101
|
Wei JY, Liu H, Li Y, Zhao D, Wang B, Wang HJ, Wang L, Wang KJ, Yue JL, Zhang HY, Li TY, Miao YJ, Wang KL, Tong PG, Zhang Z, Li ZY, Shi Z, Yao JY, Liu DX, Fang WG, Li B, Shang DS, Lyu Y, Sun HZ, Zhao WD, Chen YH. Melatonin Protects Against Cocaine-Induced Blood-Brain Barrier Dysfunction and Cognitive Impairment by Regulating miR-320a-Dependent GLUT1 Expression. J Pineal Res 2024; 76:e70002. [PMID: 39539049 DOI: 10.1111/jpi.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Cocaine abuse has been strongly linked to blood-brain barrier (BBB) dysfunction, though the exact mechanism by which cocaine disrupts the BBB remains unclear. In this study, we found that cocaine treatment reduces the expression of glucose transporter 1 (GLUT1) in brain microvascular endothelial cells, a key factor in cocaine-induced brain glucose uptake, BBB leakage, and cognitive impairment. Mechanistically, our results show that cocaine upregulates miR-320a, which in turn suppresses GLUT1 expression via the beta 2-adrenergic receptor (ADRB2). Notably, the administration of adeno-associated viruses encoding full-length GLUT1 or miR-320a inhibitors to the brain microvascular endothelium significantly mitigated cocaine-induced BBB leakage and cognitive deficits. Additionally, we discovered that melatonin, a well-known neuroprotective hormone, alleviates cocaine-induced BBB disruption and cognitive impairment. This protective effect of melatonin was mediated through the upregulation of miR-320a-dependent GLUT1 expression in brain endothelial cells via MT1 receptor-mediated inhibition of the cAMP/PKA/CREB signaling pathway. In conclusion, our findings demonstrate that cocaine downregulates brain microvascular GLUT1, leading to BBB dysfunction, and highlight melatonin as a potential therapeutic agent for treating cocaine-related complications.
Collapse
Affiliation(s)
- Jia-Yi Wei
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Hui Liu
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Yuan Li
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Dan Zhao
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Bo Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hui-Jie Wang
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Li Wang
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Kang-Ji Wang
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Jin-Li Yue
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Hong-Yan Zhang
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Tian-Yue Li
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Yi-Jue Miao
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Kai-Li Wang
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Pai-Ge Tong
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Zhuo Zhang
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Ze-Ye Li
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Zheng Shi
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Jia-Yuan Yao
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Dong-Xin Liu
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Wen-Gang Fang
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Bo Li
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - De-Shu Shang
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Yuan Lyu
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Department of Obstetrics and Gynecology, Research Center of China Medical University Birth Cohort, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hong-Zan Sun
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei-Dong Zhao
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| | - Yu-Hua Chen
- Key Laboratory of Cell Biology and Key Laboratory of Medical Cell Biology, Department of Developmental Cell Biology, Ministry of Public Health and Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|
102
|
Khodadadi M, Pirzad Jahromi G, Meftahi GH, Khodadadi H, Hadipour M, Ezami M. Crocin nano-chitosan-coated compound mitigates hippocampal blood-brain barrier disruption, anxiety, and cognitive deficits in chronic immobilization stress-induced rats. Heliyon 2024; 10:e39203. [PMID: 39640648 PMCID: PMC11620202 DOI: 10.1016/j.heliyon.2024.e39203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/07/2024] Open
Abstract
Stressful conditions can disrupt the central nervous system's normal homeostasis and physiological functions, resulting in blood-brain barrier malfunction, memory and learning impairment, anxiety, etc. Crocin is a long-investigated natural compound that has been documented to have anti-inflammation and neuroprotective effects, albeit it comes with some limitations such as low stability and bioavailability. Therefore, we aimed to overcome crocin's limitations by coating crocin with a nano-carrier (chitosan) in the chronic immobilization stress-induced rat model. Crocin was encapsulated into chitosan nanoparticles by a modified method. A total of 35 male Wistar rats were selected as our study subjects (220-250 g) which were randomly divided into 5 groups (control, stress, nanoparticle, crocin, and chitosan). Chronic immobilization stress was induced by placing rats for 2 h into a plastic bottle with specific measurements (for 14 consecutive days) to prevent animals from moving. To evaluate the memory and learning changes, we used the Barnes maze test and the Passive avoidance test followed by the evaluation of the N-methyl-D-aspartate |(NMDA) receptor subunits genes (GRIN1 and GRIN2A) expression. Anxiety levels were evaluated by elevated plus maze test. Furthermore, the changes in the expression of genes responsible for encoding the tight junction proteins of BBB including ZO1, CLDN5, and OCLN were assessed by RT-PCR. Compared to intact crocin, the administration of crocin nano-chitosan-coated compound resulted in significant improvement of specific memory and learning indicators as well as a significant reduction of anxiety levels in chronic immobilization stress-induced rats. Finally, we observed that treatment with the crocin nano-chitosan-coated compound can elevate the expression levels of the genes responsible for encoding NMDA receptor subunits, and the genes responsible for encoding the tight junction proteins of blood-brain barriers in the hippocampus.
Collapse
Affiliation(s)
- Mohsen Khodadadi
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Gila Pirzad Jahromi
- Neuroscience Research Centre, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Hossein Khodadadi
- The Polish Academy of Sciences, Institute of Genetics and Animal Biotechnology, Warsaw, Poland
| | | | - Masoud Ezami
- Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
103
|
Huang L, Zhang X, Zhang J, Li L, Zhou X, Yang T, An X. Efficacy of non-invasive brain stimulation for post-stroke sleep disorders: a systematic review and meta-analysis. Front Neurol 2024; 15:1420363. [PMID: 39539650 PMCID: PMC11557329 DOI: 10.3389/fneur.2024.1420363] [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: 04/20/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Objective This study aimed to systematically assess the clinical efficacy of non-invasive brain stimulation (NIBS) for treating post-stroke sleep disorders (PSSD). Methods We conducted thorough literature search across multiple databases, including PubMed, Web of Science, EmBase, Cochrane Library, Scopus, China Biology Medicine (CBM); China National Knowledge Infrastructure (CNKI); Technology Periodical Database (VIP), and Wanfang Database, focusing on RCTs examining NIBS for PSSD. Meta-analyses were performed using RevMan 5.4 and Stata 14. Results Eighteen articles were reviewed, including 16 on repetitive Transcranial Magnetic Stimulation (rTMS), one on Theta Burst Stimulation (TBS), and two on transcranial Direct Current Stimulation (tDCS). Meta-analysis results indicated that rTMS within NIBS significantly improved the Pittsburgh Sleep Quality Index (PSQI) score (MD = -1.85, 95% CI [-2.99, -0.71], p < 0.05), the 17-item Hamilton Depression Rating Scale (HAMD-17) score [MD = -2.85, 95% CI (-3.40, -2.30), p < 0.05], and serum brain-derived neurotrophic factor (BDNF) levels [MD = 4.19, 95% CI (2.70, 5.69), p < 0.05], while reducing the incidence of adverse reactions [RR = 0.36, 95% CI (0.23, 0.55), p < 0.05]. TBS significantly improved the PSQI score in patients with PSSD (p < 0.05). Conversely, tDCS significantly improved the HAMD-17 score in PSSD patients [MD = -1.52, 95% CI (-3.41, -0.64), p < 0.05]. Additionally, rTMS improved sleep parameters, including Stage 2 sleep (S2%) and combined Stage 3 and 4 sleep (S3 + S4%) (p < 0.05), while tDCS improved total sleep time (TST) and sleep efficiency (SE) (p < 0.05).Subgroup analysis results indicated: (1) Both LF-rTMS and HF-rTMS improved PSQI scores (p < 0.05). (2) Both rTMS combined with medication and rTMS alone improved PSQI scores (p < 0.05). Compared to the sham/blank group, the rTMS group showed improvements in SE, sleep latency (SL), S1%, S3 + S4%, and REM sleep (REM%). The rTMS combined with medication group showed improved SL compared to the medication-only group (p < 0.05). Conclusion NIBS effectively improves sleep quality, structure, depression levels, and BDNF levels in PSSD patients, while also being safe. Further investigations into the potential of NIBS in PSSD treatment may provide valuable insights for clinical applications. Systematic review registration https://www.crd.york.ac.uk/prospero/, CRD42023485317.
Collapse
Affiliation(s)
- Linyu Huang
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xingling Zhang
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Zhang
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Long Li
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianyu Zhou
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tingyu Yang
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuemei An
- Nursing Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Administrative Management Department, Deyang Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| |
Collapse
|
104
|
Wright B, King S, Suphioglu C. The Importance of Phosphoinositide 3-Kinase in Neuroinflammation. Int J Mol Sci 2024; 25:11638. [PMID: 39519189 PMCID: PMC11546674 DOI: 10.3390/ijms252111638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Neuroinflammation, characterised by the activation of immune cells in the central nervous system (CNS), plays a dual role in both protecting against and contributing to the progression of neurodegenerative diseases, such as Alzheimer's disease (AD) and multiple sclerosis (MS). This review explores the role of phosphoinositide 3-kinase (PI3K), a key enzyme involved in cellular survival, proliferation, and inflammatory responses, within the context of neuroinflammation. Two PI3K isoforms of interest, PI3Kγ and PI3Kδ, are specific to the regulation of CNS cells, such as microglia, astrocytes, neurons, and oligodendrocytes, influencing pathways, such as Akt, mTOR, and NF-κB, that control cytokine production, immune cell activation, and neuroprotection. The dysregulation of PI3K signalling is implicated in chronic neuroinflammation, contributing to the exacerbation of neurodegenerative diseases. Preclinical studies show promise in targeting neuronal disorders using PI3K inhibitors, such as AS605240 (PI3Kγ) and idelalisib (PI3Kδ), which have reduced inflammation, microglial activation, and neuronal death in in vivo models of AD. However, the clinical translation of these inhibitors faces challenges, including blood-brain barrier (BBB) permeability, isoform specificity, and long-term safety concerns. This review highlights the therapeutic potential of PI3K modulation in neuroinflammatory diseases, identifying key gaps in the current research, particularly in the need for brain-penetrating and isoform-specific inhibitors. These findings underscore the importance of future research to develop targeted therapies that can effectively modulate PI3K activity and provide neuroprotection in chronic neurodegenerative disorders.
Collapse
Affiliation(s)
- Brock Wright
- NeuroAllergy Research Laboratory (NARL), School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia; (B.W.); (S.K.)
- Centre for Sustainable Bioproducts, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
| | - Samuel King
- NeuroAllergy Research Laboratory (NARL), School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia; (B.W.); (S.K.)
- Centre for Sustainable Bioproducts, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
| | - Cenk Suphioglu
- NeuroAllergy Research Laboratory (NARL), School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia; (B.W.); (S.K.)
- Centre for Sustainable Bioproducts, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
| |
Collapse
|
105
|
Shirvalilou S, Khoei S, Afzalipour R, Ghaznavi H, Shirvaliloo M, Derakhti Z, Sheervalilou R. Targeting the undruggable in glioblastoma using nano-based intracellular drug delivery. Med Oncol 2024; 41:303. [PMID: 39470962 DOI: 10.1007/s12032-024-02546-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/17/2024] [Indexed: 11/01/2024]
Abstract
Glioblastoma (GBM) is a highly prevalent and aggressive brain tumor in adults with limited treatment response, leading to a 5-year survival rate of less than 5%. Standard therapies, including surgery, radiation, and chemotherapy, often fall short due to the tumor's location, hypoxic conditions, and the challenge of complete removal. Moreover, brain metastases from cancers such as breast and melanoma carry similarly poor prognoses. Recent advancements in nanomedicine offer promising solutions for targeted GBM therapies, with nanoparticles (NPs) capable of delivering chemotherapy drugs or radiation sensitizers across the blood-brain barrier (BBB) to specific tumor sites. Leveraging the enhanced permeability and retention effect, NPs can preferentially accumulate in tumor tissues, where compromised BBB regions enhance delivery efficiency. By modifying NP characteristics such as size, shape, and surface charge, researchers have improved circulation times and cellular uptake, enhancing therapeutic efficacy. Recent studies show that combining photothermal therapy with magnetic hyperthermia using AuNPs and magnetic NPs induces ROS-dependent apoptosis and immunogenic cell death providing dual-targeted, immune-activating approaches. This review discusses the latest NP-based drug delivery strategies, including gene therapy, receptor-mediated transport, and multi-modal approaches like photothermal-magnetic hyperthermia combinations, all aimed at optimizing therapeutic outcomes for GBM.
Collapse
Affiliation(s)
- Sakine Shirvalilou
- Finetech in Medicine Research Center, Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samideh Khoei
- Finetech in Medicine Research Center, Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Afzalipour
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
- Department of Radiology, Faculty of Para-Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Milad Shirvaliloo
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Future Science Group, Unitec House, 2 Albert Place, London, N3 1QB, UK
| | - Zahra Derakhti
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Roghayeh Sheervalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
106
|
Abbott KL, Subudhi S, Ferreira R, Gültekin Y, Steinbuch SC, Munim MB, Honeder SE, Kumar AS, Ramesh DL, Wu M, Hansen JA, Sivanand S, Riedmayr LM, Duquette M, Ali A, Henning N, Shevzov-Zebrun A, Gourgue F, Barbeau AM, Waite M, Kunchok T, Ferraro GB, Do BT, Spanoudaki V, Sánchez-Rivera FJ, Jin X, Church GM, Jain RK, Vander Heiden MG. Site of breast cancer metastasis is independent of single nutrient levels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.616714. [PMID: 39484531 PMCID: PMC11527034 DOI: 10.1101/2024.10.24.616714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Cancer metastasis is a major contributor to patient morbidity and mortality1, yet the factors that determine the organs where cancers can metastasize are incompletely understood. In this study, we quantify the absolute levels of over 100 nutrients available across multiple tissues in mice and investigate how this relates to the ability of breast cancer cells to grow in different organs. We engineered breast cancer cells with broad metastatic potential to be auxotrophic for specific nutrients and assessed their ability to colonize different organs. We then asked how tumor growth in different tissues relates to nutrient availability and tumor biosynthetic activity. We find that single nutrients alone do not define the sites where breast cancer cells can grow as metastases. Additionally, we identify purine synthesis as a requirement for tumor growth and metastasis across many tissues and find that this phenotype is independent of tissue nucleotide availability or tumor de novo nucleotide synthesis activity. These data suggest that a complex interplay of multiple nutrients within the microenvironment dictates potential sites of metastatic cancer growth, and highlights the interdependence between extrinsic environmental factors and intrinsic cellular properties in influencing where breast cancer cells can grow as metastases.
Collapse
Affiliation(s)
- Keene L. Abbott
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sonu Subudhi
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Raphael Ferreira
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Department of Genetics, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Yetiş Gültekin
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sophie C. Steinbuch
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Muhammad Bin Munim
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sophie E. Honeder
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, Vienna, Austria
| | - Ashwin S. Kumar
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Diya L. Ramesh
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michelle Wu
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob A. Hansen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sharanya Sivanand
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lisa M. Riedmayr
- Harvard Medical School, Department of Genetics, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Mark Duquette
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ahmed Ali
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nicole Henning
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anna Shevzov-Zebrun
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Florian Gourgue
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anna M. Barbeau
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Millenia Waite
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Tenzin Kunchok
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Gino B. Ferraro
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian T. Do
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Virginia Spanoudaki
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Francisco J. Sánchez-Rivera
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xin Jin
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
| | - George M. Church
- Harvard Medical School, Department of Genetics, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Rakesh K. Jain
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthew G. Vander Heiden
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
107
|
De Luca C, Virtuoso A, Papa M, Cirillo G, La Rocca G, Corvino S, Barbarisi M, Altieri R. The Three Pillars of Glioblastoma: A Systematic Review and Novel Analysis of Multi-Omics and Clinical Data. Cells 2024; 13:1754. [PMID: 39513861 PMCID: PMC11544881 DOI: 10.3390/cells13211754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma is the most fatal and common malignant brain tumor, excluding metastasis and with a median survival of approximately one year. While solid tumors benefit from newly approved drugs, immunotherapy, and prevention, none of these scenarios are opening for glioblastoma. The key to unlocking the peculiar features of glioblastoma is observing its molecular and anatomical features tightly entangled with the host's central nervous system (CNS). In June 2024, we searched the PUBMED electronic database. Data collection and analysis were conducted independently by two reviewers. Results: A total of 215 articles were identified, and 192 were excluded based on inclusion and exclusion criteria. The remaining 23 were used for collecting divergent molecular pathways and anatomical features of glioblastoma. The analysis of the selected papers revealed a multifaced tumor with extreme variability and cellular reprogramming that are observable within the same patient. All the variability of glioblastoma could be clustered into three pillars to dissect the physiology of the tumor: 1. necrotic core; 2. vascular proliferation; 3. CNS infiltration. These three pillars support glioblastoma survival, with a pivotal role of the neurovascular unit, as supported by the most recent paper published by experts in the field.
Collapse
Affiliation(s)
- Ciro De Luca
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
| | - Assunta Virtuoso
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
- ISBE Italy, SYSBIO Centre of Systems Biology, 20126 Milan, Italy
| | - Giovanni Cirillo
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
| | - Giuseppe La Rocca
- Department of Neurosurgery, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Catholic University of Rome School of Medicine, 00153 Rome, Italy;
| | - Sergio Corvino
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, Neurosurgical Clinic, University “Federico II” of Naples, 80131 Naples, Italy;
| | - Manlio Barbarisi
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy (R.A.)
| | - Roberto Altieri
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy (R.A.)
| |
Collapse
|
108
|
Młynarska E, Jakubowska P, Frąk W, Gajewska A, Sornowska J, Skwira S, Wasiak J, Rysz J, Franczyk B. Associations of Microbiota and Nutrition with Cognitive Impairment in Diseases. Nutrients 2024; 16:3570. [PMID: 39458564 PMCID: PMC11510709 DOI: 10.3390/nu16203570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Recent research highlights the growing interest in the impact of nutrition on cognitive health and function in disease, as dietary habits are increasingly recognized as crucial factors in relation to brain function. This focus is especially important given the rising prevalence of neurodegenerative diseases and the cognitive decline associated with poor dietary choices. Links are now being sought between brain function and the microbiota and gut-brain axis. Mechanisms are proposed that include low-grade chronic neuroinflammation, the influence of short-chain fatty acids, or the disruption of glial cells and transmitters in the brain. METHODS We reviewed the articles on pubmed. This is not a systematic review, but of the narrative type. We wanted to outline the issue and summarise the latest information. RESULTS The axis in question has its foundation in nutrition. It has been reported that diet, particularly the components and the timing of food intake, has an impact on cognitive processes. The Mediterranean diet is most often cited in the literature as being beneficial to health. In order to obtain a more complete view, it is worth considering other dietary patterns, even those that impair our health. CONCLUSIONS Determining what is beneficial and what is not will allow us to develop a speronized strategy for the prevention of, and fight against, cognitive impairment. Appropriately selected supplements, the functions of which we have also discussed, may prove supportive.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Paulina Jakubowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Weronika Frąk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Sornowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Sylwia Skwira
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
109
|
Wei L, Aitchison JD, Kaushansky A, Mast FD. Systems-level reconstruction of kinase phosphosignaling networks regulating endothelial barrier integrity using temporal data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.606198. [PMID: 39149238 PMCID: PMC11326140 DOI: 10.1101/2024.08.01.606198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Phosphosignaling networks control cellular processes. We built kinase-mediated regulatory networks elicited by thrombin stimulation of brain endothelial cells using two computational strategies: Temporal Pathway Synthesizer (TPS), which uses phosphoproetiomics data as input, and Temporally REsolved KInase Network Generation (TREKING), which uses kinase inhibitor screens. TPS and TREKING predicted overlapping barrier-regulatory kinases connected with unique network topology. Each strategy effectively describes regulatory signaling networks and is broadly applicable across biological systems.
Collapse
|
110
|
Munzen ME, Mathew C, Enriquez V, Minhas A, Charles-Niño CL, Saytoo D, Reguera-Gomez M, Dores MR, Martinez LR. Inhibition of RhoA Prevents Cryptococcus neoformans Capsule Glucuronoxylomannan-Stimulated Brain Endothelial Barrier Disruption. J Infect Dis 2024; 230:1042-1051. [PMID: 38622836 PMCID: PMC11481333 DOI: 10.1093/infdis/jiae187] [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: 12/13/2023] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024] Open
Abstract
Cryptococcus neoformans (Cn) is an opportunistic fungus that causes severe central nervous system (CNS) disease in immunocompromised individuals. Brain parenchyma invasion requires fungal traversal of the blood-brain barrier. In this study, we describe that Cn alters the brain endothelium by activating small GTPase RhoA, causing reorganization of the actin cytoskeleton and tight junction modulation to regulate endothelial barrier permeability. We confirm that the main fungal capsule polysaccharide glucuronoxylomannan is responsible for these alterations. We reveal a therapeutic benefit of RhoA inhibition by CCG-1423 in vivo. RhoA inhibition prolonged survival and reduced fungal burden in a murine model of disseminated cryptococcosis, supporting the therapeutic potential of targeting RhoA in the context of cryptococcal infection. We examine the complex virulence of Cn in establishing CNS disease, describing cellular components of the brain endothelium that may serve as molecular targets for future antifungal therapies to alleviate the burden of life-threatening cryptococcal CNS infection.
Collapse
Affiliation(s)
- Melissa E Munzen
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville
| | - Cristian Mathew
- Department of Biology, Hofstra University, Hempstead, New York
| | - Vanessa Enriquez
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville
| | - Amanjeet Minhas
- Department of Biology, Hofstra University, Hempstead, New York
| | | | | | - Marta Reguera-Gomez
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville
| | - Michael R Dores
- Department of Biology, Hofstra University, Hempstead, New York
| | - Luis R Martinez
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville
- Emerging Pathogens Institute
- Center for Immunology and Transplantation
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville
| |
Collapse
|
111
|
Velmurugan GV, Vekaria HJ, Hartz AMS, Bauer B, Hubbard WB. Oxidative stress alters mitochondrial homeostasis in isolated brain capillaries. Fluids Barriers CNS 2024; 21:81. [PMID: 39407313 PMCID: PMC11476969 DOI: 10.1186/s12987-024-00579-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Neurovascular deficits and blood-brain barrier (BBB) dysfunction are major hallmarks of brain trauma and neurodegenerative diseases. Oxidative stress is a prominent contributor to neurovascular unit (NVU) dysfunction and can propagate BBB disruption. Oxidative damage results in an imbalance of mitochondrial homeostasis, which can further drive functional impairment of brain capillaries. To this end, we developed a method to track mitochondrial-related changes after oxidative stress in the context of neurovascular pathophysiology as a critical endophenotype of neurodegenerative diseases. METHODS To study brain capillary-specific mitochondrial function and dynamics in response to oxidative stress, we developed an ex vivo model in which we used isolated brain capillaries from transgenic mice that express dendra2 green specifically in mitochondria (mtD2g). Isolated brain capillaries were incubated with 2,2'-azobis-2-methyl-propanimidamide dihydrochloride (AAPH) or hydrogen peroxide (H2O2) to induce oxidative stress through lipid peroxidation. Following the oxidative insult, mitochondrial bioenergetics were measured using the Seahorse XFe96 flux analyzer, and mitochondrial dynamics were measured using confocal microscopy with Imaris software. RESULTS We optimized brain capillary isolation with intact endothelial cell tight-junction and pericyte integrity. Further, we demonstrate consistency of the capillary isolation process and cellular enrichment of the isolated capillaries. Mitochondrial bioenergetics and morphology assessments were optimized in isolated brain capillaries. Finally, we found that oxidative stress significantly decreased mitochondrial respiration and altered mitochondrial morphology in brain capillaries, including mitochondrial volume and count. CONCLUSIONS Following ex vivo isolation of brain capillaries, we confirmed the stability of mitochondrial parameters, demonstrating the feasibility of this newly developed platform. We also demonstrated that oxidative stress has profound effects on mitochondrial homeostasis in isolated brain capillaries. This novel method can be used to evaluate pharmacological interventions to target oxidative stress or mitochondrial dysfunction in cerebral small vessel disease and neurovascular pathophysiology as major players in neurodegenerative disease.
Collapse
Affiliation(s)
- Gopal V Velmurugan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, USA
- Department of Neuroscience, University of Kentucky, Lexington, USA
| | - Hemendra J Vekaria
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, USA
- Department of Neuroscience, University of Kentucky, Lexington, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, USA
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, USA
| | - Björn Bauer
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, USA
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, USA
| | - W Brad Hubbard
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, USA.
- Department of Physiology, University of Kentucky, Lexington, USA.
- Lexington Veterans' Affairs Healthcare System, Lexington, USA.
| |
Collapse
|
112
|
Abtin S, Seyedaghamiri F, Aalidaeijavadi Z, Farrokhi AM, Moshrefi F, Ziveh T, Zibaii MI, Aliakbarian H, Rezaei-Tavirani M, Haghparast A. A review on the consequences of molecular and genomic alterations following exposure to electromagnetic fields: Remodeling of neuronal network and cognitive changes. Brain Res Bull 2024; 217:111090. [PMID: 39349259 DOI: 10.1016/j.brainresbull.2024.111090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/02/2024]
Abstract
The use of electromagnetic fields (EMFs) is essential in daily life. Since 1970, concerns have grown about potential health hazards from EMF. Exposure to EMF can stimulate nerves and affect the central nervous system, leading to neurological and cognitive changes. However, current research results are often vague and contradictory. These effects include changes in memory and learning through changes in neuronal plasticity in the hippocampus, synapses and hippocampal neuritis, and changes in metabolism and neurotransmitter levels. Prenatal exposure to EMFs has negative effects on memory and learning, as well as changes in hippocampal neuron density and histomorphology of hippocampus. EMF exposure also affects the structure and function of glial cells, affecting gate dynamics, ion conduction, membrane concentration, and protein expression. EMF exposure affects gene expression and may change epigenetic regulation through effects on DNA methylation, histone modification, and microRNA biogenesis, and potentially leading to biological changes. Therefore, exposure to EMFs possibly leads to changes in cellular and molecular mechanisms in central nervous system and alter cognitive function.
Collapse
Affiliation(s)
- Shima Abtin
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemehsadat Seyedaghamiri
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Aalidaeijavadi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Mohammad Farrokhi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fazel Moshrefi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayebeh Ziveh
- Laboratory of Biophysics and Molecular Biology, Departments of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | - Hadi Aliakbarian
- Faculty of Electrical Engineering, KN Toosi University of Technology, Tehran, Iran
| | | | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran; Department of Basic Sciences, Iranian Academy of Medical Sciences, Tehran, Iran.
| |
Collapse
|
113
|
Guillot P, Celle S, Barth N, Roche F, Perek N. 'Selected' Exosomes from Sera of Elderly Severe Obstructive Sleep Apnea Patients and Their Impact on Blood-Brain Barrier Function: A Preliminary Report. Int J Mol Sci 2024; 25:11058. [PMID: 39456840 PMCID: PMC11507461 DOI: 10.3390/ijms252011058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/07/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) affects a large part of the aging population. It is characterized by chronic intermittent hypoxia and associated with neurocognitive dysfunction. One hypothesis is that the blood-brain barrier (BBB) functions could be altered by exosomes. Exosomes are nanovesicles found in biological fluids. Through the study of exosomes and their content in tau and amyloid beta (Aβ), the aim of this study was to show how exosomes could be used as biomarkers of OSAS and of their cognitive disorders. Two groups of 15 volunteers from the PROOF cohort were selected: severe apnea (AHI > 30) and control (AHI < 5). After exosome isolation from blood serum, we characterized and quantified them (CD81, CD9, CD63) by western blot and ELISAs and put them 5 h in contact with an in vitro BBB model. The apparent permeability of the BBB was measured using sodium-fluorescein and TEER. Cell ELISAs were performed on tight junctions (ZO-1, claudin-5, occludin). The amount of tau and Aβ proteins found in the exosomes was quantified using ELISAs. Compared to controls, OSAS patients had a greater quantity of exosomes, tau, and Aβ proteins in their blood sera, which induced an increase in BBB permeability in the model and was reflected by a loss of tight junction' expression. Elderly patients suffering severe OSAS released more exosomes in serum from the brain compartment than controls. Such exosomes increased BBB permeability. The impact of such alterations on the risk of developing cognitive dysfunction and/or neurodegenerative diseases is questioned.
Collapse
Affiliation(s)
- Pauline Guillot
- Gérontopôle AURA, 42000 Saint-Etienne, France;
- Inserm, U1059, Sainbiose, Faculté de Médecine Jacques Lisfranc, Université de Lyon, 42000 Saint-Etienne, France; (S.C.); (F.R.); (N.P.)
- Faculté de Médecine Jacques Lisfranc, Université Jean Monnet, 42000 Saint-Etienne, France
| | - Sebastien Celle
- Inserm, U1059, Sainbiose, Faculté de Médecine Jacques Lisfranc, Université de Lyon, 42000 Saint-Etienne, France; (S.C.); (F.R.); (N.P.)
- Physiologie Clinique et de l’Exercice, Centre Visas, CHU Saint Etienne, 42000 Saint-Etienne, France
| | - Nathalie Barth
- Gérontopôle AURA, 42000 Saint-Etienne, France;
- Chaire Santé des Ainés, Ingénierie de la Prévention, Université Jen Monnet, 42000 Saint-Etienne, France
| | - Frederic Roche
- Inserm, U1059, Sainbiose, Faculté de Médecine Jacques Lisfranc, Université de Lyon, 42000 Saint-Etienne, France; (S.C.); (F.R.); (N.P.)
- Faculté de Médecine Jacques Lisfranc, Université Jean Monnet, 42000 Saint-Etienne, France
- Physiologie Clinique et de l’Exercice, Centre Visas, CHU Saint Etienne, 42000 Saint-Etienne, France
| | - Nathalie Perek
- Inserm, U1059, Sainbiose, Faculté de Médecine Jacques Lisfranc, Université de Lyon, 42000 Saint-Etienne, France; (S.C.); (F.R.); (N.P.)
- Faculté de Médecine Jacques Lisfranc, Université Jean Monnet, 42000 Saint-Etienne, France
| |
Collapse
|
114
|
Kuo PC, Weng WT, Scofield BA, Paraiso HC, Yu ICI, Yen JHJ. Ischemia-induced endogenous Nrf2/HO-1 axis activation modulates microglial polarization and restrains ischemic brain injury. Front Immunol 2024; 15:1440592. [PMID: 39469715 PMCID: PMC11513276 DOI: 10.3389/fimmu.2024.1440592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/18/2024] [Indexed: 10/30/2024] Open
Abstract
Cerebral ischemic stroke accounts for more than 80% of all stroke cases. During cerebral ischemia, reactive oxygen species produced in the ischemic brain induce oxidative stress and inflammatory responses. Nrf2 is a transcription factor responsible for regulating cellular redox balance through the induction of protective antioxidant and phase II detoxification responses. Although the induction of endogenous Nrf2/HO-1 axis activation has been observed in the ischemic brain, whether ischemia-induced endogenous Nrf2/HO-1 axis activation plays a role in modulating microglia (MG) phenotypes and restraining ischemic brain injury is not characterized and requires further exploration. To investigate that, we generated mice with Nrf2 knockdown specifically in MG to rigorously assess the role of endogenous Nrf2 activation in ischemic brain injury after stroke. Our results showed that MG-specific Nrf2 knockdown exacerbated ischemic brain injury after stroke. We found that Nrf2 knockdown altered MG phenotypes after stroke, in which increased frequency of inflammatory MG and decreased frequency of anti-inflammatory MG were detected in the ischemic brain. Moreover, we identified attenuated Nrf2/HO-1 axis activation led to increased CD68/IL-1β and suppressed CD206 expression in MG, resulting in aggravated inflammatory MG in MG-specific Nrf2 knockdown mice after stroke. Intriguingly, using type II diabetic preclinical models, we revealed that diabetic mice exhibited attenuated Nrf2/HO-1 axis activation in MG and exacerbated ischemic brain injury after stroke that phenocopy mice with MG-specific Nrf2 knockdown. Finally, the induction of exogenous Nrf2/HO-1 axis activation in MG through pharmacological approaches ameliorated ischemic brain injury in diabetic mice. In conclusion, our findings provide cellular and molecular insights demonstrating ischemia-induced endogenous Nrf2/HO-1 axis activation modulates MG phenotypes and restrains ischemic brain injury. These results further strengthen the therapeutic potential of targeting Nrf2/HO-1 axis in MG for the treatment of ischemic stroke and diabetic stroke.
Collapse
Affiliation(s)
- Ping-Chang Kuo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Wen-Tsan Weng
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Barbara A. Scofield
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Hallel C. Paraiso
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - I-Chen Ivorine Yu
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Jui-Hung Jimmy Yen
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, United States
| |
Collapse
|
115
|
Holst MR, Richner M, Arenshøj PO, Alam P, Hyldig K, Nielsen MS. Ex vivo nanoscale abluminal mapping of putative cargo receptors at the blood-brain barrier of expanded brain capillaries. Fluids Barriers CNS 2024; 21:80. [PMID: 39402596 PMCID: PMC11475543 DOI: 10.1186/s12987-024-00585-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
Receptor mediated transport of therapeutic antibodies through the blood-brain barrier (BBB) give promise for drug delivery to alleviate brain diseases. We developed a low-cost method to obtain nanoscale localization data of putative cargo receptors. We combine existing ex vivo isolation methods with expansion microscopy (ExM) to analyze receptor localizations in brain microcapillaries. Using this approach, we show how to analyze receptor localizations in endothelial cells of brain microcapillaries in relation to the abluminal marker collagen IV. By choosing the thinnest capillaries, microcapillaries for analysis, we ensure the validity of collagen IV as an abluminal marker. With this tool, we confirm transferrin receptors as well as sortilin to be both luminally and abluminally localized. Furthermore, we identify basigin to be an abluminal receptor. Our methodology can be adapted to analyze different types of isolated brain capillaries and we anticipate that this approach will be very useful for the research community to gain new insight into cargo receptor trafficking in the slim brain endothelial cells to elucidate novel paths for future drug design.
Collapse
Affiliation(s)
| | - Mette Richner
- Department of Biomedicine, Aarhus University, Aarhus C, 8000, Denmark
| | | | - Parvez Alam
- Department of Biomedicine, Aarhus University, Aarhus C, 8000, Denmark
- Laboratory of Neurological Infection and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Kathrine Hyldig
- Department of Biomedicine, Aarhus University, Aarhus C, 8000, Denmark
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, Copenhagen, 2500, Denmark
| | | |
Collapse
|
116
|
Ding Y, Palecek SP, Shusta EV. iPSC-derived blood-brain barrier modeling reveals APOE isoform-dependent interactions with amyloid beta. Fluids Barriers CNS 2024; 21:79. [PMID: 39394110 PMCID: PMC11468049 DOI: 10.1186/s12987-024-00580-2] [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: 05/21/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Three common isoforms of the apolipoprotein E (APOE) gene - APOE2, APOE3, and APOE4 - hold varying significance in Alzheimer's Disease (AD) risk. The APOE4 allele is the strongest known genetic risk factor for late-onset Alzheimer's Disease (AD), and its expression has been shown to correlate with increased central nervous system (CNS) amyloid deposition and accelerated neurodegeneration. Conversely, APOE2 is associated with reduced AD risk and lower CNS amyloid burden. Recent clinical data have suggested that increased blood-brain barrier (BBB) leakage is commonly observed among AD patients and APOE4 carriers. However, it remains unclear how different APOE isoforms may impact AD-related pathologies at the BBB. METHODS To explore potential impacts of APOE genotypes on BBB properties and BBB interactions with amyloid beta, we differentiated isogenic human induced pluripotent stem cell (iPSC) lines with different APOE genotypes into both brain microvascular endothelial cell-like cells (BMEC-like cells) and brain pericyte-like cells. We then compared the effect of different APOE isoforms on BBB-related and AD-related phenotypes. Statistical significance was determined via ANOVA with Tukey's post hoc testing as appropriate. RESULTS Isogenic BMEC-like cells with different APOE genotypes had similar trans-endothelial electrical resistance, tight junction integrity and efflux transporter gene expression. However, recombinant APOE4 protein significantly impeded the "brain-to-blood" amyloid beta 1-40 (Aβ40) transport capabilities of BMEC-like cells, suggesting a role in diminished amyloid clearance. Conversely, APOE2 increased amyloid beta 1-42 (Aβ42) transport in the model. Furthermore, we demonstrated that APOE-mediated amyloid transport by BMEC-like cells is dependent on LRP1 and p-glycoprotein pathways, mirroring in vivo findings. Pericyte-like cells exhibited similar APOE secretion levels across genotypes, yet APOE4 pericyte-like cells showed heightened extracellular amyloid deposition, while APOE2 pericyte-like cells displayed the least amyloid deposition, an observation in line with vascular pathologies in AD patients. CONCLUSIONS While APOE genotype did not directly impact general BMEC or pericyte properties, APOE4 exacerbated amyloid clearance and deposition at the model BBB. Conversely, APOE2 demonstrated a potentially protective role by increasing amyloid transport and decreasing deposition. Our findings highlight that iPSC-derived BBB models can potentially capture amyloid pathologies at the BBB, motivating further development of such in vitro models in AD modeling and drug development.
Collapse
Affiliation(s)
- Yunfeng Ding
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
117
|
Rodriguez SMB, Tataranu LG, Kamel A, Turliuc S, Rizea RE, Dricu A. Glioblastoma and Immune Checkpoint Inhibitors: A Glance at Available Treatment Options and Future Directions. Int J Mol Sci 2024; 25:10765. [PMID: 39409094 PMCID: PMC11477435 DOI: 10.3390/ijms251910765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma is known to be one of the most aggressive and fatal human cancers, with a poor prognosis and resistance to standard treatments. In the last few years, many solid tumor treatments have been revolutionized with the help of immunotherapy. However, this type of treatment has failed to improve the results in glioblastoma patients. Effective immunotherapeutic strategies may be developed after understanding how glioblastoma achieves tumor-mediated immune suppression in both local and systemic landscapes. Biomarkers may help identify patients most likely to benefit from this type of treatment. In this review, we discuss the use of immunotherapy in glioblastoma, with an emphasis on immune checkpoint inhibitors and the factors that influence clinical response. A Pubmed data search was performed for all existing information regarding immune checkpoint inhibitors used for the treatment of glioblastoma. All data evaluating the ongoing clinical trials involving the use of ICIs either as monotherapy or in combination with other drugs was compiled and analyzed.
Collapse
Affiliation(s)
- Silvia Mara Baez Rodriguez
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Ligia Gabriela Tataranu
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Amira Kamel
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Serban Turliuc
- Medical Department, University of Medicine and Pharmacy “G. T. Popa”, 700115 Iasi, Romania;
| | - Radu Eugen Rizea
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Anica Dricu
- Biochemistry Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania;
| |
Collapse
|
118
|
González-Hernández S, Sato R, Sato Y, Liu C, Li W, Liu C, Jackson S, Kubota Y, Mukouyama YS. ENDOTHELIAL PROX1 INDUCES BLOOD-BRAIN BARRIER DISRUPTION IN THE CENTRAL NERVOUS SYSTEM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.03.616513. [PMID: 39803470 PMCID: PMC11722279 DOI: 10.1101/2024.10.03.616513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The central nervous system (CNS) parenchyma has conventionally been believed to lack lymphatic vasculature, likely due to a non-permissive microenvironment that hinders the formation and growth of lymphatic endothelial cells (LECs). Recent findings of ectopic expression of LEC markers including Prospero Homeobox 1 (PROX1), a master regulator of lymphatic differentiation, and the vascular permeability marker Plasmalemma Vesicle Associated Protein (PLVAP), in certain glioblastoma and brain arteriovenous malformations (AVMs), has prompted investigation into their roles in cerebrovascular malformations, tumor environments, and blood-brain barrier (BBB) abnormalities. To explore the relationship between ectopic LEC properties and BBB disruption, we utilized endothelial cell-specific Prox1 overexpression mutants. When induced during embryonic stages of BBB formation, endothelial Prox1 expression induces hybrid blood-lymphatic phenotypes in the developing CNS vasculature. This effect is not observed when Prox1 is overexpressed during postnatal BBB maturation. Ectopic Prox1 expression leads to significant vascular malformations and enhanced vascular leakage, resulting in BBB disruption when induced during both embryonic and postnatal stages. Mechanistically, PROX1 downregulates critical BBB-associated genes, including ß-catenin and Claudin-5, which are essential for BBB development and maintenance. These findings suggest that PROX1 compromises BBB integrity by negatively regulating BBB-associated gene expression and Wnt/ß-catenin signaling.
Collapse
Affiliation(s)
- Sara González-Hernández
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ryo Sato
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yuya Sato
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Present Address: Knowledge Palette, Inc. Kobe, Hyogo, Japan
| | - Chang Liu
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Present Address: Sarepta Therapeutics, Inc. Durham, NC, USA
| | - Wenling Li
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chengyu Liu
- Transgenic Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sadhana Jackson
- Developmental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Yoshiaki Kubota
- Department of Anatomy, Institute for Advanced Medical, Research and Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yoh-suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
119
|
Li J, Long S, Zhang Y, Wei W, Yu S, Liu Q, Hui X, Li X, Wang Y. Molecular mechanisms and diagnostic model of glioma-related epilepsy. NPJ Precis Oncol 2024; 8:223. [PMID: 39363097 PMCID: PMC11450052 DOI: 10.1038/s41698-024-00721-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/22/2024] [Indexed: 10/05/2024] Open
Abstract
Epilepsy is one of the most common symptoms in patients with gliomas; however, the mechanisms underlying its interaction are not yet clear. Moreover, epidemiological studies have not accurately identified patients with glioma-related epilepsy (GRE), and there is an urgent need to identify the molecular mechanisms and markers of its occurrence. We analyzed the demographics, transcriptome, whole-genome, and methylation sequences of 997 patients with glioma, to determine the genetic differences between glioma and GRE patients and to determine the upregulated molecular function, cellular composition, biological processes involved, signaling pathways, and immune cell infiltration. Twelve machine learning algorithms were refined into 113 combinatorial algorithms for building diagnostic recognition models. A total of 342 patients with GRE were identified with WHO grade 2 (174), grade 3 (107), and grade 4 (61). The mean age of the patients with GREs, with IDH mutations (n = 217 [63%]) and 1p19q non-codeletion (n = 169 [49%]), was 38 years old. GRE molecular functions were mainly passive transmembrane transporter protein activity, ion channel activity, and gated channel activity. Cellular components were enriched in the cation-channel and transmembrane transporter complexes. Cerebral cortical development regulates the membrane potential and synaptic organization as major biological processes. The signaling pathways mainly focused on cholinergic, GABAergic, and glutamatergic synapses. LASSO, combined with Random Forest, was the best diagnostic model and identified nine diagnostic genes. This study provides new insights and future perspectives for resolving the molecular mechanisms of GRE.
Collapse
Affiliation(s)
- Jinwei Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shengrong Long
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yang Zhang
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wei Wei
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shuangqi Yu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Quan Liu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
| | - Xuhui Hui
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
120
|
Yonk MG, Lim MA, Thompson CM, Tora MS, Lakhina Y, Du Y, Hoang KB, Molinaro AM, Boulis NM, Hassaneen W, Lei K. Improving glioma drug delivery: A multifaceted approach for glioma drug development. Pharmacol Res 2024; 208:107390. [PMID: 39233056 PMCID: PMC11440560 DOI: 10.1016/j.phrs.2024.107390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/16/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Glioma is one of the most common central nervous system (CNS) cancers that can be found within the brain and the spinal cord. One of the pressing issues plaguing the development of therapeutics for glioma originates from the selective and semipermeable CNS membranes: the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB). It is difficult to bypass these membranes and target the desired cancerous tissue because the purpose of the BBB and BSCB is to filter toxins and foreign material from invading CNS spaces. There are currently four varieties of Food and Drug Administration (FDA)-approved drug treatment for glioma; yet these therapies have limitations including, but not limited to, relatively low transmission through the BBB/BSCB, despite pharmacokinetic characteristics that allow them to cross the barriers. Steps must be taken to improve the development of novel and repurposed glioma treatments through the consideration of pharmacological profiles and innovative drug delivery techniques. This review addresses current FDA-approved glioma treatments' gaps, shortcomings, and challenges. We then outline how incorporating computational BBB/BSCB models and innovative drug delivery mechanisms will help motivate clinical advancements in glioma drug delivery. Ultimately, considering these attributes will improve the process of novel and repurposed drug development in glioma and the efficacy of glioma treatment.
Collapse
Affiliation(s)
- Marybeth G Yonk
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA; College of Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Megan A Lim
- Carle Illinois College of Medicine, University of Illinois Urbana Champaign, Champaign, IL, USA; Department of Neurosurgery, Carle Foundation Hospital, Urbana, IL, USA
| | - Charee M Thompson
- Carle Illinois College of Medicine, University of Illinois Urbana Champaign, Champaign, IL, USA; College of Liberal Arts & Sciences, University of Illinois Urbana Champaign, Champaign, IL, USA
| | - Muhibullah S Tora
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yuliya Lakhina
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Kimberly B Hoang
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Nicholas M Boulis
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Wael Hassaneen
- Carle Illinois College of Medicine, University of Illinois Urbana Champaign, Champaign, IL, USA; Department of Neurosurgery, Carle Foundation Hospital, Urbana, IL, USA.
| | - Kecheng Lei
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
121
|
Yan RE, Greenfield JP. Challenges and Outlooks in Precision Medicine: Expectations Versus Reality. World Neurosurg 2024; 190:573-581. [PMID: 39425299 DOI: 10.1016/j.wneu.2024.06.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 10/21/2024]
Abstract
Recent developments in technology have led to rapid advances in precision medicine, especially due to the rise of next-generation sequencing and molecular profiling. These technological advances have led to rapid advances in research, including increased tumor subtype resolution, new therapeutic agents, and mechanistic insights. Certain therapies have even been approved for molecular biomarkers across histopathological diagnoses; however, translation of research findings to the clinic still faces a number of challenges. In this review, the authors discuss several key challenges to the clinical integration of precision medicine, including the blood-brain barrier, both a lack and excess of molecular targets, and tumor heterogeneity/escape from therapy. They also highlight a few key efforts to address these challenges, including new frontiers in drug delivery, a rapidly expanding treatment repertoire, and improvements in active response monitoring. With continued improvements and developments, the authors anticipate that precision medicine will increasingly become the gold standard for clinical care.
Collapse
Affiliation(s)
- Rachel E Yan
- Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Jeffrey P Greenfield
- Department of Neurological Surgery, NewYork-Presbyterian Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
122
|
Li N, Hao S, Cao X, Lin Y, Li Y, Dai T, Liu M. Significance of radiation therapy in frontal glioblastoma patients and exploration of optimal treatment modality: a real-world multiple-center study based on propensity score matching. Quant Imaging Med Surg 2024; 14:7576-7586. [PMID: 39429582 PMCID: PMC11485375 DOI: 10.21037/qims-23-1871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 08/15/2024] [Indexed: 10/22/2024]
Abstract
Background Glioblastoma (GBM) exhibits diffuse and invasive growth patterns, with a 5-year overall survival (OS) rate of 5-10%. In addition, approximately 40 percent of GBMs are localized in the frontal lobe, a region closely linked to essential life functions including cognition, so that it cannot be completely eradicated through surgical intervention, leading to very poor prognosis. Postoperative therapy is an essential treatment modality. The aim of this study is to explain the possible role of radiation therapy (RT) in the treatment of frontal GBM, providing more evidence for clinical application. Methods In the study, patient information pertaining to frontal GBM patients was collected from the Surveillance, Epidemiology, and End Results (SEER) database for the period 2000 to 2018 with 9,904 patients deemed appropriate for inclusion in this study. A 1:2 propensity score matching analysis was conducted to balance the non-radiotherapy and radiotherapy group. This study is a retrospective study. Results Before matching, the median OS, tumor specific survival (TSS) and hazard ratio (HR) were 3 months, 3 months and 4.408 [95% confidence interval (CI): 3.762-4.535, P<0.001] in the non-RT group compared to those of 13 months, 14 months and 2.463 (95% CI: 2.247-2.936, P<0.001) in the RT group. After matching, the median OS, TSS and HR were 3 months, 4 months and 1.433 (95% CI: 1.387-1.692, P<0.001) in the non-RT group compared to those of 8 months, 8 months and 1.427 (95% CI: 1.374-1.682, P<0.001) in the RT group. Conclusions Radiotherapy is an important local therapy, which can significantly improve the tumor-specific survival and OS of frontal GBM patients. With the arrival of the era of precision radiotherapy, the continuous progress of radiotherapy technology may bring more benefits to frontal GBM patients.
Collapse
Affiliation(s)
- Nan Li
- Department of Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shuai Hao
- Department of Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Radiotherapy, Aerospace Center Hospital, Beijing, China
| | - Xiaohui Cao
- Department of Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yufeng Lin
- Department of Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yunfang Li
- Department of Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tong Dai
- Department of Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Graduate School of Hebei Medical University, Shijiazhuang, China
| | - Ming Liu
- Department of Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Graduate School of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
123
|
Euclydes V, Braga CI, Gouveia G, Martinez RC, Camilo C, Simões SN, Martins-Jr DC, Fracolli L, Argeu A, Ferraro A, Matijasevich A, Fatori D, Miguel EC, Polanczyk GV, Brentani H. Maternal immune response during pregnancy and neurodevelopmental outcomes: A longitudinal approach. Brain Behav Immun Health 2024; 40:100832. [PMID: 39193418 PMCID: PMC11347843 DOI: 10.1016/j.bbih.2024.100832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 08/29/2024] Open
Abstract
Background and objectives The neurodevelopment of the offspring is suggested to be influenced by the maternal immune system's responses throughout pregnancy, which in turn is also vulnerable to maternal psychosocial stress conditions. Therefore, our main goal was to investigate whether maternal peripheral immunological biomarkers (IB) during two stages of gestation are associated with distinct neurodevelopmental trajectories in the first two years of life. As a second goal, we also explored the association between maternal distal (childhood) and proximal (gestation) stressful experiences and the immunological markers assessed during pregnancy. Methods Maternal childhood trauma, depressive and anxiety symptoms, and peripheral IB (IFNγ, IL-10, IL1β, IL6, IL8, TNFα, EGF, IL13, IL17, IL1Ra and IL4) were measured at baseline (8-16 weeks of pregnancy) and at 30 weeks of pregnancy in 160 women. The participants had the blood samples collected from two randomized clinical trials conducted by the same team and methods in the same community. A Principal Component Analysis (PCA) was implemented to create meaningful composite variables that describe the cytokines joint variation. Finally, linear mixed-effects modeling was used to investigate the influence of inflammatory biomarkers, maternal childhood trauma, anxiety, and depressive symptoms on Bayley's III scores trajectories. Results The IB profile during the 3rd trimester of pregnancy predicted the offspring's neurodevelopmental trajectories in the first two years of life. The components derived from PCA were important predictors and captured different immune responses, reflecting both pro- and anti-inflammatory states. Maternal stressful experiences did not correlate with the immunological markers. Although not a reliable predictor alone, maternal psychosocial stress at the 1st trimester of pregnancy interacted with the mother's immune response while predicting the neurodevelopmental scores during the first two years of life. Conclusions Our results underscore the importance of the maternal immune response during pregnancy in shaping the neurodevelopmental trajectory of the offspring. Additionally, we observed that the maternal distress at the early stages of pregnancy has an incremental effect on the neurodevelopmental outcome but depends upon the immune response.
Collapse
Affiliation(s)
- Veronica Euclydes
- Instituto e Departamento de Psiquiatria, Faculdade de Medicina FMUSP, LIM/23, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Caio I.S. Braga
- Center for Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Santo André, SP, Brazil
| | - Gisele Gouveia
- Instituto e Departamento de Psiquiatria, Faculdade de Medicina FMUSP, LIM/23, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Caroline Camilo
- Instituto e Departamento de Psiquiatria, Faculdade de Medicina FMUSP, LIM/23, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | | | - David C. Martins-Jr
- Center for Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Santo André, SP, Brazil
| | - Lislaine Fracolli
- Escola de Enfermagem, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Adriana Argeu
- Instituto e Departamento de Psiquiatria, Faculdade de Medicina FMUSP, LIM/23, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Alexandre Ferraro
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Alicia Matijasevich
- Departamento de Medicina Preventiva, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Daniel Fatori
- Instituto e Departamento de Psiquiatria, Faculdade de Medicina FMUSP, LIM/23, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Euripedes C. Miguel
- Instituto e Departamento de Psiquiatria, Faculdade de Medicina FMUSP, LIM/23, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Guilherme V. Polanczyk
- Instituto e Departamento de Psiquiatria, Faculdade de Medicina FMUSP, LIM/23, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Helena Brentani
- Instituto e Departamento de Psiquiatria, Faculdade de Medicina FMUSP, LIM/23, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
124
|
Simões JLB, de Carvalho Braga G, Eichler SW, da Silva GB, Bagatini MD. Implications of COVID-19 in Parkinson's disease: the purinergic system in a therapeutic-target perspective to diminish neurodegeneration. Purinergic Signal 2024; 20:487-507. [PMID: 38460075 PMCID: PMC11377384 DOI: 10.1007/s11302-024-09998-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 02/21/2024] [Indexed: 03/11/2024] Open
Abstract
The pathophysiology of Parkinson's disease (PD) is marked by degeneration of dopaminergic neurons in the substantia nigra. With advent of COVID-19, which is closely associated with generalized inflammation and multiple organ dysfunctions, the PD patients may develop severe conditions of disease leading to exacerbated degeneration. This condition is caused by the excessive release of pro-inflammatory markers, called cytokine storm, that is capable of triggering neurodegenerative conditions by affecting the blood-brain barrier (BBB). A possible SARS-CoV-2 infection, in serious cases, may compromise the immune system by triggering a hyperstimulation of the neuroimmune response, similar to the pathological processes found in PD. From this perspective, the inflammatory scenario triggers oxidative stress and, consequently, cellular dysfunction in the nervous tissue. The P2X7R seems to be the key mediator of the neuroinflammatory process, as it acts by increasing the concentration of ATP, allowing the influx of Ca2+ and the occurrence of mutations in the α-synuclein protein, causing activation of this receptor. Thus, modulation of the purinergic system may have therapeutic potential on the effects of PD, as well as on the damage caused by inflammation of the BBB, which may be able to mitigate the neurodegeneration caused by diseases. Considering all the processes of neuroinflammation, oxidative stress, and mitochondrial dysfunction that PD propose, we can conclude that the P2X7 antagonist acts in the prevention of viral diseases, and it also controls purinergic receptors formed by multi-target compounds directed to self-amplification circuits and, therefore, may be a viable strategy to obtain the desired disease-modifying effect. Thus, purinergic system receptor modulations have a high therapeutic potential for neurodegenerative diseases such as PD.
Collapse
Affiliation(s)
| | | | | | - Gilnei Bruno da Silva
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Margarete Dulce Bagatini
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil.
| |
Collapse
|
125
|
Thomson EL, Powell E, Gandini Wheeler-Kingshott CAM, Parker GJM. Quantification of water exchange across the blood-brain barrier using noncontrast MR fingerprinting. Magn Reson Med 2024; 92:1392-1403. [PMID: 38725240 DOI: 10.1002/mrm.30127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 07/23/2024]
Abstract
PURPOSE A method is proposed to quantify cerebral blood volume (v b $$ {v}_b $$ ) and intravascular water residence time (τ b $$ {\tau}_b $$ ) using MR fingerprinting (MRF), applied using a spoiled gradient echo sequence without the need for contrast agent. METHODS An in silico study optimized an acquisition protocol to maximize the sensitivity of the measurement tov b $$ {v}_b $$ andτ b $$ {\tau}_b $$ changes. Its accuracy in the presence of variations inT 1 , t $$ {\mathrm{T}}_{1,t} $$ ,T 1 , b $$ {\mathrm{T}}_{1,b} $$ , andB 1 $$ {\mathrm{B}}_1 $$ was evaluated. The optimized protocol (scan time of 19 min) was then tested in a exploratory healthy volunteer study (10 volunteers, mean age 24± $$ \pm $$ 3, six males) at 3 T with a repeat scan taken after repositioning to allow estimation of repeatability. RESULTS Simulations show that assuming literature values forT 1 , b $$ {\mathrm{T}}_{1,b} $$ andT 1 , t $$ {\mathrm{T}}_{1,t} $$ , no variation inB 1 $$ {\mathrm{B}}_1 $$ , while fitting onlyv b $$ {v}_b $$ andτ b $$ {\tau}_b $$ , leads to large errors in quantification ofv b $$ {v}_b $$ andτ b $$ {\tau}_b $$ , regardless of noise levels. However, simulations also show that matchingT 1 , t $$ {\mathrm{T}}_{1,t} $$ ,T 1 , b $$ {\mathrm{T}}_{1,b} $$ ,B 1 + $$ {\mathrm{B}}_1^{+} $$ ,v b $$ {v}_b $$ andτ b $$ {\tau}_b $$ , simultaneously is feasible at clinically achievable noise levels. Across the healthy volunteers, all parameter quantifications fell within the expected literature range. In addition, the maps show good agreement between hemispheres suggesting physiologically relevant information is being extracted. Expected differences between white and gray matterT 1 , t $$ {\mathrm{T}}_{1,t} $$ (p < 0.0001) andv b $$ {v}_b $$ (p < 0.0001) are observed,T 1 , b $$ {\mathrm{T}}_{1,b} $$ andτ b $$ {\tau}_b $$ show no significant differences, p = 0.4 and p = 0.6, respectively. Moderate to excellent repeatability was seen between repeat scans: mean intra-class correlation coefficient ofT 1 , t : 0 . 91 $$ {\mathrm{T}}_{1,t}:0.91 $$ ,T 1 , b : 0 . 58 $$ {\mathrm{T}}_{1,b}:0.58 $$ ,v b : 0 . 90 $$ {v}_b:0.90 $$ , andτ b : 0 . 96 $$ {\tau}_b:0.96 $$ . CONCLUSION We demonstrate that regional simultaneous quantification ofv b $$ {v}_b $$ ,τ b $$ {\tau}_b $$ ,T 1 , b , T 1 , t $$ {\mathrm{T}}_{1,b},{T}_{1,t} $$ , andB 1 + $$ {\mathrm{B}}_1^{+} $$ using MRF is feasible in vivo.
Collapse
Affiliation(s)
- Emma L Thomson
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, London, UK
| | - Elizabeth Powell
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Claudia A M Gandini Wheeler-Kingshott
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, London, UK
- Department of Brain & Behavioural Sciences, University of Pavia, Pavia, Italy
- IRCCS Mondino Foundation, Pavia, Italy
| | - Geoff J M Parker
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, London, UK
- Bioxydyn Limited, Manchester, UK
| |
Collapse
|
126
|
Kim TY, Choi JW, Park K, Kim S, Kim JF, Park TE, Seo J. Lubricant-Coated Organ-on-a-Chip for Enhanced Precision in Preclinical Drug Testing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402431. [PMID: 38934549 DOI: 10.1002/smll.202402431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/31/2024] [Indexed: 06/28/2024]
Abstract
In drug discovery, human organ-on-a-chip (organ chip) technology has emerged as an essential tool for preclinical testing, offering a realistic representation of human physiology, real-time monitoring, and disease modeling. Polydimethylsiloxane (PDMS) is commonly used in organ chip fabrication owing to its biocompatibility, flexibility, transparency, and ability to replicate features down to the nanoscale. However, the porous nature of PDMS leads to unintended absorption of small molecules, critically affecting the drug response analysis. Addressing this challenge, the precision drug testing organ chip (PreD chip) is introduced, an innovative platform engineered to minimize small molecule absorption while facilitating cell culture. This chip features a PDMS microchannel wall coated with a perfluoropolyether-based lubricant, providing slipperiness and antifouling properties. It also incorporates an ECM-coated semi-porous membrane that supports robust multicellular cultures. The PreD chip demonstrates its outstanding antifouling properties and resistance to various biological fluids, small molecule drugs, and plasma proteins. In simulating the human gut barrier, the PreD chip demonstrates highly enhanced sensitivity in tests for dexamethasone toxicity and is highly effective in assessing drug transport across the human blood-brain barrier. These findings emphasize the potential of the PreD chip in advancing organ chip-based drug testing methodologies.
Collapse
Affiliation(s)
- Tae Young Kim
- School of Electrical and Electronic Engineering, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeong-Won Choi
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Kijun Park
- School of Electrical and Electronic Engineering, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - SeungHwan Kim
- Department of Energy and Chemical Engineering, Incheon National University (INU), Incheon, 22012, South Korea
| | - Jeong F Kim
- Department of Energy and Chemical Engineering, Incheon National University (INU), Incheon, 22012, South Korea
- Innovation Center for Chemical Engineering, Incheon National University (INU), Incheon, 22012, South Korea
| | - Tae-Eun Park
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jungmok Seo
- School of Electrical and Electronic Engineering, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Lynk Solutec lnc, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
127
|
Mineiro R, Rodrigues Cardoso M, Catarina Duarte A, Santos C, Cipolla-Neto J, Gaspar do Amaral F, Costa D, Quintela T. Melatonin and brain barriers: The protection conferred by melatonin to the blood-brain barrier and blood-cerebrospinal fluid barrier. Front Neuroendocrinol 2024; 75:101158. [PMID: 39395545 DOI: 10.1016/j.yfrne.2024.101158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/29/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
The blood-brain barrier and the blood-cerebrospinal fluid barrier separate the blood from brain tissue and cerebrospinal fluid. These brain barriers are important to maintain homeostasis and complex functions by protecting the brain from xenobiotics and harmful endogenous compounds. The disruption of brain barriers is a characteristic of neurologic diseases. Melatonin is a lipophilic hormone that is mainly produced by the pineal gland. The blood-brain barrier and the blood-cerebrospinal fluid barriers are melatonin-binding sites. Among the several melatonin actions, the most characteristic one is the regulation of sleep-wake cycles, melatonin has anti-inflammatory and antioxidant properties. Since brain barriers disruption can arise from inflammation and oxidative stress, knowing the influence of melatonin on the integrity of brain barriers is extremely important. Therefore, the objective of this review is to gather and discuss the available literature about the regulation of brain barriers by melatonin.
Collapse
Affiliation(s)
- Rafael Mineiro
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Maria Rodrigues Cardoso
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana Catarina Duarte
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Cecília Santos
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Jose Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Diana Costa
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; Instituto Politécnico da Guarda, 6300-559 Guarda, Portugal.
| |
Collapse
|
128
|
Sahu B, Nookala S, Floden AM, Ambhore NS, Sathish V, Klug MG, Combs CK. House dust mite-induced asthma exacerbates Alzheimer's disease changes in the brain of the App NL-G-F mouse model of disease. Brain Behav Immun 2024; 121:365-383. [PMID: 39084541 PMCID: PMC11442016 DOI: 10.1016/j.bbi.2024.07.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 07/19/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterized by the accumulation of amyloid-β (Aβ) plaques, neuroinflammation, and neuronal death. Besides aging, various comorbidities increase the risk of AD, including obesity, diabetes, and allergic asthma. Epidemiological studies have reported a 2.17-fold higher risk of dementia in asthmatic patients. However, the molecular mechanism(s) underlying this asthma-associated AD exacerbation is unknown. This study was designed to explore house dust mite (HDM)-induced asthma effects on AD-related brain changes using the AppNL-G-F transgenic mouse model of disease. Male and female 8-9 months old C57BL/6J wild type and AppNL-G-F mice were exposed to no treatment, saline sham, or HDM extract every alternate day for 16 weeks for comparison across genotypes and treatment. Mice were euthanized at the end of the experiment, and broncho-alveolar lavage fluid (BALF), blood, lungs, and brains were collected. BALF was used to quantify immune cell phenotype, cytokine levels, total protein content, lactate dehydrogenase (LDH) activity, and total IgE. Lungs were sectioned and stained with hematoxylin and eosin, Alcian blue, and Masson's trichrome. Serum levels of cytokines and soluble Aβ1-40/42 were quantified. Brains were sectioned and immunostained for Aβ, GFAP, CD68, and collagen IV. Finally, frozen hippocampi and temporal cortices were used to perform Aβ ELISAs and cytokine arrays, respectively. HDM exposure led to increased levels of inflammatory cells, cytokines, total protein content, LDH activity, and total IgE in the BALF, as well as increased pulmonary mucus and collagen staining in both sexes and genotypes. Levels of serum cytokines increased in all HDM-exposed groups. Serum from the AppNL-G-F HDM-induced asthma group also had significantly increased soluble Aβ1-42 levels in both sexes. In agreement with this peripheral change, hippocampi from asthma-induced male and female AppNL-G-F mice demonstrated elevated Aβ plaque load and increased soluble Aβ 1-40/42 and insoluble Aβ 1-40 levels. HDM exposure also increased astrogliosis and microgliosis in both sexes of AppNL-G-F mice, as indicated by GFAP and CD68 immunoreactivity, respectively. Additionally, HDM exposure elevated cortical levels of several cytokines in both sexes and genotypes. Finally, HDM-exposed groups also showed a disturbed blood-brain-barrier (BBB) integrity in the hippocampus of AppNL-G-F mice, as indicated by decreased collagen IV immunoreactivity. HDM exposure was responsible for an asthma-like condition in the lungs that exacerbated Aβ pathology, astrogliosis, microgliosis, and cytokine changes in the brains of male and female AppNL-G-F mice that correlated with reduced BBB integrity. Defining mechanisms of asthma effects on the brain may identify novel therapeutic targets for asthma and AD.
Collapse
Affiliation(s)
- Bijayani Sahu
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA
| | - Suba Nookala
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA
| | - Angela M Floden
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA
| | - Nilesh S Ambhore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Marilyn G Klug
- Department of Population health, School of Medicine and Health Sciences, USA
| | - Colin K Combs
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA.
| |
Collapse
|
129
|
Samreen S, Khan E, Ahmad IZ. Molecular docking and molecular dynamics simulation analysis of bioactive compounds of Cichorium intybus L. seed against hepatocellular carcinoma. J Biomol Struct Dyn 2024; 42:9133-9144. [PMID: 37621217 DOI: 10.1080/07391102.2023.2250465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 08/12/2023] [Indexed: 08/26/2023]
Abstract
In this article, bioactive compounds present in Cichorium intybus L. seeds were collected from literature review and analyzed for probable remedy for hepatocellular carcinoma. Cichorium intybus L. is a traditional plant used all over the world mainly in hepatic disorders and renal diseases. This therapeutic plant has many bioactive compounds like chicoric acid, chlorogenic acid, sesquiterpne lactones, stigmasterols etc are found in seeds. Here, the target protein p53 (PDB ID: 2OCJ) which is involved in many cancerous pathways, is chosen. The preADMET study filtered out some compounds which were then subjected to molecular docking studies by Autodock tool 4.2. Afterwards, two best compounds (Esculetin and Isochlorogenic acid) were screened out on the basis of binding energy as compared to the standard compound (Doxorubicin). All these complexes were then analyzed for stability by molecular dynamics using online GROMACS tool. In the comparative simulation study, the compound Esculetin shows a stable interaction with the p53 over the 100 ns trajectory. Hepatocellular carcinoma accounts for high mortality of cancer related death worldwide. These findings suggest that these compound can be used to treat the hepatocellular carcinoma.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sadiyah Samreen
- Natural Products Laboratory, Department of Bioengineering, Integral University, Lucknow, India
| | - Elhan Khan
- Natural Products Laboratory, Department of Bioengineering, Integral University, Lucknow, India
| | - Iffat Zareen Ahmad
- Natural Products Laboratory, Department of Bioengineering, Integral University, Lucknow, India
| |
Collapse
|
130
|
Aziz N, Wal P, Patel A, Prajapati H. A comprehensive review on the pharmacological role of gut microbiome in neurodegenerative disorders: potential therapeutic targets. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7307-7336. [PMID: 38734839 DOI: 10.1007/s00210-024-03109-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Neurological disorders, including Alzheimer and Parkinson's, pose significant challenges to public health due to their complex etiologies and limited treatment options. Recent advances in research have highlighted the intricate bidirectional communication between the gut microbiome and the central nervous system (CNS), revealing a potential therapeutic avenue for neurological disorders. Thus, this review aims to summarize the current understanding of the pharmacological role of gut microbiome in neurological disorders. Mounting evidence suggests that the gut microbiome plays a crucial role in modulating CNS function through various mechanisms, including the production of neurotransmitters, neuroactive metabolites, and immune system modulation. Dysbiosis, characterized by alterations in gut microbial composition and function, has been observed in many neurological disorders, indicating a potential causative or contributory role. Pharmacological interventions targeting the gut microbiome have emerged as promising therapeutic strategies for neurological disorders. Probiotics, prebiotics, antibiotics, and microbial metabolite-based interventions have shown beneficial effects in animal models and some human studies. These interventions aim to restore microbial homeostasis, enhance microbial diversity, and promote the production of beneficial metabolites. However, several challenges remain, including the need for standardized protocols, identification of specific microbial signatures associated with different neurological disorders, and understanding the precise mechanisms underlying gut-brain communication. Further research is necessary to unravel the intricate interactions between the gut microbiome and the CNS and to develop targeted pharmacological interventions for neurological disorders.
Collapse
Affiliation(s)
- Namra Aziz
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India
| | - Pranay Wal
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India.
| | - Aman Patel
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India
| | - Harshit Prajapati
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India
| |
Collapse
|
131
|
Deng Y, Qiao Z, Zhou C, Pei Y, Xu H, Kang X, Luo J. Endothelial Myosin IIA Is Required for the Maintenance of Blood-Brain Barrier Integrity. Cells 2024; 13:1635. [PMID: 39404399 PMCID: PMC11475711 DOI: 10.3390/cells13191635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024] Open
Abstract
Brain endothelial cells (ECs) are essential elements of the blood-brain barrier (BBB), maintaining its integrity through both paracellular junctions and transcellular transport systems. Myosin IIA, a multifunctional protein, plays a significant role in various cellular processes, including cytoskeletal maintenance, cell division, and signal transduction. While Myosin IIA has been implicated in bleeding and ischemic stroke, its role in regulating BBB integrity under physiological conditions remains unclear. In this study, we investigated the impact of Myosin IIA deficiency on BBB integrity using intravenous tracer injections and models of epilepsy. Flow cytometry, Western blot, and real-time PCR were employed to isolate brain cells and assess changes in protein and mRNA levels. Additionally, immunofluorescence staining and electron microscopy were used to explore alterations in protein expression and the structure of BBB. Our results demonstrate that endothelial Myosin IIA deficiency increased BBB permeability and exacerbated symptoms in BBB-related diseases. Mechanistically, we found that Myosin IIA modulates β-catenin transcription and protein interactions. The overexpression of β-catenin in brain endothelial Myosin IIA deficiency mice improved BBB integrity and reduced disease severity. This study establishes Myosin IIA as a critical regulator of BBB integrity and suggests new therapeutic targets for vascular diseases.
Collapse
Affiliation(s)
- Yanan Deng
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, School of Future Technology, Peking University, Beijing 100871, China
| | - Ziqi Qiao
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, School of Future Technology, Peking University, Beijing 100871, China
| | - Changping Zhou
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, School of Future Technology, Peking University, Beijing 100871, China
| | - Yujun Pei
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, School of Future Technology, Peking University, Beijing 100871, China
| | - Han Xu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, School of Future Technology, Peking University, Beijing 100871, China
| | - Xuya Kang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, School of Future Technology, Peking University, Beijing 100871, China
| | - Jincai Luo
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, School of Future Technology, Peking University, Beijing 100871, China
- College of Future Technology, Peking University, Beijing 100871, China
| |
Collapse
|
132
|
Furman S, Green K, Lane TE. COVID-19 and the impact on Alzheimer's disease pathology. J Neurochem 2024; 168:3415-3429. [PMID: 37850241 PMCID: PMC11024062 DOI: 10.1111/jnc.15985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has rapidly escalated into a global pandemic that primarily affects older and immunocompromised individuals due to underlying clinical conditions and suppressed immune responses. Furthermore, COVID-19 patients exhibit a spectrum of neurological symptoms, indicating that COVID-19 can affect the brain in a variety of manners. Many studies, past and recent, suggest a connection between viral infections and an increased risk of neurodegeneration, raising concerns about the neurological effects of COVID-19 and the possibility that it may contribute to Alzheimer's disease (AD) onset or worsen already existing AD pathology through inflammatory processes given that both COVID-19 and AD share pathological features and risk factors. This leads us to question whether COVID-19 is a risk factor for AD and how these two conditions might influence each other. Considering the extensive reach of the COVID-19 pandemic and the devastating impact of the ongoing AD pandemic, their combined effects could have significant public health consequences worldwide.
Collapse
Affiliation(s)
- Susana Furman
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
| | - Kim Green
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
| | - Thomas E. Lane
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
- Department of Molecular Biology & Biochemistry, School of Biological Sciences, University of California, Irvine 92697, USA
- Center for Virus Research, University of California, Irvine 92697, USA
| |
Collapse
|
133
|
Hameed H, Sarwar HS, Younas K, Zaman M, Jamshaid M, Irfan A, Khalid M, Sohail MF. Exploring the potential of nanomedicine for gene therapy across the physicochemical and cellular barriers. Funct Integr Genomics 2024; 24:177. [PMID: 39340586 DOI: 10.1007/s10142-024-01459-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024]
Abstract
After COVID-19, a turning point in the way of pharmaceutical technology is gene therapy with beneficial potential to start a new medical era. However, commercialization of such pharmaceuticals would never be possible without the help of nanotechnology. Nanomedicine can fulfill the growing needs linked to safety, efficiency, and site-specific targeted delivery of Gene therapy-based pharmaceuticals. This review's goal is to investigate how nanomedicine may be used to transfer nucleic acids by getting beyond cellular and physicochemical barriers. Firstly, we provide a full description of types of gene therapy, their mechanism, translation, transcription, expression, type, and details of diseases with possible mechanisms that can only be treated with genes-based pharmaceuticals. Additionally, we also reviewed different types of physicochemical barriers, physiological and cellular barriers in nucleic acids (DNA/RNA) based drug delivery. Finally, we highlight the need and importance of cationic lipid-based nanomedicine/nanocarriers in gene-linked drug delivery and how nanotechnology can help to overcome the above-discussed barrier in gene therapy and their biomedical applications.
Collapse
Affiliation(s)
- Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Hafiz Shoaib Sarwar
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Komel Younas
- Faculty of Pharmacy, University Paris Saclay, 17 Avenue des sciences, 91190, Orsay, France
| | - Muhammad Zaman
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Muhammad Jamshaid
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Ali Irfan
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Maha Khalid
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Muhammad Farhan Sohail
- Department of Chemistry, SBASSE, Lahore University of Management Sciences (LUMS), Lahore, 54000, Pakistan
- Alliant College of Pharmacy and Allied Health Sciences, Lahore, 54000, Pakistan
| |
Collapse
|
134
|
Uddin N, Scott J, Nixon J, Patterson SD, Kidgell D, Pearce AJ, Waldron M, Tallent J. The effects of exercise, heat-induced hypo-hydration and rehydration on blood-brain-barrier permeability, corticospinal and peripheral excitability. Eur J Appl Physiol 2024:10.1007/s00421-024-05616-x. [PMID: 39340668 DOI: 10.1007/s00421-024-05616-x] [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: 04/12/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024]
Abstract
PURPOSE The effects of low-intensity exercise, heat-induced hypo-hydration and rehydration on maximal strength and the underlying neurophysiological mechanisms are not well understood. METHODS To assess this, 12 participants took part in a randomised crossover study, in a prolonged (3 h) submaximal (60 W) cycling protocol under 3 conditions: (i) in 45 °C (achieving ~ 5% body mass reduction), with post-exercise rehydration in 2 h (RHY2), (ii) with rehydration across 24 h (RHY24), and (iii) a euhydrated trial in 25 °C (CON). Dependent variables included maximal voluntary contractions (MVC), maximum motor unit potential (MMAX), motor evoked potential (MEPRAW) amplitude and cortical silent period (cSP) duration. Blood-brain-barrier integrity was also assessed by serum Ubiquitin Carboxyl-terminal Hydrolase (UCH-L1) concentrations. All measures were obtained immediately pre, post, post 2 h and 24 h. RESULTS During both dehydration trials, MVC (RHY2: p < 0.001, RHY24: p = 0.001) and MEPRAW (RHY2: p = 0.025, RHY24: p = 0.045) decreased from pre- to post-exercise. MEPRAW returned to baseline during RHY2 and CON, but not RHY24 (p = 0.020). MEP/MMAX ratio decreased across time for all trials (p = 0.009) and returned to baseline, except RHY24 (p < 0.026). Increased cSP (p = 0.011) was observed during CON post-exercise, but not during RHY2 and RHY24. Serum UCH-L1 increased across time for all conditions (p < 0.001) but was not significantly different between conditions. CONCLUSION Our findings demonstrate an increase in corticospinal inhibition after exercise with fluid ingestion, but a decrease in corticospinal excitability after heat-induced hypo-hydration. In addition, low-intensity exercise increases peripheral markers of blood-brain-barrier permeability.
Collapse
Affiliation(s)
- Nasir Uddin
- Faculty of Sport, Technology and Health Sciences, St Mary's University, Twickenham, UK.
- School of Sport, Rehabilitation, and Exercise Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK.
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| | - Jamie Scott
- Faculty of Sport, Technology and Health Sciences, St Mary's University, Twickenham, UK
| | - Jonathan Nixon
- Faculty of Sport, Technology and Health Sciences, St Mary's University, Twickenham, UK
| | - Stephen D Patterson
- Faculty of Sport, Technology and Health Sciences, St Mary's University, Twickenham, UK
| | - Dawson Kidgell
- Monash Exercise Neuroplasticity Research Unit, Department of Physiotherapy, School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Australia
| | - Alan J Pearce
- Swinburne Neuroimaging Facility, School of Health Science, Swinburne University of Technology, Melbourne, Australia
| | - Mark Waldron
- Applied Sport, Technology, Exercise and Medicine, College of Engineering, Swansea University, Swansea, Wales, UK
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Sippy Downs, QLD, Australia
- Welsh Institute of Performance Science, Swansea University, Swansea, UK
| | - Jamie Tallent
- School of Sport, Rehabilitation, and Exercise Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
- Monash Exercise Neuroplasticity Research Unit, Department of Physiotherapy, School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Australia
| |
Collapse
|
135
|
Cater RJ. Food for thought: The molecular basis of nutrient uptake into the brain. Science 2024; 385:1429. [PMID: 39325899 DOI: 10.1126/science.ads1320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
The molecular basis of nutrient uptake into the brain.
Collapse
Affiliation(s)
- Rosemary J Cater
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
136
|
Fu Y, Gu Z, Cao H, Zuo C, Huang Y, Song Y, Jiang Y, Wang F. The role of the gut microbiota in neurodegenerative diseases targeting metabolism. Front Neurosci 2024; 18:1432659. [PMID: 39391755 PMCID: PMC11464490 DOI: 10.3389/fnins.2024.1432659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024] Open
Abstract
In recent years, the incidence of neurodegenerative diseases (NDs) has gradually increased over the past decades due to the rapid aging of the global population. Traditional research has had difficulty explaining the relationship between its etiology and unhealthy lifestyle and diets. Emerging evidence had proved that the pathogenesis of neurodegenerative diseases may be related to changes of the gut microbiota's composition. Metabolism of gut microbiota has insidious and far-reaching effects on neurodegenerative diseases and provides new directions for disease intervention. Here, we delineated the basic relationship between gut microbiota and neurodegenerative diseases, highlighting the metabolism of gut microbiota in neurodegenerative diseases and also focusing on treatments for NDs based on gut microbiota. Our review may provide novel insights for neurodegeneration and approach a broadly applicable basis for the clinical therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Yufeng Fu
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhongya Gu
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huan Cao
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chengchao Zuo
- Department of Rehabilitation, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yaqi Huang
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Song
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yongsheng Jiang
- Cancer Center of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Furong Wang
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging (HUST), Ministry of Education, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
137
|
Zhao Y, Huang Y, Cao Y, Yang J. Astrocyte-Mediated Neuroinflammation in Neurological Conditions. Biomolecules 2024; 14:1204. [PMID: 39456137 PMCID: PMC11505625 DOI: 10.3390/biom14101204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
Astrocytes are one of the key glial types of the central nervous system (CNS), accounting for over 20% of total glial cells in the brain. Extensive evidence has established their indispensable functions in the maintenance of CNS homeostasis, as well as their broad involvement in neurological conditions. In particular, astrocytes can participate in various neuroinflammatory processes, e.g., releasing a repertoire of cytokines and chemokines or specific neurotrophic factors, which result in both beneficial and detrimental effects. It has become increasingly clear that such astrocyte-mediated neuroinflammation, together with its complex crosstalk with other glial cells or immune cells, designates neuronal survival and the functional integrity of neurocircuits, thus critically contributing to disease onset and progression. In this review, we focus on the current knowledge of the neuroinflammatory responses of astrocytes, summarizing their common features in neurological conditions. Moreover, we highlight several vital questions for future research that promise novel insights into diagnostic or therapeutic strategies against those debilitating CNS diseases.
Collapse
Affiliation(s)
- Yanxiang Zhao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- The Affiliated High School, Peking University, Beijing 100080, China
| | - Yingying Huang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ying Cao
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jing Yang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Peking University Third Hospital Cancer Center, Beijing 100191, China
| |
Collapse
|
138
|
Martínez-Espinosa I, Serrato JA, Ortiz-Quintero B. MicroRNAs in Lung Cancer Brain Metastasis. Int J Mol Sci 2024; 25:10325. [PMID: 39408656 PMCID: PMC11476622 DOI: 10.3390/ijms251910325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Brain metastasis is a significant clinical challenge for patients with advanced lung cancer, occurring in about 20-40% of cases. Brain metastasis causes severe neurological symptoms, leading to a poor prognosis and contributing significantly to lung cancer-related mortality. However, the underlying molecular mechanism behind brain metastasis remains largely unknown. MicroRNAs (miRNAs) are small, non-coding RNAs linked to several aspects of cancer progression, including metastasis. In the context of lung cancer, significant research has shown the involvement of miRNAs in regulating critical pathways related to metastatic spread to the brain. This review summarizes the scientific evidence regarding the regulatory roles of intra- and extracellular miRNAs, which specifically drive the spread of lung cancer cells to the brain. It also revises the known molecular mechanisms of brain metastasis, focusing on those from lung cancer as the primary tumor to better understand the complex mechanisms underlying this regulation. Understanding these complex regulatory mechanisms holds promise for developing novel diagnostic biomarkers and potential therapeutic strategies in brain metastasis.
Collapse
Affiliation(s)
| | | | - Blanca Ortiz-Quintero
- Department of Molecular Biomedicine and Translational Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080 Mexico City, Mexico
| |
Collapse
|
139
|
Mohamed SS, Rasheed NOA, Ibrahim WW, Shiha NA. Targeting Toll-like Receptor 4/Nuclear Factor-κB and Nrf2/Heme Oxygenase-1 Crosstalk via Trimetazidine Alleviates Lipopolysaccharide-Induced Depressive-like Behaviors in Mice. J Neuroimmune Pharmacol 2024; 19:50. [PMID: 39312021 PMCID: PMC11420337 DOI: 10.1007/s11481-024-10149-3] [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: 05/18/2024] [Accepted: 09/01/2024] [Indexed: 09/26/2024]
Abstract
Depression is a global psychiatric illness that imposes a substantial economic burden. Unfortunately, traditional antidepressants induce many side effects which limit patient compliance thus, exploring alternative therapies with fewer adverse effects became urgent. This study aimed to investigate the effect of trimetazidine (TMZ); a well-known anti-ischemic drug in lipopolysaccharide (LPS) mouse model of depression focusing on its ability to regulate toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) as well as nuclear factor erythroid 2 related factor 2 (Nrf2)/ heme oxygenase-1 (HO-1) signaling pathways. Male Swiss albino mice were injected with LPS (500 µg/kg, i.p) every other day alone or parallel with oral doses of either escitalopram (Esc) (10 mg/kg/day) or TMZ (20 mg/kg/day) for 14 days. Treatment with TMZ attenuated LPS-induced animals' despair with reduced immobility time inforced swimming test. TMZ also diminished LPS- induced neuro-inflammation via inhibition of TLR4/NF-κB pathway contrary to Nrf2/HO-1 cascade activation with consequent increase in reduced glutathione (GSH) and HO-1 levels whereas the pro-inflammatory cytokines; tumor necrosis factor-α (TNF-α) and interleukin (IL)-1β were evidently reduced. Besides, TMZ replenished brain serotonin levels via serotonin transporter (SERT) inhibition. Thus, TMZ hindered LPS-induced neuro-inflammation, oxidative stress, serotonin deficiency besides its anti-apoptotic effect which was reflected by decreased caspase-3 level. Neuroprotective effects of TMZ were confirmed by the histological photomicrographs which showed prominent neuronal survival. Here we showed that TMZ is an affluent nominee for depression management via targeting TLR4/NF-κB and Nrf2/HO-1 pathways. Future research addressing TMZ-antidepressant activity in humans is mandatory to enroll it as a novel therapeutic strategy for depression.
Collapse
Affiliation(s)
- Sarah S Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, Egypt.
| | - Nora O Abdel Rasheed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, Egypt
| | - Weam W Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, Egypt
| | - Nesma A Shiha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, Egypt
| |
Collapse
|
140
|
Chuang SH, Chen KJ, Cheng YT, Chen YS, Lin SY, Chou HY, Tsai HC. A thermo-responsive chemically crosslinked long-term-release chitosan hydrogel system increases the efficiency of synergy chemo-immunotherapy in treating brain tumors. Int J Biol Macromol 2024; 280:135894. [PMID: 39322160 DOI: 10.1016/j.ijbiomac.2024.135894] [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: 04/29/2024] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Glioblastoma multiforme (GBM) is an aggressive and common brain tumor. The blood-brain barrier prevents several treatments from reaching the tumor. This study proposes a Chemo-Immunotherapy synergy treatment chemically crosslinked hydrogel system that is injected into the tumor to treat GBM. The strategy uses doxorubicin and BMS-1 with a thermo-responsive and chemically crosslinked hydrogel for extended drug release into the affected area. The hydrogels are produced by mixing with Chitosan (Chi), modified Pluronic F-127 (PF-127) with aldehyde end group and doxorubicin and then chemically crosslinking the aldehyde and amine bonds to increase the drug retention time. PF-127-CHO/Chi, which gels at body temperatures and chemically crosslinks between PF-127-CHO and Chitosan, increases the time that the drug remains in the affected area and prevents the hydrogel from swelling and compressing surrounding tissue. The drug is released from the chemically crosslinked hydrogels, prevents tumor progression and increases survival for subjects with GBM tumors. The Synergy Chemo-Immunotherapy also allows more efficient treatment of GBM than chemotherapy. The PF-127-CHO/Chi DOX and BMS-1 group have a tumor that is 43 times smaller than the untreated group. These results show that the proposed chemically crosslinking hydrogel is an efficient intratumoral delivery platform for the treatment of tumors.
Collapse
Affiliation(s)
- Shun-Hao Chuang
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan
| | - Kuan-Ju Chen
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan
| | - Yu-Ting Cheng
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan
| | - Yu-Shuan Chen
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Taiwan; Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan
| | - Shuian-Yin Lin
- Biomedical Technology and Device Research Center, Industrial Technology Research Institute, Hsinchu, 310, Taiwan
| | - Hsiao-Ying Chou
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan
| | - Hsieh-Chih Tsai
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan; Advanced Membrane Materials Center, National Taiwan University of Science and Technology, Taipei 106, Taiwan; R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan.
| |
Collapse
|
141
|
Shamul JG, Wang Z, Gong H, Ou W, White AM, Moniz-Garcia DP, Gu S, Clyne AM, Quiñones-Hinojosa A, He X. Meta-analysis of the make-up and properties of in vitro models of the healthy and diseased blood-brain barrier. Nat Biomed Eng 2024:10.1038/s41551-024-01250-2. [PMID: 39304761 DOI: 10.1038/s41551-024-01250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
In vitro models of the human blood-brain barrier (BBB) are increasingly used to develop therapeutics that can cross the BBB for treating diseases of the central nervous system. Here we report a meta-analysis of the make-up and properties of transwell and microfluidic models of the healthy BBB and of BBBs in glioblastoma, Alzheimer's disease, Parkinson's disease and inflammatory diseases. We found that the type of model, the culture method (static or dynamic), the cell types and cell ratios, and the biomaterials employed as extracellular matrix are all crucial to recapitulate the low permeability and high expression of tight-junction proteins of the BBB, and to obtain high trans-endothelial electrical resistance. Specifically, for models of the healthy BBB, the inclusion of endothelial cells and pericytes as well as physiological shear stresses (~10-20 dyne cm-2) are necessary, and when astrocytes are added, astrocytes or pericytes should outnumber endothelial cells. We expect this meta-analysis to facilitate the design of increasingly physiological models of the BBB.
Collapse
Affiliation(s)
- James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Zhiyuan Wang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Hyeyeon Gong
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Alisa M White
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | | | - Shuo Gu
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA
| | | | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA.
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
142
|
Bassi I, Grunspan M, Hen G, Ravichandran KA, Moshe N, Gutierrez-Miranda L, Safriel SR, Kostina D, Shen A, Ruiz de Almodovar C, Yaniv K. Endolysosomal dysfunction in radial glia progenitor cells leads to defective cerebral angiogenesis and compromised blood-brain barrier integrity. Nat Commun 2024; 15:8158. [PMID: 39289367 PMCID: PMC11408700 DOI: 10.1038/s41467-024-52365-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
The neurovascular unit (NVU) is a complex multicellular structure that helps maintain cerebral homeostasis and blood-brain barrier (BBB) integrity. While extensive evidence links NVU alterations to cerebrovascular diseases and neurodegeneration, the underlying molecular mechanisms remain unclear. Here, we use zebrafish embryos carrying a mutation in Scavenger Receptor B2, a highly conserved endolysosomal protein expressed predominantly in Radial Glia Cells (RGCs), to investigate the interplay among different NVU components. Through live imaging and genetic manipulations, we demonstrate that compromised acidification of the endolysosomal compartment in mutant RGCs leads to impaired Notch3 signaling, thereby inducing excessive neurogenesis and reduced glial differentiation. We further demonstrate that alterations to the neuron/glia balance result in impaired VEGF and Wnt signaling, leading to severe vascular defects, hemorrhages, and a leaky BBB. Altogether, our findings provide insights into NVU formation and function and offer avenues for investigating diseases involving white matter defects and vascular abnormalities.
Collapse
Affiliation(s)
- Ivan Bassi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Moshe Grunspan
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Gideon Hen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Kishore A Ravichandran
- Institute for Neurovascular Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Noga Moshe
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Laura Gutierrez-Miranda
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Stav R Safriel
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daria Kostina
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Amitay Shen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Carmen Ruiz de Almodovar
- Institute for Neurovascular Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
- Schlegel Chair for Neurovascular Cell Biology, University of Bonn, Bonn, Germany
| | - Karina Yaniv
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
143
|
Wakonigg Alonso C, McElhatton F, O'Mahony B, Campbell M, Pollak TA, Stokes PRA. The blood-brain barrier in bipolar disorders: A systematic review. J Affect Disord 2024; 361:434-444. [PMID: 38897301 DOI: 10.1016/j.jad.2024.06.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/13/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Bipolar disorders (BD) are chronic, debilitating disorders. The blood-brain barrier (BBB) has been increasingly investigated in BD. This systematic review aimed to assess the available evidence on the relationship between BD and markers of BBB dysfunction. METHODS A systematic search in PubMed, Embase, PsycINFO, CINAHL and Web of Science was run where the primary outcomes were BBB markers such as S100B, albumin ratio, matrix metalloproteinase (MMP), cell adhesion molecule (CAM), and tight junction proteins. Techniques included blood, cerebrospinal fluid (CSF), post-mortem, genetic and imaging methods in BD compared to healthy controls. RESULTS 55 studies were identified, 38 of which found an association between BD and markers of BBB dysfunction. 16/29 studies found increased blood/CSF albumin ratio, S100B, CAMs or MMP levels in BD participants compared to controls. 5/19 post-mortem studies found increased levels of chondroitin sulphate proteoglycans, intercellular CAM, neurexin or claudin-5 mRNA in distinct locations throughout the brain in BD compared to controls. One imaging study identified extensive BBB leakage in 30 % of BD participants, compared to 0 % in controls. LIMITATIONS The diversity in methodologies used in the included studies makes direct comparison of results challenging. Furthermore, imaging methods are the gold standard, but only one study used them. Other markers are only indicative of BBB permeability. CONCLUSIONS This review suggests an association between BD and BBB dysfunction. Further research is needed to provide definite answers considering the existing literature's limitations, and to clarify whether this association provides a pathogenic mechanism, or is an epiphenomenon of BD.
Collapse
Affiliation(s)
- Clara Wakonigg Alonso
- Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom.
| | - Frances McElhatton
- Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom
| | - Brian O'Mahony
- Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | - Thomas A Pollak
- Dept of Psychosis Studies, Institute of Psychiatry & Psychology and Neuroscience, King's College London, United Kingdom; South London and Maudsley NHS Foundation Trust,Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, BR3 3BX, United Kingdom
| | - Paul R A Stokes
- South London and Maudsley NHS Foundation Trust,Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, BR3 3BX, United Kingdom; Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom
| |
Collapse
|
144
|
Lu S, Di John Portela I, Martino N, Bossardi Ramos R, Salinero AE, Smith RM, Zuloaga KL, Adam AP. A transient brain endothelial translatome response to endotoxin is associated with mild cognitive changes post-shock in young mice. Neuroscience 2024; 555:194-204. [PMID: 39067684 PMCID: PMC11470799 DOI: 10.1016/j.neuroscience.2024.07.041] [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: 05/16/2024] [Revised: 07/10/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Sepsis-associated encephalopathy (SAE) is associated with increased risk of long-term cognitive impairment. SAE is driven, at least in part, by brain endothelial dysfunction in response to systemic cytokine signaling. However, the mechanisms driving SAE and its consequences remain largely unknown. Here, we performed translating ribosome affinity purification and RNA-sequencing (TRAP-seq) from the brain endothelium to determine the transcriptional changes after an acute endotoxemic (LPS) challenge. LPS induced a strong acute transcriptional response in the brain endothelium that partially correlates with the whole brain transcriptional response and suggested an endothelial-specific hypoxia response. Consistent with a crucial role for IL-6, loss of the main regulator of this pathway, SOCS3, leads to a broadening of the population of genes responsive to LPS, suggesting that an overactivation of the IL-6/JAK/STAT3 pathway leads to an increased transcriptional response that could explain our prior findings of severe brain injury in these mice. To identify any potential sequelae of this acute response, we performed brain TRAP-seq following a battery of behavioral tests in mice after apparent recovery. We found that the transcriptional response returns to baseline within days post-challenge, but reductions in gene expression regulating protein translation and respiratory electron transport remained. We observed that mice that recovered from the endotoxemic shock showed mild, sex-dependent cognitive impairment, suggesting that the acute brain injury led to sustained effects. A better understanding of the transcriptional and non-transcriptional changes in response to shock is needed in order to prevent and/or revert the devastating consequences of septic shock.
Collapse
Affiliation(s)
- Shuhan Lu
- Department of Molecular and Cellular Physiology, Albany Medical College, United States
| | - Iria Di John Portela
- Department of Molecular and Cellular Physiology, Albany Medical College, United States
| | - Nina Martino
- Department of Molecular and Cellular Physiology, Albany Medical College, United States
| | - Ramon Bossardi Ramos
- Department of Molecular and Cellular Physiology, Albany Medical College, United States
| | - Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, United States
| | - Rachel M Smith
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, United States
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, United States
| | - Alejandro P Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, United States; Department of Ophthalmology, Albany Medical College, United States.
| |
Collapse
|
145
|
Kuhn E, Srinageshwar B, Story DT, Swanson D, Sharma A, Dunbar GL, Rossignol J. Delivery of PAMAM dendrimers and dendriplexes across natural barriers (blood-brain barrier and placental barrier) in healthy pregnant mice. DISCOVER NANO 2024; 19:148. [PMID: 39264474 PMCID: PMC11393257 DOI: 10.1186/s11671-024-04105-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Gene therapy is an important tool for treating fetal diseases that allows for the delivery and integration of therapeutic genes into the genome of cells carrying mutations. Nanomolecules, like PAMAM dendrimers, have recently come into wider use for carrying vectors as they have several advantages over viral vectors. Namely, (1) tunable size and surface chemistry, (2) uniform size, (3) the ability to target specific tissues, and (4) the ability to carry large biomolecules and drugs. Recently, we demonstrated that 4th generation (G4) PAMAM dendrimer with a cystamine core and a non-toxic surface having 90% -OH and 10% -NH2 groups (D-Cys) could cross the blood-brain barrier following injection into the bloodstream. In the current study, as a proof of concept, we delivered the dendrimers alone (D-Cys) tagged with Cy5.5 (D-Cys-cy5.5) to healthy pregnant C57BL/6J mice to determine the fate of these dendrimers in the pregnant mice as well as in the fetus. Systematic diffusion of the D-Cys-cy5.5 was evaluated on gestational day 17 (3 days after injection) using in vivo imaging. This revealed that the dendrimer was taken up into circulation and away from the injection site. Analysis of sections by fluorescence microscopy showed that D-Cys-cy5.5 was able to successfully cross the maternal blood-brain barrier. However, analysis of the fetal brains failed to detect dendrimers in the central nervous system (CNS). Instead, they appeared to be retained in the placenta. This is one of the first studies to analyze the distribution of surface-modified PAMAM dendrimer in the pregnant mouse and fetus following systemic injection.
Collapse
Affiliation(s)
- Eric Kuhn
- College of Medicine, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Bhairavi Srinageshwar
- College of Medicine, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Darren T Story
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Department of Psychology, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Department of Psychology, Saginaw Valley State University, University Center, MI, 48710, USA
| | - Douglas Swanson
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Ajit Sharma
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Gary L Dunbar
- College of Medicine, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Department of Psychology, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Julien Rossignol
- College of Medicine, Central Michigan University, Mount Pleasant, MI, 48859, USA.
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, 48859, USA.
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA.
| |
Collapse
|
146
|
Polyzos AA, Cheong A, Yoo JH, Blagec L, Toprani SM, Nagel ZD, McMurray CT. Base excision repair and double strand break repair cooperate to modulate the formation of unrepaired double strand breaks in mouse brain. Nat Commun 2024; 15:7726. [PMID: 39231940 PMCID: PMC11375129 DOI: 10.1038/s41467-024-51906-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 08/19/2024] [Indexed: 09/06/2024] Open
Abstract
We lack the fundamental information needed to understand how DNA damage in the brain is generated and how it is controlled over a lifetime in the absence of replication check points. To address these questions, here, we integrate cell-type and region-specific features of DNA repair activity in the normal brain. The brain has the same repair proteins as other tissues, but normal, canonical repair activity is unequal and is characterized by high base excision repair (BER) and low double strand break repair (DSBR). The natural imbalance creates conditions where single strand breaks (SSBs) can convert to double strand breaks (DSBs) and reversibly switch between states in response to oxidation both in vivo and in vitro. Our data suggest that, in a normal background of repair, SSBs and DSBs are in an equilibrium which is pushed or pulled by metabolic state. Interconversion of SSB to DSBs provides a physiological check point, which would allow the formation of unrepaired DSBs for productive functions, but would also restrict them from exceeding tolerable limits.
Collapse
Affiliation(s)
- Aris A Polyzos
- Division of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| | - Ana Cheong
- Department of Environmental Health, John B Little Centre for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jung Hyun Yoo
- Division of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Lana Blagec
- Division of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Sneh M Toprani
- Department of Environmental Health, John B Little Centre for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zachary D Nagel
- Department of Environmental Health, John B Little Centre for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Cynthia T McMurray
- Division of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
147
|
Tail M, Zhang H, Zheng G, Harms AK, Hatami M, Skutella T, Kiening K, Unterberg A, Zweckberger K, Younsi A. Sonic Hedgehog reduces inflammatory response, decreases blood-spinal cord barrier permeability, and improves locomotor function recovery in an acute spinal cord injury rat model. J Inflamm (Lond) 2024; 21:34. [PMID: 39227870 PMCID: PMC11373473 DOI: 10.1186/s12950-024-00404-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Sonic Hedgehog (Shh), extensively researched for its role in early neurogenesis and brain development, has recently been recognized for its neuroprotective potential following neuronal injuries. This study examines the immediate impact of early administered Shh on the local inflammatory response post-acute spinal cord injury in rats. METHODS Thirty-four female Wistar rats underwent either sham surgery (laminectomy; n = 10) or clip compression/contusion spinal cord injury (SCI) at the T9 level. This was followed by implantation of an osmotic pump and a subdural catheter for continuous intrathecal delivery of Shh (n = 12) or placebo (NaCl; n = 12). Locomotor function was assessed at 3- and 7-days post-injury (dpi) using the Basso, Beattie, and Bresnahan (BBB) score and the Gridwalk test. Animals were euthanized after 3 or 7 days for immunohistochemical analysis of the local inflammatory reaction and immune cell migration. RESULTS Shh-treated rats demonstrated significant hindlimb movement and coordination improvements at 7 days post-injury, compared to controls. This enhancement was accompanied by a significant reduction in both immune cell presence and blood plasma products within spinal cord lesions, suggesting Shh's dual role in modulating immune cell migration and maintaining the integrity of the blood-spinal cord barrier. Separately, these Shh-treated rats also showed an increase in M(IL-4) polarization of macrophages, further underlining the potential therapeutic impact of Shh in post-injury recovery. Notably, these effects were not evident at three days post-injury. CONCLUSION Shh application at 7 days post-injury showed immunomodulatory effects, possibly via enhanced blood-spinal cord barrier integrity, reduced immune cell migration, and increased anti-inflammatory immune cell differentiation. These mechanisms collectively contribute to enhanced locomotor recovery.
Collapse
Affiliation(s)
- Mohamed Tail
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
- Department of Neurology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Hao Zhang
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Guoli Zheng
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Anna-Kathrin Harms
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Maryam Hatami
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120, Heidelberg, Germany
| | - Thomas Skutella
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120, Heidelberg, Germany
| | - Karl Kiening
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Klaus Zweckberger
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Alexander Younsi
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany.
| |
Collapse
|
148
|
Kwak G, Grewal A, Slika H, Mess G, Li H, Kwatra M, Poulopoulos A, Woodworth GF, Eberhart CG, Ko HS, Manbachi A, Caplan J, Price RJ, Tyler B, Suk JS. Brain Nucleic Acid Delivery and Genome Editing via Focused Ultrasound-Mediated Blood-Brain Barrier Opening and Long-Circulating Nanoparticles. ACS NANO 2024; 18:24139-24153. [PMID: 39172436 DOI: 10.1021/acsnano.4c05270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
We introduce a two-pronged strategy comprising focused ultrasound (FUS)-mediated blood-brain barrier (BBB) opening and long-circulating biodegradable nanoparticles (NPs) for systemic delivery of nucleic acids to the brain. Biodegradable poly(β-amino ester) polymer-based NPs were engineered to stably package various types of nucleic acid payloads and enable prolonged systemic circulation while retaining excellent serum stability. FUS was applied to a predetermined coordinate within the brain to transiently open the BBB, thereby allowing the systemically administered long-circulating NPs to traverse the BBB and accumulate in the FUS-treated brain region, where plasmid DNA or mRNA payloads produced reporter proteins in astrocytes and neurons. In contrast, poorly circulating and/or serum-unstable NPs, including the lipid NP analogous to a platform used in clinic, were unable to provide efficient nucleic acid delivery to the brain regardless of the BBB-opening FUS. The marriage of FUS-mediated BBB opening and the long-circulating NPs engineered to copackage mRNA encoding CRISPR-associated protein 9 and single-guide RNA resulted in genome editing in astrocytes and neurons precisely in the FUS-treated brain region. The combined delivery strategy provides a versatile means to achieve efficient and site-specific therapeutic nucleic acid delivery to and genome editing in the brain via a systemic route.
Collapse
Affiliation(s)
- Gijung Kwak
- Department of Neurosurgery, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
- Medicine Institute for Neuroscience Discovery (UM-MIND), School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
| | - Angad Grewal
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Hasan Slika
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Griffin Mess
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Haolin Li
- Department of Neurosurgery, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
- Department of Chemical and Biomolecular Engineering, School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Mohit Kwatra
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Alexandros Poulopoulos
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
| | - Graeme F Woodworth
- Department of Neurosurgery, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
- Medicine Institute for Neuroscience Discovery (UM-MIND), School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
| | - Charles G Eberhart
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21287, United States
- Department of Ophthalmology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
| | - Han Seok Ko
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Amir Manbachi
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Justin Caplan
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Richard J Price
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Betty Tyler
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Jung Soo Suk
- Department of Neurosurgery, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
- Medicine Institute for Neuroscience Discovery (UM-MIND), School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
- Department of Chemical and Biomolecular Engineering, School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
149
|
Li YM, Xu XH, Ren LN, Xu XF, Dai YL, Yang RR, Jin CQ. The diagnostic value of neutrophil to lymphocyte ratio, albumin to fibrinogen ratio, and lymphocyte to monocyte ratio in Parkinson's disease: a retrospective study. Front Neurol 2024; 15:1450221. [PMID: 39286804 PMCID: PMC11402719 DOI: 10.3389/fneur.2024.1450221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Background Parkinson's disease (PD) is a prevalent disorder of the central nervous system, marked by the degeneration of dopamine (DA) neurons in the ventral midbrain. In the pathogenesis of PD, inflammation hypothesis has been concerned. This study aims to investigate clinical indicators of peripheral inflammation in PD patients and to explore the diagnostic value of neutrophil-to-lymphocyte ratio (NLR), albumin-to-fibrinogen ratio (AFR), and lymphocyte-to-monocyte ratio (LMR) in assessing PD risk. Methods This study included 186 patients with PD and 201 matched healthy controls (HC) with baseline data. Firstly, the differences of hematological indicators between PD group and healthy participants were compared and analyzed. Univariate and multivariate regression analyses were then conducted. Smooth curve fitting was applied to further validate the relationships between NLR, LMR, AFR, and PD. Subsequently, subgroup analysis was conducted in PD group according to different duration of disease and Hoehn and Yahr (H&Y) stage, comparing differences in clinical indicators. Finally, the receiver operating characteristic (ROC) curve was employed to assess the diagnostic value of NLR, LMR, and AFR in PD. Results Compared to the HC group, the PD group showed significantly higher levels of hypertension, diabetes, neutrophil count, monocyte count, CRP, homocysteine, fibrinogen, and NLR. Conversely, levels of LMR, AFR, lymphocyte count, HDL, LDL, TG, TC, uric acid, and albumin were significantly lower. The multivariate regression model indicated that NLR (OR = 1.79, 95% CI: 1.39-2.31, p < 0.001), LMR (OR = 0.75, 95% CI: 0.66-0.85, p < 0.001), and AFR (OR = 0.79, 95% CI: 0.73-0.85, p < 0.001) were significant factors associated with PD. Smooth curve fitting revealed that NLR was positively linked to PD risk, whereas AFR and LMR were inversely associated with it. In ROC curve analysis, the AUC of AFR was 0.7290, the sensitivity was 63.98%, and the specificity was 76.00%. The AUC of NLR was 0.6200, the sensitivity was 50.54%, and the specificity was 71.50%. The AUC of LMR was 0.6253, the sensitivity was 48.39%, and the specificity was 73.00%. The AUC of the combination was 0.7498, the sensitivity was 74.19%, and the specificity was 64.00%. Conclusion Our findings indicate that NLR, LMR, and AFR are significantly associated with Parkinson's disease and may serve as diagnostic markers.
Collapse
Affiliation(s)
- Yi-Ming Li
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
- Medical Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Xiao-Hu Xu
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
- Medical Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Li-Na Ren
- Medical Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Xiao-Fan Xu
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
- Medical Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Yi-Long Dai
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
- Medical Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Rui-Rui Yang
- Neurology Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Cheng-Qiang Jin
- Medical Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| |
Collapse
|
150
|
Banerjee P, Chau K, Kotla S, Davis EL, Turcios EB, Li S, Pengzhi Z, Wang G, Kolluru GK, Jain A, Cooke JP, Abe J, Le NT. A Potential Role for MAGI-1 in the Bi-Directional Relationship Between Major Depressive Disorder and Cardiovascular Disease. Curr Atheroscler Rep 2024; 26:463-483. [PMID: 38958925 DOI: 10.1007/s11883-024-01223-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW Major Depressive Disorder (MDD) is characterized by persistent symptoms such as fatigue, loss of interest in activities, feelings of sadness and worthlessness. MDD often coexist with cardiovascular disease (CVD), yet the precise link between these conditions remains unclear. This review explores factors underlying the development of MDD and CVD, including genetic, epigenetic, platelet activation, inflammation, hypothalamic-pituitary-adrenal (HPA) axis activation, endothelial cell (EC) dysfunction, and blood-brain barrier (BBB) disruption. RECENT FINDINGS Single nucleotide polymorphisms (SNPs) in the membrane-associated guanylate kinase WW and PDZ domain-containing protein 1 (MAGI-1) are associated with neuroticism and psychiatric disorders including MDD. SNPs in MAGI-1 are also linked to chronic inflammatory disorders such as spontaneous glomerulosclerosis, celiac disease, ulcerative colitis, and Crohn's disease. Increased MAGI-1 expression has been observed in colonic epithelial samples from Crohn's disease and ulcerative colitis patients. MAGI-1 also plays a role in regulating EC activation and atherogenesis in mice and is essential for Influenza A virus (IAV) infection, endoplasmic reticulum stress-induced EC apoptosis, and thrombin-induced EC permeability. Despite being understudied in human disease; evidence suggests that MAGI-1 may play a role in linking CVD and MDD. Therefore, further investigation of MAG-1 could be warranted to elucidate its potential involvement in these conditions.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
| | - Khanh Chau
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eleanor L Davis
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Estefani Berrios Turcios
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Shengyu Li
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Zhang Pengzhi
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Guangyu Wang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | | | - Abhishek Jain
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Department of Medical Physiology, School of Medicine, Texas A&M Health Science Center, Bryan, USA
| | - John P Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Junichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|