1
|
Parajuli N, Subedi K, Solone XK, Jiang A, Zhou L, Mi QS. Epigenetic Control of Alveolar Macrophages: Impact on Lung Health and Disease. Cells 2025; 14:640. [PMID: 40358164 PMCID: PMC12071345 DOI: 10.3390/cells14090640] [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: 03/22/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Alveolar macrophages (AMs) are immune cells located in the alveoli-the tiny air sacs in the lungs where gas exchange occurs. Their functions are regulated by various epigenetic mechanisms, which are essential for both healthy lung function and disease development. In the lung's microenvironment, AMs play critical roles in immune surveillance, pathogen clearance, and tissue repair. This review examines how epigenetic regulation influences AM functions and their involvement in lung diseases. Key mechanisms, such as DNA methylation, histone modifications, and non-coding RNAs, regulate gene expression in response to environmental signals. In healthy lungs, these modifications enable AMs to quickly respond to inhaled threats. However, when these processes malfunction, they could contribute to diseases such as pulmonary fibrosis, COPD, and pulmonary hypertension. By exploring how epigenetic changes affect AM polarization, plasticity, and immune responses, we can gain deeper insights into their role in lung diseases and open new avenues for treating and preventing respiratory conditions. Ultimately, understanding the epigenetic mechanisms within AMs enhances our knowledge of lung immunology and offers potential for innovative interventions to restore lung health and prevent respiratory diseases.
Collapse
Affiliation(s)
- Nirmal Parajuli
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Kalpana Subedi
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Xzaviar Kaymar Solone
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Aimin Jiang
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Li Zhou
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Department of Medicine, Henry Ford Health, Detroit, MI 48202, USA
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Department of Medicine, Henry Ford Health, Detroit, MI 48202, USA
| |
Collapse
|
2
|
Cheng X, Liang Y, Ji K, Feng M, Du X, Jiao D, Wu X, Zhong C, Cong H, Yang G. Enhanced propionate and butyrate metabolism in cecal microbiota contributes to cold-stress adaptation in sheep. MICROBIOME 2025; 13:103. [PMID: 40275300 PMCID: PMC12023611 DOI: 10.1186/s40168-025-02096-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/17/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND During cold stress, gut microbes play crucial roles in orchestrating energy metabolism to enhance environmental adaptation. In sheep, hindgut microbes ferment carbohydrates to generate short-chain fatty acids (SCFAs) as an energy source. However, the mechanisms by which hindgut microbes and their metabolites interact with the host to facilitate adaptation to cold environments remain ambiguous. Herein, we simulated a winter environment (- 20 °C) and provided a rationed diet to compare the cold adaptation mechanisms between Hulunbuir and Hu sheep. RESULTS Our findings show that cold exposure enhances SCFA metabolism in the sheep cecum. In Hu sheep, acetate, butyrate, and total SCFA concentrations increased, whereas in Hulunbuir sheep, propionate and butyrate concentrations increased, with a notable increase in total SCFAs. Notably, butyrate concentration was higher in Hulunbuir sheep than in Hu sheep under cold stress. Following cold exposure, the proinflammatory cytokine IL-1β levels increased in both breeds. In addition, Hu sheep showed increased IL-10, whereas Hulunbuir sheep exhibited elevated secretory IgA levels. The cecal microbiota responded differently, Hu sheep showed no notable changes in alpha and beta diversity, whereas Hulunbuir sheep exhibited considerable alterations. In Hu sheep, the abundance of fungi, specifically Blastocystis sp. subtype 4, decreased, and that of several Lachnospiraceae species (Roseburia hominis, Faecalicatena contorta, and Ruminococcus gnavus) involved in SCFA metabolism increased. Pathways related to carbohydrate metabolism, such as starch and sucrose metabolism, galactose metabolism, and pentose and glucuronate interconversions, were upregulated. In Hulunbuir sheep, the abundance of Treponema bryantii, Roseburia sp. 499, and Prevotella copri increased, with upregulation in pathways related to amino acid metabolism and energy metabolism. Cold exposure increased node connectivity within the symbiotic networks of both breeds, with increased network vulnerability in Hu sheep. Following cold exposure, the microbial community of Hulunbuir sheep showed a decrease in the influence of stochastic processes on community assembly, with a corresponding increase in the role of environmental selection. Conversely, no such shift was evident in Hu sheep. Further transcriptomic analysis revealed distinct regulatory mechanisms between breeds. In Hu sheep, protein synthesis, energy metabolism, and thermogenesis pathways were substantially upregulated. By contrast, Hulunbuir sheep showed considerable upregulation of immune pathways and energy conservation through reduced ribosome synthesis. Correlation analysis indicated that butyrate holds a central position in both networks, with Hulunbuir sheep exhibiting a more complex and tightly regulated network involving SCFAs, microbiota, microbial functions, and transcriptomes. Partial least squares path modeling revealed that cold exposure substantially altered the cecal microbiota and transcriptomes of Hulunbuir sheep, affecting SCFAs and cytokines. CONCLUSIONS The findings of this study suggest that under cold exposure, Hu sheep enhance acetate fermentation and rely on tissue thermogenesis for adaptation. By contrast, Hulunbuir sheep exhibit changes in microbial diversity and function, leading to increased propionate and butyrate metabolism. This may promote physiological energy conservation and innate immune defense, balancing heat loss and enhancing cold adaptation.
Collapse
Affiliation(s)
- Xindong Cheng
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanping Liang
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kaixi Ji
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Animal Husbandry and Veterinary Medicine, Shandong Academy of Agricultural Sciences/Shandong Key Laboratory of Animal Microecologics and Efficient Breeding of Livestock and Poultry, Jinan, 250100, China
| | - Mengyu Feng
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xia Du
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dan Jiao
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiukun Wu
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Extreme Environmental Microbial Resources and Engineering, Lanzhou, 730000, China
| | - Chongyue Zhong
- Dongying Animal Husbandry and Veterinary Station, Dongying, 257000, China
| | - Haitao Cong
- Shandong Huakun Rural Revitalization Institute Co., Ltd, Jinan, 250014, China
| | - Guo Yang
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
3
|
Wu Y, Zeng M, Cao B, Zhang B, Zheng X, Feng W. Material basis and mechanism of Ephedra sinica in interfering with wind-chill cold. Int Immunopharmacol 2025; 152:114432. [PMID: 40058103 DOI: 10.1016/j.intimp.2025.114432] [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: 07/01/2024] [Revised: 01/18/2025] [Accepted: 03/03/2025] [Indexed: 03/24/2025]
Abstract
Ephedra sinica has a long history in medicine, i.e., as the first medicinal plant for treating wind-chill colds. However, few studies have shown its material basis and mechanism of action, so this article mainly focuses on these two issues. Rats were stimulated by fan blowing and low temperature for 7 days, following which alkaloid extract (SWJ), non-alkaloid extract (FSWJ), and polysaccharide extract (DT) from Ephedra sinica (MH) were administered for 4 days. First, the body temperature, sweat spots, activity status, and cough-related indexes were detected to screen the pharmacodynamic material basis of MH. Then, the immune cells, ROS, MDA, GSH-PX, SOD, IL-4, IgE, TRPM8, TLR4, p-P65/P65, acetylcholine, and CHRM3 levels were detected to explore the potential mechanism of the SWJ. In addition, BEAS-2B cells were cultured at 26 °C for 12 h to establish a cell injury model induced by cold stimulation. BEAS-2B cells were co-cultured with spleen cells, and the effects of ephedrine (MHJ) and pseudoephedrine (WMHJ) on immune cells and inflammatory factors in this system were detected. The TRPM8 inhibitor (AMG-333) was added 3 h before administration to detect the effects of MHJ and WMHJ on the expression levels of TRPM8, TLR4, and P-P65/P65. Animal experiments showed that SWJ, FSWJ, and DT reduced body temperature and relieved symptoms such as sweating difficulty, listlessness, and cough, and SWJ produced the best effect. Subsequently, it was found that SWJ transformed immune cells, weakened oxidative stress, downregulated IL-4, IgE, TRPM8, TLR4, and P-P65/P65, and upregulated ACH and CHRM3. Cell experiments showed that MHJ and WMHJ upregulated the Ths and Tcs and downregulated IL-6, TNF-α, TRPM8, TLR4, and P-P65/P65. The intervention of AMG-333 affected the TRPM8 and p-P65/P65, but AMG-333 did not regulate TLR4. Taken together, alkaloid extract is the material basis for MH to improve rats' wind-chill cold and may protect rats from injury by regulating the TRPM8/TLR4-NFκB pathway and CHRM3, with ephedrine and pseudoephedrine possibly playing an important role.
Collapse
Affiliation(s)
- Yuanyuan Wu
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Mengnan Zeng
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Bing Cao
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Beibei Zhang
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Xiaoke Zheng
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China.
| | - Weisheng Feng
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China.
| |
Collapse
|
4
|
Priya A, Mol N, Singh AK, Aditya AK, Ray AK. "Unveiling the impacts of climatic cold events on the cardiovascular health in animal models". THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 971:179028. [PMID: 40073773 DOI: 10.1016/j.scitotenv.2025.179028] [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: 10/14/2024] [Revised: 02/01/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
Climate change is increasingly driving extreme weather events, leading to drastic temperature fluctuations worldwide. While overall temperatures rise, many regions are simultaneously experiencing severe cold spells that threaten the health of human populations, especially to vulnerable populations including the elderly and those with pre-existing conditions. Exposure to cold stress triggers significant physiological and biochemical disruptions. As cardiovascular diseases (CVDs) rank among the leading causes of global morbidity and mortality, the exacerbation of these conditions by cold exposure underscores critical public health challenges. The complex pathophysiological processes in cold-induced CVDs require careful analysis at an organ-system level, making animal models an ideal tool for replicating human physiological and molecular responses in a controlled environment. However, a detailed mechanism linking cold exposure and cardiovascular dysfunction remains incompletely understood, particularly in the context of animal models. Therefore, this comprehensive review aims to address and analyze from traditional rodent models to less conventional ruminants, broilers, canines, and primate animal models to understand cold stress-induced CVDs, with an extensive account of the potential molecular mechanisms and pathways such as oxidative stress, inflammation, vasomotor dysfunction, and apoptosis, along with emerging roles of cold shock proteins (CSPs), etc. We also delve into various potential therapeutic approaches and preventive measures in cold stress conditions. In conclusion, this review is the first to comprehensively address the underexplored cardiovascular complications arising from cold stress and their underlying mechanisms, particularly using animal models. Furthermore, it provides a foundation for advancing the development of more effective and targeted therapies through translational research.
Collapse
Affiliation(s)
- Anjali Priya
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Nidhi Mol
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Alok Kumar Singh
- Department of Zoology, Ramjas College, University of Delhi, New Delhi, India
| | - Abhishek Kumar Aditya
- Department of Medicine, K.D. Medical College, Hospital and Research Centre, Mathura, India
| | - Ashwini Kumar Ray
- Department of Environmental Studies, University of Delhi, New Delhi, India.
| |
Collapse
|
5
|
Fang Q, Cai Y, Chi J, Yang Y, Chen Q, Chen L, Zhang J, Ke J, Wu Y, He X. Silencing miR-155-5p alleviates hippocampal damage in kainic acid-induced epileptic rats via the Dusp14/MAPK pathway. Brain Res Bull 2024; 217:111057. [PMID: 39209069 DOI: 10.1016/j.brainresbull.2024.111057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/10/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Epilepsy with recurrent seizures is characterized by neuronal damage and glial proliferation induced by brain inflammation. Recurrent seizures can lead to changes in the microRNA (miRNA) spectrum, significantly influencing the inflammatory response of microglia. MiR-155-5p, as a pro-inflammatory miRNA, is increased in the epileptic brain. However, its specific role in acute seizures remains unknown. The study aimed to develop a new strategy for treating epilepsy by investigating how silencing of miR-155-5p initiated its anticonvulsive mechanism. The level of miR-155-5p was up-regulated in the hippocampus of epileptic immature rats induced by kainic acid (KA). The use of antago-miR-155-5p exerted significant beneficial effects on the seizure scores, brain discharges and cognition in immature rats following KA-induced epilepsy. Antago-miR-155-5p also inhibited neuron damage and microglial activation. Moreover, the silencing of miR-155-5p significantly inhibited the Dual-specificity phosphatase 14 (Dusp14)/ mitogen-activated protein kinase (MAPK) axis in vivo. MiR-155-5p interacted with dusp14 to regulate MAPK signaling way expression, verified by a dual-luciferase reporter assay. The results suggested that the silencing of miR-155-5p might reduce hippocampal damage in epileptic immature rats induced by KA via Dusp14/MAPK signaling way. This implied that miR-155-5p could serve as a therapeutic tool to prevent the development of epilepsy.
Collapse
Affiliation(s)
- Qiong Fang
- Department of Pediatrics, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, China.
| | - Yuehao Cai
- Department of Pediatrics, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, China
| | - Jiali Chi
- Department of Pediatrics, Ningde Normal University, NingDe, Ningde, Fujian 352000, China
| | - Yating Yang
- Department of Pediatrics, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, China
| | - Qiaobin Chen
- Department of Pediatrics, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, China.
| | - Libin Chen
- Department of Pediatrics, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, China
| | - Jiuyun Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, China; Department of Emergency, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, China; Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian 350001, China
| | - Jun Ke
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, China; Department of Emergency, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, China; Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian 350001, China
| | - Yanchen Wu
- Department of Pediatrics, Ningde Maternal and Child Health Hospital, Ningde, Fujian 352000, China
| | - Xiaoshuang He
- Department of Pediatrics, Fuzhou First General Hospital with Fujian Medical University, Fuzhou, Fujian 350001, China
| |
Collapse
|
6
|
Yan Q, Liu S, Sun Y, Chen C, Yang Y, Yang S, Lin M, Long J, Lin Y, Liang J, Ai Q, Chen N. CC chemokines Modulate Immune responses in Pulmonary Hypertension. J Adv Res 2024; 63:171-186. [PMID: 37926143 PMCID: PMC11380027 DOI: 10.1016/j.jare.2023.10.015] [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: 08/08/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) represents a progressive condition characterized by the remodeling of pulmonary arteries, ultimately culminating in right heart failure and increased mortality rates. Substantial evidence has elucidated the pivotal role of perivascular inflammatory factors and immune dysregulation in the pathogenesis of PH. Chemokines, a class of small secreted proteins, exert precise control over immune cell recruitment and functionality, particularly with respect to their migration to sites of inflammation. Consequently, chemokines emerge as critical drivers facilitating immune cell infiltration into the pulmonary tissue during inflammatory responses. This review comprehensively examines the significant contributions of CC chemokines in the maintenance of immune cell homeostasis and their pivotal role in regulating inflammatory responses. The central focus of this discussion is directed towards elucidating the precise immunoregulatory actions of CC chemokines concerning various immune cell types, including neutrophils, monocytes, macrophages, lymphocytes, dendritic cells, mast cells, eosinophils, and basophils, particularly in the context of pH processes. Furthermore, this paper delves into an exploration of the underlying pathogenic mechanisms that underpin the development of PH. Specifically, it investigates processes such as cellular pyroptosis, examines the intricate crosstalk between bone morphogenetic protein receptor type 2 (BMPR2) mutations and the immune response, and sheds light on key signaling pathways involved in the inflammatory response. These aspects are deemed critical in enhancing our understanding of the complex pathophysiology of PH. Moreover, this review provides a comprehensive synthesis of findings from experimental investigations targeting immune cells and CC chemokines. AIM OF REVIEW In summary, the inquiry into the inflammatory responses mediated by CC chemokines and their corresponding receptors, and their potential in modulating immune reactions, holds promise as a prospective avenue for addressing PH. The potential inhibition of CC chemokines and their receptors stands as a viable strategy to attenuate the inflammatory cascade and ameliorate the pathological manifestations of PH. Nonetheless, it is essential to acknowledge the current state of clinical trials and the ensuing progress, which regrettably appears to be less than encouraging. Substantial hurdles exist in the successful translation of research findings into clinical applications. The intention is that such emphasis could potentially foster the advancement of potent therapeutic agents presently in the process of clinical evaluation. This, in turn, may further bolster the potential for effective management of PH.
Collapse
Affiliation(s)
- Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha 410007, China
| | - Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jinping Liang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
7
|
Yuan M, Liu T, Cai A, Zhan Z, Cheng Y, Wang Q, Xia Y, Shen N, Huang P, Zou X. Emerging connectivity of programmed cell death pathways and pulmonary vascular remodelling during pulmonary hypertension. J Cell Mol Med 2024; 28:e70003. [PMID: 39153207 PMCID: PMC11330287 DOI: 10.1111/jcmm.70003] [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: 10/25/2023] [Revised: 04/08/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024] Open
Abstract
Pulmonary hypertension (PH) is a chronic progressive vascular disease characterized by abnormal pulmonary vascular resistance and pulmonary artery pressure. The major structural alteration during PH is pulmonary vascular remodelling, which is mainly caused by the imbalance between proliferation and apoptosis of pulmonary vascular cells. Previously, it was thought that apoptosis was the only type of programmed cell death (PCD). Soon afterward, other types of PCD have been identified, including autophagy, pyroptosis, ferroptosis and necroptosis. In this review, we summarize the role of the above five forms of PCD in mediating pulmonary vascular remodelling, and discuss their guiding significance for PH treatment. The current review could provide a better understanding of the correlation between PCD and pulmonary vascular remodelling, contributing to identify new PCD-associated drug targets for PH.
Collapse
Affiliation(s)
- Meng‐nan Yuan
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Ting Liu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - An‐qi Cai
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Zibo Zhan
- School of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Yi‐li Cheng
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Qi‐yue Wang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Yu‐xuan Xia
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Nong‐er Shen
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Xiao‐zhou Zou
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| |
Collapse
|
8
|
Aghajani Mir M. Illuminating the pathogenic role of SARS-CoV-2: Insights into competing endogenous RNAs (ceRNAs) regulatory networks. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 122:105613. [PMID: 38844190 DOI: 10.1016/j.meegid.2024.105613] [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: 03/07/2024] [Revised: 05/20/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
The appearance of SARS-CoV-2 in 2019 triggered a significant economic and health crisis worldwide, with heterogeneous molecular mechanisms that contribute to its development are not yet fully understood. Although substantial progress has been made in elucidating the mechanisms behind SARS-CoV-2 infection and therapy, it continues to rank among the top three global causes of mortality due to infectious illnesses. Non-coding RNAs (ncRNAs), being integral components across nearly all biological processes, demonstrate effective importance in viral pathogenesis. Regarding viral infections, ncRNAs have demonstrated their ability to modulate host reactions, viral replication, and host-pathogen interactions. However, the complex interactions of different types of ncRNAs in the progression of COVID-19 remains understudied. In recent years, a novel mechanism of post-transcriptional gene regulation known as "competing endogenous RNA (ceRNA)" has been proposed. Long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and viral ncRNAs function as ceRNAs, influencing the expression of associated genes by sequestering shared microRNAs. Recent research on SARS-CoV-2 has revealed that disruptions in specific ceRNA regulatory networks (ceRNETs) contribute to the abnormal expression of key infection-related genes and the establishment of distinctive infection characteristics. These findings present new opportunities to delve deeper into the underlying mechanisms of SARS-CoV-2 pathogenesis, offering potential biomarkers and therapeutic targets. This progress paves the way for a more comprehensive understanding of ceRNETs, shedding light on the intricate mechanisms involved. Further exploration of these mechanisms holds promise for enhancing our ability to prevent viral infections and develop effective antiviral treatments.
Collapse
Affiliation(s)
- Mahsa Aghajani Mir
- Deputy of Research and Technology, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
9
|
Santos-Gomes J, Mendes-Ferreira P, Adão R, Maia-Rocha C, Rego B, Poels M, Saint-Martin Willer A, Masson B, Provencher S, Bonnet S, Montani D, Perros F, Antigny F, Leite-Moreira AF, Brás-Silva C. Unraveling the Impact of miR-146a in Pulmonary Arterial Hypertension Pathophysiology and Right Ventricular Function. Int J Mol Sci 2024; 25:8054. [PMID: 39125620 PMCID: PMC11311781 DOI: 10.3390/ijms25158054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/17/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic disorder characterized by excessive pulmonary vascular remodeling, leading to elevated pulmonary vascular resistance and right ventricle (RV) overload and failure. MicroRNA-146a (miR-146a) promotes vascular smooth muscle cell proliferation and vascular neointimal hyperplasia, both hallmarks of PAH. This study aimed to investigate the effects of miR-146a through pharmacological or genetic inhibition on experimental PAH and RV pressure overload animal models. Additionally, we examined the overexpression of miR-146a on human pulmonary artery smooth muscle cells (hPASMCs). Here, we showed that miR-146a genic expression was increased in the lungs of patients with PAH and the plasma of monocrotaline (MCT) rats. Interestingly, genetic ablation of miR-146a improved RV hypertrophy and systolic pressures in Sugen 5415/hypoxia (SuHx) and pulmonary arterial banding (PAB) mice. Pharmacological inhibition of miR-146a improved RV remodeling in PAB-wild type mice and MCT rats, and enhanced exercise capacity in MCT rats. However, overexpression of miR-146a did not affect proliferation, migration, and apoptosis in control-hPASMCs. Our findings show that miR-146a may play a significant role in RV function and remodeling, representing a promising therapeutic target for RV hypertrophy and, consequently, PAH.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Rats
- Cell Proliferation/genetics
- Disease Models, Animal
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Monocrotaline
- Myocytes, Smooth Muscle/metabolism
- Pulmonary Arterial Hypertension/genetics
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Rats, Sprague-Dawley
- Vascular Remodeling/genetics
- Ventricular Function, Right
Collapse
Affiliation(s)
- Joana Santos-Gomes
- Cardiovascular R&D Centre–UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.S.-G.); (P.M.-F.); (R.A.); (C.M.-R.); (B.R.); (M.P.); (A.F.L.-M.)
| | - Pedro Mendes-Ferreira
- Cardiovascular R&D Centre–UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.S.-G.); (P.M.-F.); (R.A.); (C.M.-R.); (B.R.); (M.P.); (A.F.L.-M.)
- Paris-Porto Pulmonary Hypertension Collaborative Laboratory (3PH), UMR_S 999, INSERM, Université Paris-Saclay, 91190 Paris, France;
| | - Rui Adão
- Cardiovascular R&D Centre–UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.S.-G.); (P.M.-F.); (R.A.); (C.M.-R.); (B.R.); (M.P.); (A.F.L.-M.)
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
- CIBER Enfermedades Respiratorias (Ciberes), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Carolina Maia-Rocha
- Cardiovascular R&D Centre–UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.S.-G.); (P.M.-F.); (R.A.); (C.M.-R.); (B.R.); (M.P.); (A.F.L.-M.)
| | - Beatriz Rego
- Cardiovascular R&D Centre–UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.S.-G.); (P.M.-F.); (R.A.); (C.M.-R.); (B.R.); (M.P.); (A.F.L.-M.)
| | - Manu Poels
- Cardiovascular R&D Centre–UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.S.-G.); (P.M.-F.); (R.A.); (C.M.-R.); (B.R.); (M.P.); (A.F.L.-M.)
| | - Anaïs Saint-Martin Willer
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France; (A.S.-M.W.); (B.M.); (D.M.); (F.A.)
- Inserm UMR-S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Bastien Masson
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France; (A.S.-M.W.); (B.M.); (D.M.); (F.A.)
- Inserm UMR-S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec City, QC G1V 4G5, Canada; (S.P.); (S.B.)
- Department of Medicine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec City, QC G1V 4G5, Canada; (S.P.); (S.B.)
- Department of Medicine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - David Montani
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France; (A.S.-M.W.); (B.M.); (D.M.); (F.A.)
- Inserm UMR-S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Frédéric Perros
- Paris-Porto Pulmonary Hypertension Collaborative Laboratory (3PH), UMR_S 999, INSERM, Université Paris-Saclay, 91190 Paris, France;
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France; (A.S.-M.W.); (B.M.); (D.M.); (F.A.)
- Inserm UMR-S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, 69310 Pierre-Bénite, France
| | - Fabrice Antigny
- Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France; (A.S.-M.W.); (B.M.); (D.M.); (F.A.)
- Inserm UMR-S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Adelino F. Leite-Moreira
- Cardiovascular R&D Centre–UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.S.-G.); (P.M.-F.); (R.A.); (C.M.-R.); (B.R.); (M.P.); (A.F.L.-M.)
| | - Carmen Brás-Silva
- Cardiovascular R&D Centre–UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.S.-G.); (P.M.-F.); (R.A.); (C.M.-R.); (B.R.); (M.P.); (A.F.L.-M.)
- Faculty of Nutrition and Food Sciences, University of Porto, 4099-002 Porto, Portugal
| |
Collapse
|
10
|
Kosanovic D, Avdeev SN, Milovanov AP, Chernyaev AL. Northern Pulmonary Hypertension: A Forgotten Kind of Pulmonary Circulation Pathology. Life (Basel) 2024; 14:875. [PMID: 39063628 PMCID: PMC11277641 DOI: 10.3390/life14070875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Northern pulmonary hypertension (NPH) is a medical condition that is still enigmatic in non-Russian-speaking countries. The extant previous literature is mostly available in the Russian language and, therefore, not accessible to the rest of the world. The recent increased interest in climate changes and environmental effects on pulmonary circulation prompted us to summarize the knowledge from the past about the effects of cold on pulmonary vasculature. In this review, we, for the first time, describe, in detail, the pathological attributes of human NPH, a medical disorder that occurs in people living in extremely cold regions, in the English language. Briefly, NPH is characterized by the hyper-muscularization of the pulmonary arteries and de novo muscularization of the arterioles with the ultimate development of right ventricular hypertrophy. However, the profound molecular mechanisms of the NPH pathology are to be revealed in future comprehensive studies.
Collapse
Affiliation(s)
- Djuro Kosanovic
- Department of Pulmonology, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Sergey N. Avdeev
- Department of Pulmonology, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Andrey P. Milovanov
- Laboratory of Pathology of Reproduction, A. P. Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia;
| | - Andrey L. Chernyaev
- Fundamental Pulmonology Department, Federal Pulmonology Research Institute, 115682 Moscow, Russia
- Laboratory of Clinical Morphology, A. P. Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
| |
Collapse
|
11
|
Zhong Y, Liang B, Zhang X, Li J, Zeng D, Huang T, Wu J. NF-κB affected the serum levels of TNF-α and IL-1β via activation of the MAPK/NF-κB signaling pathway in rat model of acute pulmonary microthromboembolism. Pulm Circ 2024; 14:e12357. [PMID: 38584678 PMCID: PMC10995479 DOI: 10.1002/pul2.12357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 04/09/2024] Open
Abstract
Pulmonary thromboembolism caused by thrombi blocking major pulmonary artery and its branches, is a frequently encountered phenomenon and an important cause of high morbidity and mortality in lung diseases and may develop into persistent pulmonary hypertension (PH). Nuclear factor-κB (NF-κB) signaling pathway had been reported participated in the formation and development of PH by promoting inflammatory response. The aim of this study was to investigate the effects of NF-κB activation on the serum levels of tumor necrosis factor α (TNF-α) and interleukin-1β (IL-1β) in acute pulmonary microthromboembolism (APMTE) rats. Rats were randomized into five groups. APMTE group received jugular vein injection of autologous thrombus, while control group rats received normal saline injection. Pulmonary hemodynamic parameters were measured through ECHO-guided transthoracic puncture. Pulmonary vascular morphological changes were analyzed by HE. The expression changes of NF-κB and serum TNF-α、IL-1β levels were detected by enzyme-linked immunosorbent assay. Protein expression of the MAPK/NF-κB signaling pathway including p-IκBα, p-p38 MAPK, p-NF-κB p65, IκBα, p38 MAPK, and NF-κB p65 was determined using western blot analysis. Compared with control group, the expression of NF-κB in lung tissue and the levels of serum TNF-α and IL-1β rats were higher, a significant reduction in IκBα and elevation in the phosphorylation of IκBα, p38 MAPK, and NF-κB p65 were found in APMTE group rats. And UK administration reversed the APMTE-induced increase in TNF-α, IL-1β, p-IκBα, p-MAPK, and p-NF-κB protein. Furthermore, the levels of NF-κB, TNF-α, and IL-1β were positively correlated with mean pulmonary artery. And the levels of TNF-α and IL-1β were positively correlated with NF-κB. These findings suggest that the activation of MAPK/NF-κB pathway as a critical driver of increasing TNF-α and IL-1β level in APMTE rats and UK exerted protective effects against APMTE-induced PH may be related to the downregulation of the MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yanfen Zhong
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Binbin Liang
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Xiaofeng Zhang
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jingtao Li
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Decai Zeng
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Tongtong Huang
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Ji Wu
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
12
|
Lichtblau M, Reimann L, Piccari L. Pulmonary vascular disease, environmental pollution, and climate change. Pulm Circ 2024; 14:e12394. [PMID: 38933180 PMCID: PMC11205889 DOI: 10.1002/pul2.12394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Pollution and climate change constitute a combined, grave and pervasive threat to humans and to the life-support systems on which they depend. Evidence shows a strong association between pollution and climate change on cardiovascular and respiratory diseases, and pulmonary vascular disease (PVD) is no exception. An increasing number of studies has documented the impact of environmental pollution and extreme temperatures on pulmonary circulation and the right heart, on the severity and outcomes of patients with pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension (PH), on the incidence of pulmonary embolism, and the prevalence and severity of diseases associated with PH. Furthermore, the downstream consequences of climate change impair health care systems' accessibility, which could pose unique obstacles in the case of PVD patients, who require a complex and sophisticated network of health interventions. Patients, caretakers and health care professionals should thus be included in the design of policies aimed at adaptation to and mitigation of current challenges, and prevention of further climate change. The purpose of this review is to summarize the available evidence concerning the impact of environmental pollution and climate change on the pulmonary circulation, and to propose measures at the individual, healthcare and community levels directed at protecting patients with PVD.
Collapse
Affiliation(s)
- Mona Lichtblau
- Clinic of Pulmonology, Pulmonary Hypertension UnitUniversity Hospital ZurichZurichSwitzerland
| | - Lena Reimann
- Clinic of Pulmonology, Pulmonary Hypertension UnitUniversity Hospital ZurichZurichSwitzerland
| | - Lucilla Piccari
- Department of Pulmonary MedicineHospital del MarBarcelonaSpain
| |
Collapse
|
13
|
Yu X, Qin W, Cai H, Ren C, Huang S, Lin X, Tang L, Shan Z, Al-Ameer WHA, Wang L, Yan H, Chen M. Analyzing the molecular mechanism of xuefuzhuyu decoction in the treatment of pulmonary hypertension with network pharmacology and bioinformatics and verifying molecular docking. Comput Biol Med 2024; 169:107863. [PMID: 38199208 DOI: 10.1016/j.compbiomed.2023.107863] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/16/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND XueFuZhuYu (XFZY), a typical Chinese herbal formula, has remarkable clinical effects for treating Pulmonary Hypertension (PH) with unclear mechanisms. Our research involved the utilization of network pharmacology to explore the traditional Chinese herbal monomers and their related targets within XFZY for PH treatment. Furthermore, molecular docking verification was performed. METHODS The XFZY's primary active compounds, along with their corresponding targets, were both obtained from the TCMSP, ChEMBL, and UniProt databases. The target proteins relevant to PH were sifted through OMIM, GeneCards and TTD databases. The common "XFZY-PH" targets were evaluated with Disease Ontology (DO), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses with the assistance of R software. The Protein-Protein Interaction (PPI) network and compound-target-pathway network were constructed and a systematic analysis of network parameters was performed by the powerful software Cytoscape. Molecular docking was employed for assessing and verifying the interactions between the core targets and the top Chinese herbal monomer. RESULTS The screening included 297 targets of active compounds in XFZY and 8400 PH-related targets. DO analysis of the above common 268 targets indicated that the treatment of the diseases by XFZY is mediated by genes related to Chronic Obstructive Pulmonary Disease (COPD), Obstructive Lung Disease (OLD), ischemia, and myocardial infarction. The findings from molecular docking indicated that the binding energies of 57 ligand-receptor pairs in PH and 20 ligand-receptor pairs in COPD-PH were lower than -7kJ•mol-1. CONCLUSIONS This study indicates that XFZY is a promising option within traditional Chinese medicine compound preparation for combating PH, particularly in cases associated with COPD. Our demonstration of the specific molecular mechanism of XFZY anti-PH and its effective active ingredients provides a theoretical basis for better clinical application of the compound.
Collapse
Affiliation(s)
- Xiaoming Yu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Wenxiang Qin
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Haijian Cai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Chufan Ren
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Shengjing Huang
- Department of Pulmonary and Critical Care Medicine, The People's Hospital of Cangnan, The Affiliated Cangnan Hospital of Wenzhou Medical University, Wenzhou, 325800, Zhejiang, China.
| | - Xiao Lin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Lin Tang
- Alberta Institute, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Zhuohan Shan
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | | | - Liangxing Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Hanhan Yan
- Department of Pulmonary and Critical Care Medicine, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, Zhejiang, China.
| | - Mayun Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
14
|
Sun L, Liu L, Liang D, Liu L. SOCS5, targeted by miR-155-5p, plays a negative regulatory role in pulmonary hypertension through inhibiting JAK2/STAT3 signaling pathway. BMC Pulm Med 2024; 24:52. [PMID: 38267898 PMCID: PMC10809471 DOI: 10.1186/s12890-024-02857-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024] Open
Abstract
Pulmonary hypertension (PH) is a chronic pulmonary vascular disease and causes massive deaths. Here, we intended to investigate the function and mechanism of SOCS5 in PH. We engineered a hypoxia-induced PH model in mice. HE staining were implemented to evaluate pathological alterations in the lung tissues. The potential mechanism of SOCS5 in regulating hypoxia-induced pulmonary artery smooth muscle cell (PASMC) function was explored in vitro. RT-qPCR and western blot revealed that the level of SOCS5 was decreased both in PH mice and hypoxia-induced HPASMCs. Functional assays were performed for confirming the role of SOCS5 in modulating the cell phenotype and JAK2/STAT3 pathway in HPASMCs. Results revealed that overexpression of SOCS5 suppressed proliferation, migration and contraction of HPASMCs and negatively regulated the JAK2/STAT3 signaling pathway in HPASMCs under hypoxia in vitro, while knockdown of SOCS5 accelerated it. As evidenced by mechanism studies, SOCS5 was targeted and regulated by miR-155-5p, hence affecting on HPASMC proliferation, migration and contraction. These outcomes indicated that the decreased level of SOCS5 in hypoxia-induced HPASMCs promoted the cell proliferation, cell migration, and cell contraction through activating JAK2/STAT3 signaling pathway. Moreover, SOCS5 was targeted by miR-155-5p. All in all, our work hinted that miR-155-5p/SOCS5/JAK2/STAT3 axis played a crucial part in PH.
Collapse
Affiliation(s)
- Lili Sun
- Ward of Respiratory and Critical Care Department, The First Affiliated Hospital of Jinzhou Medical University, No. 2 Section 5 Renmin Street, Jinzhou, Liaoning, P.R. China
| | - Lihua Liu
- Ward of Respiratory and Critical Care Department, The First Affiliated Hospital of Jinzhou Medical University, No. 2 Section 5 Renmin Street, Jinzhou, Liaoning, P.R. China
| | - Dongxue Liang
- Ward of Respiratory and Critical Care Department, The First Affiliated Hospital of Jinzhou Medical University, No. 2 Section 5 Renmin Street, Jinzhou, Liaoning, P.R. China
| | - Linlin Liu
- Ward of Respiratory and Critical Care Department, The First Affiliated Hospital of Jinzhou Medical University, No. 2 Section 5 Renmin Street, Jinzhou, Liaoning, P.R. China.
| |
Collapse
|
15
|
He YZG, Wang YX, Ma JS, Li RN, Wang J, Lian TY, Zhou YP, Yang HP, Sun K, Jing ZC. MicroRNAs and their regulators: Potential therapeutic targets in pulmonary arterial hypertension. Vascul Pharmacol 2023; 153:107216. [PMID: 37699495 DOI: 10.1016/j.vph.2023.107216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 08/26/2023] [Accepted: 09/03/2023] [Indexed: 09/14/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a complex and progressive disease characterized by pulmonary arterial remodeling. Despite that current combination therapy has shown improvement in morbidity and mortality, a better deciphering of the underlying pathological mechanisms and novel therapeutic targets is urgently needed to combat PAH. MicroRNA, the critical element in post-transcription mechanisms, mediates cellular functions mainly by tuning downstream target gene expression. Meanwhile, upstream regulators can regulate miRNAs in synthesis, transcription, and function. In vivo and in vitro studies have suggested that miRNAs and their regulators are involved in PAH. However, the miRNA-related regulatory mechanisms governing pulmonary vascular remodeling and right ventricular dysfunction remain elusive. Hence, this review summarized the controversial roles of miRNAs in PAH pathogenesis, focused on different miRNA-upstream regulators, including transcription factors, regulatory networks, and environmental stimuli, and finally proposed the prospects and challenges for the therapeutic application of miRNAs and their regulators in PAH treatment.
Collapse
Affiliation(s)
- Yang-Zhi-Ge He
- Center for bioinformatics, National Infrastructures for Translational Medicine, Institute of Clinical Medicine & Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Yi-Xuan Wang
- Laboratory Department of Qingzhou People's Hospital, Qingzhou 262500, Shandong, China
| | - Jing-Si Ma
- Department of School of Pharmacy, Henan University, Kaifeng 475100, Henan, China
| | - Ruo-Nan Li
- Department of School of Pharmacy, Henan University, Kaifeng 475100, Henan, China
| | - Jia Wang
- Department of Medical Laboratory, Weifang Medical University, Weifang 261053, Shandong, China
| | - Tian-Yu Lian
- Medical Science Research Center, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Yu-Ping Zhou
- Department of Cardiology, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Beijing 100730, China
| | - Hao-Pu Yang
- Tsinghua University School of Medicine, Beijing 100084, China
| | - Kai Sun
- Medical Science Research Center, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China.
| | - Zhi-Cheng Jing
- Department of Cardiology, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Beijing 100730, China.
| |
Collapse
|
16
|
Huang Y, Xiong K, Wang A, Wang Z, Cui Q, Xie H, Yang T, Fan X, Jiang W, Tan X, Huang Q. Cold stress causes liver damage by inducing ferroptosis through the p38 MAPK/Drp1 pathway. Cryobiology 2023; 113:104563. [PMID: 37532122 DOI: 10.1016/j.cryobiol.2023.104563] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/29/2023] [Accepted: 07/29/2023] [Indexed: 08/04/2023]
Abstract
Acute extreme cold exposure impairs human health and even causes hypothermia which threatens human life. Liver, as a hub in metabolism and thermogenesis, is vital for cold acclimatization. Although accumulating evidence has suggested that cold exposure can cause liver damage, the underlying mechanisms remain poorly understood. This study investigated the role and underlying mechanisms of ferroptosis in cold stress-induced liver damage. To evaluate the role of ferroptosis in cold stress-induced liver damage, rats were pretreated with ferroptosis inhibitor liproxstatin-1 (Lip-1) before exposed to -10 °C for 8 h. Core body temperature was recorded. The levels of ferroptosis-related indicators were examined with the corresponding assay kits or by western blotting. Hepatic pathological changes were analyzed by hematoxylin-eosin staining and ultrastructural observation. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels were measured to assess liver function. Rats were also pretreated with p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580 or Dynamin-related protein 1 (Drp1) inhibitor Mdivi-1 to determine the underlying mechanisms. We found that Lip-1 inhibited ferroptosis, attenuated hepatic pathological damages and blocked the increased ALT and AST levels in cold-exposed rats. Moreover, Mdivi-1 inhibited mitochondrial fission and suppressed ferroptosis. Furthermore, SB203580 and Mdivi-1 administration alleviated cold stress-induced liver injury. Our results suggested that cold stress caused liver damage partially by inducing ferroptosis through the p38 MAPK/Drp1 pathway. These findings might provide an effective preventive and therapeutic target for cold stress-induced liver injury.
Collapse
Affiliation(s)
- Yujie Huang
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Shapingba District, Chongqing, 400038, PR China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, PR China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, PR China.
| | - Kun Xiong
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Shapingba District, Chongqing, 400038, PR China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, PR China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, PR China
| | - Aiping Wang
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Shapingba District, Chongqing, 400038, PR China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, PR China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, PR China
| | - Zejun Wang
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Shapingba District, Chongqing, 400038, PR China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, PR China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, PR China
| | - Qi Cui
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Shapingba District, Chongqing, 400038, PR China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, PR China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, PR China
| | - Hongchen Xie
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Shapingba District, Chongqing, 400038, PR China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, PR China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, PR China
| | - Tian Yang
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Shapingba District, Chongqing, 400038, PR China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, PR China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, PR China
| | - Xu Fan
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Shapingba District, Chongqing, 400038, PR China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, PR China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, PR China
| | - Wenjun Jiang
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Shapingba District, Chongqing, 400038, PR China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, PR China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, PR China
| | - Xiaoling Tan
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Shapingba District, Chongqing, 400038, PR China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, PR China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, PR China.
| | - Qingyuan Huang
- Department of Cold Environmental Medicine, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Shapingba District, Chongqing, 400038, PR China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, PR China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, PR China.
| |
Collapse
|
17
|
Chen C, He YS, Tao SS, Fang Y, Zhang RD, Fang X, Jiang LQ, Zhao Y, Musonye HA, Tao JH, Pan HF. Climate change and daily outpatient visits for dermatomyositis in Hefei, China: a time-series study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:101053-101063. [PMID: 37644268 DOI: 10.1007/s11356-023-29542-1] [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: 05/30/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
With the deepening of research on the correlation between meteorological factors and autoimmune diseases, the relationship between climate change and dermatomyositis (DM) has come to our attention. This study aimed to explore the short-term correlation between meteorological factors and DM outpatient visits. Daily records of hospital outpatient visits for DM, air pollutants, and meteorological factor data in Hefei from January 1, 2018 to December 31, 2021 were obtained. The mean temperature (MT), relative humidity (RH), diurnal temperature range (DTR), and temperature change between neighboring days (TCN) were used to quantify environmental temperature and humidity and their variations. And we performed a time series analysis using a generalized linear model (GLM) in combination with a distributed lag nonlinear model (DLNM). Furthermore, gender and age were further stratified for the analysis. The sensitivity analysis was also performed. A total of 4028 DM outpatient visits were recorded during this period. There were statistically significant associations of low temperature (5th, 1.5 °C), low RH (1st, 48.6%), high RH (99th, 99%), high DTR (75th, 12.6°c), and low TCN (10th, -2.7 °C) that were associated with risk of DM outpatient visits, with lag days of 30, 16, 16, 10, and 14, respectively. Moreover, women were more susceptible to high RH exposure and low TCN exposure, while the elderly were more susceptible to low temperature. This study concluded that exposure to low temperature, extreme RH, and temperature changes (especially high DTR and low TCN) was associated with an increased risk of DM outpatient visits.
Collapse
Affiliation(s)
- Cong Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
- Institute of Kidney Disease, Inflammation, and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Yi-Sheng He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
- Institute of Kidney Disease, Inflammation, and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Sha-Sha Tao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
- Institute of Kidney Disease, Inflammation, and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Yang Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
- Institute of Kidney Disease, Inflammation, and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Ruo-Di Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
- Institute of Kidney Disease, Inflammation, and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Xi Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
- Institute of Kidney Disease, Inflammation, and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Ling-Qiong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
- Institute of Kidney Disease, Inflammation, and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Yan Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
- Institute of Kidney Disease, Inflammation, and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Harry Asena Musonye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Jin-Hui Tao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of the University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
- Institute of Kidney Disease, Inflammation, and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
| |
Collapse
|
18
|
Sánchez-Gloria JL, Martínez-Olivares CE, Del Valle-Mondragón L, Cortés-Camacho F, Zambrano-Vásquez OR, Hernández-Pando R, Sánchez-Muñoz F, Sánchez-Lozada LG, Osorio-Alonso H. Allicin, an Emerging Treatment for Pulmonary Arterial Hypertension: An Experimental Study. Int J Mol Sci 2023; 24:12959. [PMID: 37629140 PMCID: PMC10454707 DOI: 10.3390/ijms241612959] [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: 07/05/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
We assessed whether allicin, through its antihypertensive and antioxidant effects, relieves vascular remodeling, endothelial function, and oxidative stress (OS), thereby improving experimental pulmonary arterial hypertension (PAH). Allicin (16 mg/kg) was administered to rats with PAH (monocrotaline 60 mg/kg). Allicin encouraged body weight gain and survival rate, and medial wall thickness and the right ventricle (RV) hypertrophy were prevented. Also, angiotensin II concentrations in the lung (0.37 ± 0.01 vs. 0.47 ± 0.06 pmoles/mL, allicin and control, respectively) and plasma (0.57 ± 0.05 vs. 0.75 ± 0.064, allicin and control respectively) and the expressions of angiotensin-converting enzyme II and angiotensin II type 1 receptor in lung tissue were maintained at normal control levels with allicin. In PAH rats treated with allicin, nitric oxide (NO) (31.72 ± 1.22 and 51.4 ± 3.45 pmoles/mL), tetrahydrobiopterin (8.43 ± 0.33 and 10.14 ± 0.70 pmoles/mL), cyclic guanosine monophosphate (5.54 ± 0.42 and 5.64 ± 0.73 pmoles/mL), and Ang-(1-7) (0.88 ± 0.23 and 0.83 ± 0.056 pmoles/mL) concentrations increased in lung tissue and plasma, respectively. In contrast, dihydrobiopterin increase was prevented in both lung tissue and plasma (5.75 ± 0.3 and 5.64 ± 0.73 pmoles/mL); meanwhile, phosphodiesterase-5 was maintained at normal levels in lung tissue. OS in PAH was prevented with allicin through the increased expression of Nrf2 in the lung. Allicin prevented the lung response to hypoxia, preventing the overexpression of HIF-1α and VEGF. Allicin attenuated the vascular remodeling and RV hypertrophy in PAH through its effects on NO-dependent vasodilation, modulation of RAS, and amelioration of OS. Also, these effects could be associated with the modulation of HIF-1α and improved lung oxygenation. The global effects of allicin contribute to preventing endothelial dysfunction, remodeling of the pulmonary arteries, and RV hypertrophy, preventing heart failure, thus favoring survival. Although human studies are needed, the data suggest that, alone or in combination therapy, allicin may be an alternative in treating PAH if we consider that, similarly to current treatments, it improves lung vasodilation and increase survival. Allicin may be considered an option when there is a lack of efficacy, and where drug intolerance is observed, to enhance the efficacy of drugs, or when more than one pathogenic mechanism must be addressed.
Collapse
Affiliation(s)
- José L. Sánchez-Gloria
- Department of Internal Medicine, Division of Nephrology, Rush University Medical Center, Chicago, IL 60612, USA;
| | - Constanza E. Martínez-Olivares
- Experimental Pathology Department, Experimental Pathology Laboratory, Instituto Nacional de Ciencia Médicas y Nutrición “Salvador Zubirán”, Mexico City 14080, Mexico; (C.E.M.-O.); (R.H.-P.)
| | - Leonardo Del Valle-Mondragón
- Departamento de Farmacología “Dr. Rafael Méndez Martínez”, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Fernando Cortés-Camacho
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (F.C.-C.); (O.R.Z.-V.); (L.G.S.-L.)
| | - Oscar R. Zambrano-Vásquez
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (F.C.-C.); (O.R.Z.-V.); (L.G.S.-L.)
| | - Rogelio Hernández-Pando
- Experimental Pathology Department, Experimental Pathology Laboratory, Instituto Nacional de Ciencia Médicas y Nutrición “Salvador Zubirán”, Mexico City 14080, Mexico; (C.E.M.-O.); (R.H.-P.)
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Laura G. Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (F.C.-C.); (O.R.Z.-V.); (L.G.S.-L.)
| | - Horacio Osorio-Alonso
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (F.C.-C.); (O.R.Z.-V.); (L.G.S.-L.)
| |
Collapse
|
19
|
Tong Y, Zhou MH, Li SP, Zhao HM, Zhang YR, Chen D, Wu YX, Pang QF. MiR-155-5p Attenuates Vascular Smooth Muscle Cell Oxidative Stress and Migration via Inhibiting BACH1 Expression. Biomedicines 2023; 11:1679. [PMID: 37371773 DOI: 10.3390/biomedicines11061679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/03/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The malfunction of vascular smooth muscle cells (VSMCs) is an initiating factor in the pathogenesis of pathological vascular remodeling, including hypertension-related vascular lesions. MicroRNAs (miRNAs) have been implicated in the pathogenesis of VSMC proliferation and migration in numerous cases of cardiovascular remodeling. The evidence for the regulatory role of miR-155-5p in the development of the cardiovascular system has been emerging. However, it was previously unclear whether miR-155-5p participated in the migration of VSMCs under hypertensive conditions. Thus, we aimed to define the exact role and action of miR-155-5p in VSMC migration by hypertension. Here, we detected that the level of miR-155-5p was lower in primary VSMCs from spontaneously hypertensive rats (SHRs). Its overexpression attenuated, while its depletion accelerated, the migration and oxidative damage of VSMCs from SHRs. Our dual-luciferase reporter assay showed that miRNA-155-5p directly targeted the 3'-untranslated region (3'-UTR) of BTB and CNC homology 1 (BACH1). The miR-155-5p mimic inhibited BACH1 upregulation in SHR VSMCs. By contrast, the deletion of miR-155-5p further elevated the upregulation of BACH1 in SHR-derived VSMCs. Importantly, the overexpression of miR-155-5p and knockdown of BACH1 had synergistic effects on the inhibition of VSMCs in hypertension. Collectively, miR-155-5p attenuates VSMC migration and ameliorates vascular remodeling in SHRs, via suppressing BACH1 expression.
Collapse
Affiliation(s)
- Ying Tong
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, China
- Department of Pathophysiology, Nanjing Medical University, Nanjing 211166, China
| | - Mei-Hui Zhou
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, China
| | - Sheng-Peng Li
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, China
| | - Hui-Min Zhao
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, China
| | - Ya-Ru Zhang
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, China
| | - Dan Chen
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, China
| | - Ya-Xian Wu
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, China
| | - Qing-Feng Pang
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, China
| |
Collapse
|
20
|
Portes AMO, Paula ABR, Miranda DCD, Resende LT, Coelho BIC, Teles MC, Jardim IABA, Natali AJ, Castrucci AMDL, Isoldi MC. A systematic review of the effects of cold exposure on pathological cardiac remodeling in mice. J Therm Biol 2023; 114:103598. [PMID: 37321023 DOI: 10.1016/j.jtherbio.2023.103598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023]
Abstract
Exposure to cold promotes cardiac remodeling, characterized by deleterious effects on structure and function, contributing to increased mortality from cardiovascular diseases. The mechanisms associated with these changes are poorly understood. This review gathers the literature data on the main alterations and mechanisms associated with the adverse cardiac structural and functional remodeling induced by cold exposure in mice. Original studies were identified by searching PubMed, Scopus, and Embase databases from January 1990 to June 2022. This systematic review was conducted in accordance with the criteria established by PRISMA and registered in PROSPERO (CRD42022350637). The risk of bias was evaluated by the SYRCLE. Eligible studies included original papers published in English that evaluated cardiac outcomes in mice submitted to short- or long-time cold exposure and had a control group at room temperature. Seventeen original articles were included in this review. Cold exposure induces pathological cardiac remodeling, characterized by detrimental structural and functional parameters, changes in metabolism and autophagy process, and increases in oxidative stress, inflammation, and apoptosis. In addition, Nppa, AT1A, Fbp3, BECN, ETA, and MT, appear to play fundamental roles in regulating cardiac remodeling. We suggest that strategies that seek to minimize the CVD risk and adverse effects of cold exposure should target these agents.
Collapse
Affiliation(s)
- Alexandre Martins Oliveira Portes
- Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil; Department of Physical Education, Federal University of Viçosa, Viçosa, Brazil.
| | | | - Denise Coutinho de Miranda
- Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil; Department of Nutrition, Governador Ozanam Coelho University Center, Uba, Brazil
| | | | | | - Maria Cecília Teles
- Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | | | - Antônio José Natali
- Department of Physical Education, Federal University of Viçosa, Viçosa, Brazil
| | - Ana Maria de Lauro Castrucci
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil; Department of Biology, University of Virginia, Charlottesville, United States
| | - Mauro César Isoldi
- Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
21
|
Liu J, Wu J, Qiao C, He Y, Xia S, Zheng Y, Lv H. Impact of chronic cold exposure on lung inflammation, pyroptosis and oxidative stress in mice. Int Immunopharmacol 2023; 115:109590. [PMID: 36577159 DOI: 10.1016/j.intimp.2022.109590] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022]
Abstract
Chronic cold exposure, which is the main inducer of lung diseases in high latitudes, affects production efficiency and restricts the development of aquaculture. Although the relationship between cold exposure and susceptibility to the lungs is widely accepted, but the influence between them has not been fully explored. The aim of this study is to understand the underlying mechanism. In the present study, the mice, which are used to establish cold stress (CS)-induced lung injury model, are exposed to cold temperature (4 °C) for 3 h each day for 4 weeks. The results indicate that the expression of heat shock protein 70 (HSP70) is augmented by cold exposure. In addition, chronic cold exposure aggravate the formation of malondialdehyde (MDA) and lead to a significant decrease in the contents of micrococcus catalase (CAT) and glutathione (GSH). Moreover, chronic cold exposure significantly exacerbates the expression of inflammation- and apoptosis-related proteins. The activation of Bax and caspase-3 are significantly augmented. However, that of Bcl-2 is decreased. These results are different from those in room team. The results show that chronic cold exposure plays an important roles in the activation of multiple signaling pathways, such as pyroptosis-related, inflammation-related and oxidative stress-regulated signaling pathways. In summary, these investigations support that chronic cold exposure increase the risk of lung injury by activating inflammation, oxidative stress and pyroptosis.
Collapse
Affiliation(s)
- Jiahe Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jingjing Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Chunyu Qiao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yuxi He
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Shijie Xia
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yuwei Zheng
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
| | - Hongming Lv
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
| |
Collapse
|
22
|
Wang C, Xing Y, Zhang J, He M, Dong J, Chen S, Wu H, Huang HY, Chou CH, Bai L, He F, She J, Su A, Wang Y, Thistlethwaite PA, Huang HD, Yuan JXJ, Yuan ZY, Shyy JYJ. MED1 Regulates BMP/TGF-β in Endothelium: Implication for Pulmonary Hypertension. Circ Res 2022; 131:828-841. [PMID: 36252121 DOI: 10.1161/circresaha.122.321532] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/26/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Dysregulated BMP (bone morphogenetic protein) or TGF-β (transforming growth factor beta) signaling pathways are imperative in idiopathic and familial pulmonary arterial hypertension (PAH) as well as experimental pulmonary hypertension (PH) in rodent models. MED1 (mediator complex subunit 1) is a key transcriptional co-activator and KLF4 (Krüppel-like factor 4) is a master transcription factor in endothelium. However, MED1 and KLF4 epigenetic and transcriptional regulations of the BMP/TGF-β axes in pulmonary endothelium and their dysregulations leading to PAH remain elusive. We investigate the MED1/KLF4 co-regulation of the BMP/TGF-β axes in endothelium by studying the epigenetic regulation of BMPR2 (BMP receptor type II), ETS-related gene (ERG), and TGFBR2 (TGF-β receptor 2) and their involvement in the PH. METHODS High-throughput screening involving data from RNA-seq, MED1 ChIP-seq, H3K27ac ChIP-seq, ATAC-seq, and high-throughput chromosome conformation capture together with in silico computations were used to explore the epigenetic and transcriptional regulation of BMPR2, ERG, and TGFBR2 by MED1 and KLF4. In vitro experiments with cultured pulmonary arterial endothelial cells (ECs) and bulk assays were used to validate results from these in silico analyses. Lung tissue from patients with idiopathic PAH, animals with experimental PH, and mice with endothelial ablation of MED1 (EC-MED1-/-) were used to study the PH-protective effect of MED1. RESULTS Levels of MED1 were decreased in lung tissue or pulmonary arterial endothelial cells from idiopathic PAH patients and rodent PH models. Mechanistically, MED1 acted synergistically with KLF4 to transactivate BMPR2, ERG, and TGFBR2 via chromatin remodeling and enhancer-promoter interactions. EC-MED1-/- mice showed PH susceptibility. In contrast, MED1 overexpression mitigated the PH phenotype in rodents. CONCLUSIONS A homeostatic regulation of BMPR2, ERG, and TGFBR2 in ECs by MED1 synergistic with KLF4 is essential for the normal function of the pulmonary endothelium. Dysregulation of MED1 and the resulting impairment of the BMP/TGF-β signaling is implicated in the disease progression of PAH in humans and PH in rodent models.
Collapse
Affiliation(s)
- Chen Wang
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
| | - Yuanming Xing
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
| | - Jiao Zhang
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA (J.Z., M.H., J.D., J.Y.-J.)
| | - Ming He
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA (J.Z., M.H., J.D., J.Y.-J.)
| | - Jianjie Dong
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA (J.Z., M.H., J.D., J.Y.-J.)
| | - Shanshan Chen
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
| | - Haoyu Wu
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
| | - Hsi-Yuan Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong-Shenzhen, Shenzhen, China (H.-Y.H., H.-D.H.)
- School of Life and Health Sciences, The Chinese University of Hong Kong-Shenzhen, Shenzhen, China (H.-Y.H., H.-D.H.)
| | - Chih-Hung Chou
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan (C.-H.C.)
| | - Liang Bai
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
| | - Fangzhou He
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
| | - Jianqing She
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
| | - Ailing Su
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
| | - Youhua Wang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China (Y.W.)
| | - Patricia A Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, CA (P.A.T.)
| | - Hsien-Da Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong-Shenzhen, Shenzhen, China (H.-Y.H., H.-D.H.)
- School of Life and Health Sciences, The Chinese University of Hong Kong-Shenzhen, Shenzhen, China (H.-Y.H., H.-D.H.)
| | - Jason X-J Yuan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA (J.X.-J.Y.)
| | - Zu-Yi Yuan
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA (J.Z., M.H., J.D., J.Y.-J.)
| |
Collapse
|
23
|
Lu L, Xiong Y, Zhou J, Wang G, Mi B, Liu G. The Therapeutic Roles of Cinnamaldehyde against Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9177108. [PMID: 36254234 PMCID: PMC9569207 DOI: 10.1155/2022/9177108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/06/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022]
Abstract
Evidence from epidemiological studies has demonstrated that the incidence and mortality of cardiovascular diseases (CVDs) increase year by year, which pose a great threat on social economy and human health worldwide. Due to limited therapeutic benefits and associated adverse effects of current medications, there is an urgent need to uncover novel agents with favorable safety and efficacy. Cinnamaldehyde (CA) is a bioactive phytochemical isolated from the stem bark of Chinese herbal medicine Cinnamon and has been suggested to possess curative roles against the development of CVDs. This integrated review intends to summarize the physicochemical and pharmacokinetic features of CA and discuss the recent advances in underlying mechanisms and potential targets responsible for anti-CVD properties of CA. The CA-related cardiovascular protective mechanisms could be attributed to the inhibition of inflammation and oxidative stress, improvement of lipid and glucose metabolism, regulation of cell proliferation and apoptosis, suppression of cardiac fibrosis, and platelet aggregation and promotion of vasodilation and angiogenesis. Furthermore, CA is likely to inhibit CVD progression via affecting other possible processes including autophagy and ER stress regulation, gut microbiota and immune homeostasis, ion metabolism, ncRNA expression, and TRPA1 activation. Collectively, experiments reported previously highlight the therapeutic effects of CA and clinical trials are advocated to offer scientific basis for the compound future applied in clinical practice for CVD prophylaxis and treatment.
Collapse
Affiliation(s)
- Li Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuan Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Juan Zhou
- Department of Cardiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430073, China
| | - Guangji Wang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
24
|
Wang XS, Peng MJ, He CT. The antihypertensive effects of Eucommia ulmoides leaf water/ethanol extracts are chlorogenic acid dependent. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
25
|
Wang G, Tao X, Peng L. miR-155-5p regulates hypoxia-induced pulmonary artery smooth muscle cell function by targeting PYGL. Bioengineered 2022; 13:12985-12997. [PMID: 35611851 PMCID: PMC9275946 DOI: 10.1080/21655979.2022.2079304] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a cardiovascular disease that has high incidence and causes massive deaths. miR-155-5p/PYGL pathway was revealed to play a crucial role in PAH by weighted gene co-expression network analysis (WGCNA). The potential mechanism of miR-155-5p in regulating hypoxia-induced pulmonary artery smooth muscle cell (PASMC) function was analyzed through in vitro experiments. Hypoxia treatment stimulated the proliferation of PASMCs and increased the expression of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α (HIF-1α). At the same time, revealed by qRT-PCR and western blot, the level of miR-155-5p was raised, and the level of PYGL was decreased in hypoxia-induced PASMCs. Through CCK-8 assay, transwell assay and flow cytometry, it was revealed that miR-155-5p inhibitor remarkably inhibited the cell proliferation and migration and decreased the proportion of hypoxia-stimulated PASMCs in S and G2/M phases. Dual-luciferase reporter system was subsequently applied to validate the straight regulation of miR-155-5p on PYGL based on the analysis of online database. Furthermore, siPYGL was revealed to reverse the influence of miR-155-5p inhibitor on hypoxia-induced PASMCs. These outcomes indicate that the increased level of miR-155-5p in hypoxia-stimulated PASMCs could enhance the cell proliferation, cell migration, and cell cycle progression by targeting PYGL directly. This study may supply novel treatment strategies for PAH.Abbreviations: PH, pulmonary hypertension; PAH, pulmonary arterial hypertension; WGCNA, weighted gene co-expression network analysis; PASMCs, pulmonary artery smooth muscle cells; VEGF, vascular endothelial growth factor; HIF-1α, hypoxia-inducible factor-1α; SMCs, smooth muscle cells; DEGs, differentially expressed genes; GEO, Gene Expression Omnibus; GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; FBS, fetal bovine serum; OD, optical density; BCA, bicinchoninic acid; PVDF, polyvinylidene fluoride; PBS, phosphate-buffered saline; BP, biological process; MF, molecular function; CC, cell component.
Collapse
Affiliation(s)
- Guowen Wang
- Department of Respiratory Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Xuefang Tao
- Department of Respiratory Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Linlin Peng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
26
|
Yang YH, Lei L, Bao YP, Zhang L. An Integrated Metabolomic Screening Platform Discovers the Potential Biomarkers of Ischemic Stroke and Reveals the Protective Effect and Mechanism of Folic Acid. Front Mol Biosci 2022; 9:783793. [PMID: 35664672 PMCID: PMC9158342 DOI: 10.3389/fmolb.2022.783793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/17/2022] [Indexed: 11/29/2022] Open
Abstract
Folic acid has a protective effect against ischemic stroke. However, the protective pharmacological mechanism remains unclear. The aim of this study is to explore the protective effect of folic acid on ischemic stroke animals by an integrated metabolomic biomarker screening platform. Based on ultra-performance liquid chromatography-tandem mass spectrometry (UPLC/MS) coupled with multivariate data analysis, the changes in metabolites and pathways were characterized. We found that the metabolic alteration involved a total of 37 metabolites, of which 26 biomarkers such as γ-aminobutyric acid, lysine, glutamate, ribose, and valine can be regulated by folic acid via metabolic pathways of amino acid metabolism, carbohydrate metabolism, fatty acid metabolism, citrate cycle, and pyruvate metabolism, which may be the potential therapeutic targets of folic acid against ischemic stroke. Folic acid as an emerging potential natural anti-fibrosis agent has significant activity in protecting against middle cerebral artery occlusion-induced rat ischemic stroke model by delaying pathological development, reversing the metabolic biomarkers, and mainly regulating the perturbation in amino acid metabolism, carbohydrate metabolism, fatty acid metabolism, citrate cycle, and pyruvate metabolism. It also showed that the integrated metabolic biomarker screening platform could provide a better understanding of the therapeutic effect and mechanism of drugs.
Collapse
Affiliation(s)
- Yan-hui Yang
- Department of Clinical Nutrition, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- *Correspondence: Yan-hui Yang,
| | - Lei Lei
- Department of Nutrition, Harbin First Hospital, Harbin, China
| | - Yin-ping Bao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Lu Zhang
- Department of Clinical Nutrition, Heilongjiang Provincial Hospital, Harbin, China
| |
Collapse
|
27
|
Chen C, Duan F, Xie Y, Wan Q, Liu H, Gong J, Huang L, Song Z. Nuciferine attenuates acute ischemic stroke in a rat model: a metabolomic approach for the mechanistic study. Mol Omics 2022; 18:765-778. [DOI: 10.1039/d2mo00158f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nuciferine is a promise therapeutic candidate for ischemic stroke. 1H NMR metabolomics was conducted in this study to further elucidate its pharmacological mechanism, which is helpful to be used as a potential treatment for stroke clinically.
Collapse
Affiliation(s)
- Chang Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Feipeng Duan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Yongyan Xie
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Quan Wan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Haiyun Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Jinpeng Gong
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Liping Huang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, P. R. China
| | - Zonghua Song
- Chinese Pharmacopoeia Commission, Beijing 100061, P. R. China
| |
Collapse
|