontitis: pathological mechanisms and therapeutic potential. J Transl Med 2024; 22:1136. [PMID: 39709490 DOI: 10.1186/s12967-024-05944-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024] Open
Abstract
Periodontal ligament fibroblasts (PDLFs) play a crucial role in the etiology of periodontitis and periodontal tissue regeneration. In healthy periodontal tissues, PDLFs maintain the homeostasis of periodontal soft and hard tissues as well as the local immune microenvironment. PDLFs also have the potential for multidirectional transdifferentiation and are involved in periodontal tissue regeneration. On the other hand, PDLFs can become dysfunctional and acquire an inflammatory phenotype to secret various inflammatory cytokines when affected by pathological factors. These cytokines further trigger immune and inflammatory events, and lead to destruction of periodontal soft and hard tissues as well as damage to the regenerative potential of PDLFs. This review summarizes the physiological functions of PDLFs. Meanwhile, this review also highlights recent insights into the pathological mechanisms driving the development of periodontitis through dysfunctional PDLFs and the negative impact on periodontal tissue regeneration. Additionally, this paper summarizes strategies for targeting PDLFs to treat periodontitis, involving blocking multiple stages of the inflammatory response induced by PDLFs and promoting the multidirectional transdifferentiation of PDLFs. Future research directions are proposed to address important questions that have not yet been answered in this field. This article provides a reference for understanding the important role of PDLFs in the pathological mechanisms of periodontitis and for developing new strategies for targeting PDLFs in periodontitis treatment.
Collapse
Affiliation(s)
- Yijie Huang
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Ying Tang
- Department of Prosthodontics, Huangpu District Dental Disease Prevention and Treatment Institute, Shanghai, 200001, China
| | - Ruiqi Zhang
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Xiao Wu
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Li Yan
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Xiling Chen
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Qianqi Wu
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Yiyan Chen
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Yingtao Lv
- Department of Prosthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Su
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China.
- Department of Periodontology, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Tang D, Li X, Zhang L, Xiao P, Nie Y, Qiu F, Cheng Z, Li W, Zhao Y. Reactive oxygen species-mediated signal transduction and utilization strategies in microalgae. BIORESOURCE TECHNOLOGY 2024; 418:132004. [PMID: 39710205 DOI: 10.1016/j.biortech.2024.132004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/10/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Reactive oxygen species (ROS) are crucial in stress perception, the integration of environmental signals, and the activation of downstream response networks. This review emphasizes ROS-mediated signaling pathways in microalgae and presents an overview of strategies for leveraging ROS. Eight distinct signaling pathways mediated by ROS in microalgae have been summarized, including the calcium signaling pathway, the target of rapamycin signaling pathway, the mitogen-activated protein kinase signaling pathway, the cyclic adenosine monophosphate/protein kinase A signaling pathway, the ubiquitin/protease pathway, the ROS-regulated transcription factors and enzymes, the endoplasmic reticulum stress, and the retrograde ROS signaling. Moreover, this review outlines three strategies for utilizing ROS: two-stage cultivation, combined stress with phytohormones, and strain engineering. The physicochemical properties of various ROS, together with their redox reactions with downstream targets, have been elucidated to reveal the role of ROS in signal transduction processes while delineating the ROS-mediated signal transduction network within microalgae.
Collapse
Affiliation(s)
- Dexin Tang
- College of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Xu Li
- College of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Lei Zhang
- College of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, PR China.
| | - Pengying Xiao
- College of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Yudong Nie
- College of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Facheng Qiu
- College of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Zhiliang Cheng
- College of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Wensheng Li
- College of Chemistry and Chemical Engineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Yongteng Zhao
- Yunnan Urban Agricultural Engineering & Technological Research Center, College of Agriculture and Life Science, Kunming University, Kunming 650214, PR China.
| |
Collapse
|
10
|
Chang J, Yao Y, Sun X, Wang W, Qian H, Liu Y, Xue C, Ye W, Jiang F. JAG1 mediates apoptosis in herpes simplex keratitis by suppressing autophagy via ROS/JAG1/NOTCH1/pULK1 signaling pathway. Cell Biol Toxicol 2024; 41:1. [PMID: 39704867 DOI: 10.1007/s10565-024-09968-0] [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: 09/03/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Herpes simplex keratitis (HSK), an ocular disease resulted from herpes simplex virus type 1 (HSV-1) infection, leads to the majority of infectious corneal blindness worldwide. The apoptosis of corneal epithelial cells (CECs) resulted from HSV-1 disrupts the epithelial barrier and exacerbates the infection; however, there is no definitive cure for HSK. Jagged1 (JAG1), one of the primary functional ligands for NOTCH receptors, plays a crucial role in regulating apoptosis and autophagy; however, its role in HSK is unclear. Our transcriptome analysis showed JAG1 was significantly upregulated in HSV-1-infected human CECs. We aimed to explore JAG1's role in regulating apoptosis in HSV-1-infected human CECs and in HSK mice. HSV-1 infection induced apoptosis and reactive oxygen species (ROS) generation in CECs. HSV-1 also activated the JAG1/NOTCH1 signaling pathway. The ROS scavenger N-acetylcysteine significantly mitigated these effects. Additionally, inhibiting the JAG1/NOTCH1 pathway with short hairpin RNA against JAG1 or a NOTCH1 inhibitor (N-[N-{3,5-difuorophenacetyl}-1-alanyl]-S-phenylglycine t-butyl ester [DAPT]) alleviated HSV-1-induced CEC apoptosis. Transmission electron microscopy and western blotting revealed that HSV-1 infection suppressed ULK1-mediated autophagy in CECs, while DAPT treatment enhanced autophagy by suppressing ULK1 phosphorylation. The activation of autophagy by rapamycin treatment markedly reduced ROS levels and apoptosis in HSV-1-infected CECs, revealing a synergistic effect between the suppressed autophagy and increased ROS levels, ultimately leading to apoptosis. Thus, HSV-1 induces CEC apoptosis by suppressing autophagy through ROS/JAG1/NOTCH1/pULK1 signaling pathway in vitro and in vivo, providing potential therapeutic targets for HSK.
Collapse
Affiliation(s)
- Jingyao Chang
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Yao Yao
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Xinghong Sun
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Nanjing, 210011, Jiangsu, China
| | - Wenzhe Wang
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Haochen Qian
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Yumeilan Liu
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Chunyan Xue
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Ophthalmology, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, Hainan, China.
| | - Wei Ye
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China.
| | - Feng Jiang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Nanjing, 210011, Jiangsu, China.
| |
Collapse
|
11
|
Fomichova O, Oliveira PF, Bernardino RL. Exploring the interplay between inflammation and male fertility. FEBS J 2024. [PMID: 39702986 DOI: 10.1111/febs.17366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/02/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Male fertility results from a complex interplay of physiological, environmental, and genetic factors. It is conditioned by the properly developed anatomy of the reproductive system, hormonal regulation balance, and the interplay between different cell populations that sustain an appropriate and functional environment in the testes. Unfortunately, the mechanisms sustaining male fertility are not flawless and their perturbation can lead to infertility. Inflammation is one of the factors that contribute to male infertility. In the testes, it can be brought on by varicocele, obesity, gonadal infections, leukocytospermia, physical obstructions or traumas, and consumption of toxic substances. As a result of prolonged or untreated inflammation, the testicular resident cells that sustain spermatogenesis can suffer DNA damage, lipid and protein oxidation, and mitochondrial dysfunction consequently leading to loss of function in affected Sertoli cells (SCs) and Leydig cells (LCs), and the formation of morphologically abnormal dysfunctional sperm cells that lay in the basis of male infertility and subfertility. This is due mainly to the production and secretion of pro-inflammatory mediators, including cytokines, chemokines, and reactive oxygen species (ROS) by local immune cells (macrophages, lymphocytes T, mast cells) and tissue-specific cells [SCs, LCs, peritubular myoid cells (PMCs) and germ cells (GCs)]. Depending on the location, duration, and intensity of inflammation, these mediators can exert their toxic effect on different elements of the testes. In this review, we discuss the most prevalent inflammatory factors that negatively affect male fertility and describe the different ways inflammation can impair male reproductive function.
Collapse
Affiliation(s)
- Oleksandra Fomichova
- UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, Portugal
| | - Raquel L Bernardino
- UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Portugal
| |
Collapse
|
12
|
Hou B, Li C, Yang F, Deng W, Hu C, Liu C, Chen Y, Xiao X, Huang X, Deng J, Xie S. Ultrasmall Antioxidant Copper Nanozyme to Enhance Stem Cell Microenvironment for Promoting Diabetic Wound Healing. Int J Nanomedicine 2024; 19:13563-13578. [PMID: 39720217 PMCID: PMC11668326 DOI: 10.2147/ijn.s487647] [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: 07/19/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Purpose Stem cell therapy is a promising approach for treating chronic diabetic wounds. However, its effectiveness is significantly limited by the high oxidative stress environment and persistent inflammation induced by diabetes. Strategies to overcome these challenges are essential to enhance the therapeutic potential of stem cell therapy. Methods Cu5.4O ultrasmall nanoparticles (Cu5.4O-USNPs), known for their excellent reactive oxygen species (ROS) scavenging properties, were utilized to protect adipose-derived stem cells (ADSCs) from oxidative stress injury. In vitro experiments were conducted to evaluate the viability, paracrine activity, and anti-inflammatory capabilities of ADSCs loaded with Cu5.4O-USNPs under oxidative stress conditions. In vivo experiments in diabetic mice were performed to assess the therapeutic effects of Cu5.4O-USNP-loaded ADSCs on wound healing, including their impact on inflammation, collagen synthesis, angiogenesis, and wound closure. Results ADSCs treated with Cu5.4O-USNPs showed significantly enhanced viability, paracrine activity, and anti-inflammatory properties under oxidative stress conditions in vitro. In diabetic mice, Cu5.4O-USNP-loaded ADSCs reduced inflammatory responses in wound tissues, promoted collagen synthesis and angiogenesis, and accelerated diabetic wound healing. These findings suggest that Cu5.4O-USNPs effectively mitigate the adverse effects of oxidative stress and inflammation, enhancing the therapeutic efficacy of ADSCs. Conclusion This study presents a simple and effective approach to improve the therapeutic potential of stem cell therapy for diabetic wounds. By incorporating Cu5.4O-USNPs, the antioxidative and anti-inflammatory capabilities of ADSCs are significantly enhanced, offering a promising strategy for ROS-related tissue repair and chronic wound healing.
Collapse
Affiliation(s)
- Biao Hou
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Chengyuan Li
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, Hunan, People’s Republic of China
| | - Fen Yang
- Department of Infectious Diseases, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Wanjun Deng
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Chao Hu
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Changxiong Liu
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Yanming Chen
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Xiangjun Xiao
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Xiongjie Huang
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Jun Deng
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Army Medical University, Chongqing, People’s Republic of China
| | - Songlin Xie
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| |
Collapse
|
13
|
Sun B, Hu M, Bock C, Shao Y, Chen H, Waiho K, Liu W, Khadka K, Xu C, Wang Y. Effects of perfluorooctanoic acid and nano titanium dioxide on the immune response and energy allocation in Mytilus coruscus. CHEMOSPHERE 2024; 370:143958. [PMID: 39701318 DOI: 10.1016/j.chemosphere.2024.143958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Perfluorooctanoic acid (PFOA) functions as a surfactant, while nano-titanium dioxide (nano-TiO2) serves as an antibacterial agent. These substances are extensively utilized in industrial production and, upon release into aquatic environments, pose significant threats to the viability and development of marine organisms. However, research into the effects of PFOA and nano-TiO2 on the immune functions and cellular energy allocation (CEA) of bivalves remains limited. To investigate the impact of PFOA and nano-TiO2 on immunity and cellular energy, we exposed Mytilus coruscus individuals to different concentrations of PFOA (2 and 200 μg/L), either alone or in combination with nano-TiO2 (0.1 mg/L, particle size: 25 nm) for 14 days. We found that the co-exposure to PFOA and nano-TiO2 had significant interactive effects on multiple immune function parameters of mussels. PFOA and nano-TiO2 notably reduced the total hemocyte count (THC), esterase activity (EST), mitochondrial number (MN), lysosomal content (LYSO), and cell viability, while concurrently elevating hemocyte mortality (HM) and reactive oxygen species (ROS) levels. Some immune-related genes, such as Tumor Necrosis Factor-alpha (TNF-α) and Myeloid Differentiation Primary Response 88 (MyD88) were downregulated, while others such as Interleukin 17 (IL-17) and Transforming Growth Factor-beta (TGF-β) were upregulated after 14-day exposure to combined pollutant exposure. Furthermore, negative effects on CEA were observed under both individual and combined pollutant stress. Therefore, PFOA and nano-TiO2 regulate cellular and humoral immunity through the regulation of immune genes as mediators, while simultaneously disrupting cellular energy metabolism. The immunotoxicity of organic and particulate pollutants, and their mixtures, thus poses a significant risk to the immune defense capabilities of mussel populations in polluted coastal environments.
Collapse
Affiliation(s)
- Bingyan Sun
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, 201306, China
| | - Menghong Hu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, 201306, China
| | - Christian Bock
- Integrative Ecophysiology, Alfred Wegener Institute, Helmholtz Centre for Polar & Marine Research, Bremerhaven, Germany
| | - Ying Shao
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, 201306, China
| | - Haodong Chen
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, 201306, China
| | - Khor Waiho
- Higher Institution Center of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries, University Malaysia Terengganu, Kuala Terengganu, Terengganu, 20000, Malaysia
| | - Wei Liu
- University of Geneva, Faculty of Sciences, Earth and Environment Sciences, Department F.-A. Forel for Environmental and Aquatic Sciences, CH-1211, Geneva, Switzerland
| | - Kiran Khadka
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, 201306, China
| | - Chaosong Xu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, 201306, China
| | - Youji Wang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, 201306, China.
| |
Collapse
|
14
|
Yang L, Hu Y, Deng H, Li Y, Zhang R, Zhang Q, Yang L, Pang H, Liu F, Fu C. Water-soluble polysaccharides from Torreya grandis nuts: Structural characterization and anti-inflammatory activity. Int J Biol Macromol 2024; 291:138935. [PMID: 39701235 DOI: 10.1016/j.ijbiomac.2024.138935] [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: 09/09/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
Torreya grandis (T. grandis) nuts are widely consumed as a functional food in China. In this study, we investigated the structural characteristics of T. grandis nuts polysaccharides and evaluated their potential biological functions with anti-inflammatory activities. Polysaccharides (TGP) were extracted from T. grandis nuts using water extraction and alcohol precipitation methods. Through a series of purification steps, three heteropolysaccharides (TGP-0a, TGP-2a, and TGP-3a) with distinct molecular weights, monosaccharide compositions, and surface morphologies were isolated. Their anti-inflammatory activities were screened, and TGP-0a was shown to be the most effective component. By combining NMR and methylation studies, TGP-0a was predominantly composed of linear α-1,4-glucan region and linear β-1,4-(gluco)mannan region. In cellular anti-inflammatory assays, TGP-0a significantly diminished the release of pro-inflammatory cytokines. Furthermore, by lowering the levels of iNOS and COX-2, TGP-0a decreased the release of inflammatory mediators (NO and ROS), thereby reducing oxidative stress and inflammatory response. In conclusion, T. grandis nut polysaccharides, particularly TGP-0a, show strong potential as natural anti-inflammatory agents for functional foods and pharmaceutical applications.
Collapse
Affiliation(s)
- Luping Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yunjie Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hongdan Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ruiyuan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qian Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Huiwen Pang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Fang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
15
|
Yang T, Lu Z, Song H, Chen Y, Jiang M, Zhan K, Zhao G. Knockout of hexokinase 2 regulates mitochondrial dysfunction and activates the NLRP3 signal pathway in the rumen epithelial cells of dairy cows. Int J Biol Macromol 2024; 289:138831. [PMID: 39701238 DOI: 10.1016/j.ijbiomac.2024.138831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
Hexokinase 2 (HK2) plays a vital role in mitochondrial homeostasis; however, the molecular mechanisms underlying its involvement in high-concentrate diet-induced damage in the ruminal epithelium of dairy cows are poorly understood. This study aimed to explore the regulatory role of HK2 in mitochondrial function and responses to inflammation in the rumen of dairy cows fed a high-concentrate diet. Our results showed that, compared with a low-concentrate (LC) diet, feeding a high-concentrate (HC) diet increased oxidative stress and reduced relative antioxidant gene expression levels and enzyme activities in the ruminal epithelium. Furthermore, the expression of genes related to mitochondrial biosynthesis and structure decreased in the HC group, concomitant with nuclear oligomerization domain (NOD)-like receptor 3 (NLRP3) signaling pathway activation, which compromised normal rumen epithelium function. Meanwhile, transcription results showed the same trend in HK2-knockout bovine rumen epithelial cells (HK2KO BRECs) related to wild-type (WT) BRECs. Notably, the knockout of HK2 aggravated mitochondrial dysfunction, resulting in the impairment of mitochondrial morphology and quality, a reduction in mitochondrial membrane potential (MMP), mitochondrial permeability transition pore (MPTP) opening, increased reactive oxygen species (ROS) generation, and decreased expression of antioxidant genes. These changes led to upregulating genes and proteins in the NLRP3 pathway and activating proinflammatory response. In addition, metabolomic results showed that knockout HK2 altered the glycerophospholipid metabolic pathway. This study provides new strategies for mitigating high-concentrate diet-induced injury in the ruminal epithelium of dairy cows.
Collapse
Affiliation(s)
- Tianyu Yang
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou, China; Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zhiqi Lu
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Ningxia Dairy Science and Innovation Center of Bright Farming Company Limited, Zhongwei, China
| | - Han Song
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yuhang Chen
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Maocheng Jiang
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Kang Zhan
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Guoqi Zhao
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
16
|
Li JM, Zhang L, Pei SL, Guo L, Shen HL, He J, Guo YY, Zhang WQ, Lin F. Copper-Based Nanoparticles for Effective Treatment Against Sepsis-Induced Lung Injury in Mice Model. Int J Nanomedicine 2024; 19:13507-13524. [PMID: 39713221 PMCID: PMC11662683 DOI: 10.2147/ijn.s488357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/06/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction Lung injury, a common complication of sepsis, arises from elevated reactive oxygen species (ROS), mitochondrial dysfunction, and cell death driven by inflammation. In this study, a novel class of ultrasmall nanoparticles (Cu4.5O USNPs) was developed to address sepsis-induced lung injury (SILI). Methods The synthesized nanoparticles were thoroughly characterized to assess their properties. In vitro experiments were conducted to determine the biologically effective concentration and elucidate the anti-inflammatory mechanism of action. These findings were further supported by in vivo studies, showcasing the material's efficacy in mitigating SILI. Results The Cu4.5O USNPs demonstrated remarkable scavenging capabilities for hydrogen peroxide (H2O2), superoxide anions (O2 -), and hydroxyl radicals (·OH), attributed to their catalase (CAT)- and superoxide dismutase (SOD)-like activities. Additionally, the nanoparticles exhibited strong anti-inflammatory effects, preserved mitochondrial homeostasis through potent ROS scavenging, and significantly reduced cell death. In vivo studies on mice further validated their protective role against SILI. The conclusion This study highlights the therapeutic potential of Cu4.5O USNPs in treating sepsis-induced lung injury by effectively scavenging ROS and reducing cell death. These findings provide compelling evidence for the future use of copper-based nanoparticles as antioxidant therapeutics.
Collapse
Affiliation(s)
- Jie-Mei Li
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi, People’s Republic of China
| | - Lu Zhang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi, People’s Republic of China
| | - Sheng-Lin Pei
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, Guangxi, People’s Republic of China
| | - Liang Guo
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, Guangxi, People’s Republic of China
| | - Hong-Lei Shen
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi, People’s Republic of China
| | - Jing He
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi, People’s Republic of China
| | - You-Yuan Guo
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi, People’s Republic of China
| | - Wei-Qing Zhang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, Guangxi, People’s Republic of China
| |
Collapse
|
17
|
Xu C, Yao X, Kong W, Mu B, Duan G, Wang J, Xu Y, Li X. Ecotoxicological risk of co-exposure to fosthiazate and microplastics on earthworms (Eisenia fetida): Integrating biochemical and transcriptomic analyses. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125053. [PMID: 39357558 DOI: 10.1016/j.envpol.2024.125053] [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: 08/01/2024] [Revised: 09/06/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
Fosthiazate (FOS) is a widely used organophosphorus insecticide effective against soil root-knot nematodes. However, its ecotoxicity to non-target soil organisms, particularly in combination with microplastics (MPs), is unclear. This study explores the toxic-effects and molecular mechanisms of co-exposure to FOS and MPs on earthworms (Eisenia fetida) using multilevel toxicity endpoints and transcriptomics. Results showed that both FOS and MPs elevated the intracellular levels of reactive oxygen species (ROS), malondialdehyde (MDA), and 8-hydroxy-2-deoxyguanosine (8-OHdG) in earthworms' cells. The superoxide dismutase (SOD) and catalase (CAT) activities followed a similar trend in all treatments, with changes observed at 14 and 28 days, indicating that co-exposure to FOS and MPs increased DNA oxidative damage. Notably, the co-exposure more significantly inhibited Ca2+-ATPase activity and exacerbated neurotoxicity compared to individual treatments, closely associated with changes in intracellular ROS levels that mediate neuroinhibition and lead to neurotoxicity. KEGG enrichment analysis revealed that MPs and FOS disrupted pathways related to metabolism, immunity, and apoptosis, while co-exposure primarily impaired endocrine and receptor pathways, showing higher toxicity. Our study offers novel insights into the ecotoxicological effects and mechanisms of pesticides and microplastics on earthworms, providing valuable data for evaluating the soil environmental health risks associated with compound pollution.
Collapse
Affiliation(s)
- Chonglin Xu
- College of Resources and Environment, Key Laboratory of Agricultural Environment, Shandong Agricultural University, Tai'an, 271000, China; State Key Lab of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Xiangfeng Yao
- College of Resources and Environment, Key Laboratory of Agricultural Environment, Shandong Agricultural University, Tai'an, 271000, China
| | - Weizheng Kong
- College of Resources and Environment, Key Laboratory of Agricultural Environment, Shandong Agricultural University, Tai'an, 271000, China
| | - Baoyan Mu
- College of Resources and Environment, Key Laboratory of Agricultural Environment, Shandong Agricultural University, Tai'an, 271000, China
| | - Guilan Duan
- State Key Lab of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Jun Wang
- College of Resources and Environment, Key Laboratory of Agricultural Environment, Shandong Agricultural University, Tai'an, 271000, China
| | - Yuxin Xu
- College of Resources and Environment, Key Laboratory of Agricultural Environment, Shandong Agricultural University, Tai'an, 271000, China.
| | - Xianxu Li
- College of Resources and Environment, Key Laboratory of Agricultural Environment, Shandong Agricultural University, Tai'an, 271000, China.
| |
Collapse
|
18
|
Chauhan A, Kamal R, Bhaita R, Thakur GS, Awasthi A. From Bench to Bedside: ROS-Responsive Nanocarriers in Cancer Therapy. AAPS PharmSciTech 2024; 26:10. [PMID: 39668268 DOI: 10.1208/s12249-024-03011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024] Open
Abstract
Reactive oxygen species (ROS) play a dual role in cancer, acting as both signaling molecules that promote tumour growth and as agents that can inhibit tumour progression through cytotoxic effects. In cancer therapy, ROS-responsive drug delivery systems take advantage of the elevated ROS levels found in tumors compared to healthy tissues. These systems are engineered to release drugs precisely in response to increased ROS levels in tumour cells, allowing targeted and controlled treatment, minimizing side effects, and enhancing therapeutic outcomes. ROS generation in cancer cells is linked to metabolic changes, mitochondrial dysfunction, and oncogenic signaling, leading to increased oxidative stress. Tumour cells manage this by upregulating antioxidant defenses to prevent ROS from reaching harmful levels. This balance between ROS production and neutralization is critical for cancer cell survival, making ROS both a challenge and an opportunity for targeted therapies. ROS also connect inflammation and cancer. Chronic inflammation leads to elevated ROS, which can damage DNA and proteins, promoting mutations and cancer development. Additionally, ROS contribute to protein degradation, affecting essential cellular functions. Therapeutic strategies targeting ROS aim to either increase ROS beyond tolerable levels for cancer cells or inhibit their antioxidant defenses. Nanocarriers responsive to ROS show great potential in improving the precision of cancer treatments by releasing drugs specifically in high ROS environments, like tumors. This review discusses the mechanisms of ROS in cancer, its role in inflammation and protein degradation, and the advances in ROS-targeted nanocarrier therapies across different cancer types.
Collapse
Affiliation(s)
- Abhishek Chauhan
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Raj Kamal
- School of Pharmacy, Desh Bhagat University, 147301, Punjab, India, Mandi Gobindgarh
| | - Rohit Bhaita
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Ankit Awasthi
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| |
Collapse
|
19
|
Zhang F, Lu L, Ma S, Sun J, Liu J, Gao N, Gou Z, Zhou Y, Lai C, Li Y, Sun M, Jiang H. Artemisinin attenuates perinatal inflammation and consequent oxidative stress in oligodendrocyte precursor cells by inhibiting IRAK-4 and IRAK-1. Int Immunopharmacol 2024; 142:113117. [PMID: 39293313 DOI: 10.1016/j.intimp.2024.113117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND The main causes of abnormal white matter development (periventricular leukomalacia) in premature infants are perinatal inflammation and the consequent oxidant/antioxidant imbalance in oligodendrocyte precursor cells (OPCs); however, the underlying mechanisms remain largely unclear. In this work, a rat model of prenatal inflammation was used to examine the mechanism by which artemisinin (ART) protects against white matter dysplasia. METHODS We established a primary OPC model and rat model of perinatal inflammation. ART was identified from the FDA-approved medicinal chemical library to be beneficial for treating OPC inflammation in model systems. Based on bioinformatics analysis of protein interactions and molecular docking analysis, we further identified the possible targets of ART and evaluated its specific effects and the underlying molecular mechanisms in vivo and in vitro. RESULTS Following inflammatory stimulation, ART strongly promoted the maturation of OPCs and the development of white matter in the brain. A Cellular thermal shift assay (CETSA) demonstrated that interleukin-1 receptor-associated kinase-4 (IRAK-4) and interleukin-1 receptor-associated kinase-1 (IRAK-1) may be targets of ART, which was consistent with the findings from molecular modelling with Autodock software. Experiments conducted both in vivo and in vitro demonstrated the activation of the IRAK-4/IRAK-1/nuclear factor kappa-B (NF-κB) pathway and the production of inflammatory factors (IL-1β, IL-6, and TNF-α) in OPCs were greatly suppressed in the group treated with ART compared to the lipopolysaccharide (LPS)-treated group. Moreover, ART dramatically decreased reactive oxygen species (ROS) levels in OPCs while increasing nuclear factor e2-related factor 2 (Nrf2) levels. CONCLUSION Our findings suggest that ART can significantly reduce OPC perinatal inflammation and consequent oxidative stress. The targeted inhibition of IRAK-4 and IRAK-1 by ART may be a potential therapeutic strategy for alleviating abnormalities in white matter development in premature newborns.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China; Animal Experiment Center, Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Liqun Lu
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan Province 610500, China
| | - Shiyi Ma
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China; Animal Experiment Center, Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Junfang Sun
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China; Animal Experiment Center, Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Jingyi Liu
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan Province 610500, China
| | - Na Gao
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan Province 610500, China
| | - Zhixian Gou
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan Province 610500, China
| | - Yue Zhou
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan Province 610500, China
| | - Chunchi Lai
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China; Animal Experiment Center, Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Yishi Li
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China; Animal Experiment Center, Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Mengya Sun
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China; Animal Experiment Center, Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Hong Jiang
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China; Animal Experiment Center, Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China.
| |
Collapse
|
20
|
Wei F, Yang W, Wang H, Song S, Ji Y, Chen Z, Zhuang Y, Dai J, Shen H. Reactive oxygen species-scavenging biomaterials for neural regenerative medicine. Biomater Sci 2024. [PMID: 39620279 DOI: 10.1039/d4bm01221f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Reactive oxygen species (ROS) are natural by-products of oxygen metabolism. As signaling molecules, ROS can regulate various physiological processes in the body. However excessive ROS may be a major cause of inflammatory diseases. In the field of neurological diseases, ROS cause neuronal apoptosis and neurodegeneration, which severely impede neuroregeneration. Currently, ROS-scavenging biomaterials are considered as a promising therapeutic strategy for neurological injuries due to their ability to scavenge excessive ROS at defects and modulate the oxidative stress microenvironment. This review provides an overview of the generation and sources of ROS, briefly describes the dangers of generating excessive ROS in nervous system diseases, and highlights the importance of scavenging excessive ROS for neuroregeneration. We have classified ROS-scavenging biomaterials into three categories based on the different mechanisms of ROS clearance. The applications of ROS-responsive biomaterials for neurological diseases, such as spinal cord injury, brain injury, and peripheral nerve injury, are also discussed. Our review contributes to the development of ROS-scavenging biomaterials in the field of neural regeneration.
Collapse
Affiliation(s)
- Feng Wei
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou 215123, China.
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Wen Yang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou 215123, China.
| | - Huiru Wang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou 215123, China.
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Saijie Song
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yuxuan Ji
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou 215123, China.
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Zhong Chen
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yan Zhuang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou 215123, China.
| | - Jianwu Dai
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou 215123, China.
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology Chinese Academy of Sciences, Beijing 100101, China
| | - He Shen
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou 215123, China.
| |
Collapse
|
21
|
Ma YX, Han YQ, Wang PZ, Wang MY, Yang GY, Li JL, Wang J, Chu BB. Porcine reproductive and respiratory syndrome virus activates lipid synthesis through a ROS-dependent AKT/PCK1/INSIG/SREBPs axis. Int J Biol Macromol 2024; 282:136720. [PMID: 39433189 DOI: 10.1016/j.ijbiomac.2024.136720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV) is a highly contagious pathogen in pigs. This study aimed to investigate the impact of PRRSV infection on cellular metabolism, particularly focusing on lipid metabolism to understand its role in promoting viral replication. We conducted a metabolic analysis on MARC-145 cells before and after PRRSV infection. Our results demonstrated that the most significant alterations in cellular metabolism, accounting for 40.8 % of total changes, were related to lipid metabolism. These changes were primarily driven by the activation of sterol regulatory-element binding proteins (SREBPs), critical regulators of lipid biosynthesis. To understand the mechanisms behind SREBPs activation by PRRSV, we investigated the involvement of upstream effectors, specifically protein kinase B (AKT) and phosphoenolpyruvate carboxykinase 1 (PCK1). Our findings indicated that PRRSV infection triggered AKT activation, leading to the subsequent activation of PCK1. Activated PCK1 then phosphorylated insulin-induced genes (INSIGs), resulting in their degradation. This degradation facilitated the translocation of SREBPs from the endoplasmic reticulum to the nucleus. Additionally, we observed that PRRSV infection stimulated the production of reactive oxygen species (ROS), which played a critical role in activating AKT. Collectively, our findings demonstrate that PRRSV enhances lipid synthesis through a ROS-dependent AKT/PCK1/INSIG/SREBPs signaling axis, which provides new insights into the metabolic strategies employed by PRRSV.
Collapse
Affiliation(s)
- Ying-Xian Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Ya-Qi Han
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Pei-Zhu Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Ming-Yang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Jian-Li Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China.
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, Henan Province, China.
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Longhu Advanced Immunization Laboratory, Zhengzhou 450046, Henan Province, China; International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, Henan Province, China.
| |
Collapse
|
22
|
Wang W, Tai S, Tao J, Yang L, Cheng X, Zhou J. Innovative hydrogel-based therapies for ischemia-reperfusion injury: bridging the gap between pathophysiology and treatment. Mater Today Bio 2024; 29:101295. [PMID: 39493810 PMCID: PMC11528235 DOI: 10.1016/j.mtbio.2024.101295] [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: 07/01/2024] [Revised: 09/21/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024] Open
Abstract
Ischemia-reperfusion injury (IRI) commonly occurs in clinical settings, particularly in medical practices such as organ transplantation, cardiopulmonary resuscitation, and recovery from acute trauma, posing substantial challenges in clinical therapies. Current systemic therapies for IRI are limited by poor drug targeting, short efficacy, and significant side effects. Owing to their exceptional biocompatibility, biodegradability, excellent mechanical properties, targeting capabilities, controlled release potential, and properties mimicking the extracellular matrix (ECM), hydrogels not only serve as superior platforms for therapeutic substance delivery and retention, but also facilitate bioenvironment cultivation and cell recruitment, demonstrating significant potential in IRI treatment. This review explores the pathological processes of IRI and discusses the roles and therapeutic outcomes of various hydrogel systems. By categorizing hydrogel systems into depots delivering therapeutic agents, scaffolds encapsulating mesenchymal stem cells (MSCs), and ECM-mimicking hydrogels, this article emphasizes the selection of polymers and therapeutic substances, and details special crosslinking mechanisms and physicochemical properties, as well as summarizes the application of hydrogel systems for IRI treatment. Furthermore, it evaluates the limitations of current hydrogel treatments and suggests directions for future clinical applications.
Collapse
Affiliation(s)
- Weibo Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Supeng Tai
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Junyue Tao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Lexing Yang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Xi Cheng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
23
|
Buranrat B, Laoprom N, Saenmanot S, Chaisri P. Antioxidant, Apoptotic, and Wound Healing Effects of Xantolis Cambodiana Extracts in Normal Human Dermal Fibroblasts. Chem Biodivers 2024; 21:e202301594. [PMID: 39235530 DOI: 10.1002/cbdv.202301594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 08/24/2024] [Accepted: 09/02/2024] [Indexed: 09/06/2024]
Abstract
Xantolis cambodiana has demonstrated significant antioxidant properties; however, the mechanisms underlying its protective effects against oxidative stress in cellular systems remain unexplored. This work investigated the efficacy of methanolic extracts in exhibiting oxidative damage and examined their mechanisms. The methanolic extract had a high phenolic content (116.89±29.01 mg GAE/g FW) and exhibited scavenging of 2,2-diphenyl-1-picrylhydrazyl radicals with an IC50 value of 42.35±9.20 μg/ml. In addition, it had the highest antioxidant activity based on ferric-reducing antioxidant power (467.45±50.74 mg AA/100 g). Normal human dermal fibroblast (NHDF) cells were pretreated with the methanolic extract in a hydrogen peroxide (H2O2; 500 mM)-induced oxidative stress model, which resulted in a significant decrease in apoptosis and autophagy. Not only did the methanolic extract reduce mitochondrial membrane potential, it also stimulated NHDF cell migration and reduced reactive oxygen species production through mitochondrial dysfunction in the H2O2-induced stress model. These findings suggested that the methanolic extract (25 μg/ml) attenuated H2O2-induced oxidative stress in NHDF cells, significantly reducing apoptosis, autophagy, and mitochondrial dysfunction. Thus, this extract has the potential to support the wound healing process due to its antioxidant activity.
Collapse
Affiliation(s)
- Benjaporn Buranrat
- Faculty of Medicine, Mahasarakham University, Talad, Muang, Maha Sarakham, 44000, Thailand
| | - Nonglak Laoprom
- Co-first author, Department of General Science, Faculty of Science and Engineering, Kasetsart University, Chalermphrakiat Sakon Nakhon Province Campus, Sakon Nakhon, 47000, Thailand
| | - Soraya Saenmanot
- Department of Public Health, Faculty of Science, Udon Thani Rajabhat University, Udon Thani, 41000, Thailand
| | - Patcharaporn Chaisri
- Department of Public Health, Faculty of Science, Udon Thani Rajabhat University, Udon Thani, 41000, Thailand
| |
Collapse
|
24
|
Ijaz MU, Imtiaz S, Hayat MF, Batool M, Al-Ghanim KA, Riaz MN. Sudachitin Alleviates Paraquat Instigated Testicular Toxicity in Albino Rats via Regulating Nrf-2/Keap-1, Inflammatory, Steroidogenic, and Histological Profile. ENVIRONMENTAL TOXICOLOGY 2024; 39:5284-5295. [PMID: 39179512 DOI: 10.1002/tox.24408] [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: 05/07/2024] [Revised: 07/14/2024] [Accepted: 08/10/2024] [Indexed: 08/26/2024]
Abstract
Paraquat (PQ) is a noxious herbicide which adversely affects the vital organs including male reproductive system. Sudachitin (SCN) is a naturally occurring flavonoid that demonstrates a wide range of biological potentials. The current study was designed to investigate the alleviative potential of SCN to avert PQ-induced testicular toxicity in rats. Forty-eight male rats (Rattus norvegicus) were apportioned into four groups including control, PQ (5 mg/kg), PQ + SCN (5 mg/kg + 30 mg/kg), and SCN (30 mg/kg) only treated group. Our findings elucidated that PQ treatment reduced the expression of nuclear factor erythroid 2-related factor 2 (Nrf-2) and its antioxidant genes as well as the activities of superoxide dismutase (SOD), catalase (CAT), glutathione reductase (GSR) and glutathione peroxidase (GPx), while elevating the levels of reactive oxygen species (ROS), and malondialdehyde (MDA). Furthermore, PQ intoxication upregulated the expressions of Keap-1 while downregulating the expression of 3-beta hydroxysteroid dehydrogenase (3β-HSD), 17-beta hydroxysteroid dehydrogenase (17β-HSD), and steroidogenic acute regulatory protein (StAR). Moreover, sperm anomalies were increased following the exposure to PQ. Besides, PQ exposure decreased the levels of plasma testosterone, luteinizing hormone (LH), and follicle stimulating hormone (FSH) while increasing the levels of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), nuclear factor-kappa B (NF-κB), interleukin-1beta (IL-1β), and cyclooxygenase-2 (COX-2). Additionally, PQ treatment escalated the expressions of cysteinyl aspartate-specific proteases-3 (Caspase-3) and Bcl-2-associated X-protein (Bax) while downregulating the expressions of B-cell lymphoma-2 (Bcl-2). Furthermore, PQ exposure disrupted the normal architecture of testicular tissues. However, SCN treatment remarkably protected the testicular tissues via regulating the aforementioned disruptions owing to its antioxidant, anti-inflammatory, and androgenic potential.
Collapse
Affiliation(s)
- Muhammad Umar Ijaz
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Sana Imtiaz
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Faisal Hayat
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Moazama Batool
- Department of Zoology, Govt. College Women University, Sialkot, Pakistan
| | - Khalid A Al-Ghanim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | | |
Collapse
|
25
|
Molina SJ, Corsi GN, Araujo Añon LC, Guelman LR. Sex-dependent effects of short-term ethanol, energy drinks and acute noise exposure on hippocampal oxidative balance and glutamate transporter EAAT-1 during rat adolescence. Neurotoxicology 2024; 105:147-157. [PMID: 39366467 DOI: 10.1016/j.neuro.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/17/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
It is known that human adolescents often consume ethanol (EtOH) alone or mixed with energy drinks (ED), especially in noisy environments. Although these agents impact the developing brain, their effects after brief exposure or when presented together remain unclear. Given that few animal studies in this subject are available, this research aimed to study the effects of a brief exposure to these stimuli on the oxidative state and EAAT-1 glutamate transporter levels in the developing rat hippocampus (HC). Adolescent Wistar rats were subjected to a two-bottle choice, limited access to drinking in the dark paradigm, for EtOH and EtOH+ED intake, for 4 days, and subsequent acute noise exposure. Next, hippocampal catalase activity, reactive oxygen species (ROS), glutaredoxin-1 (Grx-1) and glutamate transporter EAAT-1 levels were assessed. Results showed sex-dependent alterations after exposure to these stimuli: Females consuming EtOH had higher hippocampal ROS levels, which decreased when combined with noise; males showed reduced ROS levels only after noise exposure. No significant changes occurred in catalase activity, Grx-1, or EAAT-1 levels with EtOH and noise exposure in neither sex. Additionally, ED raised EtOH consumption in both sexes, normalizing ROS levels only in females when combined with EtOH. Finally, ED consumption altered Grx-1 and EAAT-1 levels in both sexes. In summary, brief exposure to these stimuli induced sex-dependent alterations, suggesting differentiated coping strategies between sexes. Whereas ED consumption may have antioxidant effects in some cases, it could also increase excitotoxicity risk. These novel findings raise questions for future research on the underlying corresponding mechanisms.
Collapse
Affiliation(s)
- Sonia Jazmín Molina
- Universidad de Buenos Aires. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBA-CONICET). Facultad de Medicina, Buenos Aires, Argentina.
| | - Gonzalo Nahuel Corsi
- Universidad de Buenos Aires. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBA-CONICET). Facultad de Medicina, Buenos Aires, Argentina
| | - Lara Candela Araujo Añon
- Universidad de Buenos Aires. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBA-CONICET). Facultad de Medicina, Buenos Aires, Argentina
| | - Laura Ruth Guelman
- Universidad de Buenos Aires. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro de Estudios Farmacológicos y Botánicos (CEFyBO, UBA-CONICET). Facultad de Medicina, Buenos Aires, Argentina; Universidad de Buenos Aires. Facultad de Medicina, 1ª Cátedra de Farmacología, Buenos Aires, Argentina
| |
Collapse
|
26
|
Abdi M, Fadaee M, Jourabchi A, Karimzadeh H, Kazemi T. Cyclophosphamide-Induced Infertility and the Impact of Antioxidants. Am J Reprod Immunol 2024; 92:e70014. [PMID: 39625043 DOI: 10.1111/aji.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
An important drawback of anticancer chemotherapy is the harm it causes to healthy cells. Cyclophosphamide (CP) is a widely used chemotherapeutic alkylating agent that is regularly used in cancer treatment. However, it can cause severe side effects, including genotoxicity, due to its ability to damage DNA. This toxicity is thought to be associated with oxidative stress induced by an excessive amount of reactive oxygen species (ROS). Therefore, there is a specific focus on the potential effects of anticancer treatments on fertility. Due to the increasing life expectancy of cancer patients, those desiring parenthood may face the negative impacts of therapies. Utilizing substances with antioxidant and cytoprotective characteristics to protect the reproductive system from harmful consequences during chemotherapy would be highly beneficial. This review introduces the physiological and pathological roles of ROS in the reproductive systems of both males and females, then we address the adverse effects of CP administration on infertility and discuss how antioxidants can reverse these effects.
Collapse
Affiliation(s)
- Morteza Abdi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirreza Jourabchi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Karimzadeh
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| |
Collapse
|
27
|
Zhu S, Wang J, Liu Y, Jin D, Luo X, Wan M, Fan Y. Multifaceted roles of NADPH oxidases in the growth and pathogenicity of Beauveria bassiana. Virulence 2024; 15:2413850. [PMID: 39377461 PMCID: PMC11469448 DOI: 10.1080/21505594.2024.2413850] [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: 04/02/2024] [Revised: 05/13/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
Reactive oxygen species (ROS), synthesized by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) complex, are vital molecules in biological cells, influencing various physiological processes such as fungal growth, development, and virulence. Beauveria bassiana, an entomopathogenic fungus, is a promising biopesticide for agricultural, forestry, and urban pest control. This study focuses on the characterization of NADPH oxidases (Noxs) in B. bassiana. Gene expression profiles of Noxs in B. bassiana (BbNoxs) were analysed using RT-qPCR. Knockout strains of single BbNoxA, BbNoxB, BbNoxR, and double BbNoxA and BbNoxB were constructed via homologous recombination, and their phenotypic characteristics were examined. Fungal virulence was evaluated using Galleria mellonella larvae, and infection structures formation and penetration ability were assessed on cicada wings. ROS production and actin assembly during fungal growth and infection were detected using staining and marker methods. Expression analysis revealed significant upregulation of BbNoxs during fungal growth and infection. Compared to the wild-type strain, single knockouts (ΔBbNoxA/B/R) and double knockout (ΔBbNoxAB) of BbNoxs exhibited reduced conidial yields, accelerated conidial germination rates. Deletion of BbNoxB or BbNoxR decreased fungal virulence compared to the WT strain in topical inoculation experiments. Additionally, loss of BbNoxB or BbNoxR impaired infection structures formation, penetration ability, ROS production, and actin aggregation during fungal infection. BbNoxs are crucial for fungal growth, development, and virulence in B. bassiana, playing essential roles in infection structures formation, penetration, ROS production, and actin assembly. Understanding their functions provides insights into B. bassiana's pathogenic mechanisms.
Collapse
Affiliation(s)
- Shengan Zhu
- College of Agronomy and Biotechnology, Southwest University, Chongqing, China
| | - Jing Wang
- College of Agronomy and Biotechnology, Southwest University, Chongqing, China
| | - Yu Liu
- Laboratory Animal Center, Southwest University, Chongqing, China
| | - Dan Jin
- College of Agronomy and Biotechnology, Southwest University, Chongqing, China
| | - Xingyou Luo
- College of Agronomy and Biotechnology, Southwest University, Chongqing, China
| | - Min Wan
- College of Agronomy and Biotechnology, Southwest University, Chongqing, China
| | - Yanhua Fan
- College of Agronomy and Biotechnology, Southwest University, Chongqing, China
| |
Collapse
|
28
|
Lei L, Jiang S, Yao Z. Antibacterial activities of Adina rubella extract enhanced by fermentation and its application in packaging films. Food Chem 2024; 460:140604. [PMID: 39111038 DOI: 10.1016/j.foodchem.2024.140604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 09/06/2024]
Abstract
Food spoilage caused by pathogens pose great threat to food safety and human health. Plastarch-based packaging films with antibacterial activities provide an effective way to control foodborne pathogens. In this study, microbial fermentation dominated by yeast was used for the first time to increase the antibacterial activity of Adina rubella extract (ARE). The best antimicrobial effect of ARE was observed by fermentation for 9 days. The minimum inhibitory concentration of ARE against Listeria monocytogenes was 3.125 mg/mL. ARE destroyed the structure of the cell wall, increased the permeability of the cell membrane, led to the leakage of nucleic acids, and induced the change of ROS level, which caused cell death of Listeria monocytogenes. ARE-based biodegradable films were prepared and their performance in pork packaging application was evaluated. The films showed effective antimicrobial properties and showed great potential for the development of safe and sustainable food packaging films.
Collapse
Affiliation(s)
- Lei Lei
- Department of Environmental Science and Engineering, Beijing Technology and Business University, Beijing 100048, China; State Environmental Protection Key Laboratory of Food Chain Pollution Control, Beijing Technology and Business University, Beijing 100048, China
| | - Shanxue Jiang
- Department of Environmental Science and Engineering, Beijing Technology and Business University, Beijing 100048, China; State Environmental Protection Key Laboratory of Food Chain Pollution Control, Beijing Technology and Business University, Beijing 100048, China; China Food Flavor and Nutrition Health Innovation Center, Beijing Technology and Business University, Beijing 100048, China.
| | - Zhiliang Yao
- Department of Environmental Science and Engineering, Beijing Technology and Business University, Beijing 100048, China; State Environmental Protection Key Laboratory of Food Chain Pollution Control, Beijing Technology and Business University, Beijing 100048, China; China Food Flavor and Nutrition Health Innovation Center, Beijing Technology and Business University, Beijing 100048, China.
| |
Collapse
|
29
|
Mostafa EMA, Atta R, Maher SA, El-Kherbetawy MK, Ameen AM. Quercetin and its potential therapeutic effects on aluminum phosphide-induced cardiotoxicity in rats: Role of NOX4, FOXO1, ERK1/2, and NF-κB. Tissue Cell 2024; 91:102622. [PMID: 39549503 DOI: 10.1016/j.tice.2024.102622] [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: 08/09/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Acute Aluminum phosphide (AlP) poisoning poses a serious global issue, yet the exact mechanisms behind AlP-induced cardiotoxicity are still not well understood. Moreover, there is no specific antidote available for AlP toxicity. Nevertheless, Quercetin (QE) has emerged as a promising therapeutic candidate in various contexts. Accordingly, our study aimed to evaluate the QE potential therapeutic effects against AlP-induced cardiotoxicity and the mechanisms underlying such effects. Rats were assigned into four groups: Group I (control group), Group II (vehicle (corn oil) group), Group III (AlP group) received a single dose of AlP (10 mg/kg body weight) dissolved in corn oil by oral gavage, and Group IV (AlP + QE group) received a single dose of QE (400 mg/kg body weight) dissolved in saline, one hour after AlP administration. AlP-induced cardiotoxicity was evidenced by the increase in cardiac troponin I (cTnI) as well as the hemodynamic, ECG, and histopathological abnormalities. The AlP group denoted a decrease of the antioxidant enzymes; catalase and SOD and an increase of the lipid peroxidation marker; MDA. This was associated with a notable increase in inflammatory cytokines (TNFα, IL-6, and IL1β), in addition to a significant upregulation of the expression of NOX4, FOXO1, ERK1/2, and NF-κB. Moreover, Caspase3, and BAX showed strong immunopositive expression, while Bcl-2 showed mild immunoexpression. On the other hand, treatment with QE showed an improvement in the cardiotoxic effects of AlP, as indicated by significant enhancements in biomarkers, functional assessments, and histopathological findings. These results suggest that QE may be a promising candidate for treating AlP-induced cardiotoxicity, attributed to its antioxidant, anti-inflammatory, and anti-apoptotic properties, particularly emphasizing the roles of NOX4, FOXO1, ERK1/2, and NF-κB.
Collapse
Affiliation(s)
- Enas M A Mostafa
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt.
| | - Rasha Atta
- Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Shymaa Ahmed Maher
- Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; Oncology Diagnostic Unit, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | | | - Angie M Ameen
- Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
30
|
de Camargo RW, Joaquim L, Machado RS, de Souza Ramos S, da Rosa LR, de Novais Junior LR, Mathias K, Maximiano L, Strickert YR, Nord R, Gava ML, Scarpari E, Martins HM, Lins EMF, Chaves JS, da Silva LE, de Oliveira MP, da Silva MR, Fernandes BB, Tiscoski ADB, Piacentini N, Santos FP, Inserra A, Bobinski F, Rezin GT, Yonamine M, Petronilho F, de Bitencourt RM. Ayahuasca Pretreatment Prevents Sepsis-Induced Anxiety-Like Behavior, Neuroinflammation, and Oxidative Stress, and Increases Brain-Derived Neurotrophic Factor. Mol Neurobiol 2024:10.1007/s12035-024-04597-4. [PMID: 39613951 DOI: 10.1007/s12035-024-04597-4] [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: 06/28/2024] [Accepted: 10/26/2024] [Indexed: 12/01/2024]
Abstract
The psychoactive decoction Ayahuasca (AYA) used for therapeutic and religious purposes by indigenous groups and peoples from Amazonian regions produces anti-inflammatory and neuroprotective effects. Thus, it may be useful to attenuate the neuroinflammation and related anxiety- and depressive-like symptoms elicited by inflammatory insults such as sepsis. Rats were pretreated for 3 days with different doses of AYA. Twenty-four hours after, cecal ligation and puncture (CLP) was performed. On days 1-4, post-CLP behavioral tests to assess anxiety-like behavior were performed. After 24-h, neuroinflammation, oxidative stress, myeloperoxidase activity, and mitochondrial metabolism were assessed in the prefrontal cortex (PFC), hippocampus (HP), and cortex. AYA pretreatment increased the time spent in the open arms of the elevated plus maze and prevented the sepsis-induced hyper-grooming and -rearing behavior, suggesting an anxiolytic effect. AYA pretreatment increased the levels of the anti-inflammatory interleukin 4, in the PFC and the cortex, and brain-derived neurotrophic factor in the cortex. Moreover, AYA pretreatment increased myeloperoxidase activity in the PFC and the HP and decreased nitrite/nitrate concentration in the PFC, HP, and cortex of septic rats, suggesting enhanced neutrophil activation and decreased nitric oxide signaling. Furthermore, AYA pretreatment prevented lipid peroxidation in the PFC, HP, and cortex of septic rats as measured by decreased levels of thiobarbituric acid reactive substances. Levels of protein carbonyls and activity of superoxide dismutase, citrate synthase, succinate dehydrogenase, and mitochondrial respiratory chain were not affected. Together, AYA represents a promising approach to prevent sepsis-induced neuroinflammatory and oxidative stress and associated anxiety-like symptoms.
Collapse
Affiliation(s)
- Rick Wilhiam de Camargo
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Larissa Joaquim
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Richard Simon Machado
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Suelen de Souza Ramos
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Lara Rodrigues da Rosa
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Linério Ribeiro de Novais Junior
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Lara Maximiano
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Yasmin Ribeiro Strickert
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Rafael Nord
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Maria Laura Gava
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Eduarda Scarpari
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Helena Mafra Martins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Elisa Mitkus Flores Lins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Jéssica Schaefer Chaves
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Larissa Espindola da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Mariana Pacheco de Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Bruna Barros Fernandes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Anita Dal Bó Tiscoski
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Natália Piacentini
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Fabiana Pereira Santos
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Antonio Inserra
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
- Previous Affiliation: Department of Psychiatry, McGill University, Montreal, Canada
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Mauricio Yonamine
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Fabrícia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Rafael Mariano de Bitencourt
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil.
| |
Collapse
|
31
|
Park JW, Kang M, Kim G, Hyun SY, Shin J, Kim SY, Lee JH, Choi WS, Lee JH, Lee K, Kim SH, Cho WS, Kim HS. The impact of atmospheric ultrafine particulate matter on IgE-mediated type 1 hypersensitivity reaction. JOURNAL OF HAZARDOUS MATERIALS 2024; 484:136705. [PMID: 39637818 DOI: 10.1016/j.jhazmat.2024.136705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/16/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
The effect of atmospheric ultrafine particulate matter (UPM) on respiratory allergic diseases has been investigated for decades; however, the precise molecular mechanisms underlying these effects remain poorly understood. In this study, we used a simulated UPM (sUPM) generated via the spark discharge method to refine black carbon, a core particle that closely mimics real-world UPM, including the size (i.e., size of agglomerates: 165 nm) and organic carbon/elemental carbon ratio (i.e., 2.62). When 25 μg/mouse of dispersed sUPM was instilled into the lungs of mice, it promoted the infiltration and degranulation response of pulmonary mast cells, and exposure to sUPM in an immunoglobulin E (IgE)-mediated passive anaphylaxis model intensified the degranulation response of peripheral mast cells. These effects of sUPM were demonstrated to amplify the downstream signaling mechanism of the high-affinity IgE receptor (FcεRI) mediated by IgE when tested using rat basophil leukemia (RBL)-2H3 and mouse bone marrow-derived mast cells (BMMCs) collected from the bone marrow of BALB/c mice. These results indicate that airborne UPM can exacerbate type 1 hypersensitivity reactions by enhancing the IgE-mediated signaling pathways within mast cells. Furthermore, this study provided mechanistic evidence on exacerbated allergic pulmonary diseases induced by UPM inhalation.
Collapse
Affiliation(s)
- Jeong Won Park
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Minseong Kang
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Gyuri Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Seung Yeun Hyun
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Juhyun Shin
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Seon Young Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Jun Ho Lee
- Department of Korean Medicine, College of Korean Medicine, Woosuk University, Jeonju 54986, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Jong-Ho Lee
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea; Department of Biomedical Sciences, College of Natural Science, Dong-A University, Busan 49315, Republic of Korea
| | - Kyuhong Lee
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup, Jeollabuk-do 56212, Republic of Korea
| | - Seok-Ho Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Wan-Seob Cho
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea.
| | - Hyuk Soon Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea; Department of Biomedical Sciences, College of Natural Science, Dong-A University, Busan 49315, Republic of Korea.
| |
Collapse
|
32
|
Xue QQ, Liu CH, Li Y. Decoding the anti-hypertensive mechanism of α-mangostin based on network pharmacology, molecular docking and experimental validation. Mol Med 2024; 30:234. [PMID: 39592923 PMCID: PMC11600633 DOI: 10.1186/s10020-024-01001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Hypertension is a leading risk factor for disability and deaths worldwide. Evidence indicates that alpha-mangostin(α-MG) can reduce blood pressure and improve target organ damage. Nonetheless, its pharmacological targets and potential mechanisms of action remain inadequately elucidated. METHOD We used SwissTargetPrediction to identify α-MG's drug targets and DisGeNET, GeneCards, CTD, and GEO databases for hypertension-related targets, and then determined antihypertensive therapeutic targets of α-MG by intersecting these targets. GO functional enrichment analysis, KEGG pathway analysis, and disease association analysis were conducted using the DAVID database and R package "clusterprofile", visualized with Cytoscape software. The binding affinity of α-MG to identified targets was confirmed through molecular docking using Autodock Vina v.1.2.2 software. The impact of α-MG on target genes was validated using an Angiotensin II-induced hypertensive mouse model and RT-qPCR. RESULTS A total of 51 potential antihypertensive therapeutic targets for α-MG were identified by intersecting 109 drug targets with 821 disease targets. Furthermore, 10 cellular component terms, 10 disease terms, and the top 20 enriched biological processes, molecular functions, and KEGG pathways related to α-MG's antihypertensive effects were documented. Molecular docking studies indicated a strong binding affinity of α-MG with the HSP90AA1 domain. In Ang II-induced hypertensive mice aorta, treatment with α-MG effectively reversed the aberrant mRNA expression of TNF, HSP90AA1, NFKB1, PPARG, SIRT1, PTGS2, and RELA. CONCLUSION Our analyses showed that TNF, HSP90AA1, NFKB1, PPARG, SIRT1, PTGS2, and RELA might be α-MG's potential therapeutic targets for hypertension, laying groundwork for further investigation into its pharmacological mechanisms and clinical uses.
Collapse
Affiliation(s)
- Qi-Qi Xue
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Ruijin 2nd Rd 197, Shanghai, 200025, China
| | - Chu-Hao Liu
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Ruijin 2nd Rd 197, Shanghai, 200025, China
| | - Yan Li
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Ruijin 2nd Rd 197, Shanghai, 200025, China.
| |
Collapse
|
33
|
Larrauri-Rodríguez KA, Leon-Chavez BA, Vallejo-Ruiz V, Peña LMP, Maycotte P. Interplay between reactive oxygen species and ERK activation in cervical cancer cells. Front Cell Dev Biol 2024; 12:1465729. [PMID: 39629272 PMCID: PMC11611811 DOI: 10.3389/fcell.2024.1465729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Among the types of cancer affecting women, cervical cancer (CC) is a public health problem with high global incidence and mortality rates. It is currently classified into three main histological types: squamous cell carcinoma (SCC), adenocarcinoma (AC), and adenosquamous (ASC) carcinoma. All of them lack a targeted therapy. The primary risk factor for CC is Human Papilloma Virus (HPV) infection, which is known to increase reactive oxygen species (ROS), contributing to malignant transformation and tumor progression. At basal levels, ROS can function as second messengers in signaling pathways, and elevated concentrations have been linked to their overactivation. One of these, the ERK pathway, is implicated in both cell proliferation and differentiation and is often dysregulated in cancer, promoting malignant transformation. Several studies have proposed antioxidant supplementation or ERK inhibitors as potential therapies. Methods In vitro studies were performed using CC cell lines. ROS levels were evaluated by flow cytometry; cellular proliferation, death and migration were evaluated using real-time microscopy; cell viability was evaluated with crystal violet staining, and phosphorylated ERK levels were evaluated by Western Blot. A bioinformatic analysis was done in a cervical cancer database. Results We elucidate part of the complex interplay between ROS and ERK pathway in CC pro-tumorigenic characteristics. Through bioinformatic analysis, we found distinct ROS and ERK activation patterns across CC tumor samples from different histological types. However, in vitro, ROS regulated migration and viability in CC, with no discernible variance based on histological classification. ERK activation, however, differed according to the histological type with SCC displaying increased ERK activation compared to AC and regulating cellular migration in SCC cells. Discussion Our study identifies a potential synergistic interaction between ROS and ERK inhibitors, highlighting the therapeutic promise of combinatorial targeting for CC treatment. These findings underscore the importance of personalized approaches aimed at improving the outcomes of CC patients.
Collapse
Affiliation(s)
- Karen Andrea Larrauri-Rodríguez
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), OOAD Puebla, Puebla, Mexico
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla (BUAP), Ciudad Universitaria, Puebla, Mexico
| | - Bertha Alicia Leon-Chavez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla (BUAP), Ciudad Universitaria, Puebla, Mexico
| | - Verónica Vallejo-Ruiz
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), OOAD Puebla, Puebla, Mexico
| | - Lourdes Millán-Perez Peña
- Centro de Química, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Ciudad Universitaria, Puebla, Mexico
| | - Paola Maycotte
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), OOAD Puebla, Puebla, Mexico
| |
Collapse
|
34
|
Shao L, Wang C, Xu G, Tu Z, Yu X, Weng C, Liu J, Jian Z. Utilizing reactive oxygen species-scavenging nanoparticles for targeting oxidative stress in the treatment of ischemic stroke: A review. Open Med (Wars) 2024; 19:20241041. [PMID: 39588390 PMCID: PMC11587925 DOI: 10.1515/med-2024-1041] [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: 05/23/2024] [Revised: 08/06/2024] [Accepted: 08/23/2024] [Indexed: 11/27/2024] Open
Abstract
Ischemic stroke, which accounts for the majority of stroke cases, triggers a complex series of pathophysiological events, prominently characterized by acute oxidative stress due to excessive production of reactive oxygen species (ROS). Oxidative stress plays a crucial role in driving cell death and inflammation in ischemic stroke, making it a significant target for therapeutic intervention. Nanomedicine presents an innovative approach to directly mitigate oxidative damage. This review consolidates existing knowledge on the role of oxidative stress in ischemic stroke and assesses the potential of various ROS-scavenging nanoparticles (NPs) as therapeutic agents. We explore the properties and mechanisms of metal, metal-oxide, and carbon-based NPs, emphasizing their catalytic activity and biocompatibility in scavenging free radicals and facilitating the delivery of therapeutic agents across the blood-brain barrier. Additionally, we address the challenges such as cytotoxicity, immunogenicity, and biodistribution that need to be overcome to translate these nanotechnologies from bench to bedside. The future of NP-based therapies for ischemic stroke holds promise, with the potential to enhance outcomes through targeted modulation of oxidative stress.
Collapse
Affiliation(s)
- Lingmin Shao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Can Wang
- Department of Neurosurgery, Ezhou Central Hospital, Ezhou, 436000, Hubei, China
| | - Gang Xu
- Department of Neurosurgery, Xiantao First People’s Hospital, Xiantao, 433000, Hubei, China
| | - Zewei Tu
- Department of Neurosurgery, Yale School of Medicine, New Haven, 06510, CT, United States of America
| | - Xinyuan Yu
- Department of Anesthesiology, Duke University Medical Center, Durham, 27710, NC, United States of America
| | - Chao Weng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jia Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| |
Collapse
|
35
|
Zivelonghi G, Melotti L, Carolo A, Venerando A, Roncoroni M, Martinelli G, Maccatrozzo L, Marzorati S, Sugni M, Patruno M. Sea food by-products valorization for biomedical applications: evaluation of their wound regeneration capabilities in an Ex vivo skin model. Front Vet Sci 2024; 11:1491385. [PMID: 39660177 PMCID: PMC11629400 DOI: 10.3389/fvets.2024.1491385] [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: 09/04/2024] [Accepted: 10/22/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction The skin is often exposed to harmful stimuli that might compromise its integrity and functionality. After an injury, the skin has a limited capability to restore its complex structure, and in the case of severe skin damage, surgical operations and rapid application of wound dressings are often required to promote optimal wound healing. Nowadays, collagen-based biomaterials are widely used in combination with bioactive molecules able to prevent excessive inflammation and possible infections. In line with a circular economy and blue biotechnology approach, it was recently demonstrated that both collagen and bioactive molecules (i.e., antioxidant compounds) can be sustainably obtained from sea food by-products and effectively used for biomaterial development. Herein, we describe and compare the application of two marine collagen-based wound dressings (CBWDs), produced with materials obtained from sea urchin food waste, for the treatment of skin lesions in a wound healing organ culture (WHOC) model. Methods The ex vivo WHOC model was set up starting from rat skin explants and the induced lesions were assigned into three different groups: control (CTRL) group, not treated, marine collagen wound dressing (MCWD) group, and antioxidants-enriched marine collagen wound dressing (A-MCWD) group. After 5 and 10 days, specimens were examined for organ maintenance and assessed for the healing process. Results Immunohistochemical results showed that both CBWDs were similarly successful in prolonging skin repair, preserving the epidermal barrier up to 5 days under static culture conditions. Histological and gene expression analysis highlighted that the A-MCWD might support and accelerate skin wound healing by exerting antioxidant activity and counteracting inflammation. Discussion Overall, these findings underline the potential of sea urchin food waste as a novel resource for the development of functional medical devices for the treatment of skin wounds.
Collapse
Affiliation(s)
- Giulia Zivelonghi
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | - Luca Melotti
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | - Anna Carolo
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | - Andrea Venerando
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - Margherita Roncoroni
- Department of Environmental Science and Policy, University of Milan, Milan, Italy
| | - Giordana Martinelli
- Department of Environmental Science and Policy, University of Milan, Milan, Italy
| | - Lisa Maccatrozzo
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | - Stefania Marzorati
- Department of Environmental Science and Policy, University of Milan, Milan, Italy
| | - Michela Sugni
- Department of Environmental Science and Policy, University of Milan, Milan, Italy
| | - Marco Patruno
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| |
Collapse
|
36
|
Ren Y, Anderson AT, Meyer G, Lauber KM, Gallucci JC, Douglas Kinghorn A. Digoxin and its Na +/K +-ATPase-targeted actions on cardiovascular diseases and cancer. Bioorg Med Chem 2024; 114:117939. [PMID: 39396465 PMCID: PMC11527570 DOI: 10.1016/j.bmc.2024.117939] [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: 08/16/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Na+/K+-ATPase (NKA) is a plasma membrane ion-transporting protein involved in the generation and maintenance of Na+ and K+ gradients across the cell membrane, which can produce a driving force for the secondary transport of metabolic substrates. NKA also regulates intracellular calcium that is responsible for modulating numerous cellular processes, while it interacts with many other proteins and functions as a signal transducer, with several signaling pathways being involved. Thus, NKA has become an important target for the treatment of human diseases. Cardiac glycosides are well-known NKA inhibitors, of which (+)-digoxin or digoxin has been long used for the treatment of congestive heart failure. Also, digoxin has exhibited potential antitumor activity, by targeting directly HIF-1α, NKA, and NF-κB. Thus, the function of NKA in human cardiovascular diseases and cancer and the therapeutic effects of digoxin on these diseases are summarized in the present review, with the correlations among digoxin, NKA, cardiovascular diseases, and cancer being discussed. Presented herein are also the antitumor potential of monosaccharide cardiac glycoside analogues of digoxin, including (-)-cryptanoside A, (-)-oleandrin, (-)-ouabain, and (+)-strebloside. It is hoped that this contribution will provide some helpful information for the design and discovery of new cardiac glycoside-type therapeutic agents for the treatment of cardiovascular diseases and cancer.
Collapse
Affiliation(s)
- Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| | - Andrew T Anderson
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Gunnar Meyer
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Kaitlyn M Lauber
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Judith C Gallucci
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
37
|
Varshavskaya KB, Barykin EP, Timoshenko RV, Kolmogorov VS, Erofeev AS, Gorelkin PV, Mitkevich VA, Makarov AA. Post-translational modifications of beta-amyloid modulate its effect on cell mechanical properties and influence cytoskeletal signaling cascades. Front Mol Neurosci 2024; 17:1501874. [PMID: 39610710 PMCID: PMC11602469 DOI: 10.3389/fnmol.2024.1501874] [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: 09/25/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024] Open
Abstract
Post-translational modifications of beta-amyloid (Aβ) play an important role in the pathogenesis of Alzheimer's disease (AD). Aβ modifications such as Ser8 phosphorylation (pS8-Aβ42) and Asp7 isomerization (iso-Aβ42) can significantly alter the properties of Aβ and have been detected in vivo. One of the reasons for the different pathogenicity of Aβ isoforms may be the activation of different signaling cascades leading to changes in the mechanical properties of cells. In this paper, we used correlative scanning ion-conductance microscopy (SICM) and Pt-nanoelectrodes to compare the effects of Aβ isoforms on the Young's modulus of SH-SY5Y cells and the level of ROS. It was found that unmodified Aβ42 resulted in the largest increase in cell Young's modulus of all isoforms after 4 h of incubation, while pS8-Aβ42 induced the greatest increase in stiffness and ROS levels after 24 h of incubation. Analysis of signaling proteins involved in the regulation of the actin cytoskeleton showed that Aβ42, pS8-Aβ42 and iso-Aβ42 have different effects on cofilin, GSK3β, LIMK, ERK and p38. This indicates that post-translational modifications of Aβ modulate its effect on neuronal cells through the activation of various signaling cascades, which affects the mechanical properties of cells.
Collapse
Affiliation(s)
| | | | - Roman V. Timoshenko
- Research Laboratory of Biophysics, National University of Science and Technology “MISIS”, Moscow, Russia
| | - Vasilii S. Kolmogorov
- Research Laboratory of Biophysics, National University of Science and Technology “MISIS”, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Alexander S. Erofeev
- Research Laboratory of Biophysics, National University of Science and Technology “MISIS”, Moscow, Russia
| | - Petr V. Gorelkin
- Research Laboratory of Biophysics, National University of Science and Technology “MISIS”, Moscow, Russia
| | | | | |
Collapse
|
38
|
Meng T, He J, Huo Q, Wang Y, Ren Q, Kang Y. Association of Stress Defense System With Fine Particulate Matter Exposure: Mechanism Analysis and Application Prospects. J Appl Toxicol 2024. [PMID: 39538419 DOI: 10.1002/jat.4724] [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: 08/01/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
The association between the stress defense system and exposure to fine particulate matter (PM2.5) is a hot topic in the field of environmental health. PM2.5 pollution is an increasingly serious issue, and its impact on health cannot be ignored. The stress defense system is an important biological mechanism for maintaining cell and internal environment homeostasis, playing a crucial role in PM2.5-induced damage and diseases. The association between PM2.5 exposure and activation of the stress defense system has been reported. Moderate PM2.5 exposure rapidly mobilizes the stress defense system, while excessive PM2.5 exposure may exceed its compensatory and coping abilities, resulting in system imbalance and dysfunction that triggers pathological changes in cells and tissues, thereby increasing the risk of chronic diseases, such as respiratory diseases, cardiovascular diseases, and cancer. This detailed review focuses on the composition, function, and regulatory mechanisms of the antioxidant defense system, autophagy system, ubiquitin-proteasome system, and inflammatory response system, which are all components of the stress defiance system. In particular, the influence of PM2.5 exposure on each of these defense systems and their roles in responding to PM2.5-induced damage was investigated to provide an in-depth understanding of the pathogenesis of PM2.5 exposure, accurately assess potential hazards, and formulate prevention and intervention strategies for health damage caused by PM2.5 exposure.
Collapse
Affiliation(s)
- Tao Meng
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
- Doctoral Innovation Station of Shanxi Province, Key Laboratory of TCM Prevention and Treatment of Dementia Disease, The Fifth People's Hospital of Datong, Datong, China
| | - Jing He
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Qianru Huo
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Yajie Wang
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Qingchun Ren
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Yihui Kang
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| |
Collapse
|
39
|
Thatikonda S, Rasoju SP, Pooladanda V, Chilvery S, Khemchandani R, Samanthula G, Godugu C. Niosomal gel improves dermal delivery of nimbolide: a promising approach for treatment of psoriasis. Nanomedicine (Lond) 2024; 19:2521-2536. [PMID: 39530550 DOI: 10.1080/17435889.2024.2405455] [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: 02/29/2024] [Accepted: 09/13/2024] [Indexed: 11/16/2024] Open
Abstract
Aim: Psoriasis is a chronic inflammatory skin disorder characterized by the excessive proliferation of keratinocytes, forming thickened skin plaques due to immune-mediated cytokine responses. Delivering drugs through this barrier to target inflamed tissues remains challenging. Nimbolide (NIM), known for its anti-inflammatory and anticancer properties, shows promise in managing psoriasis. However, its efficacy is limited by its inability to penetrate the thickened horny layer of the skin. To overcome this obstacle, we have developed Nim-loaded niosomal (Nio) formulations (NIM Nio) aimed at improving dermal delivery and achieving localized sustained release at psoriasis-affected sites.Methods: The formulation characteristics were assessed using Zeta sizer, Transmission Electron Microscopy (TEM), and High-performance liquid chromatography (HPLC). The optimized formulation was evaluated for anti-psoriatic potential compared to Nim alone by using molecular techniques such as Confocal Microscopy, Flow cytometry, enzyme-linked immunosorbent assay (ELISA), and Western blotting.Results: NIM Nio showed effective penetration into psoriatic skin, resulting in reductions in keratinocyte hyperproliferation, oxidative stress, splenomegaly, inflammatory cytokines, Psoriasis Area and Severity Index (PASI), and rete ridges compared to NIM alone.Conclusion: Our findings underscore the significant anti-proliferative, antioxidant, and anti-inflammatory properties of NIM Nio in psoriasis, demonstrating its potential as a promising therapeutic option for this challenging condition.
Collapse
Affiliation(s)
- Sowjanya Thatikonda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
- Department of Head & Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL 33612,USA
| | - Sai Prabha Rasoju
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Venkatesh Pooladanda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
- Vincent Center for Reproductive Biology, Department of Obstetrics & Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
- Obstetrics, Gynecology & Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Shrilekha Chilvery
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Rahul Khemchandani
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Gananadhamu Samanthula
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| |
Collapse
|
40
|
Wang H, He W, Liao J, Wang S, Dai X, Yu M, Xie Y, Chen Y. Catalytic Biomaterials-Activated In Situ Chemical Reactions: Strategic Modulation and Enhanced Disease Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2411967. [PMID: 39498674 DOI: 10.1002/adma.202411967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/19/2024] [Indexed: 11/07/2024]
Abstract
Chemical reactions underpin biological processes, and imbalances in critical biochemical pathways within organisms can lead to the onset of severe diseases. Within this context, the emerging field of "Nanocatalytic Medicine" leverages nanomaterials as catalysts to modulate fundamental chemical reactions specific to the microenvironments of diseases. This approach is designed to facilitate the targeted synthesis and localized accumulation of therapeutic agents, thus enhancing treatment efficacy and precision while simultaneously reducing systemic side effects. The effectiveness of these nanocatalytic strategies critically hinges on a profound understanding of chemical kinetics and the intricate interplay of reactions within particular pathological microenvironments to ensure targeted and effective catalytic actions. This review methodically explores in situ catalytic reactions and their associated biomaterials, emphasizing regulatory strategies that control therapeutic responses. Furthermore, the discussion encapsulates the crucial elements-reactants, catalysts, and reaction conditions/environments-necessary for optimizing the thermodynamics and kinetics of these reactions, while rigorously addressing both the biochemical and biophysical dimensions of the disease microenvironments to enhance therapeutic outcomes. It seeks to clarify the mechanisms underpinning catalytic biomaterials and evaluate their potential to revolutionize treatment strategies across various pathological conditions.
Collapse
Affiliation(s)
- Huijing Wang
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Wenjin He
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Jing Liao
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Shuangshuang Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xinyue Dai
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Yu Chen
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Shanghai Institute of Materdicine, Shanghai, 200051, P. R. China
| |
Collapse
|
41
|
Chen A, Huang H, Fang S, Hang Q. ROS: A "booster" for chronic inflammation and tumor metastasis. Biochim Biophys Acta Rev Cancer 2024; 1879:189175. [PMID: 39218404 DOI: 10.1016/j.bbcan.2024.189175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Reactive oxygen species (ROS) are a group of highly active molecules produced by normal cellular metabolism and play a crucial role in the human body. In recent years, researchers have increasingly discovered that ROS plays a vital role in the progression of chronic inflammation and tumor metastasis. The inflammatory tumor microenvironment established by chronic inflammation can induce ROS production through inflammatory cells. ROS can then directly damage DNA or indirectly activate cellular signaling pathways to promote tumor metastasis and development, including breast cancer, lung cancer, liver cancer, colorectal cancer, and so on. This review aims to elucidate the relationship between ROS, chronic inflammation, and tumor metastasis, explaining how chronic inflammation can induce tumor metastasis and how ROS can contribute to the evolution of chronic inflammation toward tumor metastasis. Interestingly, ROS can have a "double-edged sword" effect, promoting tumor metastasis in some cases and inhibiting it in others. This article also highlights the potential applications of ROS in inhibiting tumor metastasis and enhancing the precision of tumor-targeted therapy. Combining ROS with nanomaterials strategies may be a promising approach to enhance the efficacy of tumor treatment.
Collapse
Affiliation(s)
- Anqi Chen
- Medical College, Yangzhou University, Yangzhou 225009, China
| | - Haifeng Huang
- Department of Laboratory Medicine, The First People's Hospital of Yancheng, Yancheng 224006, China; Department of Laboratory Medicine, Yancheng Clinical Medical College of Jiangsu University, Yancheng 224006, China
| | - Sumeng Fang
- School of Mathematics, Tianjin University, Tianjin 300350, China
| | - Qinglei Hang
- Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining People's Hospital, Affiliated Hospital of Xuzhou Medical University, Suining 221200, China; Key Laboratory of Jiangsu Province University for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou 225009, China; Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
42
|
Hu S, Meng Y, Guo L, Xu X. A novel strategy to enhance inhibition of Hela cervical cancer by combining Lentinus β-glucan and autophagic flux blockage. Int J Biol Macromol 2024; 281:136309. [PMID: 39370081 DOI: 10.1016/j.ijbiomac.2024.136309] [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: 03/03/2024] [Revised: 09/23/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Lentinus β-D-glucan (LNT), derived from artificially cultured mushrooms of Lentinus edodes, shows an important yet incompletely understood biological functions in cancer. In this work, the chemical structure of the refined LNT comprising a β-D-(1, 6)-branched β-D-(1,3)-glucan was further clarified via 1D- and 2D-NMR with high resolution, and its drug resistance resulted from autophagy in human cervical cancer (CC) Hela cells besides its anti-cancer function were revealed in vitro and in vivo. In detail, LNT destroyed cellular homeostasis by significantly increasing the intracellular Ca2+ levels and promoted autophagic flux in vitro Hela cells, which was found to at least partially depend on the PI3K/Akt/mTOR-mediated pathway by up-regulating LC3-II levels and down-regulating the expression of p62, PI3K, p-Akt, and mTOR in Hela cells-transplanted BALB/c nude mice. In particular, LNT-induced autophagy led to a drug resistance against LNT-induced proliferation inhibition and apoptosis in Hela cells, and the co-treatment of autophagy inhibitors and LNT significantly enhanced the inhibition of Hela cells and tumor growth in vitro and in vivo. Therefore, the combination of LNT and autophagy inhibitors will be a novel therapeutic strategy to reduce the resistance and improve the prognosis of CC patients in the clinical.
Collapse
Affiliation(s)
- Shuqian Hu
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University; College of Chemistry and Molecular Sciences; Hubei Engineering Center of Natural Polymer-Based Medical Materials; Wuhan University, Wuhan 430072, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Yan Meng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Liang Guo
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University; College of Chemistry and Molecular Sciences; Hubei Engineering Center of Natural Polymer-Based Medical Materials; Wuhan University, Wuhan 430072, China.
| | - Xiaojuan Xu
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University; College of Chemistry and Molecular Sciences; Hubei Engineering Center of Natural Polymer-Based Medical Materials; Wuhan University, Wuhan 430072, China.
| |
Collapse
|
43
|
Zheng S, Deng R, Huang G, Ou Z, Shen Z. Screening the active ingredients of plants via molecular docking technology and evaluating their ability to reduce skin photoaging. Biogerontology 2024; 25:1115-1143. [PMID: 39312047 DOI: 10.1007/s10522-024-10125-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/29/2024] [Indexed: 10/18/2024]
Abstract
The active ingredients of plants were screened by molecular docking technology and the result were verified. According to the verification results of molecular docking, the five active ingredients were combined in equal proportions to form a compound drug. In the HaCaT photoaging model, the effects of the compound drug on antioxidant and senescence-associated secretory phenotype (SASP) factors of the NF-κB and MAPK pathways were studied via SOD and MDA kits, DCFH-DA fluorescent probes and ELISA. In the skin photoaging model, the effects of the compound drug on antioxidants and the SASP factors of the NF-κB and MAPK pathways were studied via SOD, MDA, and CAT kits and ELISA. The results revealed that the compound drug increased SOD activity, decreased the MDA content and intracellular ROS, inhibited IL-6 in the NF-κB pathway, and inhibited MMP-1 and collagen I in the MAPK pathway. The results of HE, Masson and Victoria blue skin staining revealed that the compound drug inhibited abnormal thickening of the epidermis, abnormal breaking and accumulation of collagen fibers and elastic fibers, and maintained their orderly arrangement. Moreover, the results revealed that the compound drug increased SOD, CAT and collagen I, and reduced the MDA content, the SASP factors IL-6 and TNF-α of the NF-κB pathway, and the SASP factors MMP-1 of the MAPK pathway. The above results indicate that the active ingredients of the compound drug screened by molecular docking have the potential to reduce skin photoaging.
Collapse
Affiliation(s)
- Shiqian Zheng
- Guangdong Botanical Beauty Care Biotechnology Co., Ltd, Guangzhou, China
| | - Rongrong Deng
- Guangdong Botanical Beauty Care Biotechnology Co., Ltd, Guangzhou, China
| | - Gengjiu Huang
- Guangdong Botanical Beauty Care Biotechnology Co., Ltd, Guangzhou, China
| | - Zhiwen Ou
- Guangdong Botanical Beauty Care Biotechnology Co., Ltd, Guangzhou, China
| | - Zhibin Shen
- Guangdong Botanical Beauty Care Biotechnology Co., Ltd, Guangzhou, China.
| |
Collapse
|
44
|
Xue JL, Ji JL, Zhou Y, Zhang Y, Liu BC, Ma RX, Li ZL. The multifaceted effects of mitochondria in kidney diseases. Mitochondrion 2024; 79:101957. [PMID: 39270830 DOI: 10.1016/j.mito.2024.101957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/23/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
Mitochondria serve as the primary site for aerobic respiration within cells, playing a crucial role in maintaining cellular homeostasis. To maintain homeostasis and meet the diverse demands of the cells, mitochondria have evolved intricate systems of quality control, mainly including mitochondrial dynamics, mitochondrial autophagy (mitophagy) and mitochondrial biogenesis. The kidney, characterized by its high energy requirements, is particularly abundant in mitochondria. Interestingly, the mitochondria display complex behaviors and functions. When the kidney is suffered from obstructive, ischemic, hypoxic, oxidative, or metabolic insults, the dysfunctional mitochondrial derived from the defects in the mitochondrial quality control system contribute to cellular inflammation, cellular senescence, and cell death, posing a threat to the kidney. However, in addition to causing injury to the kidney in several cases, mitochondria also exhibit protective effect on the kidney. In recent years, accumulating evidence indicated that mitochondria play a crucial role in adaptive repair following kidney diseases caused by various etiologies. In this article, we comprehensively reviewed the current understanding about the multifaceted effects of mitochondria on kidney diseases and their therapeutic potential.
Collapse
Affiliation(s)
- Jia-Le Xue
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jia-Ling Ji
- Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Zhou
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Yao Zhang
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Rui-Xia Ma
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Zuo-Lin Li
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
45
|
Singh T, Sharma K, Jena L, Kaur P, Singh S, Munshi A. Mitochondrial bioenergetics of breast cancer. Mitochondrion 2024; 79:101951. [PMID: 39218051 DOI: 10.1016/j.mito.2024.101951] [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: 04/14/2024] [Revised: 08/05/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Breast cancer cells exhibit metabolic heterogeneity based on tumour aggressiveness. Glycolysis and mitochondrial respiration are two major metabolic pathways for ATP production. The oxygen flux, oxygen tension, proton leakage, protonmotive force, inner mitochondrial membrane potential, ECAR and electrochemical proton gradient maintain metabolic homeostasis, ATP production, ROS generation, heat dissipation, and carbon flow and are referred to as "sub-domains" of mitochondrial bioenergetics. Tumour aggressiveness is influenced by these mechanisms, especially when breast cancer cells undergo metastasis. These physiological parameters for healthy mitochondria are as crucial as energy demands for tumour growth and metastasis. The instant energy demands are already elucidated under Warburg effects, while these parameters may have dual functionality to maintain cellular bioenergetics and cellular health. The tumour cell might maintain these mitochondrial parameters for mitochondrial health or avoid apoptosis, while energy production could be a second priority. This review focuses explicitly on the crosstalk between metabolic domains and the utilisation of these parameters by breast cancer cells for their progression. Some major interventions are discussed based on mitochondrial bioenergetics that need further investigation. This review highlights the pathophysiological significance of mitochondrial bioenergetics and the regulation of its sub-domains by breast tumour cells for uncontrolled proliferation.
Collapse
Affiliation(s)
- Tashvinder Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Kangan Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Laxmipriya Jena
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Prabhsimran Kaur
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India.
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India.
| |
Collapse
|
46
|
Jiang Z, Wang H, Wang X, Duo H, Tao Y, Li J, Li X, Liu J, Ni J, Wu EJ, Xiang H, Guan C, Wang X, Zhang K, Zhang P, Hou Z, Liu Y, Wang Z, Su B, Li B, Hao Y, Li B, Wu X. TMED4 facilitates regulatory T cell suppressive function via ROS homeostasis in tumor and autoimmune mouse models. J Clin Invest 2024; 135:e179874. [PMID: 39480507 DOI: 10.1172/jci179874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
Endoplasmic reticulum stress (ERS) plays crucial roles in maintaining Treg stability and function, yet the underlying mechanism remains largely unexplored. Here, we demonstrate that (Tmed4ΔTreg) mice with Treg-specific KO of ERS-related protein transmembrane p24 trafficking protein 4 (TMED4) had more Tregs with impaired Foxp3 stability, Treg signatures, and suppressive activity, which led to T cell hyperactivation and an exacerbated inflammatory phenotype and boosted antitumor immunity in mice. Mechanistically, loss of Tmed4 caused defects in ERS and a nuclear factor erythroid 2-related factor 2-related (NRF2-related) antioxidant response, which resulted in excessive ROS that reduced the Foxp3 stability and suppressive function of Tregs in an IRE1α/XBP1 axis-dependent manner. The abnormalities could be effectively rescued by the ROS scavenger, NRF2 inducer, or by forcible expression of IRE1α. Moreover, TMED4 suppressed IRE1α proteosome degradation via the ER-associated degradation (ERAD) system including the ER chaperone binding immunoglobulin protein (BIP). Our study reveals that TMED4 maintained the stability of Tregs and their suppressive function through IRE1α-dependent ROS and the NRF2-related antioxidant response.
Collapse
Affiliation(s)
- Zhenyan Jiang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Huizi Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
- Songjiang Research Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Xiaoxia Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
- Songjiang Research Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Hongrui Duo
- Research Group of Computational and Integrative Biology, College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Yuexiao Tao
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Jia Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Xin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Jiamin Liu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Jun Ni
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Emily Jiatong Wu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Hongrui Xiang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Chenyang Guan
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Xinyu Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Kun Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Key Laboratory of Emotions and Affective Disorders, SJTU-SM, Shanghai, China
| | - Peng Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Key Laboratory of Emotions and Affective Disorders, SJTU-SM, Shanghai, China
| | - Zhaoyuan Hou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, SJTU-SM, Shanghai, China
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhengting Wang
- Department of Gastroenterology, Ruijin Hospital, SJTU-SM, Shanghai, China
| | - Bing Su
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Bo Li
- Research Group of Computational and Integrative Biology, College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Youjin Hao
- Research Group of Computational and Integrative Biology, College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Xuefeng Wu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| |
Collapse
|
47
|
Comyn T, Preat T, Pavlowsky A, Plaçais PY. PKCδ is an activator of neuronal mitochondrial metabolism that mediates the spacing effect on memory consolidation. eLife 2024; 13:RP92085. [PMID: 39475218 PMCID: PMC11524582 DOI: 10.7554/elife.92085] [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] [Indexed: 11/02/2024] Open
Abstract
Relevance-based selectivity and high energy cost are two distinct features of long-term memory (LTM) formation that warrant its default inhibition. Spaced repetition of learning is a highly conserved cognitive mechanism that can lift this inhibition. Here, we questioned how the spacing effect integrates experience selection and energy efficiency at the cellular and molecular levels. We showed in Drosophila that spaced training triggers LTM formation by extending over several hours an increased mitochondrial metabolic activity in neurons of the associative memory center, the mushroom bodies (MBs). We found that this effect is mediated by PKCδ, a member of the so-called 'novel PKC' family of enzymes, which uncovers the critical function of PKCδ in neurons as a regulator of mitochondrial metabolism for LTM. Additionally, PKCδ activation and translocation to mitochondria result from LTM-specific dopamine signaling on MB neurons. By bridging experience-dependent neuronal circuit activity with metabolic modulation of memory-encoding neurons, PKCδ signaling binds the cognitive and metabolic constraints underlying LTM formation into a unified gating mechanism.
Collapse
Affiliation(s)
- Typhaine Comyn
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Thomas Preat
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Alice Pavlowsky
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Pierre-Yves Plaçais
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| |
Collapse
|
48
|
Gonçalves B, Aires A, Oliveira I, Baltazar M, Cosme F, Afonso S, Pinto T, Anjos MR, Inês A, Morais MC, Vilela A, Silva AP. From Orchard to Wellness: Unveiling the Health Effects of Sweet Cherry Nutrients. Nutrients 2024; 16:3660. [PMID: 39519493 PMCID: PMC11547742 DOI: 10.3390/nu16213660] [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: 09/27/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
This review paper explores the multifaceted relationship between sweet cherry nutrients and human health, aiming to uncover the comprehensive impact of these bioactive compounds from orchard to wellness. Furthermore, it highlights how advanced crop techniques can be pivotal in optimizing these beneficial compounds. Synthesizing existing literature, the paper examines the diverse bioactive nutrients in sweet cherries, including antioxidants, polyphenols, vitamins, and minerals, and elucidating their mechanisms of action and potential health benefits. From antioxidant properties to anti-inflammatory effects, the paper elucidates how these nutrients may mitigate chronic diseases such as cardiovascular disorders, diabetes, and neurodegenerative conditions. Additionally, it explores their role in promoting gastrointestinal health, enhancing exercise recovery, and modulating sleep patterns. The review discusses emerging research on the potential anti-cancer properties of sweet cherry compounds, highlighting their promising role in cancer prevention and treatment. Furthermore, it delves into the impact of sweet cherry consumption on metabolic health, weight management, and skin health. By providing a comprehensive overview of the current understanding of sweet cherry nutrients and their health effects, this paper offers valuable insights for researchers, healthcare professionals, and consumers interested in utilizing nature's bounty for holistic wellness.
Collapse
Affiliation(s)
- Berta Gonçalves
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Alfredo Aires
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Ivo Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Miguel Baltazar
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Fernanda Cosme
- Chemistry Research Centre-Vila Real (CQ-VR), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (F.C.); (A.I.); (A.V.)
| | - Sílvia Afonso
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Teresa Pinto
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Maria Rosário Anjos
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - António Inês
- Chemistry Research Centre-Vila Real (CQ-VR), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (F.C.); (A.I.); (A.V.)
| | - Maria Cristina Morais
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Alice Vilela
- Chemistry Research Centre-Vila Real (CQ-VR), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (F.C.); (A.I.); (A.V.)
| | - Ana Paula Silva
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| |
Collapse
|
49
|
Wang S, Nie Z, Zhu L, Wu Y, Wen Y, Deng F, Zhao L. Probiotic Characteristics and the Anti-Inflammatory Effects of Lactiplantibacillus plantarum Z22 Isolated from Naturally Fermented Vegetables. Microorganisms 2024; 12:2159. [PMID: 39597548 PMCID: PMC11596721 DOI: 10.3390/microorganisms12112159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Currently, there is increasing interest in the commercial utilization of probiotics isolated from traditional fermented food products. Therefore, this study aimed to investigate the probiotic potential of Lactiplantibacillus plantarum (L. plantarum) Z22 isolated from naturally fermented mustard. The results suggest that L. plantarum Z22 exhibits good adhesion ability, antibacterial activity, safety, and tolerance to acidic conditions and bile salts. We further determined the anti-inflammatory mechanism and properties of L. plantarum Z22 and found that L. plantarum Z22 could significantly reduce the secretion of pro-inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and the expression of the pro-inflammatory mediator cyclooxygenase-2 (COX-2) protein in LPS-induced RAW 264.7 cells. In addition, L. plantarum Z22 also effectively inhibited the signaling pathways of nuclear factor κB (NF-κB) and mitogen-activated protein kinases (MAPKs). This effect can be attributed to a decrease in the levels of reactive oxygen species (ROS) and increased heme oxygenase-1 (HO-1) expression. Moreover, whole-genome sequencing revealed that L. plantarum Z22 contains gene-encoding proteins with anti-inflammatory functions, such as beta-glucosidase (BGL) and pyruvate kinase (PK), as well as antioxidant functions, including thioredoxin reductase (TrxR), tyrosine-protein phosphatase, and ATP-dependent intracellular proteases ClpP. In summary, these results indicated that L. plantarum Z22 can serve as a potential candidate probiotic for use in fermented foods such as yogurt (starter cultures), providing a promising strategy for the development of functional foods to prevent chronic diseases.
Collapse
Affiliation(s)
- Shiyu Wang
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| | - Ziyu Nie
- College of Animal Science and Technology, Hunan Biological Electromechanical Vocational College, Changsha 410128, China;
| | - Li Zhu
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| | - Yanyang Wu
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| | - Yashi Wen
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| | - Fangming Deng
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| | - Lingyan Zhao
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| |
Collapse
|
50
|
Lu Q, Ding Y, Liu W, Liu S. Viral Infections and the Glutathione Peroxidase Family: Mechanisms of Disease Development. Antioxid Redox Signal 2024. [PMID: 39446976 DOI: 10.1089/ars.2024.0645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Significance: The glutathione peroxidase (GPx) family is recognized for its essential function in maintaining cellular redox balance and countering the overproduction of reactive oxygen species (ROS), a process intricately linked to the progression of various diseases including those spurred by viral infections. The modulation of GPx activity by viruses presents a critical juncture in disease pathogenesis, influencing cellular responses and the trajectory of infection-induced diseases. Recent Advances: Cutting-edge research has unveiled the GPx family's dynamic role in modulating viral pathogenesis. Notably, GPX4's pivotal function in regulating ferroptosis presents a novel avenue for the antiviral therapy. The discovery that selenium, an essential micronutrient for GPx activity, possesses antiviral properties has propelled us toward rethinking traditional treatment modalities. Critical Issues: Deciphering the intricate relationship between viral infections and GPx family members is paramount. Viral invasion can precipitate significant alterations in GPx function, influencing disease outcomes. The multifaceted nature of GPx activity during viral infections suggests that a deeper understanding of these interactions could yield novel insights into disease mechanisms, diagnostics, prognostics, and even chemotherapeutic resistance. Future Directions: This review aims to synthesize current knowledge on the impact of viral infections on GPx activity and expression and identify key advances. By elucidating the mechanisms through which GPx family members intersect with viral pathogenesis, we propose to uncover innovative therapeutic strategies that leverage the antioxidant properties of GPx to combat viral infections. The exploration of GPx as a therapeutic target and biomarker holds promise for the development of next-generation antiviral therapies. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Qingqing Lu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Pathogenic Biology, Qingdao University Medical College, Qingdao, China
| | - Yuan Ding
- Department of Special Examination, Qingdao Women and Children's Hospital, Qingdao University, Qingdao, China
| | - Wen Liu
- Department of Pathogenic Biology, Qingdao University Medical College, Qingdao, China
| | - Shuzhen Liu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|