1
|
Fan X, Ye J, Zhong W, Shen H, Li H, Liu Z, Bai J, Du S. The Promoting Effect of Animal Bioactive Proteins and Peptide Components on Wound Healing: A Review. Int J Mol Sci 2024; 25:12561. [PMID: 39684273 DOI: 10.3390/ijms252312561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/15/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
The skin is the first line of defense to protect the host from external environmental damage. When the skin is damaged, the wound provides convenience for the invasion of external substances. The prolonged nonhealing of wounds can also lead to numerous subsequent complications, seriously affecting the quality of life of patients. To solve this problem, proteins and peptide components that promote wound healing have been discovered in animals, which can act on key pathways involved in wound healing, such as the PI3K/AKT, TGF-β, NF-κ B, and JAK/STAT pathways. So far, some formulations for topical drug delivery have been developed, including hydrogels, microneedles, and electrospinning nanofibers. In addition, some high-performance dressings have been utilized, which also have great potential in wound healing. Here, research progress on the promotion of wound healing by animal-derived proteins and peptide components is summarized, and future research directions are discussed.
Collapse
Affiliation(s)
- Xiaoyu Fan
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jinhong Ye
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wanling Zhong
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huijuan Shen
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huahua Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhuyuan Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jie Bai
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shouying Du
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
2
|
Magnusen AF, Pandey MK. Complement System and Adhesion Molecule Skirmishes in Fabry Disease: Insights into Pathogenesis and Disease Mechanisms. Int J Mol Sci 2024; 25:12252. [PMID: 39596318 PMCID: PMC11594573 DOI: 10.3390/ijms252212252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Fabry disease is a rare X-linked lysosomal storage disorder caused by mutations in the galactosidase alpha (GLA) gene, resulting in the accumulation of globotriaosylceramide (Gb3) and its deacetylated form, globotriaosylsphingosine (Lyso-Gb3) in various tissues and fluids throughout the body. This pathological accumulation triggers a cascade of processes involving immune dysregulation and complement system activation. Elevated levels of complement 3a (C3a), C5a, and their precursor C3 are observed in the plasma, serum, and tissues of patients with Fabry disease, correlating with significant endothelial cell abnormalities and vascular dysfunction. This review elucidates how the complement system, particularly through the activation of C3a and C5a, exacerbates disease pathology. The activation of these pathways leads to the upregulation of adhesion molecules, including vascular cell adhesion molecule 1 (VCAM1), intercellular adhesion molecule 1 (ICAM1), platelet and endothelial cell adhesion molecule 1 (PECAM1), and complement receptor 3 (CR3) on leukocytes and endothelial cells. This upregulation promotes the excessive recruitment of leukocytes, which in turn exacerbates disease pathology. Targeting complement components C3a, C5a, or their respective receptors, C3aR (C3a receptor) and C5aR1 (C5a receptor 1), could potentially reduce inflammation, mitigate tissue damage, and improve clinical outcomes for individuals with Fabry disease.
Collapse
Affiliation(s)
- Albert Frank Magnusen
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
| | - Manoj Kumar Pandey
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
3
|
Ma J, Zhao Y, Cui Y, Lin H. Hypoxia Postconditioning Attenuates Hypoxia-Induced Inflammation and Endothelial Barrier Dysfunction. J Surg Res 2024; 301:413-422. [PMID: 39042975 DOI: 10.1016/j.jss.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 05/15/2024] [Accepted: 06/16/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION In recent years, a number of studies have demonstrated that hypoxia reoxygenation (HR) induced by ischemia postconditioning (IPC) reduces endothelial barrier dysfunction and inflammation in various models. When HR occurs, the P38 mitogen-activated protein kinase (P38 MAPK) breaks down the endothelial barrier. But no study has clearly clarified the effect of hypoxia postconditioning (HPC) on P38 MAPK in human dermal microvascular endothelial cells. Therefore, we investigated the function of HPC on P38 MAPK during HR in vitro. METHODS Human dermal microvascular endothelial cells were cultured in a hypoxic incubator for 8 h. Then cells were reperfused for 12 h (reoxygenation) or postconditioned by 5 min of reoxygenation and 5 min of re-hypoxia 3 times followed by 11.5 h reoxygenation. SB203580 was used as an inhibitor of P38 MAPK. Cell counting kit-8 assay kits were employed to detect cell activity. The corresponding levels of IL-6, IL-8 and IL-1β were examined via Enzyme-Linked ImmunoSorbent Assay. The endothelial barrier was evaluated using fluorescein isothiocyanate-dextran leakage assay. Western blot was used to detect claudin-5, phosphorylation of P38 MAPK (P-P38 MAPK) and P38 MAPK expression. Claudin-5 localization was studied by immunofluorescence. RESULTS HR induced endothelial barrier hyperpermeability, elevated inflammation levels, and increased the P-P38 MAPK. But HPC reduced cell injury and maintained the integrity of the endothelial barrier while inhibiting P-P38 MAPK and increasing expression of claudin-5. HPC redistributed claudin-5 in a continuous and linear pattern on the cell membrane. CONCLUSIONS HPC protects against HR induced downregulation and redistribution of claudin-5 by inhibiting P-P38 MAPK.
Collapse
Affiliation(s)
- Jiaxing Ma
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yinhua Zhao
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yue Cui
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Huang Lin
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Tao W, Min S, Chen G, He X, Meng Y, Li L, Chen J, Li Y. Tetramethylpyrazine ameliorates LPS-induced acute lung injury via the miR-369-3p/DSTN axis. Sci Rep 2024; 14:20006. [PMID: 39198493 PMCID: PMC11358269 DOI: 10.1038/s41598-024-70131-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Acute lung injury (ALI) is a severe clinical respiratory condition characterized by high rates of mortality and morbidity, for which effective treatments are currently lacking. In this study, lipopolysaccharide (LPS) was used to induce ALI mice, demonstrating the efficacy of tetramethylpyrazine (TMP) in ameliorating ALI. Subsequent we perfored high-throughput sequencing analysis and used Targetscan 8.0 and miRWalk 3.0 databases to predict the interaction between microRNAs and destrin (DSTN), ultimately identifying miR-369-3p as the focus of the investigation. The adenovirus carrying miR-369-3p was administered one week prior to LPS-induced in order to assess its potential efficacy in ameliorating ALI in mice. The findings indicated that the overexpression of miR-369-3p resulted in enhanced lung function, reduced pulmonary edema, inflammation, and permeability in LPS-induced ALI mice, while the suppression of miR-369-3p exacerbated the damage in these mice. Furthermore, the beneficial effects of TMP on LPS-induced ALI were negated by the downregulation of miR-369-3p. The results of our study demonstrate that TMP mitigates LPS-induced ALI through upregulation of miR-369-3p. Consequently, the findings of this study advocate for the clinical utilization of TMP in ALI treatment, with miR-369-3p emerging as a promising target for future ALI interventions.
Collapse
Affiliation(s)
- Weiting Tao
- Department of Pathophysiology, Bengbu Medical University, Bengbu, Anhui, China
| | - Simin Min
- Suzhou Hospital Affiliated to Anhui Medical University, Suzhou, Anhui, China
| | - Guofeng Chen
- School of Medicine and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| | - Xu He
- School of Medicine and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| | - Yuhang Meng
- School of Clinical Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Li Li
- Department of Pathophysiology, Bengbu Medical University, Bengbu, Anhui, China
| | - Jie Chen
- Department of Pathophysiology, Bengbu Medical University, Bengbu, Anhui, China
| | - Yan Li
- School of Medicine and Health Engineering, Changzhou University, Changzhou, Jiangsu, China.
| |
Collapse
|
5
|
Natarajan N, Florentin J, Johny E, Xiao H, O'Neil SP, Lei L, Shen J, Ohayon L, Johnson AR, Rao K, Li X, Zhao Y, Zhang Y, Tavakoli S, Shiva S, Das J, Dutta P. Aberrant mitochondrial DNA synthesis in macrophages exacerbates inflammation and atherosclerosis. Nat Commun 2024; 15:7337. [PMID: 39187565 PMCID: PMC11347661 DOI: 10.1038/s41467-024-51780-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 08/16/2024] [Indexed: 08/28/2024] Open
Abstract
There is a large body of evidence that cellular metabolism governs inflammation, and that inflammation contributes to the progression of atherosclerosis. However, whether mitochondrial DNA synthesis affects macrophage function and atherosclerosis pathology is not fully understood. Here we show, by transcriptomic analyzes of plaque macrophages, spatial single cell transcriptomics of atherosclerotic plaques, and functional experiments, that mitochondrial DNA (mtDNA) synthesis in atherosclerotic plaque macrophages are triggered by vascular cell adhesion molecule 1 (VCAM-1) under inflammatory conditions in both humans and mice. Mechanistically, VCAM-1 activates C/EBPα, which binds to the promoters of key mitochondrial biogenesis genes - Cmpk2 and Pgc1a. Increased CMPK2 and PGC-1α expression triggers mtDNA synthesis, which activates STING-mediated inflammation. Consistently, atherosclerosis and inflammation are less severe in Apoe-/- mice lacking Vcam1 in macrophages. Downregulation of macrophage-specific VCAM-1 in vivo leads to decreased expression of LYZ1 and FCOR, involved in STING signalling. Finally, VCAM-1 expression in human carotid plaque macrophages correlates with necrotic core area, mitochondrial volume, and oxidative damage to DNA. Collectively, our study highlights the importance of macrophage VCAM-1 in inflammation and atherogenesis pathology and proposes a self-acerbating pathway involving increased mtDNA synthesis.
Collapse
Affiliation(s)
- Niranjana Natarajan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Jonathan Florentin
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Ebin Johny
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Hanxi Xiao
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Joint CMU-Pitt PhD program in Computational Biology, Pittsburgh, PA, USA
| | - Scott Patrick O'Neil
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Liqun Lei
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Jixing Shen
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Lee Ohayon
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Aaron R Johnson
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Krithika Rao
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Xiaoyun Li
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Yanwu Zhao
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Yingze Zhang
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Sina Tavakoli
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
- University of Pittsburgh School of Medicine Department of Pharmacology & Chemical Biology, Pittsburgh, PA, USA
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Stanic B, Kokai D, Markovic Filipovic J, Tomanic T, Vukcevic J, Stojkov V, Andric N. Vascular endothelial effects of dibutyl phthalate: In vitro and in vivo evidence. Chem Biol Interact 2024; 399:111120. [PMID: 38944327 DOI: 10.1016/j.cbi.2024.111120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/24/2024] [Accepted: 06/27/2024] [Indexed: 07/01/2024]
Abstract
Dibutyl phthalate (DBP) is widely used in many consumer and personal care products. Here, we report vascular endothelial response to DBP in three different exposure scenarios: after short-term exposure (24 h) of human endothelial cells (ECs) EA.hy926 to 10-6, 10-5, and 10-4 M DBP, long-term exposure (12 weeks) of EA.hy926 cells to 10-9, 10-8, and 10-7 M DBP, and exposure of rats (28 and 90 days) to 100, 500, and 5000 mg DBP/kg food. We examined different vascular functions such as migration of ECs, adhesion of ECs to the extracellular matrix, tube formation, the morphology of rat aorta, as well as several signaling pathways involved in controlling endothelial function. Short-term in vitro exposure to DBP increased migration of ECs through G protein-coupled estrogen receptor, extracellular signal-regulated kinase 1/2, and nitric oxide (NO) signaling and decreased adhesion to gelatin. Long-term in vitro exposure to DBP transiently increased EC migration and had a bidirectional effect on EC adhesion to gelatin and tube formation. These effects were accompanied by a sustained increase in NO production and endothelial NO synthase (eNOS) and Akt activity. In vivo, exposure to DBP for 90 days decreased the aortic wall-to-lumen ratio and increased eNOS and Akt phosphorylation in ECs of rat aorta. This comparative investigation has shown that exposure to DBP may affect vascular function by altering EC migration, adhesion to gelatin, and tube formation after short- and long-term in vitro exposure and by decreasing the aortic wall-to-lumen ratio in vivo. The eNOS-NO and Akt signaling could be important in mediating the effects of DBP in long-term exposure scenarios.
Collapse
Affiliation(s)
- Bojana Stanic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia.
| | - Dunja Kokai
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | | | - Tamara Tomanic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | - Jelena Vukcevic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | - Viktor Stojkov
- University of Novi Sad, Institute of Food Technology, Serbia
| | - Nebojsa Andric
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| |
Collapse
|
7
|
Seo D, Lee CM, Apio C, Heo G, Timsina J, Kohlfeld P, Boada M, Orellana A, Fernandez MV, Ruiz A, Morris JC, Schindler SE, Park T, Cruchaga C, Sung YJ. Sex and aging signatures of proteomics in human cerebrospinal fluid identify distinct clusters linked to neurodegeneration. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.18.24309102. [PMID: 38947020 PMCID: PMC11213043 DOI: 10.1101/2024.06.18.24309102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Sex and age are major risk factors for chronic diseases. Recent studies examining age-related molecular changes in plasma provided insights into age-related disease biology. Cerebrospinal fluid (CSF) proteomics can provide additional insights into brain aging and neurodegeneration. By comprehensively examining 7,006 aptamers targeting 6,139 proteins in CSF obtained from 660 healthy individuals aged from 43 to 91 years old, we subsequently identified significant sex and aging effects on 5,097 aptamers in CSF. Many of these effects on CSF proteins had different magnitude or even opposite direction as those on plasma proteins, indicating distinctive CSF-specific signatures. Network analysis of these CSF proteins revealed not only modules associated with healthy aging but also modules showing sex differences. Through subsequent analyses, several modules were highlighted for their proteins implicated in specific diseases. Module 2 and 6 were enriched for many aging diseases including those in the circulatory systems, immune mechanisms, and neurodegeneration. Together, our findings fill a gap of current aging research and provide mechanistic understanding of proteomic changes in CSF during a healthy lifespan and insights for brain aging and diseases.
Collapse
|
8
|
Seetharaman S, Devany J, Kim HR, van Bodegraven E, Chmiel T, Tzu-Pin S, Chou WH, Fang Y, Gardel ML. Mechanosensitive FHL2 tunes endothelial function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.16.599227. [PMID: 38948838 PMCID: PMC11212908 DOI: 10.1101/2024.06.16.599227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Endothelial tissues are essential mechanosensors in the vasculature and facilitate adaptation to various blood flow-induced mechanical cues. Defects in endothelial mechanoresponses can perturb tissue remodelling and functions leading to cardiovascular disease progression. In this context, the precise mechanisms of endothelial mechanoresponses contributing to normal and diseased tissue functioning remain elusive. Here, we sought to uncover how flow-mediated transcriptional regulation drives endothelial mechanoresponses in healthy and atherosclerotic-prone tissues. Using bulk RNA sequencing, we identify novel mechanosensitive genes in response to healthy unidirectional flow (UF) and athero-prone disturbed flow (DF). We find that the transcription as well as protein expression of Four-and-a-half LIM protein 2 (FHL2) are enriched in athero-prone DF both in vitro and in vivo. We then demonstrate that the exogenous expression of FHL2 is necessary and sufficient to drive discontinuous adherens junction morphology and increased tissue permeability. This athero-prone phenotype requires the force-sensitive binding of FHL2 to actin. In turn, the force-dependent localisation of FHL2 to stress fibres promotes microtubule dynamics to release the RhoGEF, GEF-H1, and activate the Rho-ROCK pathway. Thus, we unravelled a novel mechanochemical feedback wherein force-dependent FHL2 localisation promotes hypercontractility. This misregulated mechanoresponse creates highly permeable tissues, depicting classic hallmarks of atherosclerosis progression. Overall, we highlight crucial functions for the FHL2 force-sensitivity in tuning multi-scale endothelial mechanoresponses.
Collapse
Affiliation(s)
- Shailaja Seetharaman
- Department of Physics, The University of Chicago, Chicago, IL 60637, USA
- James Franck Institute, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - John Devany
- Department of Physics, The University of Chicago, Chicago, IL 60637, USA
- James Franck Institute, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Ha Ram Kim
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| | - Emma van Bodegraven
- Department of Translational Neuroscience, Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Theresa Chmiel
- Department of Physics, The University of Chicago, Chicago, IL 60637, USA
- James Franck Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Shentu Tzu-Pin
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| | - Wen-hung Chou
- James Franck Institute, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Yun Fang
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| | - Margaret Lise Gardel
- Department of Physics, The University of Chicago, Chicago, IL 60637, USA
- James Franck Institute, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
9
|
Yaita N, Maruyama K, Hiroyasu K, Sato S. Immunogenic effects of enamel matrix derivative on human alveolar ridge mucosa-derived vascular endothelial cells under lipopolysaccharide stimulation. Odontology 2024:10.1007/s10266-024-00959-5. [PMID: 38839677 DOI: 10.1007/s10266-024-00959-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
Early peri-implant disease detection remains difficult. Enamel matrix derivative (EMD), which is used for periodontal tissue regeneration, promotes leukocyte chemotactic factor and adhesion molecule expression in vascular endothelial cells. We hypothesized that stimulating vascular endothelial cells with EMD would induce an inflammatory response in the peri-implant mucosa, enabling early peri-implant infection detection. To verify this hypothesis, we assessed the intercellular adhesion between human alveolar ridge mucosa-derived vascular endothelial cells (ARMEC) stimulated with lipopolysaccharide (LPS) and EMD and human periodontal ligament-derived vascular endothelial cells (PDLEC). Leukocyte chemotactic factors and cell adhesion molecules were investigated and we established an experimental model of peri-implant disease by stimulating ARMEC (representing the peri-implant mucosa) with Porphyromonas gingivalis-derived LPS. ARMEC and PDLEC were obtained from patients (n = 6) who visited the Nippon Dental University Niigata Hospital. The cells were divided into four subcategories, each cultured with: LPS (1 µg/mL), EMD (100 µg/mL), LPS + EMD, and pure medium. Cell viability, leukocyte chemotactic factor (interleukin-8: IL-8), adhesion molecules (intercellular adhesion molecule-1: ICAM-1), tight junction protein gene expression (zonula occludens-1: ZO-1 and Occludin), and transendothelial electrical resistance (TEER) was then determined. LPS reduced ARMEC viability, whereas simultaneous stimulation with EMD improved it. LPS and EMD stimulation enhanced IL-8 and ICAM-1 gene expression, suppressed TEER, and decreased ZO-1 and Occludin expression levels compared to that with stimulation with LPS alone. EMD stimulates leukocyte migration, increase vascular permeability, and trigger an immune response in the peri-implant mucosa, thus facilitating the early detection and treatment of peri-implant disease.
Collapse
Affiliation(s)
- Naomichi Yaita
- Field of Advanced Conservative Dentistry and Periodontology, Periodontology, Course of Clinical Science, The Nippon Dental University Graduate School Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan.
| | - Kosuke Maruyama
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Kazuhiko Hiroyasu
- Oral Implant Care Unit Niigata Hospital, The Nippon Dental University, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Soh Sato
- Field of Advanced Conservative Dentistry and Periodontology, Periodontology, Course of Clinical Science, The Nippon Dental University Graduate School Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| |
Collapse
|
10
|
Salehi A, Sprejz S, Ruehl H, Olayioye M, Cattaneo G. An imprint-based approach to replicate nano- to microscale roughness on gelatin hydrogel scaffolds: surface characterization and effect on endothelialization. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:1214-1235. [PMID: 38431849 DOI: 10.1080/09205063.2024.2322771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Biologization of biomaterials with endothelial cells (ECs) is an important step in vascular tissue engineering, aiming at improving hemocompatibility and diminishing the thrombo-inflammatory response of implants. Since subcellular topography in the scale of nano to micrometers can influence cellular adhesion, proliferation, and differentiation, we here investigate the effect of surface roughness on the endothelialization of gelatin hydrogel scaffolds. Considering the micron and sub-micron features of the different native tissues underlying the endothelium in the body, we carried out a biomimetic approach to replicate the surface roughness of tissues and analyzed how this impacted the adhesion and proliferation of human umbilical endothelial cells (HUVECs). Using an imprinting technique, nano and micro-roughness ranging from Sa= 402 nm to Sa= 8 μm were replicated on the surface of gelatin hydrogels. Fluorescent imaging of HUVECs on consecutive days after seeding revealed that microscale topographies negatively affect cell spreading and proliferation. By contrast, nanoscale roughnesses of Sa= 402 and Sa= 538 nm promoted endothelialization as evidenced by the formation of confluent cell monolayers with prominent VE-cadherin surface expression. Collectively, we present an affordable and flexible imprinting method to replicate surface characteristics of tissues on hydrogels and demonstrate how nanoscale roughness positively supports their endothelialization.
Collapse
Affiliation(s)
- Ali Salehi
- Institute of Biomedical Engineering, University of Stuttgart, Stuttgart, Germany
| | - Stefanie Sprejz
- Institute of Biomedical Engineering, University of Stuttgart, Stuttgart, Germany
| | - Holger Ruehl
- Institute for Micro Integration, University of Stuttgart, Stuttgart, Germany
| | - Monilola Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Giorgio Cattaneo
- Institute of Biomedical Engineering, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
11
|
Hu C, Lei Y, Liu X, Yu X, Geng Z, Liu Y, Yang L, Tie X, Zhou W, Li X, Zhang Y, Liang Y. Dissecting microenvironment in cystadenomas and hepatic cysts based on single nucleus RNA-sequencing data. Comput Biol Med 2024; 176:108541. [PMID: 38744012 DOI: 10.1016/j.compbiomed.2024.108541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/13/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024]
Abstract
Hepatic cystadenoma is a rare disease, accounting for about 5% of all cystic lesions, with a high tendency of malignant transformation. The preoperative diagnosis of cystadenoma is difficult, and some cystadenomas are easily misdiagnosed as hepatic cysts at first. Hepatic cyst is a relatively common liver disease, most of which are benign, but large hepatic cysts can lead to pressure on the bile duct, resulting in abnormal liver function. To better understand the difference between the microenvironment of cystadenomas and hepatic cysts, we performed single-nuclei RNA-sequencing on cystadenoma and hepatic cysts samples. In addition, we performed spatial transcriptome sequencing of hepatic cysts. Based on nucleus RNA-sequencing data, a total of seven major cell types were identified. Here we described the tumor microenvironment of cystadenomas and hepatic cysts, particularly the transcriptome signatures and regulators of immune cells and stromal cells. By inferring copy number variation, it was found that the malignant degree of hepatic stellate cells in cystadenoma was higher. Pseudotime trajectory analysis demonstrated dynamic transformation of hepatocytes in hepatic cysts and cystadenomas. Cystadenomas had higher immune infiltration than hepatic cysts, and T cells had a more complex regulatory mechanism in cystadenomas than hepatic cysts. Immunohistochemistry confirms a cystadenoma-specific T-cell immunoregulatory mechanism. These results provided a single-cell atlas of cystadenomas and hepatic cyst, revealed a more complex microenvironment in cystadenomas than in hepatic cysts, and provided new perspective for the molecular mechanisms of cystadenomas and hepatic cyst.
Collapse
Affiliation(s)
- Congxue Hu
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yongqi Lei
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xinyang Liu
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xingxin Yu
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Zhida Geng
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yu Liu
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Liyu Yang
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xuehong Tie
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Wenzhe Zhou
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xia Li
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yunpeng Zhang
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| | - Yingjian Liang
- College of the First Affiliated Hospital of Harbin Medical University, Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
12
|
Souchak J, Mohammed NBB, Lau LS, Dimitroff CJ. The role of galectins in mediating the adhesion of circulating cells to vascular endothelium. Front Immunol 2024; 15:1395714. [PMID: 38840921 PMCID: PMC11150550 DOI: 10.3389/fimmu.2024.1395714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024] Open
Abstract
Vascular cell adhesion is a complex orchestration of events that commonly feature lectin-ligand interactions between circulating cells, such as immune, stem, and tumor cells, and endothelial cells (ECs) lining post-capillary venules. Characteristically, circulating cell adherence to the vasculature endothelium is initiated through interactions between surface sialo-fucosylated glycoprotein ligands and lectins, specifically platelet (P)- or endothelial (E)-selectin on ECs or between leukocyte (L)-selectin on circulating leukocytes and L-selectin ligands on ECs, culminating in circulating cell extravasation. This lectin-ligand interplay enables the migration of immune cells into specific tissue sites to help maintain effective immunosurveillance and inflammation control, the homing of stem cells to bone marrow or tissues in need of repair, and, unfortunately, in some cases, the dissemination of circulating tumor cells (CTCs) to distant metastatic sites. Interestingly, there is a growing body of evidence showing that the family of β-galactoside-binding lectins, known as galectins, can also play pivotal roles in the adhesion of circulating cells to the vascular endothelium. In this review, we present contemporary knowledge on the significant roles of host- and/or tumor-derived galectin (Gal)-3, -8, and -9 in facilitating the adhesion of circulating cells to the vascular endothelium either directly by acting as bridging molecules or indirectly by triggering signaling pathways to express adhesion molecules on ECs. We also explore strategies for interfering with galectin-mediated adhesion to attenuate inflammation or hinder the metastatic seeding of CTCs, which are often rich in galectins and/or their glycan ligands.
Collapse
Affiliation(s)
- Joseph Souchak
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Norhan B. B. Mohammed
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
- Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Lee Seng Lau
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Charles J. Dimitroff
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| |
Collapse
|
13
|
Massey A, Stewart J, Smith C, Parvini C, McCormick M, Do K, Cartagena-Rivera AX. Mechanical properties of human tumour tissues and their implications for cancer development. NATURE REVIEWS. PHYSICS 2024; 6:269-282. [PMID: 38706694 PMCID: PMC11066734 DOI: 10.1038/s42254-024-00707-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 05/07/2024]
Abstract
The mechanical properties of cells and tissues help determine their architecture, composition and function. Alterations to these properties are associated with many diseases, including cancer. Tensional, compressive, adhesive, elastic and viscous properties of individual cells and multicellular tissues are mostly regulated by reorganization of the actomyosin and microtubule cytoskeletons and extracellular glycocalyx, which in turn drive many pathophysiological processes, including cancer progression. This Review provides an in-depth collection of quantitative data on diverse mechanical properties of living human cancer cells and tissues. Additionally, the implications of mechanical property changes for cancer development are discussed. An increased knowledge of the mechanical properties of the tumour microenvironment, as collected using biomechanical approaches capable of multi-timescale and multiparametric analyses, will provide a better understanding of the complex mechanical determinants of cancer organization and progression. This information can lead to a further understanding of resistance mechanisms to chemotherapies and immunotherapies and the metastatic cascade.
Collapse
Affiliation(s)
- Andrew Massey
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Jamie Stewart
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- These authors contributed equally: Jamie Stewart, Chynna Smith
| | - Chynna Smith
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- These authors contributed equally: Jamie Stewart, Chynna Smith
| | - Cameron Parvini
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Moira McCormick
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Kun Do
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Alexander X. Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
14
|
Maeda T, Shimamori K, Kurita H, Tokuraku K, Kuragano M. Amyloid β interferes with wound healing of brain microvascular endothelial cells by disorganizing the actin cytoskeleton. Exp Cell Res 2024; 436:113958. [PMID: 38325585 DOI: 10.1016/j.yexcr.2024.113958] [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: 11/29/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is a disease in which amyloid β (Aβ) is deposited in the cerebral blood vessels, reducing compliance, tearing and weakening of vessel walls, leading to cerebral hemorrhage. The mechanisms by which Aβ leads to focal wall fragmentation and intimal damage are not well understood. We analyzed the motility of human brain microvascular endothelial cells (hBMECs) in real-time using a wound-healing assay. We observed the suppression of cell migration by visualizing Aβ aggregation using quantum dot (QD) nanoprobes. In addition, using QD nanoprobes and a SiR-actin probe, we simultaneously observed Aβ aggregation and F-actin organization in real-time for the first time. Aβ began to aggregate at the edge of endothelial cells, reducing cell motility. In addition, Aβ aggregation disorganized the actin cytoskeleton and induced abnormal actin aggregation. Aβ aggregated actively in the anterior group, where cell motility was active. Our findings may be a first step toward explaining the mechanism by which Aβ causes vascular wall fragility, bleeding, and rebleeding in CAA.
Collapse
Affiliation(s)
- Takuma Maeda
- Graduate School of Engineering, Muroran Institute of Technology, Hokkaido, 050-8585, Japan; Department of Neurosurgery, Ohkawara Neurosurgical Hospital, Hokkaido, 050-0082, Japan; Department of Cerebrovascular Surgery, Saitama Medical University International Medical Center, Saitama, 350-1298, Japan
| | - Keiya Shimamori
- Graduate School of Engineering, Muroran Institute of Technology, Hokkaido, 050-8585, Japan
| | - Hiroki Kurita
- Department of Cerebrovascular Surgery, Saitama Medical University International Medical Center, Saitama, 350-1298, Japan
| | - Kiyotaka Tokuraku
- Graduate School of Engineering, Muroran Institute of Technology, Hokkaido, 050-8585, Japan
| | - Masahiro Kuragano
- Graduate School of Engineering, Muroran Institute of Technology, Hokkaido, 050-8585, Japan.
| |
Collapse
|
15
|
Haghayegh Jahromi N, Gkountidi AO, Collado-Diaz V, Blatter K, Bauer A, Zambounis L, Medina-Sanchez JD, Russo E, Runge P, Restivo G, Gousopoulos E, Lindenblatt N, Levesque MP, Halin C. CD112 Supports Lymphatic Migration of Human Dermal Dendritic Cells. Cells 2024; 13:424. [PMID: 38474388 DOI: 10.3390/cells13050424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/02/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Dendritic cell (DC) migration from peripheral tissues via afferent lymphatic vessels to draining lymph nodes (dLNs) is important for the organism's immune regulation and immune protection. Several lymphatic endothelial cell (LEC)-expressed adhesion molecules have thus far been found to support transmigration and movement within the lymphatic vasculature. In this study, we investigated the contribution of CD112, an adhesion molecule that we recently found to be highly expressed in murine LECs, to this process. Performing in vitro assays in the murine system, we found that transmigration of bone marrow-derived dendritic cells (BM-DCs) across or adhesion to murine LEC monolayers was reduced when CD112 was absent on LECs, DCs, or both cell types, suggesting the involvement of homophilic CD112-CD112 interactions. While CD112 was highly expressed in murine dermal LECs, CD112 levels were low in endogenous murine dermal DCs and BM-DCs. This might explain why we observed no defect in the in vivo lymphatic migration of adoptively transferred BM-DCs or endogenous DCs from the skin to dLNs. Compared to murine DCs, human monocyte-derived DCs expressed higher CD112 levels, and their migration across human CD112-expressing LECs was significantly reduced upon CD112 blockade. CD112 expression was also readily detected in endogenous human dermal DCs and LECs by flow cytometry and immunofluorescence. Upon incubating human skin punch biopsies in the presence of CD112-blocking antibodies, DC emigration from the tissue into the culture medium was significantly reduced, indicating impaired lymphatic migration. Overall, our data reveal a contribution of CD112 to human DC migration.
Collapse
Affiliation(s)
- Neda Haghayegh Jahromi
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Anastasia-Olga Gkountidi
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Victor Collado-Diaz
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Katharina Blatter
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Aline Bauer
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Lito Zambounis
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | | | - Erica Russo
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Peter Runge
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Gaetana Restivo
- Department of Dermatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Epameinondas Gousopoulos
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Nicole Lindenblatt
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| |
Collapse
|
16
|
Lechuga S, Marino-Melendez A, Naydenov NG, Zafar A, Braga-Neto MB, Ivanov AI. Regulation of Epithelial and Endothelial Barriers by Molecular Chaperones. Cells 2024; 13:370. [PMID: 38474334 PMCID: PMC10931179 DOI: 10.3390/cells13050370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
The integrity and permeability of epithelial and endothelial barriers depend on the formation of tight junctions, adherens junctions, and a junction-associated cytoskeleton. The establishment of this junction-cytoskeletal module relies on the correct folding and oligomerization of its protein components. Molecular chaperones are known regulators of protein folding and complex formation in different cellular compartments. Mammalian cells possess an elaborate chaperone network consisting of several hundred chaperones and co-chaperones. Only a small part of this network has been linked, however, to the regulation of intercellular adhesions, and the systematic analysis of chaperone functions at epithelial and endothelial barriers is lacking. This review describes the functions and mechanisms of the chaperone-assisted regulation of intercellular junctions. The major focus of this review is on heat shock protein chaperones, their co-chaperones, and chaperonins since these molecules are the focus of the majority of the articles published on the chaperone-mediated control of tissue barriers. This review discusses the roles of chaperones in the regulation of the steady-state integrity of epithelial and vascular barriers as well as the disruption of these barriers by pathogenic factors and extracellular stressors. Since cytoskeletal coupling is essential for junctional integrity and remodeling, chaperone-assisted assembly of the actomyosin cytoskeleton is also discussed.
Collapse
Affiliation(s)
- Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (S.L.); (A.M.-M.); (N.G.N.); (A.Z.); (M.B.B.-N.)
| | - Armando Marino-Melendez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (S.L.); (A.M.-M.); (N.G.N.); (A.Z.); (M.B.B.-N.)
| | - Nayden G. Naydenov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (S.L.); (A.M.-M.); (N.G.N.); (A.Z.); (M.B.B.-N.)
| | - Atif Zafar
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (S.L.); (A.M.-M.); (N.G.N.); (A.Z.); (M.B.B.-N.)
| | - Manuel B. Braga-Neto
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (S.L.); (A.M.-M.); (N.G.N.); (A.Z.); (M.B.B.-N.)
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (S.L.); (A.M.-M.); (N.G.N.); (A.Z.); (M.B.B.-N.)
| |
Collapse
|
17
|
Di Russo S, Liberati FR, Riva A, Di Fonzo F, Macone A, Giardina G, Arese M, Rinaldo S, Cutruzzolà F, Paone A. Beyond the barrier: the immune-inspired pathways of tumor extravasation. Cell Commun Signal 2024; 22:104. [PMID: 38331871 PMCID: PMC10851599 DOI: 10.1186/s12964-023-01429-1] [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: 11/06/2023] [Accepted: 12/08/2023] [Indexed: 02/10/2024] Open
Abstract
Extravasation is a fundamental step in the metastatic journey, where cancer cells exit the bloodstream and breach the endothelial cell barrier to infiltrate target tissues. The tactics cancer cells employ are sophisticated, closely reflecting those used by the immune system for tissue surveillance. Remarkably, tumor cells have been observed to form distinct associations or clusters with immune cells where neutrophils stand out as particularly crucial partners. These interactions are not accidental; they are critical for cancer cells to exploit the immune functions of neutrophils and successfully extravasate. In another strategy, tumor cells mimic the behavior and characteristics of immune cells. They release a suite of inflammatory mediators, which under normal circumstances, guide the processes of endothelium reshaping and facilitate the entry and movement of immune cells within tissues. In this review, we offer a new perspective on the tactics employed by cancer cells to extravasate and infiltrate target tissues. We delve into the myriad mechanisms that tumor cells borrow, adapt, and refine from the immune playbook. Video Abstract.
Collapse
Affiliation(s)
- Sara Di Russo
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Francesca Romana Liberati
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Agnese Riva
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Federica Di Fonzo
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Alberto Macone
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Giorgio Giardina
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Marzia Arese
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Serena Rinaldo
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Francesca Cutruzzolà
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Alessio Paone
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy.
| |
Collapse
|
18
|
Kameni LE, Januszyk M, Berry CE, Downer MA, Parker JB, Morgan AG, Valencia C, Griffin M, Li DJ, Liang NE, Momeni A, Longaker MT, Wan DC. A Review of Radiation-Induced Vascular Injury and Clinical Impact. Ann Plast Surg 2024; 92:181-185. [PMID: 37962260 DOI: 10.1097/sap.0000000000003723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
ABSTRACT The number of cancer survivors continues to increase because of advances in therapeutic modalities. Along with surgery and chemotherapy, radiotherapy is a commonly used treatment modality in roughly half of all cancer patients. It is particularly helpful in the oncologic treatment of patients with breast, head and neck, and prostate malignancies. Unfortunately, among patients receiving radiation therapy, long-term sequalae are often unavoidable, and there is accumulating clinical evidence suggesting significant radiation-related damage to the vascular endothelium. Ionizing radiation has been known to cause obliterative fibrosis and increased wall thickness in irradiated blood vessels. Clinically, these vascular changes induced by ionizing radiation can pose unique surgical challenges when operating in radiated fields. Here, we review the relevant literature on radiation-induced vascular damage focusing on mechanisms and signaling pathways involved and highlight microsurgical anastomotic outcomes after radiotherapy. In addition, we briefly comment on potential therapeutic strategies, which may have the ability to mitigate radiation injury to the vascular endothelium.
Collapse
Affiliation(s)
- Lionel E Kameni
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Michael Januszyk
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Mauricio A Downer
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Jennifer B Parker
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Annah G Morgan
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Caleb Valencia
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Michelle Griffin
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Dayan J Li
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Norah E Liang
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Arash Momeni
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Derrick C Wan
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
19
|
Culkin MC, Coons M, Bele P, Thaploo A, Georges AP, Anderson E, Browne KD, Jacovides C, Santos P, Kaplan LJ, Meaney DF, Smith DH, Pascual JL. Delayed tranexamic acid after traumatic brain injury impedes learning and memory: Early tranexamic acid is favorable but not in sham animals. J Trauma Acute Care Surg 2024; 96:26-34. [PMID: 37853567 DOI: 10.1097/ta.0000000000004155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
BACKGROUND Early but not late tranexamic acid (TXA) after TBI preserves blood-brain-barrier integrity, but it is unclear if and how dose timing affects cognitive recovery beyond hours postinjury. We hypothesized that early (1 hour post-TBI) but not late (24 hours post-TBI) TXA administration improves cognitive recovery for 14 days. METHODS CD1 male mice (n = 25) were randomized to severe TBI (injury [I], by controlled cortical impact) or sham craniotomy (S) followed by intravenous saline at 1 hour (placebo [P1]) or 30 mg/kg TXA at 1 hour (TXA1) or 24 hours (TXA24). Daily body weights, Garcia Neurological Test scores, brain/lung water content, and Morris water maze exercises quantifying swimming traffic in the platform quadrant (zone [Z] 1) and platform area (Z5) were recorded for up to 14 days. RESULTS Among injured groups, I-TXA1 demonstrated fastest weight gain for 14 days and only I-TXA1 showed rapid (day 1) normalization of Garcia Neurological Test ( p = 0.01 vs. I-P1, I-TXA24). In cumulative spatial trials, compared with I-TXA1, I-TXA24 hindered learning (distance to Z5 and % time in Z1, p < 0.05). Compared with I-TXA1, I-TXA24 showed poorer memory with less Z5 time (0.51 vs. 0.16 seconds, p < 0.01) and Z5 crossing frequency. Unexpectedly, TXA in uninjured animals (S-TXA1) displayed faster weight gain but inferior learning and memory. CONCLUSION Early TXA appears beneficial for cognitive and behavioral outcomes following TBI, although administration 24 hours postinjury consistently impairs cognitive recovery. Tranexamic acid in sham animals may lead to adverse effects on cognition.
Collapse
Affiliation(s)
- Matthew C Culkin
- From the Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery (M.C.C., M.C., P.B., A.T., C.J., P.S., L.J.K., J.L.P.), and Center for Brain Injury and Repair, Department of Neurosurgery (M.C.C., M.C., P.B., A.T., A.P.G., E.A., K.D.B., C.J., P.S., L.J.K., D.F.M., D.H.S., J.L.P.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Choi JS, Doo HM, Kim B, Lee SH, Sung S, Go G, Suarez A, Kim Y, Weon BM, Choi B, Kim HJ, Kim D. NanoIEA: A Nanopatterned Interdigitated Electrode Array-Based Impedance Assay for Real-Time Measurement of Aligned Endothelial Cell Barrier Functions. Adv Healthc Mater 2024; 13:e2301124. [PMID: 37820720 PMCID: PMC10841753 DOI: 10.1002/adhm.202301124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/18/2023] [Indexed: 10/13/2023]
Abstract
A nanopatterned interdigitated electrode array (nanoIEA)-based impedance assay is developed for quantitative real-time measurement of aligned endothelial cell (EC) barrier functions in vitro. A bioinspired poly(3,4-dihydroxy-L-phenylalanine) (poly (l-DOPA)) coating is applied to improve the human brain EC adhesion onto the Nafion nanopatterned surfaces. It is found that a poly (l-DOPA)-coated Nafion grooved nanopattern makes the human brain ECs orient along the nanopattern direction. Aligned human brain ECs on Nafion nanopatterns exhibit increased expression of genes encoding tight and adherens junction proteins. Aligned human brain ECs also have enhanced impedance and resistance versus unaligned ones. Treatment with a glycogen synthase kinase-3 inhibitor (GSK3i) further increases impedance and resistance, suggesting synergistic effects occur on the cell-cell tightness of in vitro human brain ECs via a combination of anisotropic matrix nanotopography and GSK3i treatment. It is found that this enhanced cell-cell tightness of the combined approach is accompanied by increased expression of claudin protein. These data demonstrate that the proposed nanoIEA assay integrated with poly (l-DOPA)-coated Nafion nanopatterns and interdigitated electrode arrays can make not only biomimetic aligned ECs, but also enable real-time measurement of the enhanced barrier functions of aligned ECs via tighter cell-cell junctions.
Collapse
Affiliation(s)
- Jong Seob Choi
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
- Division of Advanced Materials EngineeringKongju National UniversityCheonanChungnam31080South Korea
| | - Hyun Myung Doo
- Department of Health Sciences and TechnologySAIHSTSungkyunkwan UniversitySeoul06351South Korea
- Department of Biomedical Research CenterKorea University Guro HospitalSeoul08308South Korea
- Division of Medical Oncology, Department of Internal MedicineKorea University Guro Hospital, Korea University College of MedicineSeoul08308South Korea
| | - Byunggik Kim
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Su Han Lee
- Digital Health Care Research CenterGumi Electronics and Information Technology Research Institute (GERI)GumiGyeongbuk39253South Korea
| | - Sang‐keun Sung
- Digital Health Care Research CenterGumi Electronics and Information Technology Research Institute (GERI)GumiGyeongbuk39253South Korea
| | - Gwangjun Go
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
- Department of Mechanical EngineeringChosun UniversityGwangju61452South Korea
| | - Allister Suarez
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
| | - Yeseul Kim
- SKKU Advanced Institute of Nanotechnology (SAINT)School of Advanced Materials Science and EngineeringSungkyunkwan UniversitySuwon16419South Korea
| | - Byung Mook Weon
- SKKU Advanced Institute of Nanotechnology (SAINT)School of Advanced Materials Science and EngineeringSungkyunkwan UniversitySuwon16419South Korea
| | - Byung‐Ok Choi
- Department of Health Sciences and TechnologySAIHSTSungkyunkwan UniversitySeoul06351South Korea
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineSeoul06351South Korea
| | - Hyung Jin Kim
- School of Electrical and Electronic EngineeringUlsan CollegeUlsan44610South Korea
| | - Deok‐Ho Kim
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMD21218USA
| |
Collapse
|
21
|
Okada Y. Potential Therapeutic Strategies and Drugs That Target Vascular Permeability in Severe Infectious Diseases. Biol Pharm Bull 2024; 47:549-555. [PMID: 38432910 DOI: 10.1248/bpb.b24-00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Severe infection pathogenicity is induced by processes such as pathogen exposure, immune cell activation, inflammatory cytokine production, and vascular hyperpermeability. Highly effective drugs, such as antipathogenic agents, steroids, and antibodies that suppress cytokine function, have been developed to treat the first three processes. However, these drugs cannot completely suppress severe infectious diseases, such as coronavirus disease 2019 (COVID-19). Therefore, developing novel drugs that inhibit vascular hyperpermeability is crucial. This review summarizes the mechanisms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced vascular hyperpermeability and identifies inhibitors that increase endothelial cell (EC) junction-related proteins and determines their efficacy in COVID-19 and endotoxemia models. Analyzing the effects of SARS-CoV-2 on vascular permeability revealed that SARS-CoV-2 suppresses Claudin-5 (CLDN5) expression, which is responsible for adhesion between ECs, thereby increasing vascular permeability. Inhibiting CLDN5 function in mice induced vascular hyperpermeability and pulmonary edema. In contrast, Enhancing CLDN5 expression suppressed SARS-CoV-2-induced endothelial hyperpermeability, suggesting that SARS-CoV-2-induced vascular hyperpermeability contributes to pathological progression, which can be suppressed by upregulating EC junction proteins. Based on these results, we focused on Roundabout4 (Robo4), another EC-specific protein that stabilizes EC junctions. EC-specific Robo4 overexpression suppressed vascular hyperpermeability and mortality in lipopolysaccharide-treated mice. An ALK1 inhibitor (a molecule that increases Robo4 expression), suppressed vascular hyperpermeability and mortality in lipopolysaccharide- and SARS-CoV-2-treated mice. These results indicate that Robo4 expression-increasing drugs suppress vascular permeability and pathological phenotype in COVID-19 and endotoxemia models.
Collapse
Affiliation(s)
- Yoshiaki Okada
- Graduate School of Pharmaceutical Sciences, Osaka University
| |
Collapse
|
22
|
Zhu X, Bai H, Liu H, Wang Z, Wang Y, Zhang J, Liu J, Wang H, Wang J. A variable mineralization time and solution concentration intervene in the microstructure of biomimetic mineralized collagen and potential osteogenic microenvironment. Front Bioeng Biotechnol 2023; 11:1267912. [PMID: 38125304 PMCID: PMC10731298 DOI: 10.3389/fbioe.2023.1267912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
The absence of a conducive bone formation microenvironment between fractured ends poses a significant challenge in repairing large bone defects. A promising solution is to construct a bone formation microenvironment that mimics natural bone tissue. Biomimetic mineralized collagen possesses a chemical composition and microstructure highly similar to the natural bone matrix, making it an ideal biomimetic bone substitute material. The microstructure of biomimetic mineralized collagen is influenced by various factors, and its biomineralization and microstructure, in turn, affect its physicochemical properties and biological activity. We aimed to utilize mineralization time and solution concentration as variables and employed the polymer-induced liquid precursor strategy to fabricate mineralized collagen with diverse microstructures, to shed light on how mineralization parameters impact the material microstructure and physicochemical properties. We also investigated the influence of microstructure and physicochemical properties on cell biocompatibility and the bone-forming microenvironment. Through comprehensive characterization, we examined the physical and chemical properties of I-EMC under various mineralization conditions and assessed the in vitro and in vivo biocompatibility and osteogenic performance. By investigating the relationship between mineralization parameters, material physicochemical properties, and osteogenic performance, we revealed how microstructures influence cellular behaviors like biocompatibility and osteogenic microenvironment. Encouragingly, mineralization solutions with varying concentrations, stabilized by polyacrylic acid, successfully produced intrafibrillar and extrafibrillar mineralized collagen. Compared to non-mineralized collagen, all mineralized samples demonstrated improved bone-forming performance. Notably, samples prepared with a 1× mineralization solution exhibited relatively smooth surfaces with even mineralization. Extending the mineralization time enhanced the degree of mineralization and osteogenic performance. Conversely, samples prepared with a 2× mineralization solution had rough surfaces with large calcium phosphate particles, indicating non-uniform mineralization. Overall, our research advances the potential for commercial production of mineralized collagen protein products, characterized by dual biomimetic properties, and their application in treating various types of bone defects.
Collapse
Affiliation(s)
- Xiujie Zhu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Haotian Bai
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - He Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Zhonghan Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Yao Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Jiaxin Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Jiaqi Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Hui Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Jincheng Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
| |
Collapse
|
23
|
Song L, Shi X, Kovacs L, Han W, John J, Barman SA, Dong Z, Lucas R, Fulton DJR, Verin AD, Su Y. Calpain Promotes LPS-induced Lung Endothelial Barrier Dysfunction via Cleavage of Talin. Am J Respir Cell Mol Biol 2023; 69:678-688. [PMID: 37639326 PMCID: PMC10704117 DOI: 10.1165/rcmb.2023-0009oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023] Open
Abstract
Acute lung injury (ALI) is characterized by lung vascular endothelial cell (EC) barrier compromise resulting in increased endothelial permeability and pulmonary edema. The infection of gram-negative bacteria that produce toxins like LPS is one of the major causes of ALI. LPS activates Toll-like receptor 4, leading to cytoskeleton reorganization, resulting in lung endothelial barrier disruption and pulmonary edema in ALI. However, the signaling pathways that lead to the cytoskeleton reorganization and lung microvascular EC barrier disruption remain largely unexplored. Here we show that LPS induces calpain activation and talin cleavage into head and rod domains and that inhibition of calpain attenuates talin cleavage, RhoA activation, and pulmonary EC barrier disruption in LPS-treated human lung microvascular ECs in vitro and lung EC barrier disruption and pulmonary edema induced by LPS in ALI in vivo. Moreover, overexpression of calpain causes talin cleavage and RhoA activation, myosin light chain (MLC) phosphorylation, and increases in actin stress fiber formation. Furthermore, knockdown of talin attenuates LPS-induced RhoA activation and MLC phosphorylation and increased stress fiber formation and mitigates LPS-induced lung microvascular endothelial barrier disruption. Additionally, overexpression of talin head and rod domains increases RhoA activation, MLC phosphorylation, and stress fiber formation and enhances lung endothelial barrier disruption. Finally, overexpression of cleavage-resistant talin mutant reduces LPS-induced increases in MLC phosphorylation in human lung microvascular ECs and attenuates LPS-induced lung microvascular endothelial barrier disruption. These results provide the first evidence that calpain mediates LPS-induced lung microvascular endothelial barrier disruption in ALI via cleavage of talin.
Collapse
Affiliation(s)
| | | | - Laszlo Kovacs
- Department of Pharmacology & Toxicology
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | | | - Joseph John
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | | | - Zheng Dong
- Department of Cellular Biology and Anatomy, and
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Rudolf Lucas
- Department of Pharmacology & Toxicology
- Department of Medicine
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | - David J. R. Fulton
- Department of Pharmacology & Toxicology
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | - Alexander D. Verin
- Department of Medicine
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | - Yunchao Su
- Department of Pharmacology & Toxicology
- Department of Medicine
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| |
Collapse
|
24
|
Stierschneider A, Wiesner C. Shedding light on the molecular and regulatory mechanisms of TLR4 signaling in endothelial cells under physiological and inflamed conditions. Front Immunol 2023; 14:1264889. [PMID: 38077393 PMCID: PMC10704247 DOI: 10.3389/fimmu.2023.1264889] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Toll-like receptor 4 (TLR4) are part of the innate immune system. They are capable of recognizing pathogen-associated molecular patterns (PAMPS) of microbes, and damage-associated molecular patterns (DAMPs) of damaged tissues. Activation of TLR4 initiates downstream signaling pathways that trigger the secretion of cytokines, type I interferons, and other pro-inflammatory mediators that are necessary for an immediate immune response. However, the systemic release of pro-inflammatory proteins is a powerful driver of acute and chronic inflammatory responses. Over the past decades, immense progress has been made in clarifying the molecular and regulatory mechanisms of TLR4 signaling in inflammation. However, the most common strategies used to study TLR4 signaling rely on genetic manipulation of the TLR4 or the treatment with agonists such as lipopolysaccharide (LPS) derived from the outer membrane of Gram-negative bacteria, which are often associated with the generation of irreversible phenotypes in the target cells or unintended cytotoxicity and signaling crosstalk due to off-target or pleiotropic effects. Here, optogenetics offers an alternative strategy to control and monitor cellular signaling in an unprecedented spatiotemporally precise, dose-dependent, and non-invasive manner. This review provides an overview of the structure, function and signaling pathways of the TLR4 and its fundamental role in endothelial cells under physiological and inflammatory conditions, as well as the advances in TLR4 modulation strategies.
Collapse
Affiliation(s)
| | - Christoph Wiesner
- Department Science & Technology, Institute Biotechnology, IMC Krems University of Applied Sciences, Krems, Austria
| |
Collapse
|
25
|
Teichmann E, Blessing E, Hinz B. Non-Psychoactive Phytocannabinoids Inhibit Inflammation-Related Changes of Human Coronary Artery Smooth Muscle and Endothelial Cells. Cells 2023; 12:2389. [PMID: 37830604 PMCID: PMC10571842 DOI: 10.3390/cells12192389] [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: 08/18/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
Atherosclerosis is associated with vascular smooth muscle cell proliferation, chronic vascular inflammation, and leukocyte adhesion. In view of the cardioprotective effects of cannabinoids described in recent years, the present study investigated the impact of the non-psychoactive phytocannabinoids cannabidiol (CBD) and tetrahydrocannabivarin (THCV) on proliferation and migration of human coronary artery smooth muscle cells (HCASMC) and on inflammatory markers in human coronary artery endothelial cells (HCAEC). In HCASMC, CBD and THCV at nontoxic concentrations exhibited inhibitory effects on platelet-derived growth factor-triggered proliferation (CBD) and migration (CBD, THCV). When interleukin (IL)-1β- and lipopolysaccharide (LPS)-stimulated HCAEC were examined, both cannabinoids showed a concentration-dependent decrease in the expression of vascular cell adhesion molecule-1 (VCAM-1), which was mediated independently of classical cannabinoid receptors and was not accompanied by a comparable inhibition of intercellular adhesion molecule-1. Further inhibitor experiments demonstrated that reactive oxygen species, p38 mitogen-activated protein kinase activation, histone deacetylase, and nuclear factor κB (NF-κB) underlie IL-1β- and LPS-induced expression of VCAM-1. In this context, CBD and THCV were shown to inhibit phosphorylation of NF-κB regulators in LPS- but not IL-1β-stimulated HCAEC. Stimulation of HCAEC with IL-1β and LPS was associated with increased adhesion of monocytes, which, however, could not be significantly abolished by CBD and THCV. In summary, the results highlight the potential of the non-psychoactive cannabinoids CBD and THCV to regulate inflammation-related changes in HCASMC and HCAEC. Considering their effect on both cell types studied, further preclinical studies could address the use of CBD and THCV in drug-eluting stents for coronary interventions.
Collapse
Affiliation(s)
| | | | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany; (E.T.); (E.B.)
| |
Collapse
|
26
|
Smolinska A, Bzinkowska A, Rybkowska P, Chodkowska M, Sarnowska A. Promising Markers in the Context of Mesenchymal Stem/Stromal Cells Subpopulations with Unique Properties. Stem Cells Int 2023; 2023:1842958. [PMID: 37771549 PMCID: PMC10533301 DOI: 10.1155/2023/1842958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/11/2023] [Accepted: 08/25/2023] [Indexed: 09/30/2023] Open
Abstract
The heterogeneity of the mesenchymal stem/stromal cells (MSCs) population poses a challenge to researchers and clinicians, especially those observed at the population level. What is more, the lack of precise evidences regarding MSCs developmental origin even further complicate this issue. As the available evidences indicate several possible pathways of MSCs formation, this diverse origin may be reflected in the unique subsets of cells found within the MSCs population. Such populations differ in specialization degree, proliferation, and immunomodulatory properties or exhibit other additional properties such as increased angiogenesis capacity. In this review article, we attempted to identify such outstanding populations according to the specific surface antigens or intracellular markers. Described groups were characterized depending on their specialization and potential therapeutic application. The reports presented here cover a wide variety of properties found in the recent literature, which is quite scarce for many candidates mentioned in this article. Even though the collected information would allow for better targeting of specific subpopulations in regenerative medicine to increase the effectiveness of MSC-based therapies.
Collapse
Affiliation(s)
- Agnieszka Smolinska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Aleksandra Bzinkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Paulina Rybkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Magdalena Chodkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| |
Collapse
|
27
|
Fang J, Lu Y, Zheng J, Jiang X, Shen H, Shang X, Lu Y, Fu P. Exploring the crosstalk between endothelial cells, immune cells, and immune checkpoints in the tumor microenvironment: new insights and therapeutic implications. Cell Death Dis 2023; 14:586. [PMID: 37666809 PMCID: PMC10477350 DOI: 10.1038/s41419-023-06119-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/19/2023] [Accepted: 08/25/2023] [Indexed: 09/06/2023]
Abstract
The tumor microenvironment (TME) is a highly intricate milieu, comprising a multitude of components, including immune cells and stromal cells, that exert a profound influence on tumor initiation and progression. Within the TME, angiogenesis is predominantly orchestrated by endothelial cells (ECs), which foster the proliferation and metastasis of malignant cells. The interplay between tumor and immune cells with ECs is complex and can either bolster or hinder the immune system. Thus, a comprehensive understanding of the intricate crosstalk between ECs and immune cells is essential to advance the development of immunotherapeutic interventions. Despite recent progress, the underlying molecular mechanisms that govern the interplay between ECs and immune cells remain elusive. Nevertheless, the immunomodulatory function of ECs has emerged as a pivotal determinant of the immune response. In light of this, the study of the relationship between ECs and immune checkpoints has garnered considerable attention in the field of immunotherapy. By targeting specific molecular pathways and signaling molecules associated with ECs in the TME, novel immunotherapeutic strategies may be devised to enhance the efficacy of current treatments. In this vein, we sought to elucidate the relationship between ECs, immune cells, and immune checkpoints in the TME, with the ultimate goal of identifying novel therapeutic targets and charting new avenues for immunotherapy.
Collapse
Affiliation(s)
- Jianwen Fang
- Department of Breast Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China
| | - Yue Lu
- Department of Breast and Thyroid Surgery, First Affiliated Hospital of Huzhou University, 313000, Huzhou, China
| | - Jingyan Zheng
- Department of Breast and Thyroid Surgery, Lishui People's Hospital, The Six Affiliated Hospital of Wenzhou Medical University, 323000, Lishui, China
| | - Xiaocong Jiang
- Department of Breast Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China
| | - Haixing Shen
- Department of Breast Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China
- Department of Breast and Thyroid Surgery, Cixi People's Hospital, 315300, Cixi, China
| | - Xi Shang
- Department of Breast and Thyroid Surgery, Taizhou Hospital, Zhejiang University, 318000, Taizhou, China
| | - Yuexin Lu
- Department of Breast Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China
| | - Peifen Fu
- Department of Breast Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China.
| |
Collapse
|
28
|
Tyagi K, Rai P, Gautam A, Kaur H, Kapoor S, Suttee A, Jaiswal PK, Sharma A, Singh G, Barnwal RP. Neurological manifestations of SARS-CoV-2: complexity, mechanism and associated disorders. Eur J Med Res 2023; 28:307. [PMID: 37649125 PMCID: PMC10469568 DOI: 10.1186/s40001-023-01293-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Coronaviruses such as Severe Acute Respiratory Syndrome coronavirus (SARS), Middle Eastern Respiratory Syndrome (MERS) and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) are associated with critical illnesses, including severe respiratory disorders. SARS-CoV-2 is the causative agent of the deadly COVID-19 illness, which has spread globally as a pandemic. SARS-CoV-2 may enter the human body through olfactory lobes and interact with the angiotensin-converting enzyme2 (ACE2) receptor, further facilitating cell binding and entry into the cells. Reports have shown that the virus can pass through the blood-brain barrier (BBB) and enter the central nervous system (CNS), resulting in various disorders. Cell entry by SARS-CoV-2 largely relies on TMPRSS2 and cathepsin L, which activate S protein. TMPRSS2 is found on the cell surface of respiratory, gastrointestinal and urogenital epithelium, while cathepsin-L is a part of endosomes. AIM The current review aims to provide information on how SARS-CoV-2 infection affects brain function.. Furthermore, CNS disorders associated with SARS-CoV-2 infection, including ischemic stroke, cerebral venous thrombosis, Guillain-Barré syndrome, multiple sclerosis, meningitis, and encephalitis, are discussed. The many probable mechanisms and paths involved in developing cerebrovascular problems in COVID patients are thoroughly detailed. MAIN BODY There have been reports that the SARS-CoV-2 virus can cross the blood-brain barrier (BBB) and enter the central nervous system (CNS), where it could cause a various illnesses. Patients suffering from COVID-19 experience a range of neurological complications, including sleep disorders, viral encephalitis, headaches, dysgeusia, and cognitive impairment. The presence of SARS-CoV-2 in the cerebrospinal fluid (CSF) of COVID-19 patients has been reported. Health experts also reported its presence in cortical neurons and human brain organoids. The possible mechanism of virus infiltration into the brain can be neurotropic, direct infiltration and cytokine storm-based pathways. The olfactory lobes could also be the primary pathway for the entrance of SARS-CoV-2 into the brain. CONCLUSIONS SARS-CoV-2 can lead to neurological complications, such as cerebrovascular manifestations, motor movement complications, and cognitive decline. COVID-19 infection can result in cerebrovascular symptoms and diseases, such as strokes and thrombosis. The virus can affect the neural system, disrupt cognitive function and cause neurological disorders. To combat the epidemic, it is crucial to repurpose drugs currently in use quickly and develop novel therapeutics.
Collapse
Affiliation(s)
- Kritika Tyagi
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Prachi Rai
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Anuj Gautam
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Harjeet Kaur
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Sumeet Kapoor
- Centre for Biomedical Engineering, Indian Institute of Technology, New Delhi, India
| | - Ashish Suttee
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Pradeep Kumar Jaiswal
- Department of Biochemistry and Biophysics, Texas A & M University, College Station, TX, 77843, USA
| | - Akanksha Sharma
- Department of Biophysics, Panjab University, Chandigarh, India.
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| | | |
Collapse
|
29
|
Nan W, He Y, Wang S, Zhang Y. Molecular mechanism of VE-cadherin in regulating endothelial cell behaviour during angiogenesis. Front Physiol 2023; 14:1234104. [PMID: 37601629 PMCID: PMC10433914 DOI: 10.3389/fphys.2023.1234104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Vascular endothelial (VE)-cadherin, an endothelium-specific adhesion protein, is found in the junctions between endothelial cells (ECs). It's crucial to maintain the homogeneity of ECs. Keeping and controlling the contact between ECs is essential. In addition to its adhesive function, VE-cadherin plays important roles in vascular development, permeability, and tumour angiogenesis. Signal transfer, cytoskeletal reconstruction, and contractile integrating, which are crucial for constructing and maintaining monolayer integrity as well as for repair and regeneration, are the foundation of endothelial cell (EC) junctional dynamics. The molecular basis of adhesion junctions (AJs), which are closely related and work with actin filaments, is provided by the VE-cadherin-catenin complex. They can activate intracellular signals that drive ECs to react or communicate structural changes to junctions. An increasing number of molecules, including the vascular endothelial growth factor receptor 2 (VEGFR2) and vascular endothelial protein tyrosine phosphatase (VE-PTP), have been connected to VE-cadherin in addition to the conventional VE-cadherin-catenin complex. This review demonstrates significant progress in our understanding of the molecular mechanisms that affect VE-cadherin's function in the regulation of EC behaviour during angiogenesis. The knowledge of the molecular processes that control VE-cadherin's role in the regulation of EC behaviour during angiogenesis has recently advanced, as shown in this review.
Collapse
Affiliation(s)
- Weijin Nan
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Yuxi He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Shurong Wang
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yan Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
van der Krogt JMA, van der Meulen IJE, van Buul JD. Spatiotemporal regulation of Rho GTPase signaling during endothelial barrier remodeling. CURRENT OPINION IN PHYSIOLOGY 2023; 34:None. [PMID: 37547802 PMCID: PMC10398679 DOI: 10.1016/j.cophys.2023.100676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The vasculature is characterized by a thin cell layer that comprises the inner wall of all blood vessels, the continuous endothelium. Endothelial cells can also be found in the eye's cornea. And even though cornea and vascular endothelial (VE) cells differ from each other in structure, they both function as barriers and express similar junctional proteins such as the adherens junction VE-cadherin and tight-junction member claudin-5. How these barriers are controlled to maintain the barrier and thereby its integrity is of major interest in the development of potential therapeutic targets. An important target of endothelial barrier remodeling is the actin cytoskeleton, which is centrally coordinated by Rho GTPases that are in turn regulated by Rho-regulatory proteins. In this review, we give a brief overview of how Rho-regulatory proteins themselves are spatiotemporally regulated during the process of endothelial barrier remodeling. Additionally, we propose a roadmap for the comprehensive dissection of the Rho GTPase signaling network in its entirety.
Collapse
Affiliation(s)
| | | | - Jaap D van Buul
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, the Netherlands
- Leeuwenhoek Centre for Advanced Microscopy, section Molecular Cytology at Swammerdam Institute for Life Sciences at the University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
31
|
Greer JB, Dalsky EM, Lane RF, Hansen JD. Tire-Derived Transformation Product 6PPD-Quinone Induces Mortality and Transcriptionally Disrupts Vascular Permeability Pathways in Developing Coho Salmon. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:10940-10950. [PMID: 37467138 PMCID: PMC10399305 DOI: 10.1021/acs.est.3c01040] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/25/2023] [Accepted: 06/23/2023] [Indexed: 07/21/2023]
Abstract
Urban stormwater runoff frequently contains the car tire transformation product 6PPD-quinone, which is highly toxic to juvenile and adult coho salmon (Onchorychus kisutch). However, it is currently unclear if embryonic stages are impacted. We addressed this by exposing developing coho salmon embryos starting at the eyed stage to three concentrations of 6PPD-quinone twice weekly until hatch. Impacts on survival and growth were assessed. Further, whole-transcriptome sequencing was performed on recently hatched alevin to address the potential mechanism of 6PPD-quinone-induced toxicity. Acute mortality was not elicited in developing coho salmon embryos at environmentally measured concentrations lethal to juveniles and adults, however, growth was inhibited. Immediately after hatching, coho salmon were sensitive to 6PPD-quinone mortality, implicating a large window of juvenile vulnerability prior to smoltification. Molecularly, 6PPD-quinone induced dose-dependent effects that implicated broad dysregulation of genomic pathways governing cell-cell contacts and endothelial permeability. These pathways are consistent with previous observations of macromolecule accumulation in the brains of coho salmon exposed to 6PPD-quinone, implicating blood-brain barrier disruption as a potential pathway for toxicity. Overall, our data suggests that developing coho salmon exposed to 6PPD-quinone are at risk for adverse health events upon hatching while indicating potential mechanism(s) of action for this highly toxic chemical.
Collapse
Affiliation(s)
- Justin B. Greer
- U.S.
Geological Survey, Western Fisheries Research
Center, Seattle, Washington 98115, United States
| | - Ellie M. Dalsky
- U.S.
Geological Survey, Western Fisheries Research
Center, Seattle, Washington 98115, United States
| | - Rachael F. Lane
- U.S.
Geological Survey, Kansas Water Science
Center, Lawrence, Kansas 66049, United States
| | - John D. Hansen
- U.S.
Geological Survey, Western Fisheries Research
Center, Seattle, Washington 98115, United States
| |
Collapse
|
32
|
Chu Q, Song X, Xiao Y, Kang YJ. Alteration of endothelial permeability ensures cardiomyocyte survival from ischemic insult in the subendocardium of the heart. Exp Biol Med (Maywood) 2023; 248:1364-1372. [PMID: 37786370 PMCID: PMC10657589 DOI: 10.1177/15353702231194344] [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: 11/08/2022] [Accepted: 05/12/2023] [Indexed: 10/04/2023] Open
Abstract
Previous studies have shown that cardiomyocytes in the subendocardial region of myocardium survive from ischemic insult. This study was undertaken to explore possible mechanisms for the survival of these cardiomyocytes, focusing on changes in endothelial cells (ECs) and blood supply. C57/B6 mice were subjected to permanent ligation of left anterior descending (LAD) coronary artery to induce myocardial ischemia (MI). The hearts were harvested at 1, 4, and 7 days post MI and examined for histological changes. It was found that the survival of cardiomyocytes was associated with a preservation of ECs in the subendocardial region, as revealed by EC-specific tdTomato expression transgenic mice (Tie2tdTomato). However, the EC selective proteins, PECAM1 and VEGFR2, were significantly depressed in these ECs. Consequently, the ratio of PECAM1/tdTomato was significantly decreased, indicating a transformation from PECAM1+ ECs to PECAM1- ECs. Furthermore, EC junction protein, VE-cadherin, was not only depressed but also disassociated from PECAM1 in the same region. These changes led to an increase in EC permeability, as evidenced by increased blood infiltration in the subendocardial region. Thus, the increase in the permeability of ECs due to their transformation in the subendocardial region allows blood infiltration, creating a unique microenvironment and ensuring the survival of cardiomyocytes under ischemic conditions.
Collapse
Affiliation(s)
- Qing Chu
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China
| | - Xin Song
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China
| | - Ying Xiao
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China
| | - Y James Kang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China
- Tennessee Institute of Regenerative Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
33
|
Mulorz J, Ibing W, Cappallo M, Braß SM, Takeuchi K, Raaz U, Schellinger IN, Krott KJ, Schelzig H, Aubin H, Oberhuber A, Elvers M, Wagenhäuser MU. Ethanol Enhances Endothelial Rigidity by Targeting VE-Cadherin-Implications for Acute Aortic Dissection. J Clin Med 2023; 12:4967. [PMID: 37568369 PMCID: PMC10420172 DOI: 10.3390/jcm12154967] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
(1) Background: Acute aortic dissection (AAD) is caused by an endothelial entry tear followed by intimomedial delamination of the outer layers of the vessel wall. The established risk factors include hypertension and smoking. Another rising candidate risk factor is excessive alcohol consumption. This experimental study explores the effects of nicotine (Nic), angiotensin II (Ang II), and ethanol (EtOH) on human aortic endothelial cells (hAoEC). (2) Methods: HAoECs were exposed to Nic, Ang II, and EtOH at different dose levels. Cell migration was studied using the scratch assay and live-cell imaging. The metabolic viability and permeability capacity was investigated using the water-soluble tetrazolium (WST)-1 assay and an in vitro vascular permeability assay. Cell adherence was studied by utilizing the hanging drop assay. The transcriptional and protein level changes were analyzed by RT-qPCR, Western blotting and immunohistochemistry for major junctional complexing proteins. (3) Results: We observed reduced metabolic viability following Ang II and EtOH exposure vs. control. Further, cell adherence was enhanced by EtOH exposure prior to trituration and by all risk factors after trituration, which correlated with the increased gene and protein expression of VE-cadherin upon EtOH exposure. The cell migration capacity was reduced upon EtOH exposure vs. controls. (4) Conclusion: Marked functional changes were observed upon exposure to established and potential risk factors for AAD development in hAoECs. Our findings advocate for an enhanced mechanical rigidity in hAoECs in response to the three substances studied, which in turn might increase endothelial rigidity, suggesting a novel mechanism for developing an endothelial entry tear due to reduced deformability in response to increased shear and pulsatile stress.
Collapse
Affiliation(s)
- Joscha Mulorz
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Wiebke Ibing
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Melanie Cappallo
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
- Clinic for Cardiac Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
- CURE3D Lab, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Sönke Maximilian Braß
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Kiku Takeuchi
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Uwe Raaz
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site, 37075 Göttingen, Germany
- University Heart Center, 37075 Göttingen, Germany
| | - Isabel Nahal Schellinger
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site, 37075 Göttingen, Germany
- University Heart Center, 37075 Göttingen, Germany
| | - Kim Jürgen Krott
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Hubert Schelzig
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Hug Aubin
- Clinic for Cardiac Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
- CURE3D Lab, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Alexander Oberhuber
- Clinic for Vascular and Endovascular Surgery, University Hospital Münster, 48149 Münster, Germany
| | - Margitta Elvers
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Markus Udo Wagenhäuser
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| |
Collapse
|
34
|
Moragas LJ, Alves FDAV, Oliveira LDLS, Salomão NG, Azevedo CG, da Silva JFR, Basílio-de-Oliveira CA, Basílio-de-Oliveira R, Mohana-Borges R, de Carvalho JJ, Rosman FC, Paes MV, Rabelo K. Liver immunopathogenesis in fatal cases of dengue in children: detection of viral antigen, cytokine profile and inflammatory mediators. Front Immunol 2023; 14:1215730. [PMID: 37457689 PMCID: PMC10348746 DOI: 10.3389/fimmu.2023.1215730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Dengue virus (DENV), the etiologic agent of dengue fever illness, represents a global public health concern, mainly in tropical and subtropical areas across the globe. It is well known that this acute viral disease can progress to severe hemorrhagic stages in some individuals, however, the immunopathogenic basis of the development of more severe forms by these patients is yet to be fully understood. Objective In this context, we investigated and characterized the histopathological features as well as the cytokine profile and cell subpopulations present in liver tissues from three fatal cases of DENV in children. Methods Hematoxylin and Eosin, Periodic Acid Schiff and Picro Sirius Red staining were utilized for the histopathological analysis. Immunohistochemistry assay was performed to characterize the inflammatory response and cell expression patterns. Results Vascular dysfunctions such as hemorrhage, vascular congestion and edema associated with a mononuclear infiltrate were observedin all three cases. Liver tissues exhibited increased presence of CD68+ and TCD8+ cells as well as high expression of MMP-9, TNF-a, RANTES, VEGFR-2 mediators. Viral replication was confirmed by the detection of NS3 protein. Conclusion Taken together, these results evidenced key factors that may be involved in the development of severe alterations in liver tissues of children in response to DENV infection.
Collapse
Affiliation(s)
- Leandro Junqueira Moragas
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Felipe de Andrade Vieira Alves
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Laboratório de Ultraestrutura e Biologia Tecidual, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Natália Gedeão Salomão
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Caio Gonçalves Azevedo
- Laboratório de Ultraestrutura e Biologia Tecidual, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | - Ronaldo Mohana-Borges
- Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge José de Carvalho
- Laboratório de Ultraestrutura e Biologia Tecidual, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando Colonna Rosman
- Serviço de Anatomia Patológica, Hospital Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marciano Viana Paes
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Kíssila Rabelo
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Laboratório de Ultraestrutura e Biologia Tecidual, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
35
|
Weigel C, Bellaci J, Spiegel S. Sphingosine-1-phosphate and its receptors in vascular endothelial and lymphatic barrier function. J Biol Chem 2023; 299:104775. [PMID: 37142226 PMCID: PMC10220486 DOI: 10.1016/j.jbc.2023.104775] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023] Open
Abstract
The vascular and lymphatic systems both comprise a series of structurally distinct vessels lined with an inner layer of endothelial cells that function to provide a semipermeable barrier to blood and lymph. Regulation of the endothelial barrier is critical for maintaining vascular and lymphatic barrier homeostasis. One of the regulators of endothelial barrier function and integrity is sphingosine-1-phosphate (S1P), a bioactive sphingolipid metabolite secreted into the blood by erythrocytes, platelets, and endothelial cells and into the lymph by lymph endothelial cells. Binding of S1P to its G protein-coupled receptors, known as S1PR1-5, regulates its pleiotropic functions. This review outlines the structural and functional differences between vascular and lymphatic endothelium and describes current understanding of the importance of S1P/S1PR signaling in regulation of barrier functions. Most studies thus far have been primarily focused on the role of the S1P/S1PR1 axis in vasculature and have been summarized in several excellent reviews, and thus, we will only discuss new perspectives on the molecular mechanisms of action of S1P and its receptors. Much less is known about the responses of the lymphatic endothelium to S1P and the functions of S1PRs in lymph endothelial cells, and this is the major focus of this review. We also discuss current knowledge related to signaling pathways and factors regulated by the S1P/S1PR axis that control lymphatic endothelial cell junctional integrity. Gaps and limitations in current knowledge are highlighted together with the need to further understand the role of S1P receptors in the lymphatic system.
Collapse
Affiliation(s)
- Cynthia Weigel
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Jacqueline Bellaci
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.
| |
Collapse
|
36
|
Mahdinia E, Shokri N, Taheri AT, Asgharzadeh S, Elahimanesh M, Najafi M. Cellular crosstalk in atherosclerotic plaque microenvironment. Cell Commun Signal 2023; 21:125. [PMID: 37254185 DOI: 10.1186/s12964-023-01153-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/28/2023] [Indexed: 06/01/2023] Open
Abstract
Atherosclerosis is an underlying pathology of many vascular diseases as a result of cellular, structural and molecular dysfunctions within the sub-endothelial space. This review deals with the events involved in the formation, growth and remodeling of plaque, including the cell recruitment, cell polarization, and cell fat droplets. It also describes cross talking between endothelial cells, macrophages, and vascular smooth muscle cells, as well as the cellular pathways involved in plaque development in the plaque microenvironment. Finally, it describes the plaque structural components and the role of factors involved in the rupture and erosion of plaques in the vessel. Video Abstract.
Collapse
Affiliation(s)
- Elmira Mahdinia
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Shokri
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abdolkarim Talebi Taheri
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Asgharzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Ghazvin University of Medical Sciences, Ghazvin, Iran
| | - Mohammad Elahimanesh
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Department of Clinical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Microbial Biotechnology Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
You LJ, Li PW, Zhang WW, Feng MF, Zhao WP, Hou HM, Piao XM, Wang LB, Zhang Y. Schisandrin A ameliorates increased pulmonary capillary endothelial permeability accompanied with sepsis through inhibition of RhoA/ROCK1/MLC pathways. Int Immunopharmacol 2023; 118:110124. [PMID: 37028276 DOI: 10.1016/j.intimp.2023.110124] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Sepsis is a systemic inflammatory response, and vascular leakage associated with acute lung injury (ALI) is an important pathophysiological process during sepsis. Schisandrin A (SchA) is a bioactive lignan which has been reported to have the anti-inflammatory effects in many studies, while whether SchA can ameliorate ALI-related vascular leakage caused by sepsis is unknown. OBJECTIVE To evaluate the role and the underlying mechanism of SchA in increase of pulmonary vascular permeability induced by sepsis. METHODS The effect of SchA on pulmonary vascular permeability was examined in rat acute lung injury model. The effect of SchA on skin vascular permeability of mice was investigated through Miles assay. MTT assay was performed to detect the cell activity, and transwell assay was used to detect the effect of SchA on cell permeability. The effects of SchA on junction proteins and RhoA/ROCK1/MLC signaling pathway were manifested by immunofluorescence staining and western blot. RESULTS The administration of SchA alleviated rat pulmonary endothelial dysfunction, relieved increased permeability in the mouse skin and HUVECs induced by lipopolysaccharide (LPS). Meanwhile, SchA inhibited the formation of stress fibers, reversed the decrease of expression of ZO-1 and VE-cadherin. Subsequent experiments confirmed that SchA inhibited RhoA/ROCK1/MLC canonical pathway in rat lungs and HUVECs induced by LPS. Moreover, overexpression of RhoA reversed the inhibitory effect of SchA in HUVECs, which suggested that SchA protected the pulmonary endothelial barrier by inhibiting RhoA/ROCK1/MLC pathway. CONCLUSION In summary, our results indicate that SchA ameliorates the increase of pulmonary endothelial permeability induced by sepsis through inhibition of RhoA/ROCK1/MLC pathway, providing a potentially effective therapeutic strategy for sepsis.
Collapse
Affiliation(s)
- Li-Juan You
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Pei-Wei Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Wen-Wen Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Ming-Feng Feng
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Wei-Ping Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Hui-Min Hou
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Xian-Mei Piao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China.
| | - Li-Bo Wang
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, PR China.
| | - Yan Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China.
| |
Collapse
|
38
|
Francesca T, Elena LP, Barbara DSA, Marta DS, Marco T, Maria CA, Carmela P, Francesco M, Francesco D, Adriana C, Serena M. An analysis of the immunomodulatory properties of human spheroids from adipose-derived stem cells. Life Sci 2023; 321:121610. [PMID: 36948391 DOI: 10.1016/j.lfs.2023.121610] [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: 10/03/2022] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023]
Abstract
AIMS Current methods to induce tolerance following allotransplantation or in autoimmunity carry significant morbidity, and research is very active in investigating alternative methods which could minimize toxicity. Spheroids from adipose stem cells (SASCs) are increasingly gaining interest, they hold a great proliferative and differentiating potential. An immunomodulatory effect has not been investigated on SASCs yet. In this study, we analysed the immunomodulatory properties of SASCs and compared them to ADSCs. MAIN METHODS Adipose stem cells (SASCs and ADSCs) and peripheral blood mononuclear cells (PBMCs) were collected from healthy individuals. We analysed the cytokine production and proliferation of T cells co-cultured with adipose samples or conditioned medium. KEY FINDINGS SASCs modulated cytokines production and proliferation of heterologous and autologous T cells. In the heterologous assays, we observed a reduction of IFNγ and IL-17 production and an increase of IL-9 in γδ T cells. The soluble factors present in SASCs sovranatants were also able to induce a slight reduction of IFNγ and an increase of IL-9, IL-10 and IL-17 while they could not modulate the proliferative ability of γδ T cells. In the autologous assays, we observed a reduction of the proliferative ability of T cells in co-culture at different ratios with SASCs. Analysis of the SASCs secretome showed an increased IL-5, IL-10, IL-4 and IL-13 production compared to the ADSCs one, demonstrating greater anti-inflammatory properties. SIGNIFICANCE Our preliminary results support the idea that SASCs exert more pronounced biological immune modulation compared to the classical adherent ADSCs, especially in heterologous experimental settings.
Collapse
Affiliation(s)
- Toia Francesca
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Section of Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy; Plastic and Reconstructive Unit, Department of Oncology, Azienda Ospedaliera Universitaria Policlinico "Paolo Giaccone", 90127 Palermo, Italy
| | - Lo Presti Elena
- National Research Council (CNR)-Institute for Biomedical Research and Innovation (IRIB), 90146 Palermo, Italy
| | - Di Stefano Anna Barbara
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy.
| | - Di Simone Marta
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Trapani Marco
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Corsale Anna Maria
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Picone Carmela
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Moschella Francesco
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Dieli Francesco
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Cordova Adriana
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Section of Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy; Plastic and Reconstructive Unit, Department of Oncology, Azienda Ospedaliera Universitaria Policlinico "Paolo Giaccone", 90127 Palermo, Italy
| | - Meraviglia Serena
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| |
Collapse
|
39
|
Perrelli A, Ferraris C, Berni E, Glading AJ, Retta SF. KRIT1: A Traffic Warden at the Busy Crossroads Between Redox Signaling and the Pathogenesis of Cerebral Cavernous Malformation Disease. Antioxid Redox Signal 2023; 38:496-528. [PMID: 36047808 PMCID: PMC10039281 DOI: 10.1089/ars.2021.0263] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 12/18/2022]
Abstract
Significance: KRIT1 (Krev interaction trapped 1) is a scaffolding protein that plays a critical role in vascular morphogenesis and homeostasis. Its loss-of-function has been unequivocally associated with the pathogenesis of Cerebral Cavernous Malformation (CCM), a major cerebrovascular disease of genetic origin characterized by defective endothelial cell-cell adhesion and ensuing structural alterations and hyperpermeability in brain capillaries. KRIT1 contributes to the maintenance of endothelial barrier function by stabilizing the integrity of adherens junctions and inhibiting the formation of actin stress fibers. Recent Advances: Among the multiple regulatory mechanisms proposed so far, significant evidence accumulated over the past decade has clearly shown that the role of KRIT1 in the stability of endothelial barriers, including the blood-brain barrier, is largely based on its involvement in the complex machinery governing cellular redox homeostasis and responses to oxidative stress and inflammation. KRIT1 loss-of-function has, indeed, been demonstrated to cause an impairment of major redox-sensitive mechanisms involved in spatiotemporal regulation of cell adhesion and signaling, which ultimately leads to decreased cell-cell junction stability and enhanced sensitivity to oxidative stress and inflammation. Critical Issues: This review explores the redox mechanisms that influence endothelial cell adhesion and barrier function, focusing on the role of KRIT1 in such mechanisms. We propose that this supports a novel model wherein redox signaling forms the common link between the various pathogenetic mechanisms and therapeutic approaches hitherto associated with CCM disease. Future Directions: A comprehensive characterization of the role of KRIT1 in redox control of endothelial barrier physiology and defense against oxy-inflammatory insults will provide valuable insights into the development of precision medicine strategies. Antioxid. Redox Signal. 38, 496-528.
Collapse
Affiliation(s)
- Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Chiara Ferraris
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Elisa Berni
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Angela J. Glading
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| |
Collapse
|
40
|
Nachit M, Montemagno C, Clerc R, Ahmadi M, Briand F, Bacot S, Devoogdt N, Serdjebi C, Ghezzi C, Sulpice T, Broisat A, Leclercq IA, Perret P. Molecular imaging of liver inflammation using an anti-VCAM-1 nanobody. Nat Commun 2023; 14:1062. [PMID: 36828835 PMCID: PMC9957989 DOI: 10.1038/s41467-023-36776-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 02/09/2023] [Indexed: 02/26/2023] Open
Abstract
To date, a biopsy is mandatory to evaluate parenchymal inflammation in the liver. Here, we evaluated whether molecular imaging of vascular cell adhesion molecule-1 (VCAM-1) could be used as an alternative non-invasive tool to detect liver inflammation in the setting of chronic liver disease. To do so, we radiolabeled anti-VCAM-1 nanobody (99mTc-cAbVCAM1-5) and used single-photon emission computed tomography (SPECT) to quantify liver uptake in preclinical models of non-alcoholic fatty liver disease (NAFLD) with various degree of liver inflammation: wild-type mice fed a normal or high-fat diet (HFD), FOZ fed a HFD and C57BL6/J fed a choline-deficient or -supplemented HFD. 99mTc-cAbVCAM1-5 uptake strongly correlates with liver histological inflammatory score and with molecular inflammatory markers. The diagnostic power to detect any degree of liver inflammation is excellent (AUROC 0.85-0.99). These data build the rationale to investigate 99mTc-cAbVCAM1-5 imaging to detect liver inflammation in patients with NAFLD, a largely unmet medical need.
Collapse
Affiliation(s)
- Maxime Nachit
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | | | - Romain Clerc
- Univ. Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| | - Mitra Ahmadi
- Univ. Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| | | | - Sandrine Bacot
- Univ. Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| | - Nick Devoogdt
- Department of Medical Imaging, Laboratory of in vivo Cellular and Molecular Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | - Alexis Broisat
- Univ. Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France.
| | - Isabelle A Leclercq
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Pascale Perret
- Univ. Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| |
Collapse
|
41
|
Fan Y, Liu X, Wu J, Ni J, Liang J, Hou Y, Dou H. Small molecule compound K-7174 attenuates neuropsychiatric manifestations in lupus-prone mice. Brain Res 2023; 1801:148203. [PMID: 36521514 DOI: 10.1016/j.brainres.2022.148203] [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: 10/21/2022] [Revised: 12/03/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
The neuropsychiatric manifestations of systemic lupus erythematosus (NPSLE) present significant morbidity and mortality due to frequent non-response or adverse effects of the current clinical drugs. The disruption of the blood-brain barrier (BBB) contributes to inflammatory NPSLE disease progression. K-7174, a highly piperazine-derived compound, inhibits leukocyte adhesion and inflammatory factor expression. The present study aimed to comprehensively assess the treatment effect of neurobehavioral deficits in MRL/lpr mice, a validated neuropsychiatric lupus model. The intraperitoneal injection of K-7174 alleviated lupus-like symptoms and improved cognitive dysfunction in MRL/lpr mice. Also, it significantly attenuated neuronal degeneration and decreased serum albumin deposition in the hippocampus. Furthermore, K-7174 acted directly on the brain microvascular endothelial bEnd.3 cells and reduced the BBB permeability, manifested by inhibiting the activation of brain microvascular endothelial cells and increasing the expression of tight junctions (TJs). Notably, in vitro experiments showed that K-7174 alleviates the decreased ZO1 and Occludin expression in bEnd.3 cells caused by lactate increase, improving cell permeability via the MCT4/NKAP/CREB signaling pathway. These findings suggested that K-7174 mediates the attenuation of NPSLE in MRL/lpr mice, indicating a promising therapeutic strategy for NPSLE.
Collapse
Affiliation(s)
- Yu Fan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Xuan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Jinjin Wu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Jiali Ni
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China.
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China.
| |
Collapse
|
42
|
Kiełbowski K, Bakinowska E, Pawlik A. The Potential Role of Connexins in the Pathogenesis of Atherosclerosis. Int J Mol Sci 2023; 24:ijms24032600. [PMID: 36768920 PMCID: PMC9916887 DOI: 10.3390/ijms24032600] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/29/2022] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Connexins (Cx) are members of a protein family which enable extracellular and intercellular communication through hemichannels and gap junctions (GJ), respectively. Cx take part in transporting important cell-cell messengers such as 3',5'-cyclic adenosine monophosphate (cAMP), adenosine triphosphate (ATP), and inositol 1,4,5-trisphosphate (IP3), among others. Therefore, they play a significant role in regulating cell homeostasis, proliferation, and differentiation. Alterations in Cx distribution, degradation, and post-translational modifications have been correlated with cancers, as well as cardiovascular and neurological diseases. Depending on the isoform, Cx have been shown either to promote or suppress the development of atherosclerosis, a progressive inflammatory disease affecting large and medium-sized arteries. Cx might contribute to the progression of the disease by enhancing endothelial dysfunction, monocyte recruitment, vascular smooth muscle cell (VSMC) activation, or by inhibiting VSMC autophagy. Inhibition or modulation of the expression of specific isoforms could suppress atherosclerotic plaque formation and diminish pro-inflammatory conditions. A better understanding of the complexity of atherosclerosis pathophysiology linked with Cx could result in developing novel therapeutic strategies. This review aims to present the role of Cx in the pathogenesis of atherosclerosis and discusses whether they can become novel therapeutic targets.
Collapse
|
43
|
Wang H, Zhou Z, Xie J, Qi S, Tang J. Integration of single-cell and bulk transcriptomics reveals immune-related signatures in keloid. J Cosmet Dermatol 2023; 22:1893-1905. [PMID: 36701151 DOI: 10.1111/jocd.15649] [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: 10/22/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Keloid is a pathological dermatological condition that manifests as an overgrowth scar secondary to skin trauma. This study endeavored to excavate immune-related signatures of keloid based on single-cell RNA (scRNA) sequencing data and bulk RNA sequencing data. METHOD The keloid-relevant scRNA sequencing dataset GSE163973 and bulk RNA sequencing dataset GSE113619 were mined from the GEO database. The "Seurat" R package was utilized for data quality control, cell clustering, and investigation of marker genes of each cell cluster. The "SingleR" package helped match the marker genes of the corresponding cluster to specific cell types. Moreover, the R package "Monocle" was deployed for pseudotemporal ordering analysis, and the "clusterProfiler" was applied for functional and pathway enrichment analysis. The immune-related signatures were then identified, and potential targeted drugs were predicted via the DGIdb database. Verification of the immune-related signatures in clinical validation samples was implemented by RT-qPCR. RESULTS Totally 23 cell clusters were screened and classified into 10 cell types based on the scRNA sequencing data. The keloid group had a significantly higher endothelial cell proportion than the control group. As enrichment analysis was applied in both differentially expressed genes (DEGs) of scRNA and bulk RNA sequencing data, we found they were enriched in multiple common immune-related pathways and biological processes. Meanwhile, we acquired three immune-related signatures (VCAM1, CALCRL, and HLA-DPB1) by intersecting the above DEGs with immune-related genes (IRGs). Then, we predicted 16 drugs potentially targeting the biomarkers through the DGIdb database. Finally, the outcome of RT-qPCR of clinical validation samples further verified the results. CONCLUSION In conclusion, we analyzed the cell types and functional differences in the keloid through scRNA and bulk RNA sequencing data. We identified three immune-related signatures (VCAM1, CALCRL, and HLA-DPB1) in keloid, providing a basis for further in-depth investigation of the molecular mechanisms of keloid and exploration of therapeutic targets.
Collapse
Affiliation(s)
- Hanwen Wang
- Department of Burn Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ziheng Zhou
- Department of Burn Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Julin Xie
- Department of Burn Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shaohai Qi
- Department of Burn Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jinming Tang
- Department of Burn Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
44
|
Bkaily G, Jacques D. Morphological and Functional Remodeling of Vascular Endothelium in Cardiovascular Diseases. Int J Mol Sci 2023; 24:ijms24031998. [PMID: 36768314 PMCID: PMC9916505 DOI: 10.3390/ijms24031998] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/21/2023] Open
Abstract
The vascular endothelium plays a vital role during embryogenesis and aging and is a cell monolayer that lines the blood vessels. The immune system recognizes the endothelium as its own. Therefore, an abnormality of the endothelium exposes the tissues to the immune system and provokes inflammation and vascular diseases such as atherosclerosis. Its secretory role allows it to release vasoconstrictors and vasorelaxants as well as cardio-modulatory factors that maintain the proper functioning of the circulatory system. The sealing of the monolayer provided by adhesion molecules plays an important role in cardiovascular physiology and pathology.
Collapse
|
45
|
Upregulation of Robo4 expression by SMAD signaling suppresses vascular permeability and mortality in endotoxemia and COVID-19 models. Proc Natl Acad Sci U S A 2023; 120:e2213317120. [PMID: 36634143 PMCID: PMC9934020 DOI: 10.1073/pnas.2213317120] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
There is an urgent need to develop novel drugs to reduce the mortality from severe infectious diseases with the emergence of new pathogens, including Coronavirus disease 2019 (COVID-19). Although current drugs effectively suppress the proliferation of pathogens, immune cell activation, and inflammatory cytokine functions, they cannot completely reduce mortality from severe infections and sepsis. In this study, we focused on the endothelial cell-specific protein, Roundabout 4 (Robo4), which suppresses vascular permeability by stabilizing endothelial cells, and investigated whether enhanced Robo4 expression could be a novel therapeutic strategy against severe infectious diseases. Endothelial-specific overexpression of Robo4 suppresses vascular permeability and reduces mortality in lipopolysaccharide (LPS)-treated mice. Screening of small molecules that regulate Robo4 expression and subsequent analysis revealed that two competitive small mothers against decapentaplegic (SMAD) signaling pathways, activin receptor-like kinase 5 (ALK5)-SMAD2/3 and ALK1-SMAD1/5, positively and negatively regulate Robo4 expression, respectively. An ALK1 inhibitor was found to increase Robo4 expression in mouse lungs, suppress vascular permeability, prevent extravasation of melanoma cells, and decrease mortality in LPS-treated mice. The inhibitor suppressed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced endothelial barrier disruption and decreased mortality in mice infected with SARS-CoV-2. These results indicate that enhancing Robo4 expression is an efficient strategy to suppress vascular permeability and mortality in severe infectious diseases, including COVID-19, and that small molecules that upregulate Robo4 can be potential therapeutic agents against these diseases.
Collapse
|
46
|
Adzraku SY, Wang G, Cao C, Bao Y, Wang Y, Smith AO, Du Y, Wang H, Li Y, Xu K, Qiao J, Ju W, Zeng L. Robo4 inhibits gamma radiation-induced permeability of a murine microvascular endothelial cell by regulating the junctions. Cell Mol Biol Lett 2023; 28:2. [PMID: 36647012 PMCID: PMC9843922 DOI: 10.1186/s11658-022-00413-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/19/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Hematopoietic stem cell transplantation involves irradiation preconditioning which causes bone marrow endothelial cell dysfunction. While much emphasis is on the reconstitution of hematopoietic stem cells in the bone marrow microenvironment, endothelial cell preservation is indispensable to overcome the preconditioning damages. This study aims to ascertain the role of Roundabout 4 (Robo4) in regulating irradiation-induced damage to the endothelium. METHODS Microvascular endothelial cells were treated with γ-radiation to establish an endothelial cell injury model. Robo4 expression in the endothelial cells was manipulated employing lentiviral-mediated RNAi and gene overexpression technology before irradiation treatment. The permeability of endothelial cells was measured using qPCR, immunocytochemistry, and immunoblotting to analyze the effect on the expression and distribution of junctional molecules, adherens junctions, tight junctions, and gap junctions. Using Transwell endothelial monolayer staining, FITC-Dextran permeability, and gap junction-mediated intercellular communication (GJIC) assays, we determined the changes in endothelial functions after Robo4 gene manipulation and irradiation. Moreover, we measured the proportion of CD31 expression in endothelial cells by flow cytometry. We analyzed variations between two or multiple groups using Student's t-tests and ANOVA. RESULTS Ionizing radiation upregulates Robo4 expression but disrupts endothelial junctional molecules. Robo4 deletion causes further degradation of endothelial junctions hence increasing the permeability of the endothelial cell monolayer. Robo4 knockdown in microvascular endothelial cells increases the degradation and delocalization of ZO-1, PECAM-1, occludin, and claudin-5 molecules after irradiation. Conversely, connexin 43 expression increases after silencing Robo4 in endothelial cells to induce permeability but are readily destroyed when exposed to 10 Gy of gamma radiation. Also, Robo4 knockdown enhances Y731-VE-cadherin phosphorylation leading to the depletion and destabilization of VE-cadherin at the endothelial junctions following irradiation. However, Robo4 overexpression mitigates irradiation-induced degradation of tight junctional proteins and stabilizes claudin-5 and ZO-1 distribution. Finally, the enhanced expression of Robo4 ameliorates the irradiation-induced depletion of VE-cadherin and connexin 43, improves the integrity of microvascular endothelial cell junctions, and decreases permeability. CONCLUSION This study reveals that Robo4 maintains microvascular integrity after radiation preconditioning treatment by regulating endothelial permeability and protecting endothelial functions. Our results also provided a potential mechanism to repair the bone marrow vascular niche after irradiation by modulating Robo4 expression.
Collapse
Affiliation(s)
- Seyram Yao Adzraku
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Guozhang Wang
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Can Cao
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Yurong Bao
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Yizhou Wang
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Alhaji Osman Smith
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Yuwei Du
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Haiyang Wang
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Yue Li
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Kailin Xu
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Jianlin Qiao
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Wen Ju
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Lingyu Zeng
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| |
Collapse
|
47
|
Aghdash SN, Foroughi G. Chemical Kindling as an Experimental Model to Assess the Conventional Drugs in the Treatment of Post-traumatic Epilepsy. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:1417-1428. [PMID: 36443981 DOI: 10.2174/1871527322666221128155813] [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: 02/28/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality today, which will surpass many infectious diseases in the coming years/decades. Posttraumatic epilepsy (PTE) is one of the most common debilitating consequences of TBI. PTE is a secondary, acquired epilepsy that causes recurrent, spontaneous seizures more than a week after TBI. The extent of head injury in individuals who develop PTE is unknown; however, trauma is thought to account for 20% of symptomatic epilepsy worldwide. Understanding the mechanisms of epilepsy following TBI is crucial for the discovery of new anticonvulsant drugs for the treatment of PTE, as well as for improving the quality of life of patients with PTE. OBJECTIVE This review article explains the rationale for the usage of a chemical model to access new treatments for post-traumatic epilepsy. RESULTS There are multiple methods to control and manage PTE. The essential and available remedy for the management of epilepsy is the use of antiepileptic drugs. Antiepileptic drugs (AEDs) decrease the frequency of seizures without affecting the disease's causality. Antiepileptic drugs are administrated for the prevention and treatment of PTE; however, 30% of epilepsy patients are drug-resistant, and AED side effects are significant in PTE patients. There are different types of animal models, such as the liquid percussion model, intracortical ferric chloride injection, and cortical subincision model, to study PTE and neurophysiological mechanisms underlying the development of epilepsy after head injury. However, these animal models do not easily mimic the pathological events occurring in epilepsy. Therefore, animal models of PTE are an inappropriate tool for screening new and putatively effective AEDs. Chemical kindling is the most common animal model used to study epilepsy. There is a strong similarity between the kindling model and different types of human epilepsy. CONCLUSION Today, researchers use experimental animal models to evaluate new anticonvulsant drugs. The chemical kindling models, such as pentylenetetrazol, bicuculline, and picrotoxin-induced seizures, are important experimental models to analyze the impact of putative antiepileptic drugs.
Collapse
Affiliation(s)
- Simin Namvar Aghdash
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Golsa Foroughi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| |
Collapse
|
48
|
Rezaie J, Hosseinpour H, Rahbarghazi R, Keyhanmanesh R, Khanzadeh S, Mahdipour M, Soleimanpour J, Ahmadi M. Type 2 diabetes mellitus stimulated pulmonary vascular inflammation and exosome biogenesis in rats. Cell Biochem Funct 2023; 41:78-85. [PMID: 36335538 DOI: 10.1002/cbf.3764] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022]
Abstract
It has been shown that type 2 Diabetes Mellitus (T2DM) changes the paracrine activity of several cell types. Whether the biogenesis of exosomes is changed during diabetic conditions is the subject of debate. Here, we investigated the effect of T2M on exosome biogenesis in rat pulmonary tissue. Rats received a high-fat diet regime and a single low dose of Streptozocin to mimic the T2DM-like condition. A total of 8 weeks after induction of T2DM, rats were subjected to several analyses. Besides histological examination, vascular cell adhesion molecule 1 (VCAM-1) levels were detected using immunohistochemistry (IHC) staining. Transcription of several genes such as IL-1β, Alix, and Rab27b was calculated by real-time polymerase chain reaction assay. Using western blot analysis, intracellular CD63 levels were measured. The morphology and exosome secretion activity were assessed using acetylcholinesterase (AChE) assay and scanning electron microscopy, respectively. Histological results exhibited a moderate-to-high rate of interstitial pneumonia with emphysematous changes. IHC staining showed an increased VCAM-1 expression in the diabetic lungs compared with the normal conditions (p < .05). Likewise, we found the induction of IL-1β, and exosome-related genes Alix and Rab27b under diabetic conditions compared with the control group (p < .05). Along with these changes, protein levels of CD63 and AChE activity were induced upon the initiation of T2DM, indicating accelerated exosome biogenesis. Taken together, current data indicated the induction of exosome biogenesis in rat pulmonary tissue affected by T2DM. It seems that the induction of inflammatory niche is touted as a stimulatory factor to accelerate exosome secretion.
Collapse
Affiliation(s)
- Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Hossein Hosseinpour
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shokoufeh Khanzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Soleimanpour
- Department of Orthopedics Surgery, Shohada Teaching Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Ahmadi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
49
|
Immunopathology of Renal Tissue in Fatal Cases of Dengue in Children. Pathogens 2022; 11:pathogens11121543. [PMID: 36558877 PMCID: PMC9785549 DOI: 10.3390/pathogens11121543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Dengue virus (DENV) infection represents a worldwide public health concern and can cause damage to multiple organs, including the kidney. In this work, we investigated the histopathological changes caused by dengue virus infection along with the detection of inflammatory mediators, cytokines, and cell expression patterns in the renal tissue of three fatal cases in children. Hematoxylin and Eosin staining was performed to analyze these histopathological changes. Immunohistochemistry allowed for the detection of immunological inflammatory markers in renal tissues that were quantified and further analyzed. Vascular congestion, edema and glomerular infiltrate were observed in the three cases, in addition to the thickening of the matrix area around the glomerular capillaries and mononuclear infiltrate associated with vascular congestion in the medullary region. The renal tissues exhibited collagen deposition and high expression of CD68+ Mø, CD8+ T, CD56+ cells and MMP-9, and the cytokine profile was mainly characterized by the expression of IFN-γ and TNF-α. Additionally, the expression of RANTES, VEGFR-2 and VCAM-1 were observed. The replication of DENV was evidenced by the detection of the NS3 protein. These results contributed to clarifying the main factors that may be involved in changes in the renal tissue of fatal cases of dengue in children.
Collapse
|
50
|
Manilall A, Mokotedi L, Gunter S, Le Roux R, Fourie S, Flanagan CA, Millen AME. Increased protein phosphatase 5 expression in inflammation-induced left ventricular dysfunction in rats. BMC Cardiovasc Disord 2022; 22:539. [PMID: 36494772 PMCID: PMC9732989 DOI: 10.1186/s12872-022-02977-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Titin phosphorylation contributes to left ventricular (LV) diastolic dysfunction. The independent effects of inflammation on the molecular pathways that regulate titin phosphorylation are unclear. METHODS We investigated the effects of collagen-induced inflammation and subsequent tumor necrosis factor-α (TNF-α) inhibition on mRNA expression of genes involved in regulating titin phosphorylation in 70 Sprague-Dawley rats. LV diastolic function was assessed with echocardiography. Circulating inflammatory markers were quantified by enzyme-linked immunosorbent assay and relative LV gene expression was assessed by Taqman® polymerase chain reaction. Differences in normally distributed variables between the groups were determined by two-way analysis of variance (ANOVA), followed by Tukey post-hoc tests. For non-normally distributed variables, group differences were determined by Kruskal-Wallis tests. RESULTS Collagen inoculation increased LV relative mRNA expression of vascular cell adhesion molecule 1 (VCAM1), pentraxin 3 (PTX3), and inducible nitric oxide synthase (iNOS) compared to controls, indicating local microvascular inflammation. Collagen inoculation decreased soluble guanylate cyclase alpha-2 (sGCα2) and soluble guanylate cyclase beta-2 (sGCβ2) expression, suggesting downregulation of nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate (NO-sGC-cGMP) signaling. Inhibiting TNF-α prevented collagen-induced changes in VCAM1, iNOS, sGCα2 and sGCβ2 expression. Collagen inoculation increased protein phosphatase 5 (PP5) expression. Like LV diastolic dysfunction, increased PP5 expression was not prevented by TNF-α inhibition. CONCLUSION Inflammation-induced LV diastolic dysfunction may be mediated by a TNF-α-independent increase in PP5 expression and dephosphorylation of the N2-Bus stretch element of titin, rather than by TNF-α-induced downregulation of NO-sGC-cGMP pathway-dependent titin phosphorylation. The steady rise in number of patients with inflammation-induced diastolic dysfunction, coupled with low success rates of current therapies warrants a better understanding of the systemic signals and molecular pathways responsible for decreased titin phosphorylation in development of LV diastolic dysfunction. The therapeutic potential of inhibiting PP5 upregulation in LV diastolic dysfunction requires investigation.
Collapse
Affiliation(s)
- Ashmeetha Manilall
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Lebogang Mokotedi
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Sulè Gunter
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Regina Le Roux
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Serena Fourie
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Colleen A. Flanagan
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| | - Aletta M. E. Millen
- grid.11951.3d0000 0004 1937 1135Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| |
Collapse
|