1
|
García DM. Retinal physiology in metabolic syndrome. ADVANCES IN GENETICS 2025; 113:76-101. [PMID: 40409801 DOI: 10.1016/bs.adgen.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Obesity is increasingly recognized not only for its systemic health impacts but also for its association with visual defects and eye diseases. This chapter explores the relationship between obesity and ocular health, highlighting the mechanisms by which metabolic dysregulation influences visual outcomes. Obesity exacerbates risk factors such as hypertension, dyslipidemia, and insulin resistance, which compromise retinal and optic nerve health. Conditions like diabetic retinopathy, age-related macular degeneration, and glaucoma are discussed in the context of obesity-related inflammation, oxidative stress, and altered vascular function, focusing on the retina as one of the body's most metabolically demanding tissues. Key pathways include adipose-derived cytokines that disrupt retinal homeostasis, and the effects of insulin resistance on retinal cells and vasculature. Furthermore, this chapter covers emerging evidence on the advances of genetic factors linking diabetic retinopathy to retinal impairments. By elucidating these interactions, we aim to provide insight into preventive and therapeutic strategies that could mitigate vision loss among individuals with obesity.
Collapse
Affiliation(s)
- David Meseguer García
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
2
|
Cai C, Gu C, Meng C, He S, Thashi L, Deji D, Zheng Z, Qiu Q. Therapeutic Effects of Metformin on Central Nervous System Diseases: A Focus on Protection of Neurovascular Unit. Pharm Res 2024; 41:1907-1920. [PMID: 39375240 DOI: 10.1007/s11095-024-03777-0] [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: 02/20/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024]
Abstract
Metformin is one of the most commonly used oral hypoglycemic drugs in clinical practice, with unique roles in neurodegeneration and vascular lesions. Neurodegeneration and vasculopathy coexist in many diseases and typically affect the neurovascular unit (NVU), a minimal structural and functional unit in the central nervous system. Its components interact with one another and are indispensable for maintaining tissue homeostasis. This review focuses on retinal (diabetic retinopathy, retinitis pigmentosa) and cerebral (ischemic stroke, Alzheimer's disease) diseases to explore the effects of metformin on the NVU. Metformin has a preliminarily confirmed therapeutic effect on the retinal NUV, affecting many of its components, such as photoreceptors (cones and rods), microglia, ganglion, Müller, and vascular endothelial cells. Since it rapidly penetrates the blood-brain barrier (BBB) and accumulates in the brain, metformin also has an extensively studied neuronal protective effect in neuronal diseases. Its mechanism affects various NVU components, including pericytes, astrocytes, microglia, and vascular endothelial cells, mainly serving to protect the BBB. Regulating the inflammatory response in NVU (especially neurons and microglia) may be the main mechanism of metformin in improving central nervous system related diseases. Metformin may be a potential drug for treating diseases associated with NVU deterioration, however, more trials are needed to validate its timing, duration, dose, clinical effects, and side effects.
Collapse
Affiliation(s)
- Chunyang Cai
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, No. 1111 Xianxia Road, Changning District, Shanghai, 200050, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai engineering center for precise diagnosis and treatment of eye diseases, Shanghai, PR China
| | - Chufeng Gu
- Department of Ophthalmology, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Shengli Clinical College of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Chunren Meng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Shuai He
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, No. 1111 Xianxia Road, Changning District, Shanghai, 200050, PR China
| | - Lhamo Thashi
- Department of Ophthalmology, Shigatse People's Hospital, Shigatse, Tibet, PR China
| | - Draga Deji
- Department of Ophthalmology, Shigatse People's Hospital, Shigatse, Tibet, PR China
| | - Zhi Zheng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai engineering center for precise diagnosis and treatment of eye diseases, Shanghai, PR China.
| | - Qinghua Qiu
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, No. 1111 Xianxia Road, Changning District, Shanghai, 200050, PR China.
- Department of Ophthalmology, Shigatse People's Hospital, Shigatse, Tibet, PR China.
- High Altitude Ocular Disease Research Center of People's Hospital of Shigatse City and Tongren Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
3
|
Martins B, Pires M, Ambrósio AF, Girão H, Fernandes R. Contribution of extracellular vesicles for the pathogenesis of retinal diseases: shedding light on blood-retinal barrier dysfunction. J Biomed Sci 2024; 31:48. [PMID: 38730462 PMCID: PMC11088087 DOI: 10.1186/s12929-024-01036-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
Retinal degenerative diseases, including diabetic retinopathy (DR) and age-related macular degeneration (AMD), loom as threats to vision, causing detrimental effects on the structure and function of the retina. Central to understanding these diseases, is the compromised state of the blood-retinal barrier (BRB), an effective barrier that regulates the influx of immune and inflammatory components. Whether BRB breakdown initiates retinal distress, or is a consequence of disease progression, remains enigmatic. Nevertheless, it is an indication of retinal dysfunction and potential vision loss.The intricate intercellular dialogues among retinal cell populations remain unintelligible in the complex retinal milieu, under conditions of inflammation and oxidative stress. The retina, a specialized neural tissue, sustains a ceaseless demand for oxygen and nutrients from two vascular networks. The BRB orchestrates the exchange of molecules and fluids within this specialized region, comprising the inner BRB (iBRB) and the outer BRB (oBRB). Extracellular vesicles (EVs) are small membranous structures, and act as messengers facilitating intercellular communication in this milieu.EVs, both from retinal and peripheral immune cells, increase complexity to BRB dysfunction in DR and AMD. Laden with bioactive cargoes, these EVs can modulate the retinal microenvironment, influencing disease progression. Our review delves into the multifaceted role of EVs in retinal degenerative diseases, elucidating the molecular crosstalk they orchestrate, and their microRNA (miRNA) content. By shedding light on these nanoscale messengers, from their biogenesis, release, to interaction and uptake by target cells, we aim to deepen the comprehension of BRB dysfunction and explore their therapeutic potential, therefore increasing our understanding of DR and AMD pathophysiology.
Collapse
Affiliation(s)
- Beatriz Martins
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, 3000-548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
| | - Maria Pires
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, 3000-548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
| | - António Francisco Ambrósio
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3004-561, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, 3000-548, Portugal
| | - Henrique Girão
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3004-561, Portugal
| | - Rosa Fernandes
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal.
- University of Coimbra, Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, 3000-548, Portugal.
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal.
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3004-561, Portugal.
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, 3000-548, Portugal.
| |
Collapse
|
4
|
Vonniessen B, Tabariès S, Siegel PM. Antibody-mediated targeting of Claudins in cancer. Front Oncol 2024; 14:1320766. [PMID: 38371623 PMCID: PMC10869466 DOI: 10.3389/fonc.2024.1320766] [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: 10/12/2023] [Accepted: 01/09/2024] [Indexed: 02/20/2024] Open
Abstract
Tight junctions (TJs) are large intercellular adhesion complexes that maintain cell polarity in normal epithelia and endothelia. Claudins are critical components of TJs, forming homo- and heteromeric interaction between adjacent cells, which have emerged as key functional modulators of carcinogenesis and metastasis. Numerous epithelial-derived cancers display altered claudin expression patterns, and these aberrantly expressed claudins have been shown to regulate cancer cell proliferation/growth, metabolism, metastasis and cell stemness. Certain claudins can now be used as biomarkers to predict patient prognosis in a variety of solid cancers. Our understanding of the distinct roles played by claudins during the cancer progression has progressed significantly over the last decade and claudins are now being investigated as possible diagnostic markers and therapeutic targets. In this review, we will summarize recent progress in the use of antibody-based or related strategies for targeting claudins in cancer treatment. We first describe pre-clinical studies that have facilitated the development of neutralizing antibodies and antibody-drug-conjugates targeting Claudins (Claudins-1, -3, -4, -6 and 18.2). Next, we summarize clinical trials assessing the efficacy of antibodies targeting Claudin-6 or Claudin-18.2. Finally, emerging strategies for targeting Claudins, including Chimeric Antigen Receptor (CAR)-T cell therapy and Bi-specific T cell engagers (BiTEs), are also discussed.
Collapse
Affiliation(s)
- Benjamin Vonniessen
- Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Medicine, McGill University, Montréal, QC, Canada
| | - Sébastien Tabariès
- Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Medicine, McGill University, Montréal, QC, Canada
| | - Peter M. Siegel
- Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Medicine, McGill University, Montréal, QC, Canada
- Department of Biochemistry, McGill University, Montréal, QC, Canada
- Department of Anatomy & Cell Biology, McGill University, Montréal, QC, Canada
- Department of Oncology, McGill University, Montréal, QC, Canada
| |
Collapse
|
5
|
Li X, Owen LA, Taylor KD, Ostmo S, Chen YDI, Coyner AS, Sonmez K, Hartnett ME, Guo X, Ipp E, Roll K, Genter P, Chan RVP, DeAngelis MM, Chiang MF, Campbell JP, Rotter JI. Genome-wide association identifies novel ROP risk loci in a multiethnic cohort. Commun Biol 2024; 7:107. [PMID: 38233474 PMCID: PMC10794688 DOI: 10.1038/s42003-023-05743-9] [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: 05/07/2023] [Accepted: 12/26/2023] [Indexed: 01/19/2024] Open
Abstract
We conducted a genome-wide association study (GWAS) in a multiethnic cohort of 920 at-risk infants for retinopathy of prematurity (ROP), a major cause of childhood blindness, identifying 1 locus at genome-wide significance level (p < 5×10-8) and 9 with significance of p < 5×10-6 for ROP ≥ stage 3. The most significant locus, rs2058019, reached genome-wide significance within the full multiethnic cohort (p = 4.96×10-9); Hispanic and European Ancestry infants driving the association. The lead single nucleotide polymorphism (SNP) falls in an intronic region within the Glioma-associated oncogene family zinc finger 3 (GLI3) gene. Relevance for GLI3 and other top-associated genes to human ocular disease was substantiated through in-silico extension analyses, genetic risk score analysis and expression profiling in human donor eye tissues. Thus, we identify a novel locus at GLI3 with relevance to retinal biology, supporting genetic susceptibilities for ROP risk with possible variability by race and ethnicity.
Collapse
Affiliation(s)
- Xiaohui Li
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Leah A Owen
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, USA.
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA.
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT, USA.
- Department of Ophthalmology, University at Buffalo the State University of New York, Buffalo, NY, USA.
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Susan Ostmo
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Aaron S Coyner
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Kemal Sonmez
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | | | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Eli Ipp
- Division of Endocrinology and Metabolism, Department of Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kathryn Roll
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Pauline Genter
- Division of Endocrinology and Metabolism, Department of Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - R V Paul Chan
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Margaret M DeAngelis
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
- Department of Ophthalmology, University at Buffalo the State University of New York, Buffalo, NY, USA
- Department of Biochemistry; Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/State University of New York (SUNY), Buffalo, NY, USA
- Department of Neuroscience; Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/State University of New York (SUNY), Buffalo, NY, USA
- Department of Genetics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/State University of New York (SUNY), Buffalo, NY, USA
| | - Michael F Chiang
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
- National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - J Peter Campbell
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA.
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA.
| |
Collapse
|
6
|
Gjervan SC, Ozgoren OK, Gow A, Stockler-Ipsiroglu S, Pouladi MA. Claudin-11 in health and disease: implications for myelin disorders, hearing, and fertility. Front Cell Neurosci 2024; 17:1344090. [PMID: 38298375 PMCID: PMC10827939 DOI: 10.3389/fncel.2023.1344090] [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: 11/25/2023] [Accepted: 12/31/2023] [Indexed: 02/02/2024] Open
Abstract
Claudin-11 plays a critical role in multiple physiological processes, including myelination, auditory function, and spermatogenesis. Recently, stop-loss mutations in CLDN11 have been identified as a novel cause of hypomyelinating leukodystrophy (HLD22). Understanding the multifaceted roles of claudin-11 and the potential pathogenic mechanisms in HLD22 is crucial for devising targeted therapeutic strategies. This review outlines the biological roles of claudin-11 and the implications of claudin-11 loss in the context of the Cldn11 null mouse model. Additionally, HLD22 and proposed pathogenic mechanisms, such as endoplasmic reticulum stress, will be discussed.
Collapse
Affiliation(s)
- Sophia C. Gjervan
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Djavad Mowafaghian Centre for Brain Health, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Oguz K. Ozgoren
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Djavad Mowafaghian Centre for Brain Health, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Alexander Gow
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Sylvia Stockler-Ipsiroglu
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
- Division of Biochemical Genetics, The University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Mahmoud A. Pouladi
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Djavad Mowafaghian Centre for Brain Health, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
7
|
Castillo-González J, Ruiz JL, Serrano-Martínez I, Forte-Lago I, Ubago-Rodriguez A, Caro M, Pérez-Gómez JM, Benítez-Troncoso A, Andrés-León E, Sánchez-Navarro M, Luque RM, González-Rey E. Cortistatin deficiency reveals a dysfunctional brain endothelium with impaired gene pathways, exacerbated immune activation, and disrupted barrier integrity. J Neuroinflammation 2023; 20:226. [PMID: 37794493 PMCID: PMC10548650 DOI: 10.1186/s12974-023-02908-5] [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: 04/03/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Brain activity governing cognition and behaviour depends on the fine-tuned microenvironment provided by a tightly controlled blood-brain barrier (BBB). Brain endothelium dysfunction is a hallmark of BBB breakdown in most neurodegenerative/neuroinflammatory disorders. Therefore, the identification of new endogenous molecules involved in endothelial cell disruption is essential to better understand BBB dynamics. Cortistatin is a neuroimmune mediator with anti-inflammatory and neuroprotective properties that exerts beneficial effects on the peripheral endothelium. However, its role in the healthy and injured brain endothelium remains to be evaluated. Herein, this study aimed to investigate the potential function of endogenous and therapeutic cortistatin in regulating brain endothelium dysfunction in a neuroinflammatory/neurodegenerative environment. METHODS Wild-type and cortistatin-deficient murine brain endothelium and human cells were used for an in vitro barrier model, where a simulated ischemia-like environment was mimicked. Endothelial permeability, junction integrity, and immune response in the presence and absence of cortistatin were evaluated using different size tracers, immunofluorescence labelling, qPCR, and ELISA. Cortistatin molecular mechanisms underlying brain endothelium dynamics were assessed by RNA-sequencing analysis. Cortistatin role in BBB leakage was evaluated in adult mice injected with LPS. RESULTS The endogenous lack of cortistatin predisposes endothelium weakening with increased permeability, tight-junctions breakdown, and dysregulated immune activity. We demonstrated that both damaged and uninjured brain endothelial cells isolated from cortistatin-deficient mice, present a dysregulated and/or deactivated genetic programming. These pathways, related to basic physiology but also crucial for the repair after damage (e.g., extracellular matrix remodelling, angiogenesis, response to oxygen, signalling, and metabolites transport), are dysfunctional and make brain endothelial barrier lacking cortistatin non-responsive to any further injury. Treatment with cortistatin reversed in vitro hyperpermeability, tight-junctions disruption, inflammatory response, and reduced in vivo BBB leakage. CONCLUSIONS The neuropeptide cortistatin has a key role in the physiology of the cerebral microvasculature and its presence is crucial to develop a canonical balanced response to damage. The reparative effects of cortistatin in the brain endothelium were accompanied by the modulation of the immune function and the rescue of barrier integrity. Cortistatin-based therapies could emerge as a novel pleiotropic strategy to ameliorate neuroinflammatory/neurodegenerative disorders with disrupted BBB.
Collapse
Affiliation(s)
- Julia Castillo-González
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - José Luis Ruiz
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Ignacio Serrano-Martínez
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Irene Forte-Lago
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Ana Ubago-Rodriguez
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Marta Caro
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Jesús Miguel Pérez-Gómez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, 14004, Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | | | - Eduardo Andrés-León
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Macarena Sánchez-Navarro
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, 14004, Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Elena González-Rey
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain.
| |
Collapse
|
8
|
Robles-Osorio ML, Sabath E. Tight junction disruption and the pathogenesis of the chronic complications of diabetes mellitus: A narrative review. World J Diabetes 2023; 14:1013-1026. [PMID: 37547580 PMCID: PMC10401447 DOI: 10.4239/wjd.v14.i7.1013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/20/2023] [Accepted: 05/23/2023] [Indexed: 07/12/2023] Open
Abstract
The chronic complications of diabetes mellitus constitute a major public health problem. For example, diabetic eye diseases are the most important cause of blindness, and diabetic nephropathy is the most frequent cause of chronic kidney disease worldwide. The cellular and molecular mechanisms of these chronic complications are still poorly understood, preventing the development of effective treatment strategies. Tight junctions (TJs) are epithelial intercellular junctions located at the most apical region of cell-cell contacts, and their main function is to restrict the passage of molecules through the paracellular space. The TJs consist of over 40 proteins, and the most important are occludin, claudins and the zonula occludens. Accumulating evidence suggests that TJ disruption in different organs, such as the brain, nerves, retina and kidneys, plays a fundamental pathophysiological role in the development of chronic complications. Increased permeability of the blood-brain barrier and the blood-retinal barrier has been demonstrated in diabetic neuropathy, brain injury and diabetic retinopathy. The consequences of TJ disruption on kidney function or progression of kidney disease are currently unknown. In the present review, we highlighted the molecular events that lead to barrier dysfunction in diabetes. Further investigation of the mechanisms underlying TJ disruption is expected to provide new insights into therapeutic approaches to ameliorate the chronic complications of diabetes mellitus.
Collapse
Affiliation(s)
| | - Ernesto Sabath
- Renal and Metabolism Unit, Hospital General de Querétaro, Queretaro 76180, Mexico
- Department of Nutrition, Universidad Autónoma de Queretaro, Queretaro 76230, Mexico
| |
Collapse
|
9
|
Braunger BM, Gießl A, Schlötzer-Schrehardt U. The Blood-ocular Barriers and their Dysfunction: Anatomy, Physiology, Pathology. Klin Monbl Augenheilkd 2023; 240:650-661. [PMID: 37207638 DOI: 10.1055/a-2063-8957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Complex barriers comprise the blood-aqueous (BAB) and the blood-retinal barrier (BRB), and separate anterior and posterior eye chambers, vitreous body, and sensory retina from the circulation. They prevent pathogens and toxins from entering the eye, control movement of fluid, proteins, and metabolites, and contribute to the maintenance of the ocular immune status. Morphological correlates of blood-ocular barriers are tight junctions between neighboring endothelial and epithelial cells, which function as gatekeepers of the paracellular transport of molecules, thereby limiting their uncontrolled access to ocular chambers and tissues. The BAB is composed of tight junctions between endothelial cells of the iris vasculature, endothelial cells of Schlemm's canal inner wall, and cells of the nonpigmented ciliary epithelium. The BRB consists of tight junctions between endothelial cells of the retinal vessels (inner BRB) and epithelial cells of the retinal pigment epithelium (outer BRB). These junctional complexes respond rapidly to pathophysiological changes, thus enabling vascular leakage of blood-derived molecules and inflammatory cells into ocular tissues and chambers. Blood-ocular barrier function, which can be clinically measured by laser flare photometry or fluorophotometry, is compromised in traumatic, inflammatory, or infectious processes, but also frequently contributes to the pathophysiology of chronic diseases of the anterior eye segment and the retina, as exemplified by diabetic retinopathy and age-related macular degeneration.
Collapse
Affiliation(s)
- Barbara M Braunger
- Institut für Anatomie und Zellbiologie, Julius-Maximilians-Universität Würzburg, Medizinische Fakultät, Deutschland
| | - Andreas Gießl
- Augenklinik, Friedrich-Alexander-Universität Erlangen-Nürnberg, Medizinische Fakultät, Erlangen, Deutschland
| | - Ursula Schlötzer-Schrehardt
- Augenklinik, Friedrich-Alexander-Universität Erlangen-Nürnberg, Medizinische Fakultät, Erlangen, Deutschland
| |
Collapse
|
10
|
Menero-Valdés P, Lores-Padín A, Fernández B, Quarles CD, García M, González-Iglesias H, Pereiro R. Determination and localization of specific proteins in individual ARPE-19 cells by single cell and laser ablation ICP-MS using iridium nanoclusters as label. Talanta 2023; 253:123974. [PMID: 36195026 DOI: 10.1016/j.talanta.2022.123974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/13/2022] [Accepted: 09/25/2022] [Indexed: 12/13/2022]
Abstract
Single cell-inductively coupled plasma-mass spectrometry (sc-ICP-MS) and laser ablation (LA)-ICP-MS have been complementary employed to develop a comprehensive study of APOE and claudin-1 expression in ARPE-19 cells submitted to a glucose treatment (100 mM, 48 h) that induces oxidative stress conditions. Results were compared with control cells. The determination of the two proteins by ICP-MS was sequentially carried out using specific immunoprobes labelled with IrNCs that offer a huge amplification (1760 ± 90 atoms of Ir on average). A novel sample introduction system, the microFAST Single Cell set-up, was employed for sc-ICP-MS analysis. This introduction system resulted in a cellular transport efficiency of 85 ± 9% for ARPE-19 cells (91 ± 5% using a PtNPs standard). After the proper immunocytochemistry protocol with the specific IrNCs immunoprobes in cell suspensions (sc-ICP-MS), the mass of APOE and claudin-1 in individual ARPE-19 cells was obtained. Average detection limits per cell by sc-ICP-MS were 0.02 fg of APOE and 3 ag of claudin-1. The results of sample analyses obtained by sc-ICP-MS were validated with commercial ELISA kits. The distribution of both target proteins in individual cells (fixated in the chamber wall) was unveiled by LA-ICP-MS. The high amplification provided by the IrNCs immunoprobes allowed the identification of APOE and claudin-1 within individual ARPE-19 cells. High resolution images were obtained using a laser spot of 2 × 2 μm.
Collapse
Affiliation(s)
- Paula Menero-Valdés
- Department of Physical and Analytical Chemistry, University of Oviedo, Julian Clavería 8, Oviedo, 33006, Spain
| | - Ana Lores-Padín
- Department of Physical and Analytical Chemistry, University of Oviedo, Julian Clavería 8, Oviedo, 33006, Spain
| | - Beatriz Fernández
- Department of Physical and Analytical Chemistry, University of Oviedo, Julian Clavería 8, Oviedo, 33006, Spain.
| | - C Derrick Quarles
- Elemental Scientific, Inc., 7277 World Communications Drive, Omaha, NE, 68122, USA
| | - Montserrat García
- Instituto Oftalmológico Fernández-Vega, Avda. Dres. Fernández-Vega, 34, Oviedo, 33012, Spain; Department of Cellular Morphology and Biology, Faculty of Medicine, Julian Clavería, Oviedo, 33006, Spain
| | - Héctor González-Iglesias
- Department of Technology and Biotechnology of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain
| | - Rosario Pereiro
- Department of Physical and Analytical Chemistry, University of Oviedo, Julian Clavería 8, Oviedo, 33006, Spain.
| |
Collapse
|
11
|
Goncalves A, Antonetti DA. Transgenic animal models to explore and modulate the blood brain and blood retinal barriers of the CNS. Fluids Barriers CNS 2022; 19:86. [PMID: 36320068 PMCID: PMC9628113 DOI: 10.1186/s12987-022-00386-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022] Open
Abstract
The unique environment of the brain and retina is tightly regulated by blood-brain barrier and the blood-retinal barrier, respectively, to ensure proper neuronal function. Endothelial cells within these tissues possess distinct properties that allow for controlled passage of solutes and fluids. Pericytes, glia cells and neurons signal to endothelial cells (ECs) to form and maintain the barriers and control blood flow, helping to create the neurovascular unit. This barrier is lost in a wide range of diseases affecting the central nervous system (CNS) and retina such as brain tumors, stroke, dementia, and in the eye, diabetic retinopathy, retinal vein occlusions and age-related macular degeneration to name prominent examples. Recent studies directly link barrier changes to promotion of disease pathology and degradation of neuronal function. Understanding how these barriers form and how to restore these barriers in disease provides an important point for therapeutic intervention. This review aims to describe the fundamentals of the blood-tissue barriers of the CNS and how the use of transgenic animal models led to our current understanding of the molecular framework of these barriers. The review also highlights examples of targeting barrier properties to protect neuronal function in disease states.
Collapse
Affiliation(s)
- Andreia Goncalves
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St Rm, Ann Arbor, MI, 7317, USA
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St Rm, Ann Arbor, MI, 7317, USA.
| |
Collapse
|
12
|
Zhang S, Gan L, Cao F, Wang H, Gong P, Ma C, Ren L, Lin Y, Lin X. The barrier and interface mechanisms of the brain barrier, and brain drug delivery. Brain Res Bull 2022; 190:69-83. [PMID: 36162603 DOI: 10.1016/j.brainresbull.2022.09.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 08/25/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022]
Abstract
Three different barriers are formed between the cerebrovascular and the brain parenchyma: the blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the cerebrospinal fluid-brain barrier (CBB). The BBB is the main regulator of blood and central nervous system (CNS) material exchange. The semipermeable nature of the BBB limits the passage of larger molecules and hydrophilic small molecules, Food and Drug Administration (FDA)-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Although the complexity of the BBB affects CNS drug delivery, understanding the composition and function of the BBB can provide a platform for the development of new methods for CNS drug delivery. This review summarizes the classification of the brain barrier, the composition and role of the basic structures of the BBB, and the transport, barrier, and destruction mechanisms of the BBB; discusses the advantages and disadvantages of different drug delivery methods and prospects for future drug delivery strategies.
Collapse
Affiliation(s)
- Shanshan Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, Zhejiang Province, China
| | - Lin Gan
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Fengye Cao
- Yiyang The First Hospital of Traditional Chinese Medicine, Yiyang, Hunan Province, 413000, China
| | - Hao Wang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Peng Gong
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Congcong Ma
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Li Ren
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Yubo Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Xianming Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China.
| |
Collapse
|
13
|
Zhang F, Wang R, Huang J, Zhang H, Wang L, Zhong Z, Ruan J, Liu H. Blood–brain barrier permeability to enrofloxacin in Pengze crucian carp (
Carassius auratus var
. Pengze). Vet Med Sci 2022; 8:2404-2410. [DOI: 10.1002/vms3.918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Fan Zhang
- Department of Aquaculture College of Animal Science & Technology Jiangxi Agricultural University Nanchang P.R. China
| | - Runping Wang
- Department of Aquaculture College of Animal Science & Technology Jiangxi Agricultural University Nanchang P.R. China
| | - Jianzhen Huang
- Department of Aquaculture College of Animal Science & Technology Jiangxi Agricultural University Nanchang P.R. China
| | - Haixin Zhang
- Jiangxi Fisheries Research Institute Nanchang P.R. China
| | - Long Wang
- Bureau of Agriculture and Rural Affairs of Pengze County Pengze P.R. China
| | - Zhiwei Zhong
- Department of Aquaculture College of Animal Science & Technology Jiangxi Agricultural University Nanchang P.R. China
| | - Jiming Ruan
- Department of Aquaculture College of Animal Science & Technology Jiangxi Agricultural University Nanchang P.R. China
| | - Huazhong Liu
- College of Chemistry & Environmental Science Guangdong Ocean University Zhanjiang P.R. China
| |
Collapse
|
14
|
Herrera RA, Deshpande K, Martirosian V, Saatian B, Julian A, Eisenbarth R, Das D, Iyer M, Neman J. Cortisol promotes breast-to-brain metastasis through the blood-cerebrospinal fluid barrier. Cancer Rep (Hoboken) 2022; 5:e1351. [PMID: 33635590 PMCID: PMC9124512 DOI: 10.1002/cnr2.1351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/20/2020] [Accepted: 12/02/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Elevated basal cortisol levels are present in women with primary and metastatic breast cancer. Although cortisol's potential role in breast-to-brain metastasis has yet to be sufficiently studied, prior evidence indicates that it functions as a double-edged sword-cortisol induces breast cancer metastasis in vivo, but strengthens the blood-brain-barrier (BBB) to protect the brain from microbes and peripheral immune cells. AIMS In this study, we provide a novel examination on whether cortisol's role in tumor invasiveness eclipses its supporting role in strengthening the CNS barriers. We expanded our study to include the blood-cerebrospinal fluid barrier (BCSFB), an underexamined site of tumor entry. METHODS AND RESULTS Utilizing in vitro BBB and BCSFB models to measure barrier strength in the presence of hydrocortisone (HC). We established that lung tumor cells migrate through both CNS barriers equally while breast tumors cells preferentially migrate through the BCSFB. Furthermore, HC treatment increased breast-to-brain metastases (BBM) but not primary breast tumor migratory capacity. When examining the transmigration of breast cancer cells across the BCSFB, we demonstrate that HC induces increased traversal of BBM but not primary breast cancer. We provide evidence that HC increases tightness of the BCSFB akin to the BBB by upregulating claudin-5, a tight junction protein formerly acknowledged as exclusive to the BBB. CONCLUSION Our findings indicate, for the first time that increased cortisol levels facilitate breast-to-brain metastasis through the BCSFB-a vulnerable point of entry which has been typically overlooked in brain metastasis. Our study suggests cortisol plays a pro-metastatic role in breast-to-brain metastasis and thus caution is needed when using glucocorticoids to treat breast cancer patients.
Collapse
Affiliation(s)
- Robert A. Herrera
- Department of Molecular Microbiology and ImmunologyKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Krutika Deshpande
- Department of Neurological SurgeryKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Vahan Martirosian
- Department of Neurological SurgeryKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Behnaz Saatian
- Department of Neurological SurgeryKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Alex Julian
- Department of Neurological SurgeryKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Rachel Eisenbarth
- Department of Neurological SurgeryKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Diganta Das
- Department of Neurological SurgeryKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Mukund Iyer
- Department of Neurological SurgeryKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Josh Neman
- Department of Neurological SurgeryKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Physiology and NeuroscienceKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
- Norris Comprehensive Cancer CenterKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
- Brain Tumor CenterKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
15
|
Ouyang H, Du A, Zhou L, Zhang T, Lu B, Wang Z, Ji L. Chlorogenic acid improves diabetic retinopathy by alleviating blood-retinal-barrier dysfunction via inducing Nrf2 activation. Phytother Res 2022; 36:1386-1401. [PMID: 35133045 DOI: 10.1002/ptr.7401] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/29/2021] [Accepted: 01/14/2022] [Indexed: 12/29/2022]
Abstract
As one of the major diabetic microvascular complications, diabetic retinopathy (DR) is mainly initiated by the blood-retinal barrier (BRB) dysfunction. Chlorogenic acid (CGA) is a natural polyphenolic compound in Lonicerae Japonicae Flos, which traditionally has the beneficial function for eyes and is commonly included in many anti-diabetic formulas. In this study, the potential protective mechanism of CGA against DR was investigated. Streptozotocin (STZ) was used to induce diabetes in mice. CGA attenuated BRB dysfunction and reversed endothelial-mesenchymal transition (EndoMT) and epithelial-mesenchymal transition (EMT) in retinas in vivo. CGA inhibited microglia activation and reduced tumor necrosis factor (TNF)α release both in vivo and in vitro. CGA promoted nuclear factor erythroid 2-related factor 2 (Nrf2) activation and prevented EndoMT/EMT in TNFα-treated human retinal endothelial cells (HRECs) or retinal pigment epithelial APRE19 cells. CGA alleviated endothelial/epithelial barrier oxidative injury in HRECs or APRE19 cells stimulated with TNFα, but this effect was disappeared in cells co-incubated with Nrf2 inhibitor. Additionally, the CGA-supplied alleviation on BRB damage and EndoMT/EMT was markedly weakened in retinas from STZ-treated Nrf2 knock-out mice. All results suggest that CGA improves DR through attenuating BRB injury by reducing microglia-initiated inflammation and preventing TNFα-induced EndoMT/EMT and oxidative injury via inducing Nrf2 activation.
Collapse
Affiliation(s)
- Hao Ouyang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ao Du
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lingyu Zhou
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianyu Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhengtao Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
16
|
Nahar N, Mohamed S, Mustapha NM, Fong LS, Mohd Ishak NI. Gallic acid and myricetin-rich Labisia pumila extract mitigated multiple diabetic eye disorders in rats. J Food Biochem 2021; 45:e13948. [PMID: 34622461 DOI: 10.1111/jfbc.13948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 12/15/2022]
Abstract
Diabetes affected about a quarter of a billion people globally, and one out of four diabetics has eye or vision problems. This study investigated whether gallic acid and myricetin-rich Labisia pumila extract (LP) consumption would help prevent diabetic eye disorders and some probable biochemistry involved relating to inflammation, vascular leakage, and oxidative tension. Male rats were divided into four groups (n = 6), namely healthy control, diabetic non-treated control, and hyperglycemic rats treated with 150 or 300 mg/kg LP. Intraperitoneal injection of 60 mg/kg streptozotocin was used to induce diabetes. Rats were fed in the morning and evening. Diabetic retinopathy was graded in rats using a dilated retinal digital ophthalmoscopy. Rats were sacrificed at 12 weeks and the retina, optic nerve, cornea, lens, sclera, ciliary bodies, iris, and conjunctiva were examined histologically. The diabetic rats consuming LP for 10 weeks showed dose-dependent, histopathologically-reduced eye abnormalities (keratopathy, cataract, sclera, conjunctiva, ciliary bodies, iris, limbus, corneal edema, epithelial barrier inefficiency, shallow punctate keratitis, lower basal layer cell density, retinopathy, glaucoma, and corneal changes). The LP significantly suppressed inflammation [increased serum tumor necrosis factor-α (TNF-α), prostaglandin-E2 (PGE2)], vascular leakage [claudin-1], abnormal vascularization [vascular endothelial growth factor (VEGF)], oxidative tension [malondialdehyde/reduced glutathione ratio], and hyperglycemia [fasting blood glucose] of the diabetic rats. The LP consumption was significantly protective against diabetic eye disorders and optic nerve dysfunction which were related to inflammation, vascular leakage, abnormal vascularization, and oxidative tension, which most likely influenced eye hemorrhage and collagen cross-linkage. PRACTICAL APPLICATIONS: The study shows that gallic acid and myricetin-rich Labisia pumila (LP) leaf consumption may be used as a complementary therapy for managing diabetes (fasting blood glucose) and preventing diabetic eye disorders (keratopathy, cataract, sclera, conjunctiva, ciliary bodies, iris, limbus, corneal edema, epithelial barrier inefficiency, shallow punctate keratitis, lower basal layer cell density, retinopathy, glaucoma, and corneal abnormalities). The LP consumptions reduced the serum biomarkers for inflammation (serum tumor necrosis factor-α TNF-α; prostaglandin-E2), vascular leakage/abnormalities (claudin-1 and vascular endothelial growth factor VEGF), and oxidative tension (malondialdehyde/reduced glutathione MDA/GSH ratio). The LP was eye-protective probably by normalizing fasting blood glucose, reducing inflammation, oxidative tension, vascular leakage, and irregular vascularization.
Collapse
Affiliation(s)
- Nazmun Nahar
- Institute of Bioscience, Universiti Putra Malaysia, UPM Serdang, Serdang, Malaysia
| | - Suhaila Mohamed
- Institute of Bioscience, Universiti Putra Malaysia, UPM Serdang, Serdang, Malaysia
| | | | - Lau Seng Fong
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, UPM Serdang, Serdang, Malaysia
| | | |
Collapse
|
17
|
Pediaditakis I, Kodella KR, Manatakis DV, Le CY, Hinojosa CD, Tien-Street W, Manolakos ES, Vekrellis K, Hamilton GA, Ewart L, Rubin LL, Karalis K. Modeling alpha-synuclein pathology in a human brain-chip to assess blood-brain barrier disruption. Nat Commun 2021; 12:5907. [PMID: 34625559 PMCID: PMC8501050 DOI: 10.1038/s41467-021-26066-5] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/15/2021] [Indexed: 01/08/2023] Open
Abstract
Parkinson's disease and related synucleinopathies are characterized by the abnormal accumulation of alpha-synuclein aggregates, loss of dopaminergic neurons, and gliosis of the substantia nigra. Although clinical evidence and in vitro studies indicate disruption of the Blood-Brain Barrier in Parkinson's disease, the mechanisms mediating the endothelial dysfunction is not well understood. Here we leveraged the Organs-on-Chips technology to develop a human Brain-Chip representative of the substantia nigra area of the brain containing dopaminergic neurons, astrocytes, microglia, pericytes, and microvascular brain endothelial cells, cultured under fluid flow. Our αSyn fibril-induced model was capable of reproducing several key aspects of Parkinson's disease, including accumulation of phosphorylated αSyn (pSer129-αSyn), mitochondrial impairment, neuroinflammation, and compromised barrier function. This model may enable research into the dynamics of cell-cell interactions in human synucleinopathies and serve as a testing platform for target identification and validation of novel therapeutics.
Collapse
Affiliation(s)
- Iosif Pediaditakis
- Emulate Inc., 27 Drydock Avenue, Boston, MA, USA.
- Serqet Therapeutics, Inc. 55 Cambridge Parkway, Suite 800E, Boston, MA, 02142, USA.
| | | | | | | | | | | | - Elias S Manolakos
- Department of Informatics and Telecommunications, National and Kapodistrian University of Athens, Athens, Greece
- Northeastern University, Bouvé College of Health Sciences, Boston, MA, USA
| | - Kostas Vekrellis
- Biomedical Research Foundation of Academy of Athens, Athens, Greece
| | | | - Lorna Ewart
- Emulate Inc., 27 Drydock Avenue, Boston, MA, USA
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Katia Karalis
- Emulate Inc., 27 Drydock Avenue, Boston, MA, USA.
- Endocrine Division, Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Rd, Tarrytown, NY, 10591, USA.
| |
Collapse
|
18
|
Scalise AA, Kakogiannos N, Zanardi F, Iannelli F, Giannotta M. The blood-brain and gut-vascular barriers: from the perspective of claudins. Tissue Barriers 2021; 9:1926190. [PMID: 34152937 PMCID: PMC8489939 DOI: 10.1080/21688370.2021.1926190] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In some organs, such as the brain, endothelial cells form a robust and highly selective blood-to-tissue barrier. However, in other organs, such as the intestine, endothelial cells provide less stringent permeability, to allow rapid exchange of solutes and nutrients where needed. To maintain the structural and functional integrity of the highly dynamic blood–brain and gut–vascular barriers, endothelial cells form highly specialized cell-cell junctions, known as adherens junctions and tight junctions. Claudins are a family of four-membrane-spanning proteins at tight junctions and they have both barrier-forming and pore-forming properties. Tissue-specific expression of claudins has been linked to different diseases that are characterized by barrier impairment. In this review, we summarize the more recent progress in the field of the claudins, with particular attention to their expression and function in the blood–brain barrier and the recently described gut–vascular barrier, under physiological and pathological conditions. Abbreviations: 22q11DS 22q11 deletion syndrome; ACKR1 atypical chemokine receptor 1; AD Alzheimer disease; AQP aquaporin; ATP adenosine triphosphate; Aβ amyloid β; BAC bacterial artificial chromosome; BBB blood-brain barrier; C/EBP-α CCAAT/enhancer-binding protein α; cAMP cyclic adenosine monophosphate (or 3ʹ,5ʹ-cyclic adenosine monophosphate); CD cluster of differentiation; CNS central nervous system; DSRED discosoma red; EAE experimental autoimmune encephalomyelitis; ECV304 immortalized endothelial cell line established from the vein of an apparently normal human umbilical cord; EGFP enhanced green fluorescent protein; ESAM endothelial cell-selective adhesion molecule; GLUT-1 glucose transporter 1; GVB gut-vascular barrier; H2B histone H2B; HAPP human amyloid precursor protein; HEK human embryonic kidney; JACOP junction-associated coiled coil protein; JAM junctional adhesion molecules; LYVE1 lymphatic vessel endothelial hyaluronan receptor 1; MADCAM1 mucosal vascular addressin cell adhesion molecule 1; MAPK mitogen-activated protein kinase; MCAO middle cerebral artery occlusion; MMP metalloprotease; MS multiple sclerosis; MUPP multi-PDZ domain protein; PATJ PALS-1-associated tight junction protein; PDGFR-α platelet-derived growth factor receptor α polypeptide; PDGFR-β platelet-derived growth factor receptor β polypeptide; RHO rho-associated protein kinase; ROCK rho-associated, coiled-coil-containing protein kinase; RT-qPCR real time quantitative polymerase chain reactions; PDGFR-β soluble platelet-derived growth factor receptor, β polypeptide; T24 human urinary bladder carcinoma cells; TG2576 transgenic mice expressing the human amyloid precursor protein; TNF-α tumor necrosis factor α; WTwild-type; ZO zonula occludens.
Collapse
|
19
|
Nahar N, Mohamed S, Mustapha NM, Lau S, Ishak NIM, Umran NS. Metformin attenuated histopathological ocular deteriorations in a streptozotocin-induced hyperglycemic rat model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:457-467. [PMID: 33047165 DOI: 10.1007/s00210-020-01989-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus (DM) often causes ocular disorders leading to vision loss. Metformin is commonly prescribed for type 2 diabetes. This study assessed the effect of metformin on hyperglycemic histopathological eye abnormalities and some possible pathways involved. Male rats were divided into 3 groups (N = 6), namely, healthy control, hyperglycemic non-treated control, and hyperglycemic rats treated with 200 mg/kg metformin. Two weeks after diabetes induction by an intraperitoneal streptozotocin (60 mg streptozotocin (STZ)/kg) injection, the rats develop ocular abnormalities, and metformin (200 mg/kg) treatment was administered daily. Rats underwent dilated retinal digital ophthalmoscope examination and graded for diabetic retinopathy. Rats were sacrificed at 12 weeks, and the cornea, lens, sclera, ciliary body, iris, conjunctiva, retinal, and optic nerve were examined histologically. Rats' fasting blood glucose and body weight were monitored. Serum tumor necrosis factor-α (TNF-α), vascular endothelial growth factor (VEGF), claudin-1, and glutathione/malondialdehyde ratios were analyzed. Metformin significantly attenuated diabetes-related histopathological ocular deteriorations in the cornea, lens, sclera, ciliary body, iris, conjunctiva, retina, and optic nerve partly by restoring serum TNF-α, VEGF, claudin-1, and glutathione/malondialdehyde ratios without significantly affecting the fasting blood glucose levels or body weight in these hyperglycemic rats. Metformin attenuated hyperglycemia-associated histopathological eye deteriorations, possibly partly by ameliorating vascular leakage, oxidative stress, inflammation, and neovascularization, without affecting the fasting blood glucose levels or body weights in these STZ-induced diabetic rats.
Collapse
Affiliation(s)
- Nazmun Nahar
- UPM-MAKNA Laboratory of Cancer Research, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Suhaila Mohamed
- UPM-MAKNA Laboratory of Cancer Research, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | | | - SengFong Lau
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
| | - Nur Iliyani Mohd Ishak
- UPM-MAKNA Laboratory of Cancer Research, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Norshahira Solehah Umran
- UPM-MAKNA Laboratory of Cancer Research, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| |
Collapse
|
20
|
Han Y, Ding L, Cheng X, Zhao M, Zhao T, Guo L, Li X, Geng Y, Fan M, Liao H, Zhu L. Hypoxia Augments Cerebral Inflammation in a Dextran Sulfate Sodium-Induced Colitis Mouse Model. Front Cell Neurosci 2020; 14:611764. [PMID: 33362475 PMCID: PMC7756107 DOI: 10.3389/fncel.2020.611764] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/18/2020] [Indexed: 11/23/2022] Open
Abstract
The importance of hypoxia in the pathophysiology of inflammatory bowel disease (IBD) is increasingly being realized; also, hypoxia seems to be an important accelerator of brain inflammation, as has been reported by our group and others. IBD is a chronic intestinal disorder that leads to the development of inflammation, which is related to brain dysfunction. However, no studies have reported whether hypoxia is associated with IBD-induced neuroinflammation. Therefore, the objective of the present study was to determine whether hypoxia augments cerebral inflammation in a DSS-induced colitis mouse model. The mouse model was developed using 3% DSS for five days combined with exposure to hypoxic conditions (6,000 m) for two days. Mice were randomly divided into four groups: control group, DSS group, hypoxia group, and DSS plus hypoxia group. The results demonstrated that DSS combined with hypoxia resulted in up-regulation of colonic and plasmatic proinflammatory cytokines. Meanwhile, DSS plus hypoxia increased expression of Iba1, which is a marker of activated microglia, accompanied by increased expression of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in the brain. Moreover, the expression of tight junction proteins, such as zonula occludens-1 (ZO-1), occludin, and claudin-5, was markedly downregulated. The current study provides new insight into how hypoxia exposure induces excessive inflammatory responses andpathophysiological consequences in the brain in a DSS-induced colitis model.
Collapse
Affiliation(s)
- Ying Han
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Liping Ding
- National Nanjing Center for Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiang Cheng
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Ming Zhao
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Tong Zhao
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Liang Guo
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Xinyang Li
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Yanan Geng
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Ming Fan
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Hong Liao
- National Nanjing Center for Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Lingling Zhu
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
21
|
Retinal capillary degeneration and blood-retinal barrier disruption in murine models of Alzheimer's disease. Acta Neuropathol Commun 2020; 8:202. [PMID: 33228786 PMCID: PMC7686701 DOI: 10.1186/s40478-020-01076-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/11/2020] [Indexed: 01/17/2023] Open
Abstract
Extensive effort has been made studying retinal pathology in Alzheimer’s disease (AD) to improve early noninvasive diagnosis and treatment. Particularly relevant are vascular changes, which appear prominent in early brain pathogenesis and could predict cognitive decline. Recently, we identified platelet-derived growth factor receptor beta (PDGFRβ) deficiency and pericyte loss associated with vascular Aβ deposition in the neurosensory retina of mild cognitively impaired (MCI) and AD patients. However, the pathological mechanisms of retinal vascular changes and their possible relationships with vascular amyloidosis, pericyte loss, and blood-retinal barrier (BRB) integrity remain unknown. Here, we evaluated the retinas of transgenic APPSWE/PS1ΔE9 mouse models of AD (ADtg mice) and wild-type mice at different ages for capillary degeneration, PDGFRβ expression, vascular amyloidosis, permeability and inner BRB tight-junction molecules. Using a retinal vascular isolation technique followed by periodic acid-Schiff or immunofluorescent staining, we discovered significant retinal capillary degeneration in ADtg mice compared to age- and sex-matched wild-type mice (P < 0.0001). This small vessel degeneration reached significance in 8-month-old mice (P = 0.0035), with males more susceptible than females. Degeneration of retinal capillaries also progressively increased with age in healthy mice (P = 0.0145); however, the phenomenon was significantly worse during AD-like progression (P = 0.0001). A substantial vascular PDGFRβ deficiency (~ 50% reduction, P = 0.0017) along with prominent vascular Aβ deposition was further detected in the retina of ADtg mice, which inversely correlated with the extent of degenerated capillaries (Pearson’s r = − 0.8, P = 0.0016). Importantly, tight-junction alterations such as claudin-1 downregulation and increased BRB permeability, demonstrated in vivo by retinal fluorescein imaging and ex vivo following injection of FITC-dextran (2000 kD) and Texas Red-dextran (3 kD), were found in ADtg mice. Overall, the identification of age- and Alzheimer’s-dependent retinal capillary degeneration and compromised BRB integrity starting at early disease stages in ADtg mice could contribute to the development of novel targets for AD diagnosis and therapy.
Collapse
|
22
|
Zaghmi A, Drouin-Ouellet J, Brambilla D, Gauthier MA. Treating brain diseases using systemic parenterally-administered protein therapeutics: Dysfunction of the brain barriers and potential strategies. Biomaterials 2020; 269:120461. [PMID: 33218788 DOI: 10.1016/j.biomaterials.2020.120461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022]
Abstract
The parenteral administration of protein therapeutics is increasingly gaining importance for the treatment of human diseases. However, the presence of practically impermeable blood-brain barriers greatly restricts access of such pharmaceutics to the brain. Treating brain disorders with proteins thus remains a great challenge, and the slow clinical translation of these therapeutics may be largely ascribed to the lack of appropriate brain delivery system. Exploring new approaches to deliver proteins to the brain by circumventing physiological barriers is thus of great interest. Moreover, parallel advances in the molecular neurosciences are important for better characterizing blood-brain interfaces, particularly under different pathological conditions (e.g., stroke, multiple sclerosis, Parkinson's disease, and Alzheimer's disease). This review presents the current state of knowledge of the structure and the function of the main physiological barriers of the brain, the mechanisms of transport across these interfaces, as well as alterations to these concomitant with brain disorders. Further, the different strategies to promote protein delivery into the brain are presented, including the use of molecular Trojan horses, the formulation of nanosystems conjugated/loaded with proteins, protein-engineering technologies, the conjugation of proteins to polymers, and the modulation of intercellular junctions. Additionally, therapeutic approaches for brain diseases that do not involve targeting to the brain are presented (i.e., sink and scavenging mechanisms).
Collapse
Affiliation(s)
- A Zaghmi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada
| | - J Drouin-Ouellet
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - D Brambilla
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - M A Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada.
| |
Collapse
|
23
|
Rudraraju M, Narayanan SP, Somanath PR. Regulation of blood-retinal barrier cell-junctions in diabetic retinopathy. Pharmacol Res 2020; 161:105115. [PMID: 32750417 PMCID: PMC7755666 DOI: 10.1016/j.phrs.2020.105115] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022]
Abstract
Loss of the blood-retinal barrier (BRB) integrity and subsequent damage to the neurovascular unit in the retina are the underlying reasons for diabetic retinopathy (DR). Damage to BRB eventually leads to severe visual impairment in the absence of prompt intervention. Diabetic macular edema and proliferative DR are the advanced stages of the disease where BRB integrity is altered. Primary mechanisms contributing to BRB dysfunction include loss of cell-cell barrier junctions, vascular endothelial growth factor, advanced glycation end products-induced damage, and oxidative stress. Although much is known about the involvement of adherens and tight-junction proteins in the regulation of vascular permeability in various diseases, there is a significant gap in our knowledge on the junctional proteins expressed in the BRB and how BRB function is modulated in the diabetic retina. In this review article, we present our current understanding of the molecular composition of BRB, the changes in the BRB junctional protein turnover in DR, and how BRB functional modulation affects vascular permeability and macular edema in the diabetic retina.
Collapse
Affiliation(s)
- Madhuri Rudraraju
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, United States
| | - S Priya Narayanan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Vascular Biology Center, Augusta University, Augusta, GA 30912, United States; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Vascular Biology Center, Augusta University, Augusta, GA 30912, United States; Department of Medicine, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
24
|
Li H, Mei XY, Wang MN, Zhang TY, Zhang Y, Lu B, Sheng YC. Scutellarein alleviates the dysfunction of inner blood-retinal-barrier initiated by hyperglycemia-stimulated microglia cells. Int J Ophthalmol 2020; 13:1538-1545. [PMID: 33078102 DOI: 10.18240/ijo.2020.10.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 07/27/2020] [Indexed: 12/22/2022] Open
Abstract
AIM To investigate the alleviation of scutellarein (SN) against inner blood-retinal-barrier (iBRB) dysfunction in microglia cells stimulated by hyperglycemia and to elucidate the engaged mechanism. METHODS Microglia BV2 cells were stimulated by using 25 mmol/L D-glucose. The same concentration of mannitol (25 mmol/L) was applied as an isotonic contrast. Real-time PCR, Western-blot assay and immunofluorescence staining assay was performed. The dysfunction of iBRB in vitro was detected by using transendothelial electrical resistance (TEER) assay. Additionally, the leakage of fluorescein isothiocyanate (FITC)-conjugated dextran (70 kDa) was detected. RESULTS SN abrogated microglia BV2 cells activation and reduced the phosphorylated activation of extracellular signal-regulated protein kinase (ERK)1/2. SN also decreased the transcriptional activation of nuclear factor κB (NFκB) and the elevated expression of tumor necrosis factor α (TNFα), interleukin (IL)-6 and IL-1β in BV2 cells treated with D-glucose (25 mmol/L). SN attenuated iBRB dysfunction in human retinal endothelial cells (HRECs) or choroid-retinal endothelial RF/6A cells when those cells were treated with TNFα, IL-1β or IL-6, or co-cultured with microglia cells stimulated by D-glucose. Moreover, SN restored the decreased protein expression of tight junctions (TJs) in TNFα-treated HRECs and RF/6A cells. CONCLUSION SN not only alleviate iBRB dysfunction via directly inhibiting retinal endothelial injury caused by TNFα, IL-1β or IL-6, but also reduce the release of TNFα, IL-1β and IL-6 from microglia cells by abrogating hyperglycemia-mediated the activation of microglia cells.
Collapse
Affiliation(s)
- Han Li
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.,Center for Drug Safety Evaluation and Research, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xi-Yu Mei
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Meng-Na Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tian-Yu Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yu-Chen Sheng
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.,Center for Drug Safety Evaluation and Research, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
25
|
Valença A, Mendes-Jorge L, Bonet A, Catita J, Ramos D, Jose-Cunilleras E, Garcia M, Carretero A, Nacher V, Navarro M, Ruberte J. TIM2 modulates retinal iron levels and is involved in blood-retinal barrier breakdown. Exp Eye Res 2020; 202:108292. [PMID: 33065090 DOI: 10.1016/j.exer.2020.108292] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 12/30/2022]
Abstract
Careful control of iron availability in the retina is central to maintenance of iron homeostasis, as its imbalance is associated with oxidative stress and the progression of several retinopathies. Ferritin, known for its role in iron storage and detoxification, has also been proposed as an iron-transporter protein, through its binding to Scara5 and TIM2 membrane receptors. In this study, the presence and iron-related functions of TIM2 in the mouse retina were investigated. Our results revealed for the first time the presence of TIM2 receptors in the mouse retina, mainly in Müller cells. Experimental TIM2 downregulation in the mouse retina promoted, probably due to a compensatory mechanism, Scara5 overexpression that increased retinal ferritin uptake and induced iron overload. Consecutive reactive oxygen species (ROS) overproduction and vascular endothelial growth factor (VEGF) overexpression led to impaired paracellular and transcellular endothelial transport characterized by tight junction degradation and increased caveolae number. In consequence, blood-retinal barrier (BRB) breakdown and retinal edema were observed. Altogether, these results point to TIM2 as a new modulator of retinal iron homeostasis and as a potential target to counteract retinopathy.
Collapse
Affiliation(s)
- Andreia Valença
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477, Lisbon, Portugal; CBATEG - Center for Animal Biotechnology and Gene Therapy, Autonomous University of Barcelona, C/ de La Vall Morona, 08193, Bellaterra (Cerdanyola Del Vallès), Spain
| | - Luísa Mendes-Jorge
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477, Lisbon, Portugal; CBATEG - Center for Animal Biotechnology and Gene Therapy, Autonomous University of Barcelona, C/ de La Vall Morona, 08193, Bellaterra (Cerdanyola Del Vallès), Spain
| | - Aina Bonet
- CBATEG - Center for Animal Biotechnology and Gene Therapy, Autonomous University of Barcelona, C/ de La Vall Morona, 08193, Bellaterra (Cerdanyola Del Vallès), Spain; Department of Animal Health and Anatomy, Faculty of Veterinary, Autonomous University of Barcelona, Travessera Del Turons, 08193, Bellaterra (Cerdanyola Del Vallès), Spain
| | - Joana Catita
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477, Lisbon, Portugal; CBATEG - Center for Animal Biotechnology and Gene Therapy, Autonomous University of Barcelona, C/ de La Vall Morona, 08193, Bellaterra (Cerdanyola Del Vallès), Spain; Faculty of Veterinary Medicine, Lusófona University, Campo Grande 376, 1749-024, Lisbon, Portugal
| | - David Ramos
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477, Lisbon, Portugal; CBATEG - Center for Animal Biotechnology and Gene Therapy, Autonomous University of Barcelona, C/ de La Vall Morona, 08193, Bellaterra (Cerdanyola Del Vallès), Spain
| | - Eduard Jose-Cunilleras
- Department of Animal Medicine and Surgery, Faculty of Veterinary, Autonomous University of Barcelona, Travessera Del Turons, 08193, Bellaterra (Cerdanyola Del Vallès), Spain
| | - Miguel Garcia
- CBATEG - Center for Animal Biotechnology and Gene Therapy, Autonomous University of Barcelona, C/ de La Vall Morona, 08193, Bellaterra (Cerdanyola Del Vallès), Spain
| | - Ana Carretero
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477, Lisbon, Portugal; CBATEG - Center for Animal Biotechnology and Gene Therapy, Autonomous University of Barcelona, C/ de La Vall Morona, 08193, Bellaterra (Cerdanyola Del Vallès), Spain; Department of Animal Health and Anatomy, Faculty of Veterinary, Autonomous University of Barcelona, Travessera Del Turons, 08193, Bellaterra (Cerdanyola Del Vallès), Spain
| | - Victor Nacher
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477, Lisbon, Portugal; CBATEG - Center for Animal Biotechnology and Gene Therapy, Autonomous University of Barcelona, C/ de La Vall Morona, 08193, Bellaterra (Cerdanyola Del Vallès), Spain; Department of Animal Health and Anatomy, Faculty of Veterinary, Autonomous University of Barcelona, Travessera Del Turons, 08193, Bellaterra (Cerdanyola Del Vallès), Spain
| | - Marc Navarro
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477, Lisbon, Portugal; CBATEG - Center for Animal Biotechnology and Gene Therapy, Autonomous University of Barcelona, C/ de La Vall Morona, 08193, Bellaterra (Cerdanyola Del Vallès), Spain; Department of Animal Health and Anatomy, Faculty of Veterinary, Autonomous University of Barcelona, Travessera Del Turons, 08193, Bellaterra (Cerdanyola Del Vallès), Spain
| | - Jesús Ruberte
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477, Lisbon, Portugal; CBATEG - Center for Animal Biotechnology and Gene Therapy, Autonomous University of Barcelona, C/ de La Vall Morona, 08193, Bellaterra (Cerdanyola Del Vallès), Spain; Department of Animal Health and Anatomy, Faculty of Veterinary, Autonomous University of Barcelona, Travessera Del Turons, 08193, Bellaterra (Cerdanyola Del Vallès), Spain.
| |
Collapse
|
26
|
Inflammation: major denominator of obesity, Type 2 diabetes and Alzheimer's disease-like pathology? Clin Sci (Lond) 2020; 134:547-570. [PMID: 32167154 DOI: 10.1042/cs20191313] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 02/08/2023]
Abstract
Adipose tissue is an active metabolic organ that contributes to processes such as energy storage and utilization and to the production of a number of metabolic agents, such as adipokines, which play a role in inflammation. In this review, we try to elucidate the connections between peripheral inflammation at obesity and Type 2 diabetes and the central inflammatory process. Multiple lines of evidence highlight the importance of peripheral inflammation and its link to neuroinflammation, which can lead to neurodegenerative diseases such as dementia, Alzheimer's disease (AD) and Parkinson's disease. In addition to the accumulation of misfolded amyloid beta (Aβ) peptide and the formation of the neurofibrillary tangles of hyperphosphorylated tau protein in the brain, activated microglia and reactive astrocytes are the main indicators of AD progression. They were found close to Aβ plaques in the brains of both AD patients and rodent models of Alzheimer's disease-like pathology. Cytokines are key players in pro- and anti-inflammatory processes and are also produced by microglia and astrocytes. The interplay of seemingly unrelated pathways between the periphery and the brain could, in fact, have a common denominator, with inflammation in general being a key factor affecting neuronal processes in the brain. An increased amount of white adipose tissue throughout the body seems to be an important player in pro-inflammatory processes. Nevertheless, other important factors should be studied to elucidate the pathological processes of and the relationship among obesity, Type 2 diabetes and neurodegenerative diseases.
Collapse
|
27
|
Tikiyani V, Babu K. Claudins in the brain: Unconventional functions in neurons. Traffic 2020; 20:807-814. [PMID: 31418988 DOI: 10.1111/tra.12685] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 08/09/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022]
Abstract
Bonafide claudin proteins are functional and structural components of tight junctions and are largely responsible for barrier formation across epithelial and endothelial membranes. However, current advances in the understanding of claudin biology have revealed their unexpected functions in the brain. Apart from maintaining blood-brain barriers in the brain, other functions of claudins in neurons and at synapses have been largely elusive and are just coming to light. In this review, we summarize the functions of claudins in the brain and their association in neuronal diseases. Further, we go on to cover some recent studies that show that claudins play signaling functions in neurons by regulating trafficking of postsynaptic receptors and controlling dendritic morphogenesis in the model organism Caenorhabditis elegans.
Collapse
Affiliation(s)
- Vina Tikiyani
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali, Punjab, India
| | - Kavita Babu
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali, Punjab, India.,Centre for Neuroscience (CNS), Indian Institute of Science (IISc), Bangalore, India
| |
Collapse
|
28
|
Moura RP, Pacheco C, Pêgo AP, des Rieux A, Sarmento B. Lipid nanocapsules to enhance drug bioavailability to the central nervous system. J Control Release 2020; 322:390-400. [PMID: 32247807 DOI: 10.1016/j.jconrel.2020.03.042] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/15/2022]
Abstract
The central nervous system (CNS), namely the brain, still remains as the hardest area of the human body to achieve adequate concentration levels of most drugs, mainly due to the limiting behavior of its physical and biological defenses. Lipid nanocapsules emerge as a versatile platform to tackle those barriers, and efficiently delivery different drug payloads due to their numerous advantages. They can be produced in a fast, solvent-free and scalable-up process, and their properties can be fine-tuned for to make an optimal brain drug delivery vehicle. Moreover, lipid nanocapsule surface modification can further improve their bioavailability towards the central nervous system. Coupling these features with alternative delivery methods that stem to disrupt or fully circumvent the blood-brain barrier may fully harness the therapeutic advance that lipid nanocapsules can supply to current treatment options. Thus, this review intends to critically address the development of lipid nanocapsules, as well as to highlight the key features that can be modulated to ameliorate their properties towards the central nervous system delivery, mainly through intravenous methods, and how the pathological microenvironment of the CNS can be taken advantage of. The different routes to promote drug delivery towards the brain parenchyma are also discussed, as well as the synergetic effect that can be obtained by combining modified lipid nanocapsules with new/smart administration routes.
Collapse
Affiliation(s)
- Rui Pedro Moura
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Catarina Pacheco
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Ana Paula Pêgo
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Anne des Rieux
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
29
|
Claudins: New Players in Human Fertility and Reproductive System Cancers. Cancers (Basel) 2020; 12:cancers12030711. [PMID: 32197343 PMCID: PMC7140004 DOI: 10.3390/cancers12030711] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/15/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022] Open
Abstract
Claudins are major integral proteins of tight junctions (TJs), the apical cell-cell adhesions that enable maintaining polarity of epithelial cells, their differentiation, and cell signaling. A number of studies have indicated that claudins might play a crucial role in both physiology and pathogenesis. Their tissue-specific expression was originally linked to the development of different types of cancer and triggered a hope to use them as diagnostic or prognostic markers. However, it seems that their expression is more complex than that, and undoubtedly, claudins participate in one of the most important molecular events in cells. This review summarizes the recent research evaluating the role of claudins in fertility and the most common endocrine-dependent cancers in the reproductive system and highlights the crucial role of claudins both in human fertility and the most common cancers.
Collapse
|
30
|
Mechanistic insights into autocrine and paracrine roles of endothelial GABA signaling in the embryonic forebrain. Sci Rep 2019; 9:16256. [PMID: 31700116 PMCID: PMC6838150 DOI: 10.1038/s41598-019-52729-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/18/2019] [Indexed: 12/24/2022] Open
Abstract
The developing cerebral cortex uses a complex developmental plan involving angiogenesis, neurogenesis and neuronal migration. Our recent studies have highlighted the importance of endothelial cell secreted GABA signaling in the embryonic forebrain and established novel autonomous links between blood vessels and the origin of neuropsychiatric diseases. A GABA pathway operates in both endothelial cells and GABAergic neurons of the embryonic telencephalon; however, while the neuronal GABA pathway has been extensively studied, little is known about the endothelial GABA pathway. Our recently generated Vgat endothelial cell knockout mouse model that blocks GABA release from endothelial cells, serves as a new tool to study how endothelial GABA signaling shapes angiogenesis and neurovascular interactions during prenatal development. Quantitative gene expression profiling reveals that the endothelial GABA signaling pathway influences genes connected to specific processes like endothelial cell proliferation, differentiation, migration, tight junction formation, vascular sprouting and integrity. It also shows how components of the neuronal GABA pathway, for instance receptor mediated signaling, cell cycle related components and transcription factors are affected in the absence of endothelial GABA release. Taken together, our findings delineate the close relationship between vascular and nervous systems that begin early in embryogenesis establishing their future interactions and interdependence.
Collapse
|
31
|
Małkiewicz MA, Szarmach A, Sabisz A, Cubała WJ, Szurowska E, Winklewski PJ. Blood-brain barrier permeability and physical exercise. J Neuroinflammation 2019; 16:15. [PMID: 30678702 PMCID: PMC6345022 DOI: 10.1186/s12974-019-1403-x] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
In this narrative review, a theoretical framework on the crosstalk between physical exercise and blood-brain barrier (BBB) permeability is presented. We discuss the influence of physical activity on the factors affecting BBB permeability such as systemic inflammation, the brain renin-angiotensin and noradrenergic systems, central autonomic function and the kynurenine pathway. The positive role of exercise in multiple sclerosis and Alzheimer’s disease is described. Finally, the potential role of conditioning as well as the effect of exercise on BBB tight junctions is outlined. There is a body of evidence that regular physical exercise diminishes BBB permeability as it reinforces antioxidative capacity, reduces oxidative stress and has anti-inflammatory effects. It improves endothelial function and might increase the density of brain capillaries. Thus, physical training can be emphasised as a component of prevention programs developed for patients to minimise the risk of the onset of neuroinflammatory diseases as well as an augmentation of existing treatment. Unfortunately, despite a sound theoretical background, it remains unclear as to whether exercise training is effective in modulating BBB permeability in several specific diseases. Further research is needed as the impact of exercise is yet to be fully elucidated.
Collapse
Affiliation(s)
- Marta A Małkiewicz
- Department of Human Physiology, Faculty of Health Sciences, Medical University of Gdansk, Tuwima Str. 15, 80-210, Gdansk, Poland.,Department of Psychiatry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Arkadiusz Szarmach
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Agnieszka Sabisz
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Wiesław J Cubała
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Edyta Szurowska
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Paweł J Winklewski
- Department of Human Physiology, Faculty of Health Sciences, Medical University of Gdansk, Tuwima Str. 15, 80-210, Gdansk, Poland. .,2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland. .,Department of Clinical Anatomy and Physiology, Faculty of Health Sciences, Pomeranian University of Slupsk, Slupsk, Poland.
| |
Collapse
|
32
|
Mei X, Zhang T, Ouyang H, Lu B, Wang Z, Ji L. Scutellarin alleviates blood-retina-barrier oxidative stress injury initiated by activated microglia cells during the development of diabetic retinopathy. Biochem Pharmacol 2019; 159:82-95. [DOI: 10.1016/j.bcp.2018.11.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/13/2018] [Indexed: 12/27/2022]
|
33
|
Tikiyani V, Li L, Sharma P, Liu H, Hu Z, Babu K. Wnt Secretion Is Regulated by the Tetraspan Protein HIC-1 through Its Interaction with Neurabin/NAB-1. Cell Rep 2018; 25:1856-1871.e6. [PMID: 30428353 PMCID: PMC6258899 DOI: 10.1016/j.celrep.2018.10.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/25/2018] [Accepted: 10/11/2018] [Indexed: 12/22/2022] Open
Abstract
The aberrant regulation of Wnt secretion is implicated in various neurological diseases. However, the mechanisms of Wnt release are still largely unknown. Here we describe the role of a C. elegans tetraspan protein, HIC-1, in maintaining normal Wnt release. We show that HIC-1 is expressed in cholinergic synapses and that mutants in hic-1 show increased levels of the acetylcholine receptor AChR/ACR-16. Our results suggest that HIC-1 maintains normal AChR/ACR-16 levels by regulating normal Wnt release from presynaptic neurons, as hic-1 mutants show an increase in secreted Wnt from cholinergic neurons. We further show that HIC-1 affects Wnt secretion by modulating the actin cytoskeleton through its interaction with the actin-binding protein NAB-1. In summary, we describe a protein, HIC-1, that functions as a neuromodulator by affecting postsynaptic AChR/ACR-16 levels by regulating presynaptic Wnt release from cholinergic motor neurons.
Collapse
Affiliation(s)
- Vina Tikiyani
- Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli PO 140306, Punjab, India
| | - Lei Li
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), University of Queensland, Upland Road 79, St. Lucia, QLD 4072, Australia
| | - Pallavi Sharma
- Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli PO 140306, Punjab, India
| | - Haowen Liu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), University of Queensland, Upland Road 79, St. Lucia, QLD 4072, Australia
| | - Zhitao Hu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), University of Queensland, Upland Road 79, St. Lucia, QLD 4072, Australia
| | - Kavita Babu
- Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli PO 140306, Punjab, India.
| |
Collapse
|
34
|
Lower genital tract cytokine profiles in South African women living with HIV: influence of mucosal sampling. Sci Rep 2018; 8:12203. [PMID: 30111808 PMCID: PMC6093917 DOI: 10.1038/s41598-018-30663-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/03/2018] [Indexed: 11/25/2022] Open
Abstract
Measurement of cytokines in the lower female genital tract offer insight into risk for HIV infection and reproductive complications. However, few studies have systematically compared mucosal collection methods or whether collection order matters. We compared longitudinal cytokine profiles in matched genital samples collected from women living with HIV using menstrual cup (MC), endocervical swabs (ECS) and swab-enriched cervicovaginal lavage (eCVL). Samples were collected at enrollment [MC:ECS:eCVL], 3-months (ECS:eCVL:MC) and 6-months (eCVL:MC:ECS) and concentrations of 28 cytokines determined by Luminex. Cytokine clustering was assessed using Principle Component Analysis (PCA), Partial Least Squares Discriminant Analysis (PLSDA) and factor analysis. Generally, higher cytokine concentrations were detected in MC samples, followed by ECS and eCVL, irrespective of study visit or sampling order. Factor analysis and PCA identified ECS to be inferior for measuring regulatory cytokines and IP-10 than eCVL or MC. Although concentrations differed, the majority of cytokines correlated between methods. Sampling order influenced cytokine concentrations marginally, and cytokines clustered more strongly by method than study visit. Variance in profiles was lowest in MC, suggesting greater consistency of sampling compared to other methods. We conclude that MC sampling offered advantages over other methods for detecting cytokines in women, with order marginally influencing profiles.
Collapse
|
35
|
The Claudin-like Protein HPO-30 Is Required to Maintain LAChRs at the C. elegans Neuromuscular Junction. J Neurosci 2018; 38:7072-7087. [PMID: 29950505 PMCID: PMC6083452 DOI: 10.1523/jneurosci.3487-17.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 12/22/2022] Open
Abstract
Communications across chemical synapses are primarily mediated by neurotransmitters and their postsynaptic receptors. There are diverse molecular systems to localize and regulate the receptors at the synapse. Here, we identify HPO-30, a member of the claudin superfamily of membrane proteins, as a positive regulator for synaptic localization of levamisole-dependent AChRs (LAChRs) at the Caenorhabditis elegans neuromuscular junction (NMJ). The HPO-30 protein localizes at the NMJ and shows genetic and physical association with the LAChR subunits LEV-8, UNC-29, and UNC-38. Using genetic and electrophysiological assays in the hermaphrodite C. elegans, we demonstrate that HPO-30 functions through Neuroligin at the NMJ to maintain postsynaptic LAChR levels at the synapse. Together, this work suggests a novel function for a tight junction protein in maintaining normal receptor levels at the NMJ. SIGNIFICANCE STATEMENT Claudins are a large superfamily of membrane proteins. Their role in maintaining the functional integrity of tight junctions has been widely explored. Our experiments suggest a critical role for the claudin-like protein, HPO-30, in maintaining synaptic levamisole-dependent AChR (LAChR) levels. LAChRs contribute to <20% of the acetylcholine-mediated conductance in adult Caenorhabditis elegans; however, they play a significant functional role in worm locomotion. This study provides a new perspective in the study of LAChR physiology.
Collapse
|
36
|
Samanta P, Wang Y, Fuladi S, Zou J, Li Y, Shen L, Weber C, Khalili-Araghi F. Molecular determination of claudin-15 organization and channel selectivity. J Gen Physiol 2018; 150:949-968. [PMID: 29915162 PMCID: PMC6028499 DOI: 10.1085/jgp.201711868] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 03/08/2018] [Accepted: 05/04/2018] [Indexed: 12/28/2022] Open
Abstract
Members of the claudin family form tight junctions between adjacent epithelial and endothelial cells. Samanta et al. build an atomic model of claudin-15 using molecular dynamics simulations and conclude that four claudin-15 molecules each contribute an aspartic acid residue to form a selectivity filter. Tight junctions are macromolecular structures that traverse the space between adjacent cells in epithelia and endothelia. Members of the claudin family are known to determine tight junction permeability in a charge- and size-selective manner. Here, we use molecular dynamics simulations to build and refine an atomic model of claudin-15 channels and study its transport properties. Our simulations indicate that claudin-15 forms well-defined channels for ions and molecules and otherwise “seals” the paracellular space through hydrophobic interactions. Ionic currents, calculated from simulation trajectories of wild-type as well as mutant channels, reflect in vitro measurements. The simulations suggest that the selectivity filter is formed by a cage of four aspartic acid residues (D55), contributed by four claudin-15 molecules, which creates a negative electrostatic potential to favor cation flux over anion flux. Charge reversal or charge ablation mutations of D55 significantly reduce cation permeability in silico and in vitro, whereas mutations of other negatively charged pore amino acid residues have a significantly smaller impact on channel permeability and selectivity. The simulations also indicate that water and small ions can pass through the channel, but larger cations, such as tetramethylammonium, do not traverse the pore. Thus, our model provides an atomic view of claudin channels, their transport function, and a potential three-dimensional organization of its selectivity filter.
Collapse
Affiliation(s)
| | - Yitang Wang
- Department of Pathology, The University of Chicago, Chicago, IL.,Department of Surgery, The University of Chicago, Chicago, IL
| | - Shadi Fuladi
- Department of Physics, University of Illinois, Chicago, IL
| | - Jinjing Zou
- Department of Pathology, The University of Chicago, Chicago, IL
| | - Ye Li
- Department of Pathology, The University of Chicago, Chicago, IL
| | - Le Shen
- Department of Pathology, The University of Chicago, Chicago, IL .,Department of Surgery, The University of Chicago, Chicago, IL
| | | | | |
Collapse
|
37
|
Lv J, Hu W, Yang Z, Li T, Jiang S, Ma Z, Chen F, Yang Y. Focusing on claudin-5: A promising candidate in the regulation of BBB to treat ischemic stroke. Prog Neurobiol 2018; 161:79-96. [PMID: 29217457 DOI: 10.1016/j.pneurobio.2017.12.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/20/2017] [Accepted: 12/03/2017] [Indexed: 12/11/2022]
Abstract
Claudin-5 is a tight junction (TJ) protein in the blood-brain barrier (BBB) that has recently attracted increased attention. Numerous studies have demonstrated that claudin-5 regulates the integrity and permeability of the BBB. Increased claudin-5 expression plays a neuroprotective role in neurological diseases, particularly in cerebral ischemic stroke. Moreover, claudin-5 might be a potential marker for early hemorrhagic transformation detection in ischemic stroke. In light of the distinctive effects of claudin-5 on the nervous system, we present the elaborate network of roles that claudin-5 plays in ischemic stroke. In this review, we first introduce basic knowledge regarding the BBB and the claudin family, the characterization and regulation of claudin-5, and association between claudin-5 and other TJ proteins. Subsequently, we describe BBB dysfunction and neuron-specific drivers of pathogenesis of ischemic stroke, including inflammatory disequilibrium and oxidative stress. Furthermore, we summarize promising ischemic stroke treatments that target the BBB via claudin-5, including modified rt-PA therapy, pharmacotherapy, hormone treatment, receptor-targeted therapy, gene therapy, and physical therapy. This review highlights recent advances and provides a comprehensive summary of claudin-5 in the regulation of the BBB and may be helpful for drug design and clinical therapy for treatment of ischemic stroke.
Collapse
Affiliation(s)
- Jianjun Lv
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China; Department of Immunology, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhi Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Fulin Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
38
|
Alrashdi SF, Deliyanti D, Talia DM, Wilkinson-Berka JL. Endothelin-2 Injures the Blood-Retinal Barrier and Macroglial Müller Cells: Interactions with Angiotensin II, Aldosterone, and NADPH Oxidase. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:805-817. [PMID: 29248456 DOI: 10.1016/j.ajpath.2017.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/20/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
Abstract
Although increasing evidence indicates that endothelin-2 (Edn2) has distinct roles in tissue pathology, including inflammation, glial cell dysfunction, and angiogenesis, its role in the retina and the factors that regulate its actions are not fully understood. We hypothesized that Edn2 damages the blood-retinal barrier (BRB) and that this is mediated by interactions with the renin-angiotensin-aldosterone system and reactive oxygen species derived from NADPH oxidase (Nox). C57BL/6J mice received an intravitreal injection of Edn2 or control vehicle to examine the blood pressure-independent effects of Edn2. Mice administered Edn2 were randomized to receive by intraperitoneal injection treatments that inhibited the Edn type a receptor, Edn type b receptor, angiotensin type 1 receptor, mineralocorticoid receptor, or Nox isoforms 1 to 4. One month later, mice administered Edn2 exhibited breakdown of the BRB with increased vascular leakage, vascular endothelial growth factor expression, and infiltrating macrophages (Ly6C+CD45highCD11b+). Further, macroglial Müller cells, which influence the integrity of the BRB and prevent retinal edema, became gliotic and expressed increased levels of water (aquaporin-4) and ion (Kir4.1) channels. This Edn2-mediated retinopathy was reduced by all treatments. Complementary in vitro studies in cultured Müller cells supported these findings and demonstrated the importance of reactive oxygen species in mediating these events. In conclusion, Edn2 has detrimental effects on the BRB and Müller cells that involve interactions with the renin-angiotensin aldosterone system and Nox1/4.
Collapse
Affiliation(s)
- Saeed F Alrashdi
- Department of Diabetes, Monash University, Melbourne, Victoria, Australia
| | - Devy Deliyanti
- Department of Diabetes, Monash University, Melbourne, Victoria, Australia
| | - Dean M Talia
- Department of Diabetes, Monash University, Melbourne, Victoria, Australia
| | | |
Collapse
|
39
|
Zenaro E, Piacentino G, Constantin G. The blood-brain barrier in Alzheimer's disease. Neurobiol Dis 2017; 107:41-56. [PMID: 27425887 PMCID: PMC5600438 DOI: 10.1016/j.nbd.2016.07.007] [Citation(s) in RCA: 490] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/06/2016] [Accepted: 07/13/2016] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder characterized by the pathological accumulation of amyloid beta (Aβ) peptides and neurofibrillary tangles containing hyperphosphorylated neuronal tau protein. AD pathology is also characterized by chronic brain inflammation, which promotes disease pathogenesis. In this context, the blood-brain barrier (BBB), a highly specialized endothelial cell membrane that lines cerebral microvessels, represents the interface between neural cells and circulating cells of the immune system. The BBB thus plays a key role in the generation and maintenance of chronic inflammation during AD. The BBB operates within the neurovascular unit (NVU), which includes clusters of glial cells, neurons and pericytes. The NVU becomes dysfunctional during AD, and each of its components may undergo functional changes that contribute to neuronal injury and cognitive deficit. In transgenic animals with AD-like pathology, recent studies have shown that circulating leukocytes migrate through the activated brain endothelium when certain adhesion molecules are expressed, penetrating into the brain parenchyma, interacting with the NVU components and potentially affecting their structural integrity and functionality. Therefore, migrating immune system cells in cerebral vessels act in concert with the modified BBB and may be integrated into the dysfunctional NVU. Notably, blocking the adhesion mechanisms controlling leukocyte-endothelial interactions inhibits both Aβ deposition and tau hyperphosphorylation, and reduces memory loss in AD models. The characterization of molecular mechanisms controlling vascular inflammation and leukocyte trafficking could therefore help to determine the basis of BBB dysfunction during AD and may lead to the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Elena Zenaro
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134 Verona, Italy
| | - Gennj Piacentino
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134 Verona, Italy
| | - Gabriela Constantin
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134 Verona, Italy.
| |
Collapse
|
40
|
Hui F, Nguyen CTO, He Z, Vingrys AJ, Gurrell R, Fish RL, Bui BV. Retinal and Cortical Blood Flow Dynamics Following Systemic Blood-Neural Barrier Disruption. Front Neurosci 2017; 11:568. [PMID: 29075176 PMCID: PMC5643486 DOI: 10.3389/fnins.2017.00568] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/28/2017] [Indexed: 12/11/2022] Open
Abstract
To consider whether imaging retinal vasculature may be used as a marker for cortical vessels, we compared fluorescein angiography flow dynamics before and after pharmacological disruption of blood-neural barriers. Sodium fluorescein (1%, 200 μl/kg) was intravenously delivered in anesthetized adult Long Evans rats (n = 44, brain = 18, retina = 26). In the brain cohort, a cranial window was created to allow direct visualization of surface cortical vessels. Video fluorescein angiography was captured using a rodent retinal camera at 30 frames/second and fluorescence intensity profiles were evaluated for the time to reach 50% brightness (half-rise), 50% decay (half-fall), and the plateau level of remnant fluorescence (offset, %). Cortical vessels fluoresced earlier (artery half-rise: 5.6 ± 0.2 s) and decayed faster (half-fall: 10.3 ± 0.2 s) compared to retinal vasculature. Cortical vessels also had a considerably higher offset, particularly in the capillaries/extravascular space (41.4 ± 2.7%) whereas pigment in the retina reduces such residual fluorescence. In a sub-cohort of animals, sodium deoxycholate (DOC, 0.06 M dissolved in sterile saline, 1 mL) was delivered intravenously to cause simultaneous disruption of the blood-brain and blood-retinal barriers. A separate group received saline as vehicle control. Fluorescein angiography was re-measured at 6 and 24 h after drug infusion and evaluated by comparing flow dynamics to the upper quartile (75%) of the control group. Retinal vasculature was more sensitive to DOC-induced disruption with a higher fluorescence offset at 6 h (47.3 ± 10.6%). A delayed effect was seen in cortical vessels with a higher offset evident only at 24 h (65.6 ± 10.1%). Here we have developed a method to quantitatively compare fluorescein angiography dynamics in the retina and superficial cortical vessels. Our results show that systemic disruption of blood-neural barriers causes vascular leakage in both tissues but earlier in the retina suggesting that pharmacological blood-neural barrier disruption may be detected earlier in the eye than in cortical vasculature.
Collapse
Affiliation(s)
- Flora Hui
- Department of Optometry and Vision Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Christine T. O. Nguyen
- Department of Optometry and Vision Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Zheng He
- Department of Optometry and Vision Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Algis J. Vingrys
- Department of Optometry and Vision Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Rachel Gurrell
- Neuroscience and Pain Research Unit, Pfizer, Cambridge, United Kingdom
| | - Rebecca L. Fish
- Neuroscience and Pain Research Unit, Pfizer, Cambridge, United Kingdom
| | - Bang V. Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
41
|
Bulldan A, Malviya VN, Upmanyu N, Konrad L, Scheiner-Bobis G. Testosterone/bicalutamide antagonism at the predicted extracellular androgen binding site of ZIP9. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2402-2414. [PMID: 28943399 DOI: 10.1016/j.bbamcr.2017.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 10/18/2022]
Abstract
ZIP9 is a Zn2+ transporter, testosterone receptor, and mediator of signaling events through G-proteins. Despite these pivotal properties, however, its physiological and pathophysiological significance has not yet been comprehensively addressed. Using a cell line that lacks the classical androgen receptor we show that ZIP9-mediated phosphorylation of Erk1/2, CREB, or ATF-1 and expression of claudin-5 and zonula occludens-1 by testosterone can be completely antagonized by bicalutamide (Casodex), an anti-androgen of significant clinical impact. Computational modeling and docking experiments with ZIP9 reveal typical characteristics of ZIP transporters and an extracellular binding site for testosterone capable of accommodating bicalutamide. The presence of this site is verified by our demonstration that the membrane-impermeable testosterone analogue T-BSA-FITC labels the membrane only when ZIP9 is expressed and that this labeling is completely prevented by bicalutamide. The study connects structural features of ZIP9 to its functions and indicates a possible relevance of ZIP9 as a pharmacological target.
Collapse
Affiliation(s)
- Ahmed Bulldan
- Institute for Veterinary-Physiology and -Biochemistry, School of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany
| | | | - Neha Upmanyu
- Institute for Veterinary-Physiology and -Biochemistry, School of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany
| | - Lutz Konrad
- Department of Obstetrics and Gynecology, Faculty of Medicine, Justus-Liebig-University, Giessen, Germany
| | - Georgios Scheiner-Bobis
- Institute for Veterinary-Physiology and -Biochemistry, School of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany.
| |
Collapse
|
42
|
Nguyen CTO, Hui F, Charng J, Velaedan S, van Koeverden AK, Lim JKH, He Z, Wong VHY, Vingrys AJ, Bui BV, Ivarsson M. Retinal biomarkers provide "insight" into cortical pharmacology and disease. Pharmacol Ther 2017; 175:151-177. [PMID: 28174096 DOI: 10.1016/j.pharmthera.2017.02.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The retina is an easily accessible out-pouching of the central nervous system (CNS) and thus lends itself to being a biomarker of the brain. More specifically, the presence of neuronal, vascular and blood-neural barrier parallels in the eye and brain coupled with fast and inexpensive methods to quantify retinal changes make ocular biomarkers an attractive option. This includes its utility as a biomarker for a number of cerebrovascular diseases as well as a drug pharmacology and safety biomarker for the CNS. It is a rapidly emerging field, with some areas well established, such as stroke risk and multiple sclerosis, whereas others are still in development (Alzheimer's, Parkinson's, psychological disease and cortical diabetic dysfunction). The current applications and future potential of retinal biomarkers, including potential ways to improve their sensitivity and specificity are discussed. This review summarises the existing literature and provides a perspective on the strength of current retinal biomarkers and their future potential.
Collapse
Affiliation(s)
- Christine T O Nguyen
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia.
| | - Flora Hui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Jason Charng
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Shajan Velaedan
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Anna K van Koeverden
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Jeremiah K H Lim
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Zheng He
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Vickie H Y Wong
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Algis J Vingrys
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Magnus Ivarsson
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| |
Collapse
|
43
|
Molins B, Pascual A, Méndez, Llorenç V, Zarranz-Ventura J, Mesquida M, Adán A, Martorell J. C-reactive protein isoforms differentially affect outer blood-retinal barrier integrity and function. Am J Physiol Cell Physiol 2016; 312:C244-C253. [PMID: 28003224 DOI: 10.1152/ajpcell.00057.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 12/16/2016] [Accepted: 12/20/2016] [Indexed: 11/22/2022]
Abstract
The retinal pigment epithelium (RPE) forms the outer blood-retinal barrier (oBRB) and is the prime target of early age-related macular degeneration (AMD). C-reactive protein (CRP), a serum biomarker for chronic inflammation and AMD, presents two different isoforms, monomeric (mCRP) and pentameric (pCRP), that may have a different effect on inflammation and barrier function in the RPE. The results reported in this study suggest that mCRP but not pCRP impairs RPE functionality by increasing paracellular permeability and disrupting the tight junction proteins ZO-1 and occludin in RPE cells. Additionally, we evaluated the effect of drugs commonly used in clinical settings on mCRP-induced barrier dysfunction. We found that a corticosteroid (methylprednisolone) and an anti-VEGF agent (bevacizumab) prevented mCRP-induced ARPE-19 barrier disruption and IL-8 production. Furthermore, bevacizumab was also able to revert mCRP-induced IL-8 increase after mCRP stimulation. In conclusion, the presence of mCRP within retinal tissue may lead to disruption of the oBRB, an effect that may be modified in the presence of corticosteroids or anti-VEGF drugs.
Collapse
Affiliation(s)
- Blanca Molins
- Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain; .,Institut Clínic d'Oftalmologia, Hospital Clínic de Barcelona, Barcelona, Spain; and
| | | | - Méndez
- Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain.,Department of Chemical Engineering and Material Sciences, IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain
| | - Victor Llorenç
- Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain.,Institut Clínic d'Oftalmologia, Hospital Clínic de Barcelona, Barcelona, Spain; and
| | - Javier Zarranz-Ventura
- Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain.,Institut Clínic d'Oftalmologia, Hospital Clínic de Barcelona, Barcelona, Spain; and
| | - Marina Mesquida
- Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain.,Institut Clínic d'Oftalmologia, Hospital Clínic de Barcelona, Barcelona, Spain; and
| | - Alfredo Adán
- Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain.,Institut Clínic d'Oftalmologia, Hospital Clínic de Barcelona, Barcelona, Spain; and
| | - Jordi Martorell
- Department of Chemical Engineering and Material Sciences, IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain
| |
Collapse
|
44
|
Wu H, Rodriguez AR, Spur BW, Venkataraman V. An Acute Retinal Model for Evaluating Blood Retinal Barrier Breach and Potential Drugs for Treatment. J Vis Exp 2016. [PMID: 27684428 DOI: 10.3791/54619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A low-cost, easy-to-use and powerful model system is established to evaluate potential treatments that could ameliorate blood retinal barrier breach. An inflammatory factor, histamine, is demonstrated to compromise vessel integrity in the cultured retina through positive staining of IgG outside of the blood vessels. The effects of histamine itself and those of candidate drugs for potential treatments, such as lipoxin A4, are assessed using three parameters: blood vessel leakage via IgG immunostaining, activation of Müller cells via GFAP staining and change in neuronal dendrites through staining for MAP2. Furthermore, the layered organization of the retina allows a detailed analysis of the processes of Müller and ganglion cells, such as changes in width and continuity. While the data presented is with swine retinal culture, the system is applicable to multiple species. Thus, the model provides a reliable tool to investigate the early effects of compromised retinal vessel integrity on different cell types and also to evaluate potential drug candidates for treatment.
Collapse
Affiliation(s)
- Hao Wu
- Graduate School of Biomedical Sciences, Rowan University
| | - Ana R Rodriguez
- Department of Cell Biology, Rowan University School of Osteopathic Medicine
| | - Bernd W Spur
- Department of Cell Biology, Rowan University School of Osteopathic Medicine
| | - Venkat Venkataraman
- Graduate School of Biomedical Sciences, Rowan University; Department of Cell Biology, Rowan University School of Osteopathic Medicine;
| |
Collapse
|
45
|
Incretin-Based Therapies for Diabetic Complications: Basic Mechanisms and Clinical Evidence. Int J Mol Sci 2016; 17:ijms17081223. [PMID: 27483245 PMCID: PMC5000621 DOI: 10.3390/ijms17081223] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 07/16/2016] [Accepted: 07/25/2016] [Indexed: 02/07/2023] Open
Abstract
An increase in the rates of morbidity and mortality associated with diabetic complications is a global concern. Glycemic control is important to prevent the development and progression of diabetic complications. Various classes of anti-diabetic agents are currently available, and their pleiotropic effects on diabetic complications have been investigated. Incretin-based therapies such as dipeptidyl peptidase (DPP)-4 inhibitors and glucagon-like peptide-1 receptor agonists (GLP-1RA) are now widely used in the treatment of patients with type 2 diabetes. A series of experimental studies showed that incretin-based therapies have beneficial effects on diabetic complications, independent of their glucose-lowering abilities, which are mediated by anti-inflammatory and anti-oxidative stress properties. Based on these findings, clinical studies to assess the effects of DPP-4 inhibitors and GLP-1RA on diabetic microvascular and macrovascular complications have been performed. Several but not all studies have provided evidence to support the beneficial effects of incretin-based therapies on diabetic complications in patients with type 2 diabetes. We herein discuss the experimental and clinical evidence of incretin-based therapy for diabetic complications.
Collapse
|
46
|
Nanoscale effects in dendrimer-mediated targeting of neuroinflammation. Biomaterials 2016; 101:96-107. [PMID: 27267631 DOI: 10.1016/j.biomaterials.2016.05.044] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/20/2016] [Accepted: 05/24/2016] [Indexed: 12/27/2022]
Abstract
Neuroinflammation, mediated by activated microglia and astrocytes, plays a key role in the pathogenesis of many neurological disorders. Systemically-administered dendrimers target neuroinflammation and deliver drugs with significant efficacy, without the need for ligands. Elucidating the nanoscale aspects of targeting neuroinflammation will enable superior nanodevices for eventual translation. Using a rabbit model of cerebral palsy, we studied the in vivo contributions of dendrimer physicochemical properties and disease pathophysiology on dendrimer brain uptake, diffusion, and cell specific localization. Neutral dendrimers move efficiently within the brain parenchyma and rapidly localize in glial cells in regions of injury. Dendrimer uptake is also dependent on the extent of blood-brain-barrier breakdown, glial activation, and disease severity (mild, moderate, or severe), which can lend the dendrimer to be used as an imaging biomarker for disease phenotype. This new understanding of the in vivo mechanism of dendrimer-mediated delivery in a clinically-relevant rabbit model provides greater opportunity for clinical translation of targeted brain injury therapies.
Collapse
|
47
|
Ghani M, Reitz C, Cheng R, Vardarajan BN, Jun G, Sato C, Naj A, Rajbhandary R, Wang LS, Valladares O, Lin CF, Larson EB, Graff-Radford NR, Evans D, De Jager PL, Crane PK, Buxbaum JD, Murrell JR, Raj T, Ertekin-Taner N, Logue M, Baldwin CT, Green RC, Barnes LL, Cantwell LB, Fallin MD, Go RCP, Griffith PA, Obisesan TO, Manly JJ, Lunetta KL, Kamboh MI, Lopez OL, Bennett DA, Hendrie H, Hall KS, Goate AM, Byrd GS, Kukull WA, Foroud TM, Haines JL, Farrer LA, Pericak-Vance MA, Lee JH, Schellenberg GD, St George-Hyslop P, Mayeux R, Rogaeva E. Association of Long Runs of Homozygosity With Alzheimer Disease Among African American Individuals. JAMA Neurol 2016; 72:1313-23. [PMID: 26366463 DOI: 10.1001/jamaneurol.2015.1700] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Mutations in known causal Alzheimer disease (AD) genes account for only 1% to 3% of patients and almost all are dominantly inherited. Recessive inheritance of complex phenotypes can be linked to long (>1-megabase [Mb]) runs of homozygosity (ROHs) detectable by single-nucleotide polymorphism (SNP) arrays. OBJECTIVE To evaluate the association between ROHs and AD in an African American population known to have a risk for AD up to 3 times higher than white individuals. DESIGN, SETTING, AND PARTICIPANTS Case-control study of a large African American data set previously genotyped on different genome-wide SNP arrays conducted from December 2013 to January 2015. Global and locus-based ROH measurements were analyzed using raw or imputed genotype data. We studied the raw genotypes from 2 case-control subsets grouped based on SNP array: Alzheimer's Disease Genetics Consortium data set (871 cases and 1620 control individuals) and Chicago Health and Aging Project-Indianapolis Ibadan Dementia Study data set (279 cases and 1367 control individuals). We then examined the entire data set using imputed genotypes from 1917 cases and 3858 control individuals. MAIN OUTCOMES AND MEASURES The ROHs larger than 1 Mb, 2 Mb, or 3 Mb were investigated separately for global burden evaluation, consensus regions, and gene-based analyses. RESULTS The African American cohort had a low degree of inbreeding (F ~ 0.006). In the Alzheimer's Disease Genetics Consortium data set, we detected a significantly higher proportion of cases with ROHs greater than 2 Mb (P = .004) or greater than 3 Mb (P = .02), as well as a significant 114-kilobase consensus region on chr4q31.3 (empirical P value 2 = .04; ROHs >2 Mb). In the Chicago Health and Aging Project-Indianapolis Ibadan Dementia Study data set, we identified a significant 202-kilobase consensus region on Chr15q24.1 (empirical P value 2 = .02; ROHs >1 Mb) and a cluster of 13 significant genes on Chr3p21.31 (empirical P value 2 = .03; ROHs >3 Mb). A total of 43 of 49 nominally significant genes common for both data sets also mapped to Chr3p21.31. Analyses of imputed SNP data from the entire data set confirmed the association of AD with global ROH measurements (12.38 ROHs >1 Mb in cases vs 12.11 in controls; 2.986 Mb average size of ROHs >2 Mb in cases vs 2.889 Mb in controls; and 22% of cases with ROHs >3 Mb vs 19% of controls) and a gene-cluster on Chr3p21.31 (empirical P value 2 = .006-.04; ROHs >3 Mb). Also, we detected a significant association between AD and CLDN17 (empirical P value 2 = .01; ROHs >1 Mb), encoding a protein from the Claudin family, members of which were previously suggested as AD biomarkers. CONCLUSIONS AND RELEVANCE To our knowledge, we discovered the first evidence of increased burden of ROHs among patients with AD from an outbred African American population, which could reflect either the cumulative effect of multiple ROHs to AD or the contribution of specific loci harboring recessive mutations and risk haplotypes in a subset of patients. Sequencing is required to uncover AD variants in these individuals.
Collapse
Affiliation(s)
- Mahdi Ghani
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Christiane Reitz
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, New York3Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, New York4
| | - Rong Cheng
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Badri Narayan Vardarajan
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Gyungah Jun
- Department of Medicine (Biomedical Genetics), Boston University, Boston, Massachusetts6Department of Biostatistics, Boston University, Boston, Massachusetts7Department of Ophthalmology, Boston University, Boston, Massachusetts
| | - Christine Sato
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Adam Naj
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida
| | - Ruchita Rajbhandary
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Otto Valladares
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Chiao-Feng Lin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Eric B Larson
- Department of Medicine, University of Washington, Seattle11Group Health Research Institute, Group Health, Seattle, Washington
| | - Neill R Graff-Radford
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida13Department of Neurology, Mayo Clinic, Jacksonville, Florida
| | - Denis Evans
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Philip L De Jager
- Program in Translational Neuropsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts16Harvard Medical School, Boston, Massachusetts17Program in Medical and Population Genetics, The Broad Institute, Cambridge, Ma
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle
| | - Joseph D Buxbaum
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York19Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, New York20Department of Neuroscience, Mount Sinai School of Medicine, New York, New York2
| | - Jill R Murrell
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis
| | | | - Nilufer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida13Department of Neurology, Mayo Clinic, Jacksonville, Florida
| | - Mark Logue
- Department of Medicine (Biomedical Genetics), Boston University, Boston, Massachusetts
| | - Clinton T Baldwin
- Department of Medicine (Biomedical Genetics), Boston University, Boston, Massachusetts
| | - Robert C Green
- Harvard Medical School, Boston, Massachusetts23Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts24Partners Center for Personalized Genetic Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lisa L Barnes
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois26Department of Behavioral Sciences, Rush University Medical Center, Chicago, Illinois
| | - Laura B Cantwell
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - M Daniele Fallin
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, Maryland
| | - Rodney C P Go
- School of Public Health, University of Alabama at Birmingham
| | | | | | - Jennifer J Manly
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, New York4Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Kathryn L Lunetta
- Department of Biostatistics, Boston University, Boston, Massachusetts
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania32Alzheimer's Disease Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Oscar L Lopez
- Alzheimer's Disease Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David A Bennett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois33Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Hugh Hendrie
- Indiana University Center for Aging Research, Indianapolis35Department of Psychiatry, Indiana University School of Medicine, Indianapolis36Regenstrief Institute Inc, Indianapolis, Indiana
| | - Kathleen S Hall
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis
| | - Alison M Goate
- Hope Center Program on Protein Aggregation and Neurodegeneration, Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Goldie S Byrd
- Department of Biology, North Carolina A & T University, Greensboro
| | - Walter A Kukull
- National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle
| | - Tatiana M Foroud
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, Illinois
| | - Jonathan L Haines
- Vanderbilt Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University, Boston, Massachusetts6Department of Biostatistics, Boston University, Boston, Massachusetts7Department of Ophthalmology, Boston University, Boston, Massachusetts41Department of Neurology, Bo
| | | | - Joseph H Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, New York3Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, New York4
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Peter St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, New York3Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, New York4
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
48
|
Mruk DD, Cheng CY. The Mammalian Blood-Testis Barrier: Its Biology and Regulation. Endocr Rev 2015; 36:564-91. [PMID: 26357922 PMCID: PMC4591527 DOI: 10.1210/er.2014-1101] [Citation(s) in RCA: 442] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 09/03/2015] [Indexed: 12/31/2022]
Abstract
Spermatogenesis is the cellular process by which spermatogonia develop into mature spermatids within seminiferous tubules, the functional unit of the mammalian testis, under the structural and nutritional support of Sertoli cells and the precise regulation of endocrine factors. As germ cells develop, they traverse the seminiferous epithelium, a process that involves restructuring of Sertoli-germ cell junctions, as well as Sertoli-Sertoli cell junctions at the blood-testis barrier. The blood-testis barrier, one of the tightest tissue barriers in the mammalian body, divides the seminiferous epithelium into 2 compartments, basal and adluminal. The blood-testis barrier is different from most other tissue barriers in that it is not only comprised of tight junctions. Instead, tight junctions coexist and cofunction with ectoplasmic specializations, desmosomes, and gap junctions to create a unique microenvironment for the completion of meiosis and the subsequent development of spermatids into spermatozoa via spermiogenesis. Studies from the past decade or so have identified the key structural, scaffolding, and signaling proteins of the blood-testis barrier. More recent studies have defined the regulatory mechanisms that underlie blood-testis barrier function. We review here the biology and regulation of the mammalian blood-testis barrier and highlight research areas that should be expanded in future studies.
Collapse
Affiliation(s)
- Dolores D Mruk
- Center for Biomedical Research, Population Council, New York, New York 10065
| | - C Yan Cheng
- Center for Biomedical Research, Population Council, New York, New York 10065
| |
Collapse
|
49
|
Li CH, Shyu MK, Jhan C, Cheng YW, Tsai CH, Liu CW, Lee CC, Chen RM, Kang JJ. Gold Nanoparticles Increase Endothelial Paracellular Permeability by Altering Components of Endothelial Tight Junctions, and Increase Blood-Brain Barrier Permeability in Mice. Toxicol Sci 2015; 148:192-203. [PMID: 26272951 DOI: 10.1093/toxsci/kfv176] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Gold nanoparticles (Au-NPs) are being increasingly used as constituents in cosmetics, biosensors, bioimaging, photothermal therapy, and targeted drug delivery. This elevated exposure to Au-NPs poses systemic risks in humans, particularly risks associated with the biodistribution of Au-NPs and their potent interaction with biological barriers. We treated human umbilical vein endothelial cells with Au-NPs and comprehensively examined the expression levels of tight junction (TJ) proteins such as occludin, claudin-5, junctional adhesion molecules, and zonula occludens-1 (ZO-1), as well as endothelial paracellular permeability and the intracellular signaling required for TJ organization. Moreover, we validated the effects of Au-NPs on the integrity of TJs in mouse brain microvascular endothelial cells in vitro and obtained direct evidence of their influence on blood-brain barrier (BBB) permeability in vivo. Treatment with Au-NPs caused a pronounced reduction of PKCζ-dependent threonine phosphorylation of occludin and ZO-1, which resulted in the instability of endothelial TJs and led to proteasome-mediated degradation of TJ components. This impairment in the assembly of TJs between endothelial cells increased the permeability of the transendothelial paracellular passage and the BBB. Au-NPs increased endothelial paracellular permeability in vitro and elevated BBB permeability in vivo. Future studies must investigate the direct and indirect toxicity caused by Au-NP-induced endothelial TJ opening and thereby address the double-edged-sword effect of Au-NPs.
Collapse
Affiliation(s)
- Ching-Hao Li
- *Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan;
| | - Ming-Kwang Shyu
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng Jhan
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Wen Cheng
- School of Pharmacy, Taipei Medicine University, Taipei, Taiwan
| | - Chi-Hao Tsai
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Wei Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Chen Lee
- Department of Microbiology and Immunology, School of Medicine, China Medicine University, Taichung, Taiwan
| | - Ruei-Ming Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Cell Physiology and Molecular Image Research Center, Taipei Medical University's Wan-Fang Hospital, Taipei, Taiwan; and Anesthetics Toxicology Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Jaw-Jou Kang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan;
| |
Collapse
|
50
|
Blecharz KG, Colla R, Rohde V, Vajkoczy P. Control of the blood-brain barrier function in cancer cell metastasis. Biol Cell 2015; 107:342-71. [PMID: 26032862 DOI: 10.1111/boc.201500011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/22/2015] [Indexed: 12/25/2022]
Abstract
Cerebral metastases are the most common brain neoplasms seen clinically in the adults and comprise more than half of all brain tumours. Actual treatment options for brain metastases that include surgical resection, radiotherapy and chemotherapy are rarely curative, although palliative treatment improves survival and life quality of patients carrying brain-metastatic tumours. Chemotherapy in particular has also shown limited or no activity in brain metastasis of most tumour types. Many chemotherapeutic agents used systemically do not cross the blood-brain barrier (BBB), whereas others may transiently weaken the BBB and allow extravasation of tumour cells from the circulation into the brain parenchyma. Increasing evidence points out that the interaction between the BBB and tumour cells plays a key role for implantation and growth of brain metastases in the central nervous system. The BBB, as the tightest endothelial barrier, prevents both early detection and treatment by creating a privileged microenvironment. Therefore, as observed in several in vivo studies, precise targetting the BBB by a specific transient opening of the structure making it permeable for therapeutic compounds, might potentially help to overcome this difficult clinical problem. Moreover, a better understanding of the molecular features of the BBB, its interrelation with metastatic tumour cells and the elucidation of cellular mechanisms responsible for establishing cerebral metastasis must be clearly outlined in order to promote treatment modalities that particularly involve chemotherapy. This in turn would substantially expand the survival and quality of life of patients with brain metastasis, and potentially increase the remission rate. Therefore, the focus of this review is to summarise the current knowledge on the role and function of the BBB in cancer metastasis.
Collapse
Affiliation(s)
- Kinga G Blecharz
- Department of Experimental Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, 10119, Germany
| | - Ruben Colla
- Department of Neurosurgery, Göttingen University Medical Center, Göttingen, 37070, Germany
| | - Veit Rohde
- Department of Neurosurgery, Göttingen University Medical Center, Göttingen, 37070, Germany
| | - Peter Vajkoczy
- Department of Experimental Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, 10119, Germany.,Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, 13353, Germany
| |
Collapse
|