1
|
Lozano-López DA, Hernández-Ortega LD, González-Mariscal L, Díaz-Coránguez M, Pinto-Dueñas DC, Castañeda-Arellano R. Preserving Blood-Brain Barrier Integrity in Ischemic Stroke: a Review on MSCs-sEVs Content and Potential Molecular Targets. Mol Neurobiol 2025:10.1007/s12035-025-04956-9. [PMID: 40259172 DOI: 10.1007/s12035-025-04956-9] [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: 11/26/2024] [Accepted: 04/13/2025] [Indexed: 04/23/2025]
Abstract
Ischemic stroke (IS) is a life-threatening condition that constitutes the second leading cause of death globally. Despite its high impact on public health, there is a shortage of treatments due to the complexity of the cellular and molecular mechanisms implicated. One main limiting factor for successful IS therapeutic intervention is stroke-induced blood-brain barrier (BBB) damage, particularly over tight junction proteins (TJs). BBB disruption is a well-established feature of IS, accelerating ischemic tissue damage and worsening prognosis. In recent years, mesenchymal stem cells (MSCs) and their small extracellular vesicles (MSCs-sEVs) have emerged as promising therapeutic interventions for several neurological disorders, including IS. However, its effects on BBB repair after IS are not completely understood. In this review, we will discuss novel experimental evidence of MSCs-sEVs effects in BBB protection and highlight the relevance of molecules reported in MSCs-sEVs, their potential cellular and molecular targets, and putative mechanisms implicated in BBB repair, providing a promising research avenue that may translate into effective therapeutic strategies for IS.
Collapse
Affiliation(s)
- David Arturo Lozano-López
- Pharmacology Laboratory, Center for Multidisciplinary Health Research, University Center of Tonalá, University of Guadalajara, Guadalajara, México
| | - Luis Daniel Hernández-Ortega
- Molecular Biology Laboratory, Center for Multidisciplinary Health Research, University Center of Tonalá, University of Guadalajara, Guadalajara, México
| | - Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, México
| | - Mónica Díaz-Coránguez
- Department of Pharmacobiology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, México
| | - Diana Cristina Pinto-Dueñas
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, México
| | - Rolando Castañeda-Arellano
- Pharmacology Laboratory, Center for Multidisciplinary Health Research, University Center of Tonalá, University of Guadalajara, Guadalajara, México.
| |
Collapse
|
2
|
Antonetti DA, Lin CM, Shanmugam S, Hager H, Cao M, Liu X, Dreffs A, Habash A, Abcouwer SF. Diabetes Renders Photoreceptors Susceptible to Retinal Ischemia-Reperfusion Injury. Invest Ophthalmol Vis Sci 2024; 65:46. [PMID: 39570639 PMCID: PMC11585066 DOI: 10.1167/iovs.65.13.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/03/2024] [Indexed: 11/22/2024] Open
Abstract
Purpose Studies have suggested that photoreceptors (PR) are altered by diabetes, contributing to diabetic retinopathy (DR) pathology. Here, we explored the effect of diabetes on retinal ischemic injury. Methods Retinal ischemia-reperfusion (IR) injury was caused by elevation of intraocular pressure in 10-week-old BKS db/db type 2 diabetes mellitus (T2DM) mice or C57BL/6J mice at 4 or 12 weeks after streptozotocin (STZ)-induced type 1 diabetes mellitus (T1DM), and respective nondiabetic controls. Retinal neurodegeneration was evaluated by retinal layer thinning, TUNEL staining, and neuron loss. Vascular permeability was evaluated as retinal accumulation of circulating fluorescent albumin. The effects of pretreatment with a sodium-glucose co-transporter (SGLT1/2) inhibitor, phlorizin, were examined. Results Nondiabetic control mice exhibited no significant outer retinal layer thinning or PR loss after IR injury. In contrast, db/db mice exhibited significant outer retina thinning (49%, P < 0.0001), loss of PR nuclei (45%, P < 0.05) and inner segment (IS) length decline (45%, P < 0.0001). STZ-induced diabetic mice at 4 weeks showed progressive thinning of the outer retina (55%, by 14 days, P < 0.0001) and 4.3-fold greater number of TUNEL+ cells in the outer nuclear layer (ONL) than injured retinas of control mice (P < 0.0001). After 12 weeks of diabetes, the retinas exhibited similar outer layer thinning and PR loss after IR. Diabetes also delayed restoration of the blood-retinal barrier after IR injury. Phlorizin reduced outer retinal layer thinning from 49% to 3% (P < 0.0001). Conclusions Diabetes caused PR to become highly susceptible to IR injury. The ability of phlorizin pretreatment to block outer retinal thinning after IR suggests that the effects of diabetes on PR are readily reversible.
Collapse
Affiliation(s)
- David A. Antonetti
- Department of Ophthalmology and Visual Sciences, University of Michigan, Michigan Medicine, Kellogg Eye Center, Ann Arbor, Michigan, United States
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Cheng-Mao Lin
- Department of Ophthalmology and Visual Sciences, University of Michigan, Michigan Medicine, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Sumathi Shanmugam
- Department of Ophthalmology and Visual Sciences, University of Michigan, Michigan Medicine, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Heather Hager
- Department of Ophthalmology and Visual Sciences, University of Michigan, Michigan Medicine, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Manjing Cao
- Department of Ophthalmology and Visual Sciences, University of Michigan, Michigan Medicine, Kellogg Eye Center, Ann Arbor, Michigan, United States
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuwen Liu
- Department of Ophthalmology and Visual Sciences, University of Michigan, Michigan Medicine, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Alyssa Dreffs
- Department of Ophthalmology and Visual Sciences, University of Michigan, Michigan Medicine, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Adam Habash
- Department of Ophthalmology and Visual Sciences, University of Michigan, Michigan Medicine, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Steven F. Abcouwer
- Department of Ophthalmology and Visual Sciences, University of Michigan, Michigan Medicine, Kellogg Eye Center, Ann Arbor, Michigan, United States
| |
Collapse
|
3
|
Duan Y, Liu D, Yu H, Zhang S, Xia Y, Du Z, Qin Y, Wang Y, Ma X, Liu H, Du Y. Transcription and post-translational mechanisms: dual regulation of adiponectin-mediated Occludin expression in diabetes. Cell Biosci 2024; 14:126. [PMID: 39354565 PMCID: PMC11443667 DOI: 10.1186/s13578-024-01306-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Occludin, a crucial component of tight junctions, has emerged as a promising biomarker for the diagnosis of acute ischemic disease, highlighting its significant potential in clinical applications. In the diabetes, Occludin serves as a downstream target gene intricately regulated by the adiponectin (APN) signaling pathway. However, the specific mechanism by which adiponectin regulates Occludin expression remains unclear. METHODS AND RESULTS Endothelial-specific Ocln knockdown reduced APN-mediated blood flow recovery after femoral artery ligation and nullified APN's protection against high-fat diet (HFD)-triggered apoptosis and angiogenesis inhibition in vivo. Mechanically, we have meticulously elucidated APN's regulatory role in Occludin expression through a comprehensive analysis spanning transcriptional and post-translational dimensions. Foxo1 has been elucidated as a crucial transcriptional regulator of Occludin that is modulated by the APN/APPL1 signaling axis, as evidenced by validation through ChIP-qPCR assays and Western blot analysis. APN hindered Occludin degradation via the ubiquitin-proteasome pathway. Mass spectrometry analysis has recently uncovered a novel phosphorylation site, Tyr467, on Occludin. This site responds to APN, playing a crucial role in inhibiting Occludin ubiquitination by APN. The anti-apoptotic and pro-angiogenic effects of APN were attenuated in vitro and in vivo following Foxo1 knockdown or expression of a non-phosphorylatable mutant, OccludinY467A. Clinically, elevated plasma concentrations of Occludin were observed in patients with diabetes. A significant negative correlation was found between Occludin levels and APN concentrations. CONCLUSION Our study proposes that APN modulates Occludin expression through mechanisms involving both transcriptional and post-translational interactions, thereby conferring a protective effect on endothelial integrity within diabetic vasculature.
Collapse
Affiliation(s)
- Yanru Duan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Demin Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Huahui Yu
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, People's Republic of China
| | - Shihan Zhang
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing Key Laboratory of Pediatric Hematology Oncology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, 100045, People's Republic of China
| | - Yihua Xia
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Zhiyong Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, People's Republic of China
| | - Yanwen Qin
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, People's Republic of China
| | - Yajing Wang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, People's Republic of China.
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing, China.
| | - Yunhui Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, People's Republic of China.
| |
Collapse
|
4
|
Jaykumar AB, Plumber S, Binns D, Wichaidit C, Luby-Phelps K, Cobb MH. SMURF1/2 are novel regulators of WNK1 stability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606092. [PMID: 39131382 PMCID: PMC11312594 DOI: 10.1101/2024.07.31.606092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Angiogenesis is essential for remodeling and repairing existing vessels, and this process requires signaling pathways including those controlled by transforming growth factor beta (TGF-β). We have previously reported crosstalk between TGF-β and the protein kinase With No lysine (K) 1 (WNK1). Homozygous disruption of the gene encoding WNK1 results in lethality in mice near embryonic day E12 due to impaired angiogenesis and this defect can be rescued by endothelial-specific expression of an activated form of the WNK1 substrate kinase Oxidative Stress-Responsive 1 (OSR1). However, molecular processes regulated via a collaboration between TGF-β and WNK1/OSR1 are not well understood. Here we show that WNK1 interacts with the E3 ubiquitin ligases SMURF1/2. In addition, we discovered that WNK1 regulates SMURF1/2 protein stability and vice versa. We also demonstrate that WNK1 activity regulates TGF-β receptor levels, in turn, controlling TGF-β signaling.
Collapse
Affiliation(s)
| | - Sakina Plumber
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Derk Binns
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | | | | | - Melanie H. Cobb
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| |
Collapse
|
5
|
Zhang XD, Yu WH, Liu MM, Liu R, Wu H, Wang Z, Hai CX. Pentoxifylline inhibits phosgene-induced lung injury via improving hypoxia. Drug Chem Toxicol 2023; 46:1100-1107. [PMID: 36220803 DOI: 10.1080/01480545.2022.2131811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/04/2022] [Accepted: 08/29/2022] [Indexed: 11/03/2022]
Abstract
Inhalation of high concentrations of phosgene often causes pulmonary edema, which obstructs the airway and causes tissue hypoxia. There is currently no specific antidote. This study was performed to investigate the effect behind pentoxifylline (PTX) treatment for phosgene-induced lung injury in rat models. Rats were exposed to phosgene. The protein levels of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), and occludin proteins in lung tissue were determined. The effect of both prophylactic and therapeutic administration of PTX (50 mg/kg and 100 mg/kg) was evaluated. The lung permeability index and HIF-1α protein level increased, the arterial blood oxygenation index (PaO2/FIO2 ratio) and occludin protein level decreased significantly 6 h after phosgene exposure (P < 0.05). PTX exerted protective effects by HIF-1α-VEGF-occludin signaling pathway to some extent. Moreover, prophylactic, but not therapeutic administration of PTX (100 mg/kg), exhibited a significant protective effect. Pretreatment with PTX protected against phosgene-induced lung injury, possibly by inhibiting differential expression of HIF-1α, VEGF, and occludin.
Collapse
Affiliation(s)
- Xiao-di Zhang
- Department of Toxicology, School of Public Health, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, China
| | - Wei-Hua Yu
- Department of Toxicology, School of Public Health, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, China
| | - Meng-Meng Liu
- Department of Health Service, Logistics College of Chinese People's Armed Police Force, Tianjin, China
| | - Rui Liu
- Department of Toxicology, School of Public Health, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, China
| | - Hao Wu
- Department of Toxicology, School of Public Health, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhao Wang
- Department of Toxicology, School of Public Health, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, China
| | - Chun-Xu Hai
- Department of Toxicology, School of Public Health, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
6
|
Torices S, Daire L, Simon S, Naranjo O, Mendoza L, Teglas T, Fattakhov N, Adesse D, Toborek M. Occludin: a gatekeeper of brain Infection by HIV-1. Fluids Barriers CNS 2023; 20:73. [PMID: 37840143 PMCID: PMC10577960 DOI: 10.1186/s12987-023-00476-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023] Open
Abstract
Compromised structure and function of the blood-brain barrier (BBB) is one of the pathological hallmarks of brain infection by HIV-1. BBB damage during HIV-1 infection has been associated with modified expression of tight junction (TJ) proteins, including occludin. Recent evidence indicated occludin as a redox-sensitive, multifunctional protein that can act as both an NADH oxidase and influence cellular metabolism through AMPK kinase. One of the newly identified functions of occludin is its involvement in regulating HIV-1 infection. Studies suggest that occludin expression levels and the rate of HIV-1 infection share a reverse, bidirectional relationship; however, the mechanisms of this relationship are unclear. In this review, we describe the pathways involved in the regulation of HIV-1 infection by occludin. We propose that occludin may serve as a potential therapeutic target to control HIV-1 infection and to improve the lives of people living with HIV-1.
Collapse
Affiliation(s)
- Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street Miami, Miami, FL, 11336, USA
| | - Leah Daire
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street Miami, Miami, FL, 11336, USA
| | - Sierra Simon
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street Miami, Miami, FL, 11336, USA
| | - Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street Miami, Miami, FL, 11336, USA
| | - Luisa Mendoza
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street Miami, Miami, FL, 11336, USA
| | - Timea Teglas
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street Miami, Miami, FL, 11336, USA
| | - Nikolai Fattakhov
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street Miami, Miami, FL, 11336, USA
| | - Daniel Adesse
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street Miami, Miami, FL, 11336, USA
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street Miami, Miami, FL, 11336, USA.
| |
Collapse
|
7
|
Srivastava AK, Venkata BS, Sweat YY, Rizzo HR, Jean-François L, Zuo L, Kurgan KW, Moore P, Shashikanth N, Smok I, Sachleben JR, Turner JR, Meredith SC. Serine 408 phosphorylation is a molecular switch that regulates structure and function of the occludin α-helical bundle. Proc Natl Acad Sci U S A 2022; 119:e2204618119. [PMID: 35969745 PMCID: PMC9407527 DOI: 10.1073/pnas.2204618119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/11/2022] [Indexed: 11/18/2022] Open
Abstract
Occludin is a tetramembrane-spanning tight junction protein. The long C-terminal cytoplasmic domain, which represents nearly half of occludin sequence, includes a distal bundle of three α-helices that mediates interactions with other tight junction components. A short unstructured region just proximal to the α-helical bundle is a phosphorylation hotspot within which S408 phosphorylation acts as molecular switch that modifies tight junction protein interactions and barrier function. Here, we used NMR to define the effects of S408 phosphorylation on intramolecular interactions between the unstructured region and the α-helical bundle. S408 pseudophosphorylation affected conformation at hinge sites between the three α-helices. Further studies using paramagnetic relaxation enhancement and microscale thermophoresis indicated that the unstructured region interacts with the α-helical bundle. These interactions between the unstructured domain are enhanced by S408 phosphorylation and allow the unstructured region to obstruct the binding site, thereby reducing affinity of the occludin tail for zonula occludens-1 (ZO-1). Conversely, S408 dephosphorylation attenuates intramolecular interactions, exposes the binding site, and increases the affinity of occludin binding to ZO-1. Consistent with an increase in binding to ZO-1, intravital imaging and fluorescence recovery after photobleaching (FRAP) analyses of transgenic mice demonstrated increased tight junction anchoring of enhanced green fluorescent protein (EGFP)-tagged nonphosphorylatable occludin relative to wild-type EGFP-occludin. Overall, these data define the mechanisms by which S408 phosphorylation modifies occludin tail conformation to regulate tight junction protein interactions and paracellular permeability.
Collapse
Affiliation(s)
| | | | - Yan Y. Sweat
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, 02115
| | - Heather R. Rizzo
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, 02115
| | - Léa Jean-François
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, 02115
| | - Li Zuo
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, 02115
- Anhui Medical University, Hefei, China, 230032
| | | | - Patrick Moore
- Department of Pathology, The University of Chicago, Chicago, IL 60637
| | - Nitesh Shashikanth
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, 02115
| | - Izabela Smok
- Department of Pathology, The University of Chicago, Chicago, IL 60637
| | | | - Jerrold R. Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, 02115
| | | |
Collapse
|
8
|
WNK1 collaborates with TGF-β in endothelial cell junction turnover and angiogenesis. Proc Natl Acad Sci U S A 2022; 119:e2203743119. [PMID: 35867836 PMCID: PMC9335306 DOI: 10.1073/pnas.2203743119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Angiogenesis is essential for growth of new blood vessels, remodeling existing vessels, and repair of damaged vessels, and these require reorganization of endothelial cell-cell junctions through a partial endothelial-mesenchymal transition. Homozygous disruption of the gene encoding the protein kinase WNK1 results in lethality in mice near embryonic day (E) 12 due to impaired angiogenesis. This angiogenesis defect can be rescued by endothelial-specific expression of an activated form of the WNK1 substrate kinase OSR1. We show that inhibition of WNK1 kinase activity not only prevents sprouting of endothelial cells from aortic slices but also vessel extension in inhibitor-treated embryos ex vivo. Mutations affecting TGF-β signaling also result in abnormal vascular development beginning by E10 and, ultimately, embryonic lethality. Previously, we demonstrated cross-talk of WNK1 with TGF-β-regulated SMAD signaling, and OSR1 was identified as a component of the TGF-β interactome. However, molecular events jointly regulated by TGF-β and WNK1/OSR1 have not been delineated. Here, we show that inhibition of WNK1 promotes TGF-β-dependent degradation of the tyrosine kinase receptor AXL, which is involved in TGF-β-mediated cell migration and angiogenesis. We also show that interaction between OSR1 and occludin, a protein associated with endothelial tight junctions, is an essential step to enable tight junction turnover. Furthermore, we show that these phenomena are WNK1 dependent, and sensitive to TGF-β. These findings demonstrate intimate connections between WNK1/OSR1 and multiple TGF-β-sensitive molecules controlling angiogenesis and suggest that WNK1 may modulate many TGF-β-regulated functions.
Collapse
|
9
|
Jung JU, Jaykumar AB, Cobb MH. WNK1 in Malignant Behaviors: A Potential Target for Cancer? Front Cell Dev Biol 2022; 10:935318. [PMID: 35813203 PMCID: PMC9257110 DOI: 10.3389/fcell.2022.935318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Metastasis is the major cause of mortality in cancer patients. Analyses of mouse models and patient data have implicated the protein kinase WNK1 as one of a handful of genes uniquely linked to a subset of invasive cancers. WNK1 signaling pathways are widely implicated in the regulation of ion co-transporters and in controlling cell responses to osmotic stress. In this review we will discuss its actions in tumor malignancy in human cancers and present evidence for its function in invasion, migration, angiogenesis and mesenchymal transition.
Collapse
Affiliation(s)
| | | | - Melanie H. Cobb
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
10
|
Yang F, Liu X, He J, Xian S, Yang P, Mai Z, Li M, Liu Y, Zhang X. Occludin facilitates tumour angiogenesis in bladder cancer by regulating IL8/STAT3 through STAT4. J Cell Mol Med 2022; 26:2363-2376. [PMID: 35224833 PMCID: PMC8995457 DOI: 10.1111/jcmm.17257] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 01/10/2023] Open
Abstract
Bladder cancer (BLCA) is a common genitourinary cancer in patients, and tumour angiogenesis is indispensable for its occurrence and development. However, the indepth mechanism of tumour angiogenesis in BLCA remains elusive. According to recent studies, the tight junction protein family member occludin (OCLN) is expressed at high levels in BLCA tissues and correlates with a poor prognosis. Downregulation of OCLN inhibits tumour angiogenesis in BLCA cells and murine xenografts, whereas OCLN overexpression exerts the opposite effect. Mechanistically, the RT‐qPCR analysis and Western blotting results showed that OCLN increased interleukin‐8 (IL8) and p‐signal transducer and activator of transcription 3 (STAT3) levels to promote BLCA angiogenesis. RNA sequencing analysis and dual‐luciferase reporter assays indicated that OCLN regulated IL8 transcriptional activity via the transcription factor STAT4. In summary, our results provide new perspectives on OCLN, as this protein participates in the development of BLCA angiogenesis by activating the IL8/STAT3 pathway via STAT4 and may serve as a novel and unique therapeutic target.
Collapse
Affiliation(s)
- Fan Yang
- Department of Pathology The Fifth Affiliated Hospital of Sun Yat‐Sen University Zhuhai China
| | - Xue‐Qi Liu
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat‐Sen University Sun Yat‐Sen University Shenzhen China
| | - Jian‐Zhong He
- Department of Pathology The Fifth Affiliated Hospital of Sun Yat‐Sen University Zhuhai China
| | - Shi‐Ping Xian
- Department of Pathology The Fifth Affiliated Hospital of Sun Yat‐Sen University Zhuhai China
| | - Peng‐Fei Yang
- Department of Pathology The Fifth Affiliated Hospital of Sun Yat‐Sen University Zhuhai China
| | - Zhi‐Ying Mai
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat‐Sen University Sun Yat‐Sen University Shenzhen China
| | - Miao Li
- Department of Hematology The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital Shenzhen China
| | - Ye Liu
- Department of Pathology The Fifth Affiliated Hospital of Sun Yat‐Sen University Zhuhai China
| | - Xing‐Ding Zhang
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat‐Sen University Sun Yat‐Sen University Shenzhen China
| |
Collapse
|
11
|
It is time for a moonshot to find “Cures” for diabetic retinal disease. Prog Retin Eye Res 2022; 90:101051. [DOI: 10.1016/j.preteyeres.2022.101051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/19/2022] [Accepted: 01/31/2022] [Indexed: 12/13/2022]
|
12
|
Ali M, Falkenhain K, Njiru BN, Murtaza-Ali M, Ruiz-Uribe NE, Haft-Javaherian M, Catchers S, Nishimura N, Schaffer CB, Bracko O. VEGF signalling causes stalls in brain capillaries and reduces cerebral blood flow in Alzheimer's mice. Brain 2022; 145:1449-1463. [PMID: 35048960 PMCID: PMC9150081 DOI: 10.1093/brain/awab387] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/09/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Increased incidence of stalled capillary blood flow caused by adhesion of
leucocytes to the brain microvascular endothelium leads to a 17%
reduction of cerebral blood flow and exacerbates short-term memory loss in
multiple mouse models of Alzheimer’s disease. Here, we report that vascular endothelial growth factor (VEGF) signalling at the
luminal side of the brain microvasculature plays an integral role in the
capillary stalling phenomenon of the APP/PS1 mouse model. Administration of the anti-mouse VEGF-A164 antibody, an isoform that inhibits
blood–brain barrier hyperpermeability, reduced the number of stalled
capillaries within an hour of injection, leading to an immediate increase in
average capillary blood flow but not capillary diameter. VEGF-A inhibition also
reduced the overall endothelial nitric oxide synthase protein concentrations,
increased occludin levels and decreased the penetration of circulating Evans
Blue dye across the blood–brain barrier into the brain parenchyma,
suggesting increased blood–brain barrier integrity. Capillaries prone to
neutrophil adhesion after anti-VEGF-A treatment also had lower occludin
concentrations than flowing capillaries. Taken together, our findings demonstrate that VEGF-A signalling in APP/PS1 mice
contributes to aberrant endothelial nitric oxide synthase /occludin-associated
blood–brain barrier permeability, increases the incidence of capillary
stalls, and leads to reductions in cerebral blood flow. Reducing leucocyte
adhesion by inhibiting luminal VEGF signalling may provide a novel and
well-tolerated strategy for improving brain microvascular blood flow in
Alzheimer’s disease patients.
Collapse
Affiliation(s)
- Muhammad Ali
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Kaja Falkenhain
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Brendah N Njiru
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Muhammad Murtaza-Ali
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Nancy E Ruiz-Uribe
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | | | | | - Nozomi Nishimura
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Oliver Bracko
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| |
Collapse
|
13
|
Liu XQ, Shao XR, Liu Y, Dong ZX, Chan SH, Shi YY, Chen SN, Qi L, Zhong L, Yu Y, Lv T, Yang PF, Li LY, Wang XB, Zhang XD, Li X, Zhao W, Sehgal L, Li M, Zhang XD. Tight junction protein 1 promotes vasculature remodeling via regulating USP2/TWIST1 in bladder cancer. Oncogene 2022; 41:502-514. [PMID: 34782718 DOI: 10.1038/s41388-021-02112-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022]
Abstract
Bladder cancer (BLCA) is the most common malignant tumor of the urinary system and is characterized by high metastatic rates and poor prognosis. The expression of tight junction protein 1 (TJP1) is associated with bladder cancer invasion; however, the mechanism by which TJP1 affects vasculature remodeling remains unknown. In this study, we found that TJP1 expression correlated with tumor angiogenesis and poor overall survival in clinical samples. Furthermore, TJP1 overexpression promoted tumor angiogenesis in BLCA cells and stimulated recruitment of macrophages to tumors by upregulating CCL2 expression. Mechanistically, TJP1 interacted with TWIST1 and enhanced the transcriptional activity of CCL2. The impairment of tumor angiogenesis caused by knockdown of TJP1 was dramatically rescued by overexpression of TWIST1. Furthermore, TJP1 recruited USP2, which deubiquitinated TWIST1, thereby protecting TWIST1 from proteasome-mediated protein degradation. In conclusion, our results suggest that TJP1 controls angiogenesis in BLCA via TWIST1-dependent regulation of CCL2. We demonstrate that TJP1 functions as a scaffold for the interaction between USP2 and TWIST1 and this may provide potential therapeutic targets in bladder cancer.
Collapse
Affiliation(s)
- Xue-Qi Liu
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xin-Rong Shao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ye Liu
- Department of Pathology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Zhao-Xia Dong
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Sze-Hoi Chan
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yuan-Yuan Shi
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Shu-Na Chen
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Lin Qi
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Li Zhong
- State Key Laboratory of Oncology in South China, Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yue Yu
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ting Lv
- Department of Pathology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Peng-Fei Yang
- Department of Pathology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Li-Yan Li
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xiao-Bin Wang
- Andrology section, Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xu-Dong Zhang
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xin Li
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Wenxue Zhao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Lalit Sehgal
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Miao Li
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China. .,Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China.
| | - Xing-Ding Zhang
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
14
|
Ali M, Bracko O. VEGF Paradoxically Reduces Cerebral Blood Flow in Alzheimer’s Disease Mice. Neurosci Insights 2022; 17:26331055221109254. [PMID: 35873789 PMCID: PMC9298729 DOI: 10.1177/26331055221109254] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/19/2022] Open
Abstract
Vascular dysfunction plays a critical role in the development of Alzheimer’s disease. Cerebral blood flow reductions of 10% to 25% present early in disease pathogenesis. Vascular Endothelial Growth Factor-A (VEGF-A) drives angiogenesis, which typically addresses blood flow reductions and global hypoxia. However, recent evidence suggests aberrant VEGF-A signaling in Alzheimer’s disease may undermine its physiological angiogenic function. Instead of improving cerebral blood flow, VEGF-A contributes to brain capillary stalls and blood flow reductions, likely accelerating cognitive decline. In this commentary, we explore the evidence for pathological VEGF signaling in Alzheimer’s disease, and discuss its implications for disease therapy.
Collapse
Affiliation(s)
- Muhammad Ali
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oliver Bracko
- Department of Biology, University of Miami, Coral Gables, FL, USA
| |
Collapse
|
15
|
Abcouwer SF, Shanmugam S, Muthusamy A, Lin CM, Kong D, Hager H, Liu X, Antonetti DA. Inflammatory resolution and vascular barrier restoration after retinal ischemia reperfusion injury. J Neuroinflammation 2021; 18:186. [PMID: 34446062 PMCID: PMC8394696 DOI: 10.1186/s12974-021-02237-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/11/2021] [Indexed: 02/08/2023] Open
Abstract
Background Several retinal pathologies exhibit both inflammation and breakdown of the inner blood-retinal barrier (iBRB) resulting in vascular permeability, suggesting that treatments that trigger resolution of inflammation may also promote iBRB restoration. Methods Using the mouse retinal ischemia-reperfusion (IR) injury model, we followed the time course of neurodegeneration, inflammation, and iBRB disruption and repair to examine the relationship between resolution of inflammation and iBRB restoration and to determine if minocycline, a tetracycline derivative shown to reverse microglial activation, can hasten these processes. Results A 90-min ischemic insult followed by reperfusion in the retina induced cell apoptosis and inner retina thinning that progressed for approximately 2 weeks. IR increased vascular permeability within hours, which resolved between 3 and 4 weeks after injury. Increased vascular permeability coincided with alteration and loss of endothelial cell tight junction (TJ) protein content and disorganization of TJ protein complexes. Shunting of blood flow away from leaky vessels and dropout of leaky capillaries were eliminated as possible mechanisms for restoring the iBRB. Repletion of TJ protein contents occurred within 2 days after injury, long before restoration of the iBRB. In contrast, the eventual re-organization of TJ complexes at the cell border coincided with restoration of the barrier. A robust inflammatory response was evident a 1 day after IR and progressed to resolution over the 4-week time course. The inflammatory response included a rapid and transient infiltration of granulocytes and Ly6C+ classical inflammatory monocytes, a slow accumulation of Ly6Cneg monocyte/macrophages, and activation, proliferation, and mobilization of resident microglia. Extravasation of the majority of CD45+ leukocytes occurred from the superficial plexus. The presence of monocyte/macrophages and increased numbers of microglia were sustained until the iBRB was eventually restored. Intervention with minocycline to reverse microglial activation at 1 week after injury promoted early restoration of the iBRB coinciding with decreased expression of mRNAs for the microglial M1 markers TNF-α, IL-1β, and Ptgs2 (Cox-2) and increased expression of secreted serine protease inhibitor Serpina3n mRNA. Conclusions These results suggest that iBRB restoration occurs as TJ complexes are reorganized and that resolution of inflammation and restoration of the iBRB following retinal IR injury are functionally linked. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02237-5.
Collapse
Affiliation(s)
- Steven F Abcouwer
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA.
| | - Sumathi Shanmugam
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | | | - Cheng-Mao Lin
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Dejuan Kong
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Heather Hager
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Xuwen Liu
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA.,Department of Molecular and Integrative Physiology, Ann Arbor, MI, 48109, USA
| |
Collapse
|
16
|
Goncalves A, Dreffs A, Lin CM, Sheskey S, Hudson N, Keil J, Campbell M, Antonetti DA. Vascular Expression of Permeability-Resistant Occludin Mutant Preserves Visual Function in Diabetes. Diabetes 2021; 70:1549-1560. [PMID: 33883214 PMCID: PMC8336002 DOI: 10.2337/db20-1220] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 04/01/2021] [Indexed: 12/20/2022]
Abstract
Diabetic retinopathy is one of the leading causes of vision loss and blindness. Extensive preclinical and clinical evidence exists for both vascular and neuronal pathology. However, the relationship of these changes in the neurovascular unit and impact on vision remains to be determined. Here, we investigate the role of tight junction protein occludin phosphorylation at S490 in modulating barrier properties and its impact on visual function. Conditional vascular expression of the phosphorylation-resistant Ser490 to Ala (S490A) form of occludin preserved tight junction organization and reduced vascular endothelial growth factor (VEGF)-induced permeability and edema formation after intraocular injection. In the retinas of streptozotocin-induced diabetic mice, endothelial-specific expression of the S490A form of occludin completely prevented diabetes-induced permeability to labeled dextran and inhibited leukostasis. Importantly, vascular-specific expression of the occludin mutant completely blocked the diabetes-induced decrease in visual acuity and contrast sensitivity. Together, these results reveal that occludin acts to regulate barrier properties downstream of VEGF in a phosphorylation-dependent manner and that loss of inner blood-retinal barrier integrity induced by diabetes contributes to vision loss.
Collapse
Affiliation(s)
- Andreia Goncalves
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI
| | - Alyssa Dreffs
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI
| | - Cheng-Mao Lin
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI
| | - Sarah Sheskey
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI
| | - Natalie Hudson
- Neurovascular Genetics Unit, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Jason Keil
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI
| | - Matthew Campbell
- Neurovascular Genetics Unit, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI
| |
Collapse
|
17
|
Karska-Basta I, Pociej-Marciak W, Chrząszcz M, Kubicka-Trząska A, Dębicka-Kumela M, Gawęcki M, Romanowska-Dixon B, Sanak M. Imbalance in the Levels of Angiogenic Factors in Patients with Acute and Chronic Central Serous Chorioretinopathy. J Clin Med 2021; 10:1087. [PMID: 33807809 PMCID: PMC7961803 DOI: 10.3390/jcm10051087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/19/2021] [Accepted: 02/26/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The pathogenesis of central serous chorioretinopathy (CSC) remains a subject of intensive research. We aimed to determine correlations between plasma levels of selected angiogenic factors and different forms of CSC. METHODS Eighty patients were enrolled in the study including 30 with a chronic form of CSC, 30 with acute CSC, and 20 controls. Presence of active CSC was determined by fluorescein angiography (FA), indocyanine green angiography (ICGA), and swept-source optical coherence tomography (SS-OCT). Plasma concentrations of angiopoietin-1, endostatin, fibroblast growth factor, placental growth factor (PlGF), platelet-derived growth factor (PDGF-AA), thrombospondin-2, vascular endothelial growth factor (VEGF), VEGF-D, and pigment epithelium-derived factor were measured, and the results were compared between groups. Additionally, mean choroidal thickness (CT) was measured in all patients. RESULTS Levels of angiopoietin-1 (p = 0.008), PlGF (p = 0.045), and PDGF-AA (p = 0.033) differed significantly between the three groups. Compared with the controls, VEGF (p = 0.024), PlGF (p = 0.013), and PDGF-AA (p = 0.012) were downregulated in the whole CSC group, specifically PDGF-AA (p = 0.002) in acute CSC and angiopoietin-1 (p = 0.007) in chronic CSC. An inverse correlation between mean CT and VEGF levels was noted in CSC patients (rho = -0.27, p = 0.044). CONCLUSIONS Downregulated angiopoietin-1, VEGF, PDGF-AA, and PlGF levels may highlight the previously unknown role of the imbalanced levels of proangiogenic and antiangiogenic factors in the pathogenesis of CSC. Moreover, downregulated VEGF levels may suggest that choroidal neovascularization in CSC is associated with arteriogenesis rather than angiogenesis.
Collapse
Affiliation(s)
- Izabella Karska-Basta
- Department of Ophthalmology, Faculty of Medicine, Clinic of Ophthalmology and Ocular Oncology, Jagiellonian University Medical College, 31-070 Krakow, Poland; (W.P.-M.); (M.C.); (A.K.-T.); (M.D.-K.); (B.R.-D.)
| | - Weronika Pociej-Marciak
- Department of Ophthalmology, Faculty of Medicine, Clinic of Ophthalmology and Ocular Oncology, Jagiellonian University Medical College, 31-070 Krakow, Poland; (W.P.-M.); (M.C.); (A.K.-T.); (M.D.-K.); (B.R.-D.)
| | - Michał Chrząszcz
- Department of Ophthalmology, Faculty of Medicine, Clinic of Ophthalmology and Ocular Oncology, Jagiellonian University Medical College, 31-070 Krakow, Poland; (W.P.-M.); (M.C.); (A.K.-T.); (M.D.-K.); (B.R.-D.)
| | - Agnieszka Kubicka-Trząska
- Department of Ophthalmology, Faculty of Medicine, Clinic of Ophthalmology and Ocular Oncology, Jagiellonian University Medical College, 31-070 Krakow, Poland; (W.P.-M.); (M.C.); (A.K.-T.); (M.D.-K.); (B.R.-D.)
| | - Magdalena Dębicka-Kumela
- Department of Ophthalmology, Faculty of Medicine, Clinic of Ophthalmology and Ocular Oncology, Jagiellonian University Medical College, 31-070 Krakow, Poland; (W.P.-M.); (M.C.); (A.K.-T.); (M.D.-K.); (B.R.-D.)
| | - Maciej Gawęcki
- Dobry Wzrok Ophthalmological Clinic, 80-402 Gdansk, Poland;
| | - Bożena Romanowska-Dixon
- Department of Ophthalmology, Faculty of Medicine, Clinic of Ophthalmology and Ocular Oncology, Jagiellonian University Medical College, 31-070 Krakow, Poland; (W.P.-M.); (M.C.); (A.K.-T.); (M.D.-K.); (B.R.-D.)
| | - Marek Sanak
- Molecular Biology and Clinical Genetics Unit, Department of Internal Medicine, Jagiellonian University Medical College Faculty of Medicine, 31-066 Krakow, Poland;
| |
Collapse
|
18
|
Karska‐Basta I, Pociej‐Marciak W, Chrząszcz M, Kubicka‐Trząska A, Romanowska‐Dixon B, Sanak M. Altered plasma cytokine levels in acute and chronic central serous chorioretinopathy. Acta Ophthalmol 2021; 99:e222-e231. [PMID: 32701204 PMCID: PMC7984262 DOI: 10.1111/aos.14547] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE To evaluate plasma levels of selected cytokines and investigate their correlation with choroidal thickness (CT) in patients with acute and chronic central serous chorioretinopathy (CSC). METHODS We enrolled 30 patients with acute CSC, 30 patients with chronic CSC and 20 controls. Plasma concentrations of 12 cytokines, interleukins IL-8, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-10 and IL-12 p70, granulocyte-macrophage colony-stimulating factor, interferon-γ, tumour necrosis factor-α (TNF-α) and vascular endothelial growth factor (VEGF), were measured using multiplex immunoassays. Differences in cytokine levels between groups were assessed. We also investigated correlations between cytokine levels and CT using swept-source optical coherence tomography, as well as an association between plasma cytokine profile and systemic hypertension. RESULTS We noted differences in IL-6 (p = 0.005), IL-10 (p = 0.03), IL-12 p70 (p = 0.028) and VEGF (p = 0.029) levels between groups. Pro-inflammatory IL-12 p70 and multidirectional IL-10 cytokines were upregulated, while pro-angiogenic VEGF was downregulated in chronic CSC as compared with controls (p = 0.005, p = 0.025 and p = 0.027, respectively). Interleukin-6 (IL-6) was upregulated in acute and chronic CSC (p = 0.030 and p = 0.005, respectively). Interleukin-5 (IL-5), IL-6 and IL-12 levels correlated with mean CT in acute CSC (p = 0.008, p = 0.003 and p = 0.044, respectively), while IL-8, IL-6 and TNF-α plasma levels correlated with hypertension in chronic CSC (p = 0.005, p = 0.033 and p = 0.001, respectively). CONCLUSION We provided new evidence for the possible role of plasma cytokines in the pathogenesis of CSC. Our results suggest that IL-6 may be important in the pathophysiology of acute and chronic CSC. The association between inflammatory response and hypertension in patients with CSC was also confirmed.
Collapse
Affiliation(s)
- Izabella Karska‐Basta
- Faculty of MedicineDepartment of OphthalmologyClinic of Ophthalmology and Ocular OncologyJagiellonian University Medical CollegeKrakówPoland
| | - Weronika Pociej‐Marciak
- Faculty of MedicineDepartment of OphthalmologyClinic of Ophthalmology and Ocular OncologyJagiellonian University Medical CollegeKrakówPoland
| | - Michał Chrząszcz
- Faculty of MedicineDepartment of OphthalmologyClinic of Ophthalmology and Ocular OncologyJagiellonian University Medical CollegeKrakówPoland
| | - Agnieszka Kubicka‐Trząska
- Faculty of MedicineDepartment of OphthalmologyClinic of Ophthalmology and Ocular OncologyJagiellonian University Medical CollegeKrakówPoland
| | - Bożena Romanowska‐Dixon
- Faculty of MedicineDepartment of OphthalmologyClinic of Ophthalmology and Ocular OncologyJagiellonian University Medical CollegeKrakówPoland
| | - Marek Sanak
- Faculty of MedicineDepartment of Internal MedicineMolecular Biology and Clinical Genetics UnitJagiellonian University Medical CollegeKrakówPoland
| |
Collapse
|
19
|
ALS-causing SOD1 mutants regulate occludin phosphorylation/ubiquitination and endocytic trafficking via the ITCH/Eps15/Rab5 axis. Neurobiol Dis 2021; 153:105315. [PMID: 33636390 DOI: 10.1016/j.nbd.2021.105315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/10/2021] [Accepted: 02/22/2021] [Indexed: 01/08/2023] Open
Abstract
It is increasingly recognized that blood-spinal cord barrier (BSCB) breakdown is a hallmark of amyotrophic lateral sclerosis (ALS). BSCB integrity is disrupted prior to disease onset. Occludin, as the functional component of the endothelial barrier, is downregulated in mouse models expressing ALS-linked superoxide dismutase-1 (SOD1) mutants. However, the molecular mechanisms underlying the regulation of occludin expression remain elusive. Here, using SOD1G93A transgenic mice and endothelial cells expressing SOD1 mutants of different biochemical characteristics, we found that the SOD1 mutation disrupted endothelial barrier integrity and that the occludin expression level was downregulated with disease progression. Our mechanistic studies revealed that abnormal reactive oxygen species (ROS) in mutant SOD1-expressing cells induced occludin phosphorylation, which facilitated the subsequent occludin ubiquitination mediated by the E3 ligase ITCH. Moreover, ubiquitinated occludin interacted with Eps15 to initiate its internalization, then trafficked to Rab5-positive vesicles and be degraded by proteasomes, resulting in a reduction in cell surface localization and total abundance. Notably, either ITCH or Eps15 knockdown was sufficient to rescue occludin degradation and ameliorate endothelial barrier disruption. In conclusion, our study reveals a novel mechanism of occludin degradation mediated by ALS-causing SOD1 mutants and demonstrates a role for occludin in regulating BSCB integrity.
Collapse
|
20
|
González-Mariscal L, Miranda J, Gallego-Gutiérrez H, Cano-Cortina M, Amaya E. Relationship between apical junction proteins, gene expression and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183278. [PMID: 32240623 DOI: 10.1016/j.bbamem.2020.183278] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022]
Abstract
The apical junctional complex (AJC) is a cell-cell adhesion system present at the upper portion of the lateral membrane of epithelial cells integrated by the tight junction (TJ) and the adherens junction (AJ). This complex is crucial to initiate and stabilize cell-cell adhesion, to regulate the paracellular transit of ions and molecules and to maintain cell polarity. Moreover, we now consider the AJC as a hub of signal transduction that regulates cell-cell adhesion, gene transcription and cell proliferation and differentiation. The molecular components of the AJC are multiple and diverse and depending on the cellular context some of the proteins in this complex act as tumor suppressors or as promoters of cell transformation, migration and metastasis outgrowth. Here, we describe these new roles played by TJ and AJ proteins and their potential use in cancer diagnostics and as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico.
| | - Jael Miranda
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Helios Gallego-Gutiérrez
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Misael Cano-Cortina
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Elida Amaya
- Department of Physiology, Biophysics and Neuroscience, Center of Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| |
Collapse
|
21
|
Bendriem RM, Singh S, Aleem AA, Antonetti DA, Ross ME. Tight junction protein occludin regulates progenitor Self-Renewal and survival in developing cortex. eLife 2019; 8:49376. [PMID: 31794381 PMCID: PMC6890460 DOI: 10.7554/elife.49376] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 11/18/2019] [Indexed: 12/31/2022] Open
Abstract
Occludin (OCLN) mutations cause human microcephaly and cortical malformation. A tight junction component thought absent in neuroepithelium after neural tube closure, OCLN isoform-specific expression extends into corticogenesis. Full-length and truncated isoforms localize to neuroprogenitor centrosomes, but full-length OCLN transiently localizes to plasma membranes while only truncated OCLN continues at centrosomes throughout neurogenesis. Mimicking human mutations, full-length OCLN depletion in mouse and in human CRISPR/Cas9-edited organoids produce early neuronal differentiation, reduced progenitor self-renewal and increased apoptosis. Human neural progenitors were more severely affected, especially outer radial glial cells, which mouse embryonic cortex lacks. Rodent and human mutant progenitors displayed reduced proliferation and prolonged M-phase. OCLN interacted with mitotic spindle regulators, NuMA and RAN, while full-length OCLN loss impaired spindle pole morphology, astral and mitotic microtubule integrity. Thus, early corticogenesis requires full-length OCLN to regulate centrosome organization and dynamics, revealing a novel role for this tight junction protein in early brain development.
Collapse
Affiliation(s)
- Raphael M Bendriem
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States.,Graduate School of Medical Sciences, Weill Cornell Medicine, New York, United States
| | - Shawn Singh
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States
| | | | - David A Antonetti
- Kellogg Eye Center, Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, United States
| | - M Elizabeth Ross
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States.,Graduate School of Medical Sciences, Weill Cornell Medicine, New York, United States
| |
Collapse
|
22
|
Díaz-Coránguez M, Liu X, Antonetti DA. Tight Junctions in Cell Proliferation. Int J Mol Sci 2019; 20:E5972. [PMID: 31783547 PMCID: PMC6928848 DOI: 10.3390/ijms20235972] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/22/2019] [Accepted: 11/22/2019] [Indexed: 12/23/2022] Open
Abstract
Tight junction (TJ) proteins form a continuous intercellular network creating a barrier with selective regulation of water, ion, and solutes across endothelial, epithelial, and glial tissues. TJ proteins include the claudin family that confers barrier properties, members of the MARVEL family that contribute to barrier regulation, and JAM molecules, which regulate junction organization and diapedesis. In addition, the membrane-associated proteins such as MAGUK family members, i.e., zonula occludens, form the scaffold linking the transmembrane proteins to both cell signaling molecules and the cytoskeleton. Most studies of TJ have focused on the contribution to cell-cell adhesion and tissue barrier properties. However, recent studies reveal that, similar to adherens junction proteins, TJ proteins contribute to the control of cell proliferation. In this review, we will summarize and discuss the specific role of TJ proteins in the control of epithelial and endothelial cell proliferation. In some cases, the TJ proteins act as a reservoir of critical cell cycle modulators, by binding and regulating their nuclear access, while in other cases, junctional proteins are located at cellular organelles, regulating transcription and proliferation. Collectively, these studies reveal that TJ proteins contribute to the control of cell proliferation and differentiation required for forming and maintaining a tissue barrier.
Collapse
Affiliation(s)
| | | | - David A. Antonetti
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI 48105, USA; (M.D.-C.); (X.L.)
| |
Collapse
|
23
|
Cong X, Kong W. Endothelial tight junctions and their regulatory signaling pathways in vascular homeostasis and disease. Cell Signal 2019; 66:109485. [PMID: 31770579 DOI: 10.1016/j.cellsig.2019.109485] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022]
Abstract
Endothelial tight junctions (TJs) regulate the transport of water, ions, and molecules through the paracellular pathway, serving as an important barrier in blood vessels and maintaining vascular homeostasis. In endothelial cells (ECs), TJs are highly dynamic structures that respond to multiple external stimuli and pathological conditions. Alterations in the expression, distribution, and structure of endothelial TJs may lead to many related vascular diseases and pathologies. In this review, we provide an overview of the assessment methods used to evaluate endothelial TJ barrier function both in vitro and in vivo and describe the composition of endothelial TJs in diverse vascular systems and ECs. More importantly, the direct phosphorylation and dephosphorylation of TJ proteins by intracellular kinases and phosphatases, as well as the signaling pathways involved in the regulation of TJs, including and the protein kinase C (PKC), PKA, PKG, Ras homolog gene family member A (RhoA), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K)/Akt, and Wnt/β-catenin pathways, are discussed. With great advances in this area, targeting endothelial TJs may provide novel treatment for TJ-related vascular pathologies.
Collapse
Affiliation(s)
- Xin Cong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| |
Collapse
|
24
|
Li R, Qi Y, Jiang M, Zhang T, Wang H, Wang L, Han M. Primary tumor-secreted VEGF induces vascular hyperpermeability in premetastatic lung via the occludin phosphorylation/ubiquitination pathway. Mol Carcinog 2019; 58:2316-2326. [PMID: 31553086 DOI: 10.1002/mc.23120] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/03/2019] [Accepted: 09/10/2019] [Indexed: 12/31/2022]
Abstract
Primary tumor can induce the formation of premetastatic niche. The hyperpermeability of the vessels in the premetastatic niche is the first step in the development of metastasis. However, the cellular and molecular mechanisms of vascular hyperpermeability remain to be elucidated. In this study, 4T1 breast cells were injected into the breasts of mice to establish a tumor model. Our results showed that primary tumors induced hyperpermeability of the vessels in the premetastatic lung. Subsequent studies showed that the level of vascular endothelial growth factor (VEGF) was elevated in the tumor-bearing mice serum and the levels of tight junction (TJ) proteins occludin and ZO-1 were decreased in the premetastatic lung. In vitro studies demonstrated that VEGF increased the permeability of dextran and decreased the levels of occludin and ZO-1 in human umbilical vein endothelial cells. Moreover, the hyperpermeability of vessels and the degradation of occludin was blocked by bevacizumab. Overexpression of occludin alleviated the VEGF-induced hyperpermeability. Further investigations revealed that VEGF-induced occludin phosphorylation at Ser-490 and ubiquitination. Finally, we showed that VEGF accelerated the process of occludin degradation through the ubiquitin-proteasome system. In conclusion, primary tumor-secrete VEGF induce the occludin phosphorylation/ubiquitination and downregulation, resulting in the disruption of TJs and hyperpermeability of vessels in premetastatic lung. The occludin phosphorylation/ubiquitination pathway may be the mechanism of VEGF-induced vascular hyperpermeability in the lung premetastatic niche.
Collapse
Affiliation(s)
- Ranran Li
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yana Qi
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Man Jiang
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Tiehong Zhang
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Hongwei Wang
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Liguang Wang
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Mingyong Han
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
25
|
Ragelle H, Goncalves A, Kustermann S, Antonetti DA, Jayagopal A. Organ-On-A-Chip Technologies for Advanced Blood-Retinal Barrier Models. J Ocul Pharmacol Ther 2019; 36:30-41. [PMID: 31140899 PMCID: PMC6985766 DOI: 10.1089/jop.2019.0017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/02/2019] [Indexed: 12/16/2022] Open
Abstract
The blood-retinal barrier (BRB) protects the retina by maintaining an adequate microenvironment for neuronal function. Alterations of the junctional complex of the BRB and consequent BRB breakdown in disease contribute to a loss of neuronal signaling and vision loss. As new therapeutics are being developed to prevent or restore barrier function, it is critical to implement physiologically relevant in vitro models that recapitulate the important features of barrier biology to improve disease modeling, target validation, and toxicity assessment. New directions in organ-on-a-chip technology are enabling more sophisticated 3-dimensional models with flow, multicellularity, and control over microenvironmental properties. By capturing additional biological complexity, organs-on-chip can help approach actual tissue organization and function and offer additional tools to model and study disease compared with traditional 2-dimensional cell culture. This review describes the current state of barrier biology and barrier function in ocular diseases, describes recent advances in organ-on-a-chip design for modeling the BRB, and discusses the potential of such models for ophthalmic drug discovery and development.
Collapse
Affiliation(s)
- Héloïse Ragelle
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Andreia Goncalves
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Harbor, Michigan
| | - Stefan Kustermann
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - David A. Antonetti
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Harbor, Michigan
| | - Ashwath Jayagopal
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
26
|
Kunze R, Marti HH. Angioneurins - Key regulators of blood-brain barrier integrity during hypoxic and ischemic brain injury. Prog Neurobiol 2019; 178:101611. [PMID: 30970273 DOI: 10.1016/j.pneurobio.2019.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022]
Abstract
The loss of blood-brain barrier (BBB) integrity leading to vasogenic edema and brain swelling is a common feature of hypoxic/ischemic brain diseases such as stroke, but is also central to the etiology of other CNS disorders. In the past decades, numerous proteins, belonging to the family of angioneurins, have gained increasing attention as potential therapeutic targets for ischemic stroke, but also other CNS diseases attributed to BBB dysfunction. Angioneurins encompass mediators that affect both neuronal and vascular function. Recently, increasing evidence has been accumulated that certain angioneurins critically determine disease progression and outcome in stroke among others through multifaceted effects on the compromised BBB. Here, we will give a concise overview about the family of angioneurins. We further describe the most important cellular and molecular components that contribute to structural integrity and low permeability of the BBB under steady-state conditions. We then discuss BBB alterations in ischemic stroke, and highlight underlying cellular and molecular mechanisms. For the most prominent angioneurin family members including vascular endothelial growth factors, angiopoietins, platelet-derived growth factors and erythropoietin, we will summarize current scientific literature from experimental studies in animal models, and if available from clinical trials, on the following points: (i) spatiotemporal expression of these factors in the healthy and hypoxic/ischemic CNS, (ii) impact of loss- or gain-of-function during cerebral hypoxia/ischemia for BBB integrity and beyond, and (iii) potential underlying molecular mechanisms. Moreover, we will highlight novel therapeutic strategies based on the activation of endogenous angioneurins that might improve BBB dysfuntion during ischemic stroke.
Collapse
Affiliation(s)
- Reiner Kunze
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany.
| | - Hugo H Marti
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany
| |
Collapse
|
27
|
Atypical Protein Kinase C: Breaking Down Barriers in Ocular Disease? THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2142-2146. [PMID: 30220553 DOI: 10.1016/j.ajpath.2018.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 11/21/2022]
Abstract
This commentary highlights the article by Lin et al that demonstrates the therapeutic potential of small-molecule atypical protein kinase C inhibitors in inflammatory ocular disease.
Collapse
|
28
|
Keep RF, Andjelkovic AV, Xiang J, Stamatovic SM, Antonetti DA, Hua Y, Xi G. Brain endothelial cell junctions after cerebral hemorrhage: Changes, mechanisms and therapeutic targets. J Cereb Blood Flow Metab 2018; 38:1255-1275. [PMID: 29737222 PMCID: PMC6092767 DOI: 10.1177/0271678x18774666] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/09/2018] [Indexed: 11/15/2022]
Abstract
Vascular disruption is the underlying cause of cerebral hemorrhage, including intracerebral, subarachnoid and intraventricular hemorrhage. The disease etiology also involves cerebral hemorrhage-induced blood-brain barrier (BBB) disruption, which contributes an important component to brain injury after the initial cerebral hemorrhage. BBB loss drives vasogenic edema, allows leukocyte extravasation and may lead to the entry of potentially neurotoxic and vasoactive compounds into brain. This review summarizes current information on changes in brain endothelial junction proteins in response to cerebral hemorrhage (and clot-related factors), the mechanisms underlying junction modification and potential therapeutic targets to limit BBB disruption and, potentially, hemorrhage occurrence. It also addresses advances in the tools that are now available for assessing changes in junctions after cerebral hemorrhage and the potential importance of such junction changes. Recent studies suggest post-translational modification, conformational change and intracellular trafficking of junctional proteins may alter barrier properties. Understanding how cerebral hemorrhage alters BBB properties beyond changes in tight junction protein loss may provide important therapeutic insights to prevent BBB dysfunction and restore normal function.
Collapse
Affiliation(s)
- Richard F Keep
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Ann Arbor, MI, USA
| | - Anuska V Andjelkovic
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Ann Arbor, MI, USA
| | - Jianming Xiang
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
| | | | - David A Antonetti
- Department of Molecular and Integrative Physiology, University of Michigan Ann Arbor, MI, USA
- Department of Ophthalmology & Visual Science Medical School, University of Michigan Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
| |
Collapse
|
29
|
The expected characteristics of an in vitro human Blood Brain Barrier model derived from cell lines, for studying how ABC transporters influence drug permeability. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
30
|
Morgan SV, Garwood CJ, Jennings L, Simpson JE, Castelli LM, Heath PR, Mihaylov SR, Vaquéz-Villaseñor I, Minshull TC, Ince PG, Dickman MJ, Hautbergue GM, Wharton SB. Proteomic and cellular localisation studies suggest non-tight junction cytoplasmic and nuclear roles for occludin in astrocytes. Eur J Neurosci 2018; 47:1444-1456. [PMID: 29738614 PMCID: PMC6079634 DOI: 10.1111/ejn.13933] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 04/16/2018] [Indexed: 12/16/2022]
Abstract
Occludin is a component of tight junctions, which are essential structural components of the blood–brain barrier. However, occludin is expressed in cells without tight junctions, implying additional functions. We determined the expression and localisation of occludin in astrocytes in cell culture and in human brain tissue, and sought novel binding partners using a proteomic approach. Expression was investigated by immunocytochemistry and immunoblotting in the 1321N1 astrocytoma cell line and ScienCell human primary astrocytes, and by immunohistochemistry in human autopsy brain tissue. Recombinant N‐ and C‐terminal occludin was used to pull‐down proteins from 1321N1 cell lysates and protein‐binding partners identified by mass spectrometry analysis. Occludin was expressed in both the cytoplasm and nucleus of astrocytes in vitro and in vivo. Mass spectrometry identified binding to nuclear and cytoplasmic proteins, particularly those related to RNA metabolism and nuclear function. Occludin is expressed in several subcellular compartments of brain cell‐types that do not form tight junctions and the expression patterns in cell culture reflect those in human brain tissue, indicating they are suitable model systems. Proteomic analysis suggests that occludin has novel functions in neuroepithelial cells that are unrelated to tight junction formation. Further research will establish the roles of these functions in both cellular physiology and in disease states.
Collapse
Affiliation(s)
- Sarah V Morgan
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Claire J Garwood
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Luke Jennings
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Lydia M Castelli
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Simeon R Mihaylov
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | | - Thomas C Minshull
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Paul G Ince
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Mark J Dickman
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Guillaume M Hautbergue
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| |
Collapse
|
31
|
Mechanisms of macular edema: Beyond the surface. Prog Retin Eye Res 2017; 63:20-68. [PMID: 29126927 DOI: 10.1016/j.preteyeres.2017.10.006] [Citation(s) in RCA: 413] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 10/24/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023]
Abstract
Macular edema consists of intra- or subretinal fluid accumulation in the macular region. It occurs during the course of numerous retinal disorders and can cause severe impairment of central vision. Major causes of macular edema include diabetes, branch and central retinal vein occlusion, choroidal neovascularization, posterior uveitis, postoperative inflammation and central serous chorioretinopathy. The healthy retina is maintained in a relatively dehydrated, transparent state compatible with optimal light transmission by multiple active and passive systems. Fluid accumulation results from an imbalance between processes governing fluid entry and exit, and is driven by Starling equation when inner or outer blood-retinal barriers are disrupted. The multiple and intricate mechanisms involved in retinal hydro-ionic homeostasis, their molecular and cellular basis, and how their deregulation lead to retinal edema, are addressed in this review. Analyzing the distribution of junction proteins and water channels in the human macula, several hypotheses are raised to explain why edema forms specifically in the macular region. "Pure" clinical phenotypes of macular edema, that result presumably from a single causative mechanism, are detailed. Finally, diabetic macular edema is investigated, as a complex multifactorial pathogenic example. This comprehensive review on the current understanding of macular edema and its mechanisms opens perspectives to identify new preventive and therapeutic strategies for this sight-threatening condition.
Collapse
|
32
|
Van Itallie CM, Anderson JM. Phosphorylation of tight junction transmembrane proteins: Many sites, much to do. Tissue Barriers 2017; 6:e1382671. [PMID: 29083946 DOI: 10.1080/21688370.2017.1382671] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Phosphorylation is a dynamic post-translational modification that can alter protein structure, localization, protein-protein interactions and stability. All of the identified tight junction transmembrane proteins can be multiply phosphorylated, but only in a few cases are the consequences of phosphorylation at specific sites well characterized. The goal of this review is to highlight some of the best understood examples of phosphorylation changes in the integral membrane tight junction proteins in the context of more general overview of the effects of phosphorylation throughout the proteome. We expect as that structural information for the tight junction proteins becomes more widely available and the molecular modeling algorithms improve, so will our understanding of the relevance of phosphorylation changes at single and multiple sites in tight junction proteins.
Collapse
Affiliation(s)
- Christina M Van Itallie
- a National Heart, Lung and Blood Institute , National Institutes of Health , Bethesda , MD , USA
| | - James M Anderson
- a National Heart, Lung and Blood Institute , National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
33
|
Kanayasu-Toyoda T, Ishii-Watabe A, Kikuchi Y, Kitagawa H, Suzuki H, Tamura H, Tada M, Suzuki T, Mizuguchi H, Yamaguchi T. Occludin as a functional marker of vascular endothelial cells on tube-forming activity. J Cell Physiol 2017; 233:1700-1711. [PMID: 28681912 DOI: 10.1002/jcp.26082] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 07/05/2017] [Indexed: 01/03/2023]
Abstract
Cell therapy using endothelial progenitor cells (EPCs) is a promising strategy for the treatment of ischemic diseases. Two types of EPCs have been identified: early EPCs and late EPCs. Late EPCs are able to form tube structure by themselves, and have a high proliferative ability. The functional marker(s) of late EPCs, which relate to their therapeutic potential, have not been fully elucidated. Here we compared the gene expression profiles of several human cord blood derived late EPC lines which exhibit different tube formation activity, and we observed that the expression of occludin (OCLN) in these lines correlated with the tube formation ability, suggesting that OCLN is a candidate functional marker of late EPCs. When OCLN was knocked down by transfecting siRNA, the tube formation on Matrigel, the S phase + G2 /M phase in the cell cycle, and the spheroid-based sprouting of late EPCs were markedly reduced, suggesting the critical role of OCLN in tube formation, sprouting, and proliferation. These results indicated that OCLN plays a novel role in neovascularization and angiogenesis.
Collapse
Affiliation(s)
- Toshie Kanayasu-Toyoda
- Department of Pharmacology, Nihon Pharmaceutical University, Komuro, Inamachi, Kita-adachigun, Saitama, Japan.,Division of Microbiology, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Yutaka Kikuchi
- Division of Microbiology, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Hiroko Kitagawa
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Hiroko Suzuki
- The Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo, Japan
| | - Hiroomi Tamura
- The Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo, Japan
| | - Minoru Tada
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Takuo Suzuki
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Teruhide Yamaguchi
- Department of Pharmacology, Nihon Pharmaceutical University, Komuro, Inamachi, Kita-adachigun, Saitama, Japan.,Division of Microbiology, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| |
Collapse
|
34
|
Díaz-Coránguez M, Ramos C, Antonetti DA. The inner blood-retinal barrier: Cellular basis and development. Vision Res 2017; 139:123-137. [PMID: 28619516 DOI: 10.1016/j.visres.2017.05.009] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 01/01/2023]
Abstract
The blood-retinal barrier (BRB) regulates transport across retinal capillaries maintaining proper neural homeostasis and protecting the neural tissue from potential blood borne toxicity. Loss of the BRB contributes to the pathophysiology of a number of blinding retinal diseases including diabetic retinopathy. In this review, we address the basis of the BRB, including the molecular mechanisms that regulate flux across the retinal vascular bed. The routes of transcellular and paracellular flux are described as well as alterations in these pathways in response to permeabilizing agents in diabetes. Finally, we provide information on exciting new studies that help to elucidate the process of BRB development or barriergenesis and how understanding this process may lead to new opportunities for barrier restoration in diabetic retinopathy.
Collapse
Affiliation(s)
- Mónica Díaz-Coránguez
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States
| | - Carla Ramos
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|