1
|
Liu J, Chen X, Liu J, Peng C, Wang F, Huang X, Li S, Liu Y, Shou W, Cao D, Li X. Prenatal Inflammatory Exposure Predisposes Offspring to Chronic Kidney Diseases Via the Activation of the eIF2α-ATF4 Pathway. Inflammation 2024:10.1007/s10753-024-02084-5. [PMID: 38913145 DOI: 10.1007/s10753-024-02084-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2024] [Indexed: 06/25/2024]
Abstract
It has recently become more recognized that renal diseases in adults can originate from adverse intrauterine (maternal) environmental exposures. Previously, we found that prenatal lipopolysaccharide (LPS) exposure can result in chronic renal inflammation, which leads to renal damage in older offspring rats. To test whether prenatal inflammatory exposure predisposes offspring to renal damage, a mouse model of oral adenine consumption-induced chronic kidney disease (CKD) was applied to offspring from prenatal LPS-treated mothers (offspring-pLPS) and age-matched control offspring of prenatal saline-treated mothers (offspring-pSaline). We found that offspring-pLPS mice presented with more severe renal collagen deposition and renal dysfunction after 4 weeks of adenine consumption than sex- and treatment-matched offspring-pSaline controls. To illustrate the underlying molecular mechanism, we subjected offspring-pLPS and offspring-pSaline kidneys to genome-wide transcriptomic analysis. Bioinformatic analysis of the sequencing data, together with further experimental confirmation, revealed a strong activation of the PERK-eIF2α-ATF4-mediated unfolded protein response (UPR) in offspring-pLPS kidneys, which likely contributed to the CKD predisposition seen in offspring-pLPS mice. More importantly, the specific eIF2α-ATF4 signaling inhibitor ISIRB was able to prevent adenine-induced CKD in the offspring-pLPS mice. Our findings suggest that the eIF2α-ATF4-mediated UPR, but not PERK, is likely the major disease-causing pathway in prenatal inflammatory exposure-induced CKD predisposition. Our study also suggests that targeting this signaling pathway is a potentially promising approach for CKD treatment.
Collapse
Affiliation(s)
- Jie Liu
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China
| | - Xin Chen
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China
| | - Jie Liu
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China
| | - Cuiping Peng
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China
| | - Fangjie Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaoyong Huang
- Institute of Immunology, PLA, Army Medical University, Chongqing, China
| | - Shuhui Li
- Department of Clinical Biochemistry, College of Pharmacy, Army Medical University, Chongqing, China
| | - Ying Liu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Weinian Shou
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dayan Cao
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China.
| | - Xiaohui Li
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China.
| |
Collapse
|
2
|
Fu Y, Zhang HJ, Zhou W, Lai ZQ, Dong YF. The protective effects of sophocarpine on sepsis-induced cardiomyopathy. Eur J Pharmacol 2023; 950:175745. [PMID: 37146706 DOI: 10.1016/j.ejphar.2023.175745] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/10/2023] [Accepted: 04/24/2023] [Indexed: 05/07/2023]
Abstract
This investigation elucidates the impact of sophocarpine treatment on lipopolysaccharide (LPS) stimulated sepsis-induced cardiomyopathy (SIC) via in vivo and in vitro experiments. Echocardiography, ELISA, TUNEL, Western blotting experiments, and Hematoxylin/Eosin, Dihydroethidium, and Immunohistochemistry staining assays, were carried out to identify associated indicators. The echocardiography revealed that sophocarpine treatment alleviated LPS-induced cardiac dysfunction as indicated by fractional shortening shortened and improved ejection fraction. Heart injury biomarkers, such as creatine kinase, lactate dehydrogenase, and creatine kinase-MB, were assessed, and indicated that sophocarpine treatment could alleviate LPS-induced upregulation of these indices. Furthermore, different experimental protocols revealed that sophocarpine treatment inhibits LPS-induced pathological alterations and decreases LPS-stimulated inflammatory cytokines, IL-1β, monocyte chemoattractant protein-1, IL-6, NOD-like receptor protein-3, and TNF-α, increase. Apoptotic proteins such as cytochrome-c, Bax, and cleaved-caspase-3 were increased, and Bcl-2 was alleviated after LPS stimulation; however, these effects were inhibited by sophocarpine treatment. Decreased antioxidant proteins [superoxide dismutase-1 (SOD-1) and SOD-2] induced by LPS stimulation were upregulated by sophocarpine treatment. LPS upregulated autophagic proteins such as Beclin-1 and the ratio of microtubule-associated protein 1A/1B-light chain 3 (LC3)-II/LC3-I and downregulated sequestosome 1 (SQSTM1, or P62), sophocarpine therapy reversed these effects. Moreover, it was indicated that sophocarpine treatment inhibited the Toll-like receptor-4 (TLR-4)/nuclear transcription factor-kappa B (NF-κB) signaling pathway and activated nuclear factor erythroid 2-related factor-2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway. In conclusion, sophocarpine treatment could alleviate LPS-trigger SIC by repressing oxidative stress, autophagy, inflammation, and apoptosis via TLR-4/NF-κB inhibition and Nrf2/HO-1 signaling pathway activation, implicating the potential of sophocarpine as a new therapeutic approach against SIC.
Collapse
Affiliation(s)
- Yang Fu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang of Jiangxi, China; Jiangxi Key Laboratory of Molecular Medicine, China
| | - Hong-Jin Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang of Jiangxi, China; Jiangxi Key Laboratory of Molecular Medicine, China
| | - Wei Zhou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang of Jiangxi, China; Jiangxi Key Laboratory of Molecular Medicine, China
| | - Ze-Qun Lai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang of Jiangxi, China; Jiangxi Key Laboratory of Molecular Medicine, China
| | - Yi-Fei Dong
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang of Jiangxi, China; Jiangxi Key Laboratory of Molecular Medicine, China.
| |
Collapse
|
3
|
Selle J, Bohl K, Höpker K, Wilke R, Dinger K, Kasper P, Abend B, Schermer B, Müller RU, Kurschat C, Nüsken KD, Nüsken E, Meyer D, Savai Pullamsetti S, Schumacher B, Dötsch J, Alcazar MAA. Perinatal Obesity Sensitizes for Premature Kidney Aging Signaling. Int J Mol Sci 2023; 24:ijms24032508. [PMID: 36768831 PMCID: PMC9916864 DOI: 10.3390/ijms24032508] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/08/2022] [Accepted: 12/18/2022] [Indexed: 01/31/2023] Open
Abstract
Chronic Kidney Disease (CKD), a global health burden, is strongly associated with age-related renal function decline, hypertension, and diabetes, which are all frequent consequences of obesity. Despite extensive studies, the mechanisms determining susceptibility to CKD remain insufficiently understood. Clinical evidence together with prior studies from our group showed that perinatal metabolic disorders after intrauterine growth restriction or maternal obesity adversely affect kidney structure and function throughout life. Since obesity and aging processes converge in similar pathways we tested if perinatal obesity caused by high-fat diet (HFD)-fed dams sensitizes aging-associated mechanisms in kidneys of newborn mice. The results showed a marked increase of γH2AX-positive cells with elevated 8-Oxo-dG (RNA/DNA damage), both indicative of DNA damage response and oxidative stress. Using unbiased comprehensive transcriptomics we identified compartment-specific differentially-regulated signaling pathways in kidneys after perinatal obesity. Comparison of these data to transcriptomic data of naturally aged kidneys and prematurely aged kidneys of genetic modified mice with a hypomorphic allele of Ercc1, revealed similar signatures, e.g., inflammatory signaling. In a biochemical approach we validated pathways of inflammaging in the kidneys after perinatal obesity. Collectively, our initial findings demonstrate premature aging-associated processes as a consequence of perinatal obesity that could determine the susceptibility for CKD early in life.
Collapse
Affiliation(s)
- Jaco Selle
- Translational Experimental Pediatrics—Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, University Hospital Cologne, 50931 Cologne, Germany
| | - Katrin Bohl
- Department of Medicine II, Nephrology Research Laboratory, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - Katja Höpker
- Department of Medicine II, Nephrology Research Laboratory, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Department of Pediatric and Adolescent Medicine, University Hospital Cologne, 50931 Cologne, Germany
| | - Rebecca Wilke
- Translational Experimental Pediatrics—Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, University Hospital Cologne, 50931 Cologne, Germany
| | - Katharina Dinger
- Translational Experimental Pediatrics—Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, University Hospital Cologne, 50931 Cologne, Germany
| | - Philipp Kasper
- Department of Gastroenterology and Hepatology, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - Bastian Abend
- Translational Experimental Pediatrics—Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, University Hospital Cologne, 50931 Cologne, Germany
| | - Bernhard Schermer
- Department of Medicine II, Nephrology Research Laboratory, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - Roman-Ulrich Müller
- Department of Medicine II, Nephrology Research Laboratory, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - Christine Kurschat
- Department of Medicine II, Nephrology Research Laboratory, University Hospital of Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - Kai-Dietrich Nüsken
- Department of Pediatric and Adolescent Medicine, University Hospital Cologne, 50931 Cologne, Germany
| | - Eva Nüsken
- Department of Pediatric and Adolescent Medicine, University Hospital Cologne, 50931 Cologne, Germany
| | - David Meyer
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Institute for Genome Stability in Aging and Disease, Medical Faculty, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - Soni Savai Pullamsetti
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), Universities of Gießen and Marburg Lung Centre (UGMLC), Cardiopulmonary Institute (CPI), Member of the German Center of Lung Research (DZL), 35392 Gießen, Germany
| | - Björn Schumacher
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Institute for Genome Stability in Aging and Disease, Medical Faculty, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatric and Adolescent Medicine, University Hospital Cologne, 50931 Cologne, Germany
| | - Miguel A. Alejandre Alcazar
- Translational Experimental Pediatrics—Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, University Hospital Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
- Institute for Lung Health (ILH), Universities of Gießen and Marburg Lung Centre (UGMLC), Cardiopulmonary Institute (CPI), Member of the German Center of Lung Research (DZL), 35392 Gießen, Germany
- Correspondence: ; Tel.: +49-221-478-96876; Fax: +49-221-478-46868
| |
Collapse
|
4
|
Sophocarpine Alleviates Isoproterenol-Induced Kidney Injury by Suppressing Inflammation, Apoptosis, Oxidative Stress and Fibrosis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227868. [PMID: 36431969 PMCID: PMC9694211 DOI: 10.3390/molecules27227868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
One of the most common diseases affecting people and leading to high morbidity is kidney injury. The alleviation of inflammation and apoptosis is considered a potential therapeutic approach for kidney injury. Sophocarpine (SOP), a tetracyclic quinolizidine alkaloid, exhibits various beneficial biological properties. To investigate the effects of SOP on isoproterenol (ISO)-induced kidney injury, we randomly divided mice into four groups: Control, ISO, ISO+SOP (20 mg/kg) and ISO+SOP (40 mg/kg). SOP was administered intraperitoneally to the mice over two weeks, accompanied by intraperitoneal stimulation of ISO (10 mg/kg) for another four weeks. After the mice were sacrificed, several methods such as ELISA, staining (H&E, TUNEL, DHE and Masson) and Western blotting were applied to detect the corresponding indicators. The kidney injury serum biomarkers SCr and BUN increased after the ISO challenge, while this effect was reversed by treatment with SOP. Pathological changes induced by ISO were also reversed by treatment with SOP in the staining. The inflammatory cytokines IL-β, IL-6, TNF-α, MCP-1 and NLRP3 increased after the challenge with ISO, while they were decreased by treatment with SOP. The apoptotic proteins cleaved-caspase-3 and Bax increased, while Bcl-2 decreased, after the challenge with ISO, and these effects were reversed by treatment with SOP. The antioxidant proteins SOD-1 and SOD-2 decreased after being stimulated by ISO, while they increased after the treatment with SOP. The fibrotic proteins collagen I, collagen III, α-SMA, fibronectin, MMP-2 and MMP-9 increased after the challenge with ISO, while they decreased after the treatment with SOP. We further discovered that the TLR-4/NF-κB and TGF-β1/Smad3 signaling pathways were suppressed, while the Nrf2/HO-1 signaling pathway was activated. In summary, SOP could alleviate ISO-induced kidney injury by inhibiting inflammation, apoptosis, oxidative stress and fibrosis. The molecular mechanisms were suppression of the TLR-4/NF-κB and TGF-β1/Smad3 signaling pathways and activation of the Nrf2/HO-1 signaling pathway, indicating that SOP might serve as a novel therapeutic strategy for kidney injury.
Collapse
|
5
|
Hoogenboom LA, Lely AT, Kemp MW, Saito M, Jobe AH, Wolfs TGAM, Schreuder MF. Chorioamnionitis Causes Kidney Inflammation, Podocyte Damage, and Pro-fibrotic Changes in Fetal Lambs. Front Pediatr 2022; 10:796702. [PMID: 35444963 PMCID: PMC9013807 DOI: 10.3389/fped.2022.796702] [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: 10/17/2021] [Accepted: 03/01/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Perinatal complications, such as prematurity and intrauterine growth restriction, are associated with increased risk of chronic kidney disease. Although often associated with reduced nephron endowment, there is also evidence of increased susceptibility for sclerotic changes and podocyte alterations. Preterm birth is frequently associated with chorioamnionitis, though studies regarding the effect of chorioamnionitis on the kidney are scarce. In this study, we aim to unravel the consequences of premature birth and/or perinatal inflammation on kidney development using an ovine model. METHODS In a preterm sheep model, chorioamnionitis was induced by intra-amniotic injection of lipopolysaccharide (LPS) at either 2, 8, or 15 days prior to delivery. Control animals received intra-amniotic injections of sterile saline. All lambs were surgically delivered at 125 days' gestation (full term is 150 days) and immediately euthanized for necropsy. Kidneys were harvested and processed for staining with myeloperoxidase (MPO), Wilms tumor-1 (WT1) and alpha-smooth muscle actine (aSMA). mRNA expression of tumor necrosis factor alpha (TNFA), Interleukin 10 (IL10), desmin (DES), Platelet derived growth factor beta (PDGFB), Platelet derived growth factor receptor beta (PDGFRB), synaptopodin (SYNPO), and transforming growth factor beta (TGFB) was measured using quantitative PCR. RESULTS Animals with extended (but not acute) LPS exposure had an inflammatory response in the kidney. MPO staining was significantly increased after 8 and 15 days (p = 0.003 and p = 0.008, respectively). Expression of TNFA (p = 0.016) and IL10 (p = 0.026) transcripts was increased, peaking on day 8 after LPS exposure. Glomerular aSMA and expression of TGFB was increased on day 8, suggesting pro-fibrotic mesangial activation, however, this was not confirmed with PDFGB or PDGFRB. The number of WT1 positive nuclei in the glomerulus, as well as expression of synaptopodin, decreased, indicating podocyte injury. CONCLUSION We report that, in an ovine model of prematurity, LPS-induced chorioamnionitis leads to inflammation of the immature kidney. In addition, this process was associated with podocyte injury and there are markers to support pro-fibrotic changes to the glomerular mesangium. These data suggest a potential important role for antenatal inflammation in the development of preterm-associated kidney disease, which is frequent.
Collapse
Affiliation(s)
- Lieke A Hoogenboom
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, Netherlands.,Department of Pediatric Nephrology, Radboudumc Amalia Children's Hospital, Nijmegen, Netherlands
| | - A Titia Lely
- Department of Obstetrics, Wilhelmina Children's Hospital Birth Center, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Matthew W Kemp
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan.,Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, WA, Australia.,Women and Infants Research Foundation, Perth, WA, Australia.,Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Masatoshi Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Alan H Jobe
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Tim G A M Wolfs
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands.,Department of Biomedical Engineering (BMT), Maastricht University, Maastricht, Netherlands
| | - Michiel F Schreuder
- Department of Pediatric Nephrology, Radboudumc Amalia Children's Hospital, Nijmegen, Netherlands
| |
Collapse
|
6
|
Wu J, Fu Y, Wu YX, Wu ZX, Wang ZH, Li P. Lycorine ameliorates isoproterenol-induced cardiac dysfunction mainly via inhibiting inflammation, fibrosis, oxidative stress and apoptosis. Bioengineered 2021; 12:5583-5594. [PMID: 34515620 PMCID: PMC8806515 DOI: 10.1080/21655979.2021.1967019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Alleviating cardiac dysfunction improves the prognosis of heart failure patients. Lycorine is an alkaloid with several beneficial biological properties. Here, we used mice to evaluate the effect of lycorine on cardiac dysfunction elicited by isoproterenol. Mice were divided into four groups: control, lycorine, isoproterenol, and isoproterenol + lycorine. Mice in the combined group were treated daily with 10 mg/kg isoproterenol intraperitoneally for 2 weeks and 5 mg/kg lycorine was given simultaneously intraperitoneally for 4 weeks. Cardiac structure and function were assessed by echocardiography, hematoxylin and eosin staining, and Masson's trichrome staining. Isoproterenol-induced cardiac dysfunction and histopathological injury that was significantly improved by treatment with lycorine. Western blotting and the quantitative real-time polymerase chain reaction were used to explore the molecular mechanisms of these effects. Levels of the inflammatory cytokines, interleukin (IL)-1β, IL-6, and tumor necrosis factor-α, were increased by treatment with isoproterenol; these increases were significantly reduced by lycorine, with involvement of the NF-κB signaling pathway. The fibrotic factors, collagen I and collagen III, were increased by isoproterenol and decreased by treatment with lycorine through inhibiting activation of the Smad signaling pathway. In addition, lycorine alleviated oxidative stress as evidenced by a reduction in total reactive oxygen species in the isoproterenol + lycorine group compared to the isoproterenol group. Lycorine exerted an anti-apoptotic effect as evidenced by upregulating Bcl-2 and downregulating Bax. Overall, our findings demonstrate that lycorine protects against cardiac dysfunction induced by isoproterenol by inhibiting inflammation, fibrosis, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Ji Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yang Fu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ying-xing Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zu-xiang Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhen-hua Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ping Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- CONTACT Ping Li Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi330006, China
| |
Collapse
|
7
|
Tsosura TVS, Mattera MSDLC, Chiba FY, Carnevali ACN, Belardi BE, Dos Santos RM, Cintra LTA, Lopes FL, Scaramele NF, Matsushita DH. Effect of maternal apical periodontitis on the final step of insulin signalling and inflammatory pathway in the adult male offspring of rats. Int Endod J 2021; 54:2113-2124. [PMID: 34389996 DOI: 10.1111/iej.13610] [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: 05/20/2021] [Accepted: 08/11/2021] [Indexed: 11/28/2022]
Abstract
AIM To evaluate the final step of insulin signalling, inflammatory pathway (related to the inhibition of insulin signalling), peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1α) protein content and DNA methylation in the Slc2a4 gene promoter region in the skeletal muscle of adult male offspring of rats with apical periodontitis (AP) in a single tooth or in four teeth. METHODOLOGY Female Wistar rats were distributed into three groups: a control group, a group with one tooth with AP and a group with four teeth with AP. Thirty days after induction of AP, female rats from all groups were mated with healthy male rats. When male offspring reached 75 days of age, the following analyses were performed in the gastrocnemius muscle (GM): insulin-stimulated Akt serine and threonine phosphorylation status; NF-κB p50 and p65 subunits phosphorylation status; GLUT4, TNF-α and PGC-1α protein content by Western blotting; GLUT4 and TNF-α gene expression by real-time polymerase chain reaction (PCR); and DNA methylation in the Slc2a4 gene promoter region by restriction digestion and real-time PCR. Analysis of variance was performed, followed by Tukey's post hoc test. p values <.05 were considered to be statistically significant. RESULTS Maternal AP in four teeth decreased insulin-stimulated Akt serine and threonine phosphorylation status, reduced GLUT4 gene expression and its protein content, and increased NF-κB p50 and p65 subunits phosphorylation status in the GM of adult offspring. There were no alterations in the parameters analysed in the GM of adult offspring of rats with AP in a single tooth. In addition, maternal AP did not affect TNF-α gene expression and its protein content, PGC-1α protein content and DNA methylation in the Slc2a4 gene promoter region in the GM of adult offspring. CONCLUSIONS Maternal AP in four teeth was associated with impairment in the final step of insulin signalling in the GM of adult male offspring in rats. An increase in NF-κB activity may be involved in this decrease in insulin signalling. This study demonstrates the impact of maternal AP on the health of offspring, demonstrating the importance of maintaining adequate maternal oral health to prevent diseases in adult offspring in rats.
Collapse
Affiliation(s)
- Thaís Verônica Saori Tsosura
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil.,Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF)/Sociedade Brasileira de Fisiologia (SBFis), School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| | | | - Fernando Yamamoto Chiba
- Department of Preventive and Restorative Dental, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| | | | - Bianca Elvira Belardi
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil.,Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF)/Sociedade Brasileira de Fisiologia (SBFis), School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| | - Rodrigo Martins Dos Santos
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil.,Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF)/Sociedade Brasileira de Fisiologia (SBFis), School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| | - Luciano Tavares Angelo Cintra
- Department of Preventive and Restorative Dental, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| | - Flávia Lombardi Lopes
- Department of Production and Animal Health, School of Veterinary Medicine, São Paulo State University (UNESP), Araçatuba, Brazil
| | - Natália Francisco Scaramele
- Department of Production and Animal Health, School of Veterinary Medicine, São Paulo State University (UNESP), Araçatuba, Brazil
| | - Doris Hissako Matsushita
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil.,Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF)/Sociedade Brasileira de Fisiologia (SBFis), School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| |
Collapse
|
8
|
Kuiper-Makris C, Selle J, Nüsken E, Dötsch J, Alejandre Alcazar MA. Perinatal Nutritional and Metabolic Pathways: Early Origins of Chronic Lung Diseases. Front Med (Lausanne) 2021; 8:667315. [PMID: 34211985 PMCID: PMC8239134 DOI: 10.3389/fmed.2021.667315] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Lung development is not completed at birth, but expands beyond infancy, rendering the lung highly susceptible to injury. Exposure to various influences during a critical window of organ growth can interfere with the finely-tuned process of development and induce pathological processes with aberrant alveolarization and long-term structural and functional sequelae. This concept of developmental origins of chronic disease has been coined as perinatal programming. Some adverse perinatal factors, including prematurity along with respiratory support, are well-recognized to induce bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease that is characterized by arrest of alveolar and microvascular formation as well as lung matrix remodeling. While the pathogenesis of various experimental models focus on oxygen toxicity, mechanical ventilation and inflammation, the role of nutrition before and after birth remain poorly investigated. There is accumulating clinical and experimental evidence that intrauterine growth restriction (IUGR) as a consequence of limited nutritive supply due to placental insufficiency or maternal malnutrition is a major risk factor for BPD and impaired lung function later in life. In contrast, a surplus of nutrition with perinatal maternal obesity, accelerated postnatal weight gain and early childhood obesity is associated with wheezing and adverse clinical course of chronic lung diseases, such as asthma. While the link between perinatal nutrition and lung health has been described, the underlying mechanisms remain poorly understood. There are initial data showing that inflammatory and nutrient sensing processes are involved in programming of alveolarization, pulmonary angiogenesis, and composition of extracellular matrix. Here, we provide a comprehensive overview of the current knowledge regarding the impact of perinatal metabolism and nutrition on the lung and beyond the cardiopulmonary system as well as possible mechanisms determining the individual susceptibility to CLD early in life. We aim to emphasize the importance of unraveling the mechanisms of perinatal metabolic programming to develop novel preventive and therapeutic avenues.
Collapse
Affiliation(s)
- Celien Kuiper-Makris
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jaco Selle
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Eva Nüsken
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Miguel A. Alejandre Alcazar
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Member of the German Centre for Lung Research (DZL), Institute for Lung Health, University of Giessen and Marburg Lung Centre (UGMLC), Gießen, Germany
| |
Collapse
|
9
|
Hoogenboom LA, Wolfs TGAM, Hütten MC, Peutz-Kootstra CJ, Schreuder MF. Prematurity, perinatal inflammatory stress, and the predisposition to develop chronic kidney disease beyond oligonephropathy. Pediatr Nephrol 2021; 36:1673-1681. [PMID: 32880745 PMCID: PMC8172498 DOI: 10.1007/s00467-020-04712-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/28/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022]
Abstract
Prematurity and perinatal stress, such as intrauterine growth restriction (IUGR) and chorioamnionitis, are pathological processes creating an impaired intrauterine environment. These intrauterine factors are associated with the development of proteinuria, hypertension, and chronic kidney disease (CKD) later in life. Initially, this was thought to be secondary to oligonephropathy, subsequent glomerular hypertrophy, and hyperfiltration, leading to glomerulosclerosis, a further decrease in nephron number, and finally CKD. Nowadays, there is increasing evidence that prematurity and perinatal stress affect not only nephron endowment but also the maturation of podocytes and vasculogenesis. IUGR is associated with podocyte damage and an aggravated course of nephrotic syndrome. Moreover, preterm birth and IUGR are known to cause upregulation of the postnatal renin-angiotensin system, resulting in hypertension. Chorioamnionitis causes damage to the glomeruli, thereby predisposing to the development of glomerulosclerosis. This review aims to summarize current knowledge on the influence of prematurity, IUGR, and chorioamnionitis on the development of different glomerular structures. After summarizing human and experimental data on low nephron number in general, a specific focus on the current understanding of podocyte and glomerular capillary formation in relation to prematurity and different causes of perinatal stress is presented.
Collapse
Affiliation(s)
- Lieke A. Hoogenboom
- grid.412966.e0000 0004 0480 1382Department of Pediatrics, Maastricht University Medical Centre+, Maastricht, The Netherlands ,grid.461578.9Department of Pediatric Nephrology, Radboudumc Amalia Children’s Hospital, Nijmegen, The Netherlands
| | - Tim G. A. M. Wolfs
- grid.412966.e0000 0004 0480 1382Department of Pediatrics, Maastricht University Medical Centre+, Maastricht, The Netherlands ,grid.5012.60000 0001 0481 6099Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands ,grid.5012.60000 0001 0481 6099Department of Biomedical Engineering (BMT), Maastricht University, Maastricht, The Netherlands
| | - Matthias C. Hütten
- grid.5012.60000 0001 0481 6099Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands ,grid.412966.e0000 0004 0480 1382Department of Neonatology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Carine J. Peutz-Kootstra
- grid.412966.e0000 0004 0480 1382Department of Pathology, School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Michiel F. Schreuder
- grid.461578.9Department of Pediatric Nephrology, Radboudumc Amalia Children’s Hospital, Nijmegen, The Netherlands
| |
Collapse
|
10
|
Pan Z, Yang K, Wang H, Xiao Y, Zhang M, Yu X, Xu T, Bai T, Zhu H. MFAP4 deficiency alleviates renal fibrosis through inhibition of NF-κB and TGF-β/Smad signaling pathways. FASEB J 2020; 34:14250-14263. [PMID: 32905637 DOI: 10.1096/fj.202001026r] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/22/2020] [Accepted: 08/03/2020] [Indexed: 12/19/2022]
Abstract
Renal fibrosis, which is characterized by excessive extracellular matrix (ECM) accumulation in the renal tubulointerstitium, can lead to chronic kidney disease (CKD). The role of microfiber-associated protein 4 (MFAP4), which is an ECM protein that interacts with elastin and collagen, in renal fibrosis has not been investigated. The aim of this study was to examine the role of MFAP4 in the pathogenesis of renal fibrosis and the underlying mechanism using in vivo and in vitro models. The MFAP4-/- mice were subjected to unilateral ureteral obstruction (UUO) to elucidate the role of MFAP4 in renal fibrosis in vivo. Compared to the wild-type mice, the MFAP4-/- mice exhibited decreased protein expression of p-p65 and p-IKBα and ECM deposition after UUO. The MFAP4-/- mice exhibited attenuated nuclear translocation of p65 (the hub subunit of nuclear factor (NF)-κB signaling pathway), suppressed activation of transforming growth factor (TGF)-β/Smad pathways, and downregulated expression of fibronectin, collagen I, and plasminogen activator inhibitor-1. The knockdown of MFAP4 mitigated the TGF-β-induced upregulated expression of fibronectin, collagen I, and plasminogen activator inhibitor-1 in the human proximal tubular epithelial cells (HK-2). Compared to the HK-2 cells transfected with sh-MFAP4, the HK-2 cells co-transfected with sh-MFAP4 and Ad-MFAP4 exhibited severe inflammatory response and increased fibrosis-related proteins expression. Mechanistically, the knockdown of MFAP4 inhibited the activation of NF-κB and TGF-β/Smad signaling pathways and downregulated the expression of fibrosis-related proteins. The findings of this study indicate that MFAP4 is involved in UUO-induced renal fibrosis through regulation of NF-κB and TGF-β/Smad pathways.
Collapse
Affiliation(s)
- Zhou Pan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Kang Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Huibo Wang
- Department of Cardiology, Three Gorges University People's Hospital, The First People's Hospital of Yichang, Yichang, P.R. China
| | - Yusha Xiao
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Ming Zhang
- Department of Orthopedics Trauma, Puren Hospital, Wuhan University of Science and Technology, Wuhan, P.R. China
| | - Xi Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Tao Xu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Tao Bai
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Hengcheng Zhu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| |
Collapse
|
11
|
Hu H, Fu Y, Li M, Xia H, Liu Y, Sun X, Hu Y, Song F, Cheng X, Li P, Wu Y. Interleukin-35 pretreatment attenuates lipopolysaccharide-induced heart injury by inhibition of inflammation, apoptosis and fibrotic reactions. Int Immunopharmacol 2020; 86:106725. [PMID: 32679538 DOI: 10.1016/j.intimp.2020.106725] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/07/2020] [Accepted: 06/17/2020] [Indexed: 12/14/2022]
Abstract
Previous studies have demonstrated that targeting inflammation is a promising strategy for treating lipopolysaccharide (LPS)-induced sepsis and related heart injury. Interleukin-35 (IL-35), which consists of two subunits, Epstein-Barr virus-induced gene 3 (EBI3) and p35, is an immunosuppressive cytokine of the IL-12 family and exhibits strong anti-inflammatory activity. However, the role of IL-35 in LPS-induced heart injury reains obscure. In this study, we explored the role of IL-35 in heart injury induced by LPS and its potential mechanisms. Mice were treated with a plasmid encoding IL-35 (pIL-35) and then injected intraperitoneally (ip) with LPS (10 mg/kg). Cardiac function was assessed by echocardiography 12 h later. LPS apparently decreased the expression of EBI3 and p35 and caused cardiac dysfunction and pathological changes, which were significantly improved by pIL-35 pretreatment. Moreover, pIL-35 pretreatment significantly decreased the levels of cardiac proinflammatory cytokines including TNF-α, IL-6, and IL-1β, and the NLRP3 inflammasome. Furthermore, decreased number of apoptotic myocardial cells, increased BCL-2 levels and decreased BAX levels inhibited apoptosis, and LPS-induced upregulation of the expression of cardiac pro-fibrotic genes (MMP2 and MMP9) and fibrotic factor (Collagen type I) was inhibited. Further investigation indicated that pIL-35 pretreatment might suppressed the activation of the cardiac NF-κBp65 and TGF-β1/Smad2/3 signaling pathways in LPS-treated mice. Similar cardioprotective effects of IL-35 pretreatment were observed in mouse myocardial fibroblasts challenged with LPS in vitro. In summary, IL-35 pretreatment can attenuate cardiac inflammation, apoptosis, and fibrotic reactions induced by LPS, implicating IL-35 as a promising therapeutic target in sepsis-related cardiac injury.
Collapse
Affiliation(s)
- Huan Hu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yang Fu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Huasong Xia
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yue Liu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiaopei Sun
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yang Hu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Fulin Song
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiaoshu Cheng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ping Li
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
12
|
Cao HY, Li D, Wang YP, Lu HX, Sun J, Li HB. The protection of NF-κB inhibition on kidney injury of systemic lupus erythematosus mice may be correlated with lncRNA TUG1. Kaohsiung J Med Sci 2020; 36:354-362. [PMID: 31930775 DOI: 10.1002/kjm2.12183] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/27/2019] [Indexed: 12/29/2022] Open
Abstract
We aimed to know the effect of nuclear factor-kappa B (NF-κB) inhibition on the kidney injury of systemic lupus erythematosus (SLE) mice. Pristane-induced SLE mice were treated with pyrrolidine dithiocarbamate (PDTC, 50 or 100 mg/kg), a NF-κB inhibitor. Histopathological changes were observed by hematoxylin & eosin, Masson and periodic schiff-methenamine stainings. Long noncoding RNA Taurine upregulated gene 1 (LncRNA TUG1) was measured by real-time reverse transcription PCR, NF-κB p65 expression by western blotting, levels of inflammatory cytokines, antinuclear antibodies (ANA), and antidouble stranded DNA (anti-dsDNA) by enzyme-linked immunosorbent assay, and the deposition of IgG and C3 by immunofluorescence. The kidney of SLE mice exhibited interstitial inflammatory cell infiltration, interstitial fibrous proliferation, glomerular mesangial proliferation, and crescent formation, which was mitigated after PDTC administration. The levels of BUN, Cr, ANA, and anti-dsDNA and the pro-inflammatory factors in SLE mice were increased with obvious deposition of IgG and C3, but they were also reversed by PDTC. Furthermore, the NF-κB p65 expression in the nucleus in the SLE mice was decreased with the up-regulation of TUG1 expression and NF-κB p65 expression in the cytoplasm after PDTC treatment. Correlation analysis revealed the negative correlation between the TUG1 expression and NF-κB p65 in the nucleus in the kidney tissues. NF-κB inhibition with PDTC protected against the kidney injury of pristine-induced SLE mice possibly via up-regulating lncRNA TUG1, and further clinical studies are needed to clarify whether NF-κB inhibition may be a therapeutic modality for the kidney injury of SLE.
Collapse
Affiliation(s)
- Hai-Yu Cao
- Department of Dermatology, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei Province, China
| | - Dong Li
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun-Peng Wang
- Department of General Medicine, The Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Hui-Xiu Lu
- Department of Dermatology, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei Province, China
| | - Jing Sun
- Department of Dermatology, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei Province, China
| | - Hai-Bin Li
- Department of General Medicine, The Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
13
|
Watkins JM, von Chamier M, Brown MB, Reyes L, Hayward LF. Prenatal infection with Mycoplasma pulmonis in rats exaggerates the angiotensin II pressor response in adult offspring. Am J Physiol Regul Integr Comp Physiol 2019; 318:R338-R350. [PMID: 31850818 DOI: 10.1152/ajpregu.00194.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Exposure to different stressors in utero is linked to adult diseases such as obesity and hypertension. In this study, the impact of prenatal infection (PNI) on adult body weight and cardiovascular function was evaluated using a naturally occurring rodent pathogen, Mycoplasma pulmonis (MP). Pregnant Sprague-Dawley rats were infected with MP on gestational day 14 and gave birth naturally. Adult PNI offspring weighed more than controls, but resting mean arterial pressure (MAP) was unchanged. Subcutaneous injection of angiotensin II (10 μg/kg) elicited a rise in MAP that was greater in both male and female PNI offspring compared with controls (P < 0.03). The accompanying reflex bradycardia was similar to the controls, suggesting that PNI induced baroreflex dysfunction. Subcutaneous nicotine administration, a potent cardiorespiratory stimulus, also elicited a transient rise in MAP that was generally greater in the PNI group, but the change in MAP from baseline was only significant in the PNI females compared with controls (P < 0.03). Elevated body weight and cardiovascular reactivity in the PNI offspring was associated with an increase in the ratio of hypothalamic corticotrophin-releasing hormone receptors type 1 to type 2 gene expression in both sexes compared with controls. These findings support previous studies demonstrating that PNI induces alterations in cardiovascular function and body weight. Yet, unlike previous studies utilizing other models of PNI (e.g., endotoxin), MP PNI did not induce resting hypertension. Thus, our study provides a foundation for future studies evaluating the cardiovascular risks of offspring exposed to microbial challenges in utero.
Collapse
Affiliation(s)
- J M Watkins
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - M von Chamier
- Department of Infectious Disease and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - M B Brown
- Department of Infectious Disease and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - L Reyes
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - L F Hayward
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
14
|
Role of ESAT-6 in renal injury by regulating microRNA-155 expression via TLR4/MyD88 signaling pathway in mice with Mycobacterium tuberculosis infection. Biosci Rep 2017; 37:BSR20170021. [PMID: 28655852 PMCID: PMC5529202 DOI: 10.1042/bsr20170021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 02/06/2023] Open
Abstract
The study aims to investigate the underlying mechanism involved in the early secretory antigenic target-6 (ESAT-6) in renal injury through regulation of the expression of miR-155 through the oll-like receptor (TLR)-4 (TLR4)/myeloid differentiation factor 88 (MyD88) signaling pathway in Mycobacterium tuberculosis (MTB)-infected mice. Sixty C57BL/6 mice with MTB-induced renal injury were randomly assigned into control, MTB, mimic, inhibitor, inhibitor + ESAT6, and inhibitor + ESAT6 + TAK242 groups. Body weight, the ratio of kidney weight to body weight (Kw/Bw), blood urea nitrogen (BUN), and serum creatinine (Scr) of mice were measured. Flow cytometry was used to detect renal activation in mice. Expressions of miR-155 and ESAT6 were detected by quantitative real-time PCR (qRT-PCR), and Western blotting was used to examine the expressions of ESAT6, TLR4, and MyD88. Expressions of tumor necrosis factor-α (TNF-α), interleukin-17 (IL-17), and interferon-γ (IFN-γ) were measured by qRT-PCR and ELISA. Compared with the control group, the BUN and Scr levels as well as the expression levels of miR-155, TLR4, MyD88, TNF-α, IL-17, and IFN-γ increased, while Kw/Bw decreased in the MTB and mimic groups. In comparison with the MTB group, the above indexes except Kw/Bw were elevated in the mimic group, but were reduced in the inhibitor group, while the Kw/Bw dropped in the mimic group but increased in the inhibitor group. Compared with the inhibitor group, the Kw/Bw decreased while the rest of the indexes increased in the inhibitor + ESAT6 group. ESAT6 may induce renal injury by promoting miR-155 expression through the TLR-4/MyD88 signaling pathway in MTB-infected mice.
Collapse
|
15
|
Shen P, Yang X, Jiang J, Wang X, Liang T, He L. Wedelolactone from Eclipta alba inhibits lipopolysaccharide-enhanced cell proliferation of human renal mesangial cells via NF-κB signaling pathway. Am J Transl Res 2017; 9:2132-2142. [PMID: 28559966 PMCID: PMC5446498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/09/2017] [Indexed: 06/07/2023]
Abstract
Mesangial cells of glomerulus which could produce and degrade several ECMs, take part in the repair and update of mesangial matrix and GBM, regulate glomerular filtration rate, secret cytokines and phagocytose immune complexes contribute a lot to physiological functions and pathological reactions of glomerular. There inflammation response of abnormal proliferation induced by LPS could lead to renal damage. Herein, wedelolactone, an active chemical constituent extracted from leaves of Eclipta alba, was used to explore if it could be an effective inhibitor of the proliferative response of HRMCs. The effects of different concentration wedelolactone on the secretion of cytokines, cell viability and NF-κB pathway were all detected by qPCR, western blotting and ELISA. The results indicated that wedelolactone could inhibit the abnormal proliferation of HRMCs via regulating the activity of several key members of NF-κB signaling pathway.
Collapse
Affiliation(s)
- Peicheng Shen
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai, P. R. China
| | - Xuejun Yang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai, P. R. China
| | - Jian Jiang
- Shanghai University of Traditional Chinese MedicineShanghai, P. R. China.
| | - Xianxian Wang
- Shanghai University of Traditional Chinese MedicineShanghai, P. R. China.
| | - Tingyu Liang
- Shanghai University of Traditional Chinese MedicineShanghai, P. R. China.
| | - Liqun He
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai, P. R. China
| |
Collapse
|
16
|
Xianchu L, Lan PZ, Qiufang L, Yi L, Xiangcheng R, Wenqi H, Yang D. Naringin protects against lipopolysaccharide-induced cardiac injury in mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 48:1-6. [PMID: 27716530 DOI: 10.1016/j.etap.2016.09.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/06/2016] [Accepted: 09/11/2016] [Indexed: 06/06/2023]
Abstract
Previous research has demonstrated that lipopolysaccharide (LPS) can induce sepsis and lead to myocardial dysfunction. Naringin has various biological activities in LPS-induced sepsis. In this study, our aim was to investigate the effects of Naringin on LPS-induced cardiac injury and clarify its potential mechanism. We found that in vivo treatment with Naringin significantly ameliorated body weight loss, and attenuated cardiac histopathological changes after LPS challenge. Furthermore, Naringin inhibited LPS-induced increase of TNF-α, IL-1β and IL-6 activities to alleviate inflammatory response in heart. Moreover, Naringin supplement dramatically increased SOD levels, and prevented MDA levels to ameliorate oxidative stress compared with the LPS group in heart. Lastly, treatment with Naringin also significantly decreased the ratio of BAX to BCL-2 to resist apoptosis in heart. It is concluded that Naringin may be a promising therapeutic agent on LPS-induced cardiac injury by anti-inflammatory, anti-oxidant and anti-apoptotic effects.
Collapse
Affiliation(s)
- Liu Xianchu
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Professor Zheng Lan
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China.
| | - Li Qiufang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Liu Yi
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Ruan Xiangcheng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Hou Wenqi
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Ding Yang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| |
Collapse
|