1
|
Zhang M, Xiang C, Niu R, He X, Luo W, Liu W, Gu R. Liposomes as versatile agents for the management of traumatic and nontraumatic central nervous system disorders: drug stability, targeting efficiency, and safety. Neural Regen Res 2025; 20:1883-1899. [PMID: 39254548 DOI: 10.4103/nrr.nrr-d-24-00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/28/2024] [Indexed: 09/11/2024] Open
Abstract
Various nanoparticle-based drug delivery systems for the treatment of neurological disorders have been widely studied. However, their inability to cross the blood-brain barrier hampers the clinical translation of these therapeutic strategies. Liposomes are nanoparticles composed of lipid bilayers, which can effectively encapsulate drugs and improve drug delivery across the blood-brain barrier and into brain tissue through their targeting and permeability. Therefore, they can potentially treat traumatic and nontraumatic central nervous system diseases. In this review, we outlined the common properties and preparation methods of liposomes, including thin-film hydration, reverse-phase evaporation, solvent injection techniques, detergent removal methods, and microfluidics techniques. Afterwards, we comprehensively discussed the current applications of liposomes in central nervous system diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, traumatic brain injury, spinal cord injury, and brain tumors. Most studies related to liposomes are still in the laboratory stage and have not yet entered clinical trials. Additionally, their application as drug delivery systems in clinical practice faces challenges such as drug stability, targeting efficiency, and safety. Therefore, we proposed development strategies related to liposomes to further promote their development in neurological disease research.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | | | | | | | | | | | | |
Collapse
|
2
|
Teglas T, Marcos AC, Torices S, Toborek M. Circadian control of polycyclic aromatic hydrocarbon-induced dysregulation of endothelial tight junctions and mitochondrial bioenergetics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 952:175886. [PMID: 39218115 PMCID: PMC11444715 DOI: 10.1016/j.scitotenv.2024.175886] [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: 04/23/2024] [Revised: 08/05/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
The study evaluates the impact of environmental toxicants, such as polycyclic aromatic hydrocarbons (PAHs), on circadian regulations and functions of brain endothelial cells, which form the main structural element of the blood-brain barrier (BBB). PAH are lipophilic and highly toxic environmental pollutants that accumulate in human and animal tissues. Environmental factors related to climate change, such as an increase in frequency and intensity of wildfires or enhanced strength of hurricanes or tropical cyclones, may lead to redistribution of these toxicants and enhanced human exposure. These natural disasters are also associated with disruption of circadian rhythms in affected populations, linking increased exposure to environmental toxicants to alterations of circadian rhythm pathways. Several vital physiological processes are coordinated by circadian rhythms, and disruption of the circadian clock can contribute to the development of several diseases. The blood-brain barrier (BBB) is crucial for protecting the brain from blood-borne harmful substances, and its integrity is influenced by circadian rhythms. Exposure of brain endothelial cells to a human and environmentally-relevant PAH mixture resulted in dose-dependent alterations of expression of critical circadian modulators, such as Clock, Bmal1, Cry1/2, and Per1/2. Moreover, silencing of the circadian Clock gene potentiated the impact of PAHs on the expression of the main tight junction genes and proteins (namely, claudin-5, occludin, JAM-2, and ZO-2), as well as mitochondrial bioenergetics. Findings from this study contribute to a better understanding of pathological influence of PAH-induced health effects, especially those related to circadian rhythm disruption.
Collapse
Affiliation(s)
- Timea Teglas
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA
| | - Anne Caroline Marcos
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA; Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland.
| |
Collapse
|
3
|
Mondkar PP, Seo HS, Lodge TP, Azarin SM. Diblock Copolymers of Poly(ethylene oxide)- b-poly(propylene oxide) Stabilize a Blood-Brain Barrier Model under Oxidative Stress. Mol Pharm 2024; 21:5646-5660. [PMID: 39400078 DOI: 10.1021/acs.molpharmaceut.4c00608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
The blood-brain barrier (BBB) is a highly restrictive barrier at the interface between the brain and the vascular system. Even under BBB dysfunction, it is extremely difficult to deliver therapies across the barrier, limiting the options for treatment of neurological injuries and disorders. To circumvent these challenges, there is interest in developing therapies that directly engage with the damaged BBB to restore its function. Previous studies revealed that poloxamer 188 (P188), a water-soluble triblock copolymer of poly(ethylene oxide) (PEO) and poly(propylene oxide) (PPO), partially mitigated BBB dysfunction in vivo. In the context of stabilization of the damaged BBB, the mechanism of action of PEO-PPO block copolymers is unknown, and there has been minimal exploration of polymers beyond P188. In this study, a human-based in vitro BBB model under oxidative stress was used to investigate polymer-BBB interactions since oxidative stress is closely linked with BBB dysfunction in many neurological injuries and disorders. PEO-PPO block copolymers of varied numbers of chemically distinct blocks, PEO block length, and functionality of the end group of the PPO block were assessed for their efficacy in improving key physiological readouts associated with BBB dysfunction. While treatment with P188 did not mitigate damage in the in vitro BBB model, treatment with three diblock copolymers improved barrier integrity under oxidative stress to a similar extent. Of the considered variations in the block copolymer design, the reduction in the number of chemically distinct blocks had the strongest influence on therapeutic function. The demonstrated efficacy of three alternative PEO-PPO diblock copolymers in this work reveals the potential of these polymers as a class of therapeutics that directly treat the damaged BBB, expanding the options for treatment of neurological injuries and disorders.
Collapse
Affiliation(s)
- Pranati P Mondkar
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Hannah S Seo
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Timothy P Lodge
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
4
|
Bai R, Ge X. Blood-brain barrier disruption following brain injury: Implications for clinical practice. Histol Histopathol 2024; 39:1435-1441. [PMID: 38618940 DOI: 10.14670/hh-18-740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The blood-brain barrier (BBB) plays a critical role in regulating the exchange of substances between peripheral blood and the central nervous system and in maintaining the stability of the neurovascular unit in neurological diseases. To guide clinical treatment and basic research on BBB protection following brain injury, this manuscript reviews how BBB disruption develops and influences neural recovery after stroke and traumatic brain injury (TBI). By summarizing the pathological mechanisms of BBB damage, we underscore the critical role of promoting BBB repair in managing brain injury. We also emphasize the potential for personalized and precise therapeutic strategies and the need for continued research and innovation. From this, broadening insights into the mechanisms of BBB disruption and repair could pave the way for breakthroughs in the treatment of brain injury-related diseases.
Collapse
Affiliation(s)
- Ruojing Bai
- Department of Geriatrics, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China
| | - Xintong Ge
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, PR China
- Tianjin Geriatrics Institute, Tianjin, PR China.
| |
Collapse
|
5
|
Jiang H, Zhang C, Lin M, Yin Y, Deng S, Liu W, Zhuo B, Tian G, Du Y, Meng Z. Deciphering the mechanistic impact of acupuncture on the neurovascular unit in acute ischemic stroke: Insights from basic research in a narrative review. Ageing Res Rev 2024; 101:102536. [PMID: 39384155 DOI: 10.1016/j.arr.2024.102536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Ischemic stroke(IS), a severe acute cerebrovascular disease, not only imposes a heavy economic burden on society but also presents numerous challenges in treatment. During the acute phase, while thrombolysis and thrombectomy serve as primary treatments, these approaches are restricted by a narrow therapeutic window. During rehabilitation, commonly used neuroprotective agents struggle with their low drug delivery efficiency and inadequate preclinical testing, and the long-term pharmacological and toxicity effects of nanomedicines remain undefined. Meanwhile, acupuncture as a therapeutic approach is widely acknowledged for its effectiveness in treating IS and has been recommended by the World Health Organization (WHO) as an alternative and complementary therapy, even though its exact mechanisms remain unclear. This review aims to summarize the known mechanisms of acupuncture and electroacupuncture (EA) in the treatment of IS. Research shows that acupuncture treatment mainly protects the neurovascular unit through five mechanisms: 1) reducing neuronal apoptosis and promoting neuronal repair and proliferation; 2) maintaining the integrity of the blood-brain barrier (BBB); 3) inhibiting the overactivation and polarization imbalance of microglia; 4) regulating the movement of vascular smooth muscle (VSM) cells; 5) promoting the proliferation of oligodendrocyte precursors. Through an in-depth analysis, this review reveals the multi-level, multi-dimensional impact of acupuncture treatment on the neurovascular unit (NVU) following IS, providing stronger evidence and a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Hailun Jiang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Chao Zhang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mengxuan Lin
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yu Yin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shizhe Deng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bifang Zhuo
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Guang Tian
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yuzheng Du
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Zhihong Meng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
6
|
Wang YH, Liao JM, Jan MS, Wang M, Su HH, Tsai WH, Liu PH, Tsuei YS, Huang SS. Prophylactic use of probiotics as an adjunctive treatment for ischemic stroke via the gut-spleen-brain axis. Brain Behav Immun 2024; 123:784-798. [PMID: 39442634 DOI: 10.1016/j.bbi.2024.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 10/11/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024] Open
Abstract
A growing body of research has focused on the role of spleen in orchestrating brain injury through the peripheral immune system following stroke, highlighting the brain-spleen axis as a potential target for mitigating neuronal damage during stroke. The gut microbiota plays a pivotal role in the bidirectional communication between the gut and the brain. Several studies have suggested that probiotic supplements hold promise as a strategic approach to maintaining a balanced intestinal microecology, reducing the apoptosis of intestinal epithelial cells, protecting the intestinal mucosal and blood-brain barrier (BBB), enhancing both intestinal and systemic immune functions, and thereby potentially affecting the pathogenesis and progression of ischemic stroke. In this study, we aimed to clarify the neuroprotective effects of supplementation with Lactobacillus, specifically Limosilactobacillus reuteri GMNL-89 (G89) and Lacticaseibacillus paracasei GMNL-133 (G133) on ischemic stroke and investigate how G89 and G133 modulate the communication mechanisms between the gut, brain, and spleen following ischemic stroke. We explored the neuroprotection and the underlying mechanisms of Lactobacillus supplementation in C57BL/6 mice subjected to permanent middle cerebral artery occlusion. Our results revealed that oral treatment with G89 or G133 alone, as well as oral administration combining G89 and G133, significantly decreased the infarct volume and improved the neurological function in mice with ischemic stroke. Moreover, G89 treatment alone preserved the tight junction integrity of gut barrier, while G133 alone and the combined treatment of G89 and G133 would significantly decreased the BBB permeability, and thereby significantly attenuated stroke-induced local and systemic inflammatory responses. Both G89 and G133 regulated cytotoxic T cells, and the balance between T helper 1 cells and T helper 2 cells in the spleen following ischemic stroke. Additionally, the combined administration of G89 and G133 improved the gut dysbiosis and significantly increased the concentration of short-chain fatty acids. In conclusion, our findings suggest that G89 and G133 may be used as nutrient supplements, holding promise as a prospective approach to combat ischemic stroke by modulating the gut-spleen-brain axis.
Collapse
Affiliation(s)
- Yi-Hsin Wang
- Department of Physiology, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Jiuan-Miaw Liao
- Department of Physiology, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Shiou Jan
- Department of Health Industry Technology Management, Chung Shan Medical University, Taichung, Taiwan
| | - Meilin Wang
- Department of Microbiology and Immunology, School of Medicine, Chung-Shan Medical University, Taichung, Taiwan
| | - Hsing-Hui Su
- Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan
| | - Wan-Hua Tsai
- Research and Development Department, GenMont Biotech Incorporation, Tainan 741014, Taiwan
| | - Pei-Hsun Liu
- Department & Institute of Physiology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yuang-Seng Tsuei
- College of Medicine, National Chung Hsing University, Taichung, Taiwan; Department of Surgical Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Neurosurgery, Taichung Verterans General Hospital, Taichung, Taiwan
| | - Shiang-Suo Huang
- Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
7
|
Kaddoura R, Abdalbari K, Kadom M, Badla BA, Hijleh AA, Hanifa M, AlAshkar M, Asbaita M, Othman D, Faraji H, AlBakri O, Tahlak S, Hijleh AA, Kabbani R, Resen M, Abdalbari H, Du Plessis SS, Omolaoye TS. Post-Meningitic Syndrome: Pathophysiology and Consequences of Streptococcal Infections on the Central Nervous System. Int J Mol Sci 2024; 25:11053. [PMID: 39456835 PMCID: PMC11507220 DOI: 10.3390/ijms252011053] [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: 07/16/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Streptococcus species represent a significant global cause of meningitis, leading to brain damage through bacterial virulence factors and the host inflammatory response. Upon entering the central nervous system (CNS), excessive inflammation leads to various neurological and psychological complications. This review explores the pathophysiological mechanisms and associated outcomes of streptococcal meningitis, particularly its short- and long-term neurological sequelae. Neurological symptoms, such as cognitive impairment, motor deficits, and sensory loss, are shown to vary in severity, with children being particularly susceptible to lasting complications. Among survivors, hearing loss, cognitive decline, and cranial nerve palsies emerge as the most frequently reported complications. The findings highlight the need for timely intervention, including neurorehabilitation strategies that focus on optimizing recovery and mitigating long-term disabilities. Future recommendations emphasize improving early diagnosis, expanding vaccine access, and personalizing rehabilitation protocols to enhance patient outcomes. As a novel contribution, this review proposes the term "post-meningitic syndrome" to showcase the broad spectrum of CNS complications that persist following streptococcal meningitis, providing a framework for a future clinical and research focus.
Collapse
Affiliation(s)
- Rachid Kaddoura
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Karim Abdalbari
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Mhmod Kadom
- Faculty of Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland;
| | - Beshr Abdulaziz Badla
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Amin Abu Hijleh
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Mohamed Hanifa
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Masa AlAshkar
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Mohamed Asbaita
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Deema Othman
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Hanan Faraji
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Orjwan AlBakri
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Sara Tahlak
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Amir Abu Hijleh
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Raneem Kabbani
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Murtadha Resen
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Helmi Abdalbari
- Faculty of Medicine, University of Nicosia, P.O. Box 24005, Nicosia 1700, Cyprus;
| | - Stefan S. Du Plessis
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| | - Temidayo S. Omolaoye
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (K.A.); (B.A.B.); (A.A.H.); (M.H.); (M.A.); (M.A.); (D.O.); (H.F.); (O.A.); (S.T.); (A.A.H.); (R.K.); (M.R.); (S.S.D.P.)
| |
Collapse
|
8
|
Yang JT, Kuo YC, Lee KC, De S, Chen YY. Resveratrol and ceftriaxone encapsulated in hybrid nanoparticles to prevent dopaminergic neurons from degeneration for Parkinson's disease treatment. BIOMATERIALS ADVANCES 2024; 166:214065. [PMID: 39426178 DOI: 10.1016/j.bioadv.2024.214065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
The purpose of this study is to evaluate the influence of phospholipid-polymer nanoparticles (PNPs) on mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling of dopaminergic neurons in degenerated brain. Resveratrol (RES)- and ceftriaxone (CEF)-entrapped PNPs with surface leptin (Lep) and transferrin (Tf) were fabricated to rescue both 1-methyl-4-phenylpyridinium (MPP+)-insulted SH-SY5Y cells and Wistar rats. Based on PNPs, anti-apoptosis of RES and CEF, and targeting of Lep and Tf were investigated. Experimental results revealed that 20-30 % alginic acid (Alg) yielded the maximal particle size, physical stability and entrapment efficiency of CEF, and the minimal release percentage of CEF. Increasing Alg content in PNPs decreased the entrapment efficiency of RES, and facilitated the release of RES. Optimized PNP composition was about 40 % Alg, 15 % phosphatidylserine and 45 % poly-ε-caprolactone. Lep-Tf-PNPs ameliorated brain permeability of RES and CEF without jeopardizing the blood-brain barrier, and promoted the viability of MPP+-insulted SH-SY5Y cells. Immunofluorescence images and western blots of MPP+-insulted SH-SY5Y cells showed that Lep-Tf-RES-CEF-PNPs upregulated dopamine transporter, tyrosine hydroxylase, B-cell lymphoma 2 (Bcl-2), cyclic AMP response element-binding protein and ERK5 expressions, and downregulated Bcl-2-associated X protein (Bax), α-synuclein (α-syn), phosphorylated tau protein (p-tau), c-Jun N-terminal kinase and ERK1/2 expressions. Lep-Tf-RES-CEF-PNPs unveiled a strong capacity to recover Bcl-2, Bax, α-syn and p-tau levels from MPP+ injury in the substantia nigra of rats. Hence, Lep-Tf-RES-CEF-PNPs can retard α-syn fibril formation, prevent tau protein from phosphorylation, and moderate MAPK/ERK and phosphatidylinositol 3-kinase/protein kinase B, and are promising for brain- and neuron-targeted pharmacotherapy to manage Parkinson's disease.
Collapse
Affiliation(s)
- Jen-Tsung Yang
- Department of Neurosurgery, Chang Gung Memorial Hospital, 6, West Sec., Chia-Pu Road, Chia-Yi 61363, Taiwan, ROC; College of Medicine, Chang Gung University, 259, Wenhua 1st Road, Tao-Yuan 33302, Taiwan, ROC
| | - Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC; Advanced Institute of Manufacturing with High-tech Innovations, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC.
| | - Kuan-Chun Lee
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| | - Sourav De
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC; Department of Pharmaceutical Technology, Eminent College of Pharmaceutical Technology, Barasat, West Bengal 700126, India
| | - Yu-Yin Chen
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| |
Collapse
|
9
|
Greene C, Rebergue N, Fewell G, Janigro D, Godfrin Y, Campbell M, Lemarchant S. NX210c drug candidate peptide strengthens mouse and human blood-brain barriers. Fluids Barriers CNS 2024; 21:76. [PMID: 39334382 PMCID: PMC11438064 DOI: 10.1186/s12987-024-00577-x] [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: 06/05/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Alterations of blood-brain barrier (BBB) and blood-spinal cord barrier have been documented in various animal models of neurodegenerative diseases and in patients. Correlations of these alterations with functional deficits suggest that repairing barriers integrity may represent a disease-modifying approach to prevent neuroinflammation and neurodegeneration induced by the extravasation of blood components into the parenchyma. Here, we screened the effect of a subcommissural organ-spondin-derived peptide (NX210c), known to promote functional recovery in several models of neurological disorders, on BBB integrity in vitro and in vivo. METHODS In vitro, bEnd.3 endothelial cell (EC) monolayers and two different primary human BBB models containing EC, astrocytes and pericytes, in static and microfluidic conditions, were treated with NX210c (1-100 µM), or its vehicle, for 4 h and up to 5 days. Tight junction (TJ) protein levels, permeability to dextrans and transendothelial electrical resistance (TEER) were evaluated. In vivo, young and old mice (3- and 21-month-old, respectively) were treated daily intraperitoneally with NX210c at 10 mg/kg or its vehicle for 5 days and their brains collected at day 6 to measure TJ protein levels by immunohistochemistry. RESULTS NX210c induced an increase in claudin-5 protein expression after 24-h and 72-h treatments in mouse EC. Occludin level was also increased after a 24-h treatment. Accordingly, NX210c decreased by half the permeability of EC to a 40-kDa FITC-dextran and increased TEER. In the human static BBB model, NX210c increased by ∼ 25% the TEER from 3 to 5 days. NX210c also increased TEER in the human 3D dynamic BBB model after 4 h, which was associated with a reduced permeability to a 4-kDa FITC-dextran. In line with in vitro results, after only 5 days of daily treatments in mice, NX210c restored aging-induced reduction of claudin-5 and occludin levels in the hippocampus, and also in the cortex for occludin. CONCLUSIONS In summary, we have gathered preclinical data showing the capacity of NX210c to strengthen BBB integrity. Through this property, NX210c holds great promises of being a disease-modifying treatment for several neurological disorders with high unmet medical needs.
Collapse
Affiliation(s)
- Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | | | | | | | - Yann Godfrin
- Axoltis Pharma, 60 avenue Rockefeller, Lyon, 69008, France
- Godfrin Life-Sciences, Caluire-et-Cuire, 69300, France
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | | |
Collapse
|
10
|
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
|
11
|
Vita SM, Cruise SC, Gilpin NW, Molina PE. HISTOLOGICAL COMPARISON OF REPEATED MILD WEIGHT DROP AND LATERAL FLUID PERCUSSION INJURY MODELS OF TRAUMATIC BRAIN INJURY IN FEMALE AND MALE RATS. Shock 2024; 62:398-409. [PMID: 38813916 DOI: 10.1097/shk.0000000000002395] [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: 05/31/2024]
Abstract
ABSTRACT In preclinical traumatic brain injury (TBI) research, the animal model should be selected based on the research question and outcome measures of interest. Direct side-by-side comparisons of different injury models are essential for informing such decisions. Here, we used immunohistochemistry to compare the outcomes from two common models of TBI, lateral fluid percussion (LFP) and repeated mild weight drop (rmWD) in adult female and male Wistar rats. Specifically, we measured the effects of LFP and rmWD on markers of cerebrovascular and tight junction disruption, neuroinflammation, mature neurons, and perineuronal nets in the cortical site of injury, cortex adjacent to injury, dentate gyrus, and the CA 2/3 area of the hippocampus. Animals were randomized into the LFP or rmWD group. On day 1, the LFP group received a craniotomy, and on day 4, injury (or sham procedure; randomly assigned). The rmWD animals underwent either injury or isoflurane only (randomly assigned) on each of those 4 days. Seven days after injury, brains were harvested for analysis. Overall, our observations revealed that the most significant disruptions were evident in response to LFP, followed by craniotomy only, whereas rmWD animals showed the least residual changes compared with isoflurane-only controls, supporting consideration of rmWD as a mild injury. LFP led to longer-lasting disruptions, perhaps more representative of moderate TBI. We also report that craniotomy and LFP produced greater disruptions in females relative to males. These findings will assist the field in the selection of animal models based on target severity of postinjury outcomes and support the inclusion of both sexes and appropriate control groups.
Collapse
Affiliation(s)
| | - Shealan C Cruise
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | | | | |
Collapse
|
12
|
Carús-Cadavieco M, González de la Fuente S, Berenguer López I, Serrano-Lope MA, Aguado B, Guix F, Palomer E, Dotti CG. Loss of Cldn5 -and increase in Irf7-in the hippocampus and cerebral cortex of diabetic mice at the early symptomatic stage. Nutr Diabetes 2024; 14:64. [PMID: 39147772 PMCID: PMC11327336 DOI: 10.1038/s41387-024-00325-y] [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: 11/17/2023] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/17/2024] Open
Abstract
Analyzing changes in gene expression within specific brain regions of individuals with Type 2 Diabetes (T2DM) who do not exhibit significant cognitive deficits can yield valuable insights into the mechanisms underlying the progression towards a more severe phenotype. In this study, transcriptomic analysis of the cortex and hippocampus of mice with long-term T2DM revealed alterations in the expression of 28 genes in the cerebral cortex and 15 genes in the hippocampus. Among these genes, six displayed consistent changes in both the cortex and hippocampus: Interferon regulatory factor 7 (Irf7), Hypoxia-inducible factor 3 alpha (Hif-3α), period circadian clock 2 (Per2), xanthine dehydrogenase (Xdh), and Transforming growth factor β-stimulated clone 22/TSC22 (Tsc22d3) were upregulated, while Claudin-5 (Cldn5) was downregulated. Confirmation of these changes was achieved through RT-qPCR. At the protein level, CLDN5 and IRF7 exhibited similar alterations, with CLDN5 being downregulated and IRF7 being upregulated. In addition, the hippocampus and cortex of the T2DM mice showed decreased levels of IκBα, implying the involvement of NF-κB pathways as well. Taken together, these results suggest that the weakening of the blood-brain barrier and an abnormal inflammatory response via the Interferon 1 and NF-κB pathways underlie cognitive impairment in individuals with long-standing T2DM.
Collapse
Affiliation(s)
- Marta Carús-Cadavieco
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Madrid, Spain
| | | | - Inés Berenguer López
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Madrid, Spain
| | - Miguel A Serrano-Lope
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Madrid, Spain
| | - Begoña Aguado
- Genomics and NGS Facility, Centro de Biología Molecular Severo Ochoa (CBM) CSIC-UAM, Madrid, Spain
| | - Francesc Guix
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Madrid, Spain
- Department of Bioengineering, Institut Químic de Sarrià (IQS) - Universitat Ramón Llull (URL), Barcelona, Spain
| | - Ernest Palomer
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Madrid, Spain.
| | - Carlos G Dotti
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Madrid, Spain.
| |
Collapse
|
13
|
D'Aversa E, Salvatori F, Vaccarezza M, Antonica B, Grisafi M, Singh AV, Secchiero P, Zauli G, Tisato V, Gemmati D. circRNAs as Epigenetic Regulators of Integrity in Blood-Brain Barrier Architecture: Mechanisms and Therapeutic Strategies in Multiple Sclerosis. Cells 2024; 13:1316. [PMID: 39195206 DOI: 10.3390/cells13161316] [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: 07/03/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 08/29/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory neurodegenerative disease leading to progressive demyelination and neuronal loss, with extensive neurological symptoms. As one of the most widespread neurodegenerative disorders, with an age onset of about 30 years, it turns out to be a socio-health and economic issue, thus necessitating therapeutic interventions currently unavailable. Loss of integrity in the blood-brain barrier (BBB) is one of the distinct MS hallmarks. Brain homeostasis is ensured by an endothelial cell-based monolayer at the interface between the central nervous system (CNS) and systemic bloodstream, acting as a selective barrier. MS results in enhanced barrier permeability, mainly due to the breakdown of tight (TJs) and adherens junctions (AJs) between endothelial cells. Specifically, proinflammatory mediator release causes failure in cytoplasmic exposure of junctions, resulting in compromised BBB integrity that enables blood cells to cross the barrier, establishing iron deposition and neuronal impairment. Cells with a compromised cytoskeletal protein network, fiber reorganization, and discontinuous junction structure can occur, resulting in BBB dysfunction. Recent investigations on spatial transcriptomics have proven circularRNAs (circRNAs) to be powerful multi-functional molecules able to epigenetically regulate transcription and structurally support proteins. In the present review, we provide an overview of the recent role ascribed to circRNAs in maintaining BBB integrity/permeability via cytoskeletal stability. Increased knowledge of the mechanisms responsible for impairment and circRNA's role in driving BBB damage and dysfunction might be helpful for the recognition of novel therapeutic targets to overcome BBB damage and unrestrained neurodegeneration.
Collapse
Affiliation(s)
- Elisabetta D'Aversa
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Francesca Salvatori
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Mauro Vaccarezza
- Curtin Medical School & Curtin Health Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Bianca Antonica
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Miriana Grisafi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Paola Secchiero
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Giorgio Zauli
- Research Department, King Khaled Eye Specialistic Hospital, Riyadh 11462, Saudi Arabia
| | - Veronica Tisato
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- University Strategic Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Donato Gemmati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- University Strategic Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Centre Haemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
14
|
Rajkumar RP. Revisiting a hypothesis: the neurovascular unit as a link between major depression and neurodegenerative disorders. Front Cell Neurosci 2024; 18:1455606. [PMID: 39157756 PMCID: PMC11327082 DOI: 10.3389/fncel.2024.1455606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Affiliation(s)
- Ravi Philip Rajkumar
- Department of Psychiatry, Jawaharlal Institute of Postgraduate Medical Education, Pondicherry, India
| |
Collapse
|
15
|
Melecchi A, Canovai A, Amato R, Dal Monte M, Filippi L, Bagnoli P, Cammalleri M. Agonism of β3-Adrenoceptors Inhibits Pathological Retinal Angiogenesis in the Model of Oxygen-Induced Retinopathy. Invest Ophthalmol Vis Sci 2024; 65:34. [PMID: 39186263 PMCID: PMC11361380 DOI: 10.1167/iovs.65.10.34] [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: 05/03/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024] Open
Abstract
Purpose In response to hypoxia, sympathetic fibers to the retina activate β-adrenoceptors (β-ARs) that play an important role in the regulation of vascular and neuronal functions. We investigated the role of β3-AR using the mouse model of oxygen-induced retinopathy (OIR). Methods Mouse pups were exposed to 75% oxygen at postnatal day 7 (PD7) followed by a return to room air at PD12. The β3-AR preferential agonist BRL37344 was subcutaneously administered once daily at different times after the return to room air. At PD17, the OIR mice underwent flash and pattern electroretinogram. After sacrifice, retinal wholemounts were used for vessel staining or immunohistochemistry for astrocytes, Müller cells, or retinal ganglion cells (RGCs). In retinal homogenates, the levels of markers associated with neovascularization (NV), the blood-retinal barrier (BRB), or astrocytes were determined by western blot, and quantitative reverse-transcription polymerase chain reaction was used to assess β3-AR messenger. Administration of the β3-AR antagonist SR59230A was performed to verify BRL37344 selectivity. Results β3-AR expression is upregulated in response to hypoxia, but its increase is prevented by BRL37344, which counteracts NV by inhibiting the pro-angiogenic pathway, activating the anti-angiogenic pathway, recovering BRB-associated markers, triggering nitric oxide production, and favoring revascularization of the central retina through recovered density of astrocytes that ultimately counteracts NV in the midperiphery. Vasculature rescue prevents dysfunctional retinal activity and counteracts OIR-associated retinal ganglion cell loss. Conclusions β3-AR has emerged as a crucial intermediary in hypoxia-dependent NV, suggesting a role of β3-AR agonists in the treatment of proliferative retinopathies.
Collapse
Affiliation(s)
| | | | - Rosario Amato
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Luca Filippi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Neonatology Unit, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Paola Bagnoli
- Department of Biology, University of Pisa, Pisa, Italy
| | | |
Collapse
|
16
|
Meng L, Zhou M, Wang Y, Pan Y, Chen Z, Wu B, Zhao Y. CD177 on neutrophils engages stress-related behavioral changes in male mice. Brain Behav Immun 2024; 120:403-412. [PMID: 38871062 DOI: 10.1016/j.bbi.2024.06.011] [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: 10/26/2023] [Revised: 05/14/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024] Open
Abstract
Persistent psychological stress can affect immune homeostasis and is a key factor in the development of depression. Many efforts are focused on the identifcation of pathways that link the immune system and mood disorders. Here, we found that psychological stress caused an increase in the frequency of brain-associated neutrophils and the level of neutrophil-specific antigen CD177 on peripheral neutrophils in male mice. Upregulated levels of blood CD177 are associated with depression in humans. Neutrophil depletion or Cd177 deficiency protected mice from stress-induced behavioral deficits. Importantly, adoptive transfer of CD177+ neutrophils from stressed mice increased the frequency of brain-associated leukocytes, including neutrophils, and caused behavioral defects in naive mice. These effects may be related to the endothelial adhesion advantage of CD177+ neutrophils and the interference of serine protease on endothelial junction. Our findings suggest a critical link between circulating CD177+ neutrophils and psychological stress-driven behavioral disorder.
Collapse
Affiliation(s)
- Ling Meng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mi Zhou
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunpeng Wang
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yiming Pan
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Zheng Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Wu
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Yan Zhao
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
17
|
Chen ZB, Zhu XP, Zheng W, Xiang Y, Huang YK, Fang HJ, Deng AJ, Yi FR, Chen HW, Han DQ, Lv SQ. Relationship between the sodium fluorescein yellow fluorescence boundary and the actual boundary of high-grade gliomas during surgical resection. Br J Neurosurg 2024; 38:825-832. [PMID: 34542381 DOI: 10.1080/02688697.2021.1976392] [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: 04/15/2020] [Revised: 07/01/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Resection of high-grade glioma with sodium fluorescein can improve the resection rate of the glioma and improve survival. However, it is unclear whether the yellow fluorescence boundary of the high-grade glioma is consistent with the actual boundary of the tumor. This study explores the yellow fluorescence boundary and the actual tumor boundary in high-grade glioma surgery. METHODS This is a retrospective analysis of 10 patients with high-grade gliomas who underwent tumor visualization with sodium fluorescein. After staining of the tumor, random selections of both developed and non-developed yellow fluorescent border tissue at the fluorescence chromogenic boundary were made, followed by pathological examination. Claudin-5, an important component of the tight connections between vascular endothelial cells, was assessed by immunohistochemistry and qRT-PCR in the tumor and surrounding tissues in order to determine the tumor cell content of the tissue, blood-brain barrier damage, and vascular proliferation. The yellow fluorescence boundary was compared with the actual tumor boundary and the results analyzed. RESULTS Tumor cells were still detected outside the yellow fluorescence boundary during high-grade glioma surgery (P < 0.05). Claudin-5 expression was higher in high-grade gliomas than in adjacent normal tissues (P < 0.05), while disconnected Claudin-5 expression was associated with intraoperative yellow fluorescence imaging (r = 0.67). CONCLUSIONS There is a difference between the yellow fluorescence boundary and the actual boundary of the tumor in high-grade glioma, and there are glioma cell infiltrations in the brain tissue of the undeveloped yellow fluorescent border. To ensure patient recovery and function, it is recommended that tumor resection be expanded based on yellow fluorescence visualization. Claudin-5 is overall up-regulated in high-grade gliomas, but some Claudin-5 expression is disconnected. This Claudin-5 expression pattern may be related to the development of yellow fluorescence.
Collapse
Affiliation(s)
- Ze-Bo Chen
- Department of Neurosurgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, Hunan, China
| | - Xiao-Peng Zhu
- Department of Neurosurgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, Hunan, China
| | - Wei Zheng
- Department of Neurosurgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, Hunan, China
| | - Yan Xiang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yong-Kai Huang
- Department of Neurosurgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, Hunan, China
| | - Hong-Jun Fang
- Department of Neurosurgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, Hunan, China
| | - Ai-Jun Deng
- Department of Neurosurgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, Hunan, China
| | - Fu-Rong Yi
- Department of Neurosurgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, Hunan, China
| | - Hui-Wei Chen
- Department of Neurosurgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, Hunan, China
| | - De-Qing Han
- Department of Neurosurgery, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, Hunan, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
18
|
Gruenbaum BF, Schonwald A, Boyko M, Zlotnik A. The Role of Glutamate and Blood-Brain Barrier Disruption as a Mechanistic Link between Epilepsy and Depression. Cells 2024; 13:1228. [PMID: 39056809 PMCID: PMC11275034 DOI: 10.3390/cells13141228] [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: 06/18/2024] [Revised: 07/10/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Epilepsy is associated with substantial neuropsychiatric impairments that persist long after the onset of the condition, significantly impacting quality of life. The goal of this review was to uncover how the pathological consequences of epilepsy, such as excessive glutamate release and a disrupted blood-brain barrier (BBB), contribute to the emergence of neuropsychiatric disorders. We hypothesize that epilepsy induces a dysfunctional BBB through hyperexcitation, which then further amplifies post-ictal glutamate levels and, thus, triggers neurodegenerative and neuropsychiatric processes. This review identifies the determinants of glutamate concentration levels in the brain and explores potential therapeutic interventions that restore BBB integrity. Our focus on therapeutic BBB restoration is guided by the premise that it may improve glutamate regulation, consequently mitigating the neurotoxicity that contributes to the onset of neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Benjamin F. Gruenbaum
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Matthew Boyko
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel; (M.B.); (A.Z.)
| | - Alexander Zlotnik
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel; (M.B.); (A.Z.)
| |
Collapse
|
19
|
Morys J, Małecki A, Nowacka-Chmielewska M. Stress and the gut-brain axis: an inflammatory perspective. Front Mol Neurosci 2024; 17:1415567. [PMID: 39092201 PMCID: PMC11292226 DOI: 10.3389/fnmol.2024.1415567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/24/2024] [Indexed: 08/04/2024] Open
Abstract
The gut-brain axis (GBA) plays a dominant role in maintaining homeostasis as well as contributes to mental health maintenance. The pathways that underpin the axis expand from macroscopic interactions with the nervous system, to the molecular signals that include microbial metabolites, tight junction protein expression, or cytokines released during inflammation. The dysfunctional GBA has been repeatedly linked to the occurrence of anxiety- and depressive-like behaviors development. The importance of the inflammatory aspects of the altered GBA has recently been highlighted in the literature. Here we summarize current reports on GBA signaling which involves the immune response within the intestinal and blood-brain barrier (BBB). We also emphasize the effect of stress response on altering barriers' permeability, and the therapeutic potential of microbiota restoration by probiotic administration or microbiota transplantation, based on the latest animal studies. Most research performed on various stress models showed an association between anxiety- and depressive-like behaviors, dysbiosis of gut microbiota, and disruption of intestinal permeability with simultaneous changes in BBB integrity. It could be postulated that under stress conditions impaired communication across BBB may therefore represent a significant mechanism allowing the gut microbiota to affect brain functions.
Collapse
Affiliation(s)
| | | | - Marta Nowacka-Chmielewska
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, Katowice, Poland
| |
Collapse
|
20
|
Alshammari MA, Alshehri AO, Alqahtani F, Khan MR, Bakhrebah MA, Alasmari F, Alshammari TK, Alsharari SD. Increased Permeability of the Blood-Brain Barrier in a Diabetic Mouse Model ( Leprdb/db Mice). Int J Mol Sci 2024; 25:7768. [PMID: 39063010 PMCID: PMC11276738 DOI: 10.3390/ijms25147768] [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] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) is linked to multiple complications, including cognitive impairment, and the prevalence of memory-related neurodegenerative diseases is higher in T2DM patients. One possible theory is the alteration of the microvascular and macrovascular environment of the blood-brain barrier (BBB). In this study, we employed different approaches, including RT-PCR, functional pharmacokinetic studies using sodium fluorescein (NaFL), and confocal microscopy, to characterize the functional and molecular integrity of the BBB in a T2DM animal model, leptin receptor-deficient mutant mice (Leprdb/db mice). As a result, VCAM-1, ICAM-1, MMP-9, and S100b (BBB-related markers) dysregulation was observed in the Leprdb/db animal model compared to littermate wild-type mice. The brain concentration of sodium fluorescein (NaFL) increased significantly in Leprdb/db untreated mice compared to insulin-treated mice. Therefore, the permeability of NaFL was higher in Leprdb/db control mice than in all remaining groups. Identifying the factors that increase the BBB in Leprdb/db mice will provide a better understanding of the BBB microvasculature and present previously undescribed findings of T2DM-related brain illnesses, filling knowledge gaps in this emerging field of research.
Collapse
Affiliation(s)
- Musaad A. Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia; (F.A.); (M.R.K.); (F.A.); (T.K.A.); (S.D.A.)
| | - Abdulaziz O. Alshehri
- Department of Pharmacology and Toxicology (Graduate Student), Pharmacy College, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia; (F.A.); (M.R.K.); (F.A.); (T.K.A.); (S.D.A.)
| | - Mohammad R. Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia; (F.A.); (M.R.K.); (F.A.); (T.K.A.); (S.D.A.)
| | - Muhammed A. Bakhrebah
- Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia;
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia; (F.A.); (M.R.K.); (F.A.); (T.K.A.); (S.D.A.)
| | - Tahani K. Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia; (F.A.); (M.R.K.); (F.A.); (T.K.A.); (S.D.A.)
| | - Shakir D. Alsharari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia; (F.A.); (M.R.K.); (F.A.); (T.K.A.); (S.D.A.)
| |
Collapse
|
21
|
Helgudóttir SS, Johnsen KB, Routhe LG, Rasmussen CLM, Thomsen MS, Moos T. Upregulation of Transferrin Receptor 1 (TfR1) but Not Glucose Transporter 1 (GLUT1) or CD98hc at the Blood-Brain Barrier in Response to Valproic Acid. Cells 2024; 13:1181. [PMID: 39056763 PMCID: PMC11275047 DOI: 10.3390/cells13141181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Transferrin receptor 1 (TfR1), glucose transporter 1 (GLUT1), and CD98hc are candidates for targeted therapy at the blood-brain barrier (BBB). Our objective was to challenge the expression of TfR1, GLUT1, and CD98hc in brain capillaries using the histone deacetylase inhibitor (HDACi) valproic acid (VPA). METHODS Primary mouse brain capillary endothelial cells (BCECs) and brain capillaries isolated from mice injected intraperitoneally with VPA were examined using RT-qPCR and ELISA. Targeting to the BBB was performed by injecting monoclonal anti-TfR1 (Ri7217)-conjugated gold nanoparticles measured using ICP-MS. RESULTS In BCECs co-cultured with glial cells, Tfrc mRNA expression was significantly higher after 6 h VPA, returning to baseline after 24 h. In vivo Glut1 mRNA expression was significantly higher in males, but not females, receiving VPA, whereas Cd98hc mRNA expression was unaffected by VPA. TfR1 increased significantly in vivo after VPA, whereas GLUT1 and CD98hc were unchanged. The uptake of anti-TfR1-conjugated nanoparticles was unaltered by VPA despite upregulated TfR expression. CONCLUSIONS VPA upregulates TfR1 in brain endothelium in vivo and in vitro. VPA does not increase GLUT1 and CD98hc proteins. The increase in TfR1 does not result in higher anti-TfR1 antibody targetability, suggesting targeting sufficiently occurs with available transferrin receptors without further contribution from accessory VPA-induced TfR1.
Collapse
Affiliation(s)
- Steinunn Sara Helgudóttir
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark
| | - Kasper Bendix Johnsen
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Lyngby, Denmark
| | - Lisa Greve Routhe
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark
| | | | - Maj Schneider Thomsen
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark
| | - Torben Moos
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark
| |
Collapse
|
22
|
Kumar Nelson V, Jha NK, Nuli MV, Gupta S, Kanna S, Gahtani RM, Hani U, Singh AK, Abomughaid MM, Abomughayedh AM, Almutary AG, Iqbal D, Al Othaim A, Begum SS, Ahmad F, Mishra PC, Jha SK, Ojha S. Unveiling the impact of aging on BBB and Alzheimer's disease: Factors and therapeutic implications. Ageing Res Rev 2024; 98:102224. [PMID: 38346505 DOI: 10.1016/j.arr.2024.102224] [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: 08/29/2023] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 05/12/2024]
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative condition that has devastating effects on individuals, often resulting in dementia. AD is primarily defined by the presence of extracellular plaques containing insoluble β-amyloid peptide (Aβ) and neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau protein (P-tau). In addition, individuals afflicted by these age-related illnesses experience a diminished state of health, which places significant financial strain on their loved ones. Several risk factors play a significant role in the development of AD. These factors include genetics, diet, smoking, certain diseases (such as cerebrovascular diseases, obesity, hypertension, and dyslipidemia), age, and alcohol consumption. Age-related factors are key contributors to the development of vascular-based neurodegenerative diseases such as AD. In general, the process of aging can lead to changes in the immune system's responses and can also initiate inflammation in the brain. The chronic inflammation and the inflammatory mediators found in the brain play a crucial role in the dysfunction of the blood-brain barrier (BBB). Furthermore, maintaining BBB integrity is of utmost importance in preventing a wide range of neurological disorders. Therefore, in this review, we discussed the role of age and its related factors in the breakdown of the blood-brain barrier and the development of AD. We also discussed the importance of different compounds, such as those with anti-aging properties, and other compounds that can help maintain the integrity of the blood-brain barrier in the prevention of AD. This review builds a strong correlation between age-related factors, degradation of the BBB, and its impact on AD.
Collapse
Affiliation(s)
- Vinod Kumar Nelson
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India.
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | - Mohana Vamsi Nuli
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Sandeep Kanna
- Department of pharmaceutics, Chalapathi Institute of Pharmaceutical Sciences, Chalapathi Nagar, Guntur 522034, India
| | - Reem M Gahtani
- Departement of Clinical Laboratory Sciences, King Khalid University, Abha, Saudi Arabia
| | - Umme Hani
- Department of pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Arun Kumar Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ali M Abomughayedh
- Pharmacy Department, Aseer Central Hospital, Ministry of Health, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, P.O. Box 59911, United Arab Emirates
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Ayoub Al Othaim
- Department of Medical Laboratory Sciences, College of Applied Medical Science, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - S Sabarunisha Begum
- Department of Biotechnology, P.S.R. Engineering College, Sivakasi 626140, India
| | - Fuzail Ahmad
- Respiratory Care Department, College of Applied Sciences, Almaarefa University, Diriya, Riyadh, 13713, Saudi Arabia
| | - Prabhu Chandra Mishra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, India.
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, P.O. Box 15551, United Arab Emirates
| |
Collapse
|
23
|
Manukjan N, Chau S, Caiment F, van Herwijnen M, Smeets HJ, Fulton D, Ahmed Z, Blankesteijn WM, Foulquier S. Wnt7a Decreases Brain Endothelial Barrier Function Via β-Catenin Activation. Mol Neurobiol 2024; 61:4854-4867. [PMID: 38147228 PMCID: PMC11236883 DOI: 10.1007/s12035-023-03872-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/11/2023] [Indexed: 12/27/2023]
Abstract
The blood-brain barrier consists of tightly connected endothelial cells protecting the brain's microenvironment from the periphery. These endothelial cells are characterized by specific tight junction proteins such as Claudin-5 and Occludin, forming the endothelial barrier. Disrupting these cells might lead to blood-brain barrier dysfunction. The Wnt/β-catenin signaling pathway can regulate the expression of these tight junction proteins and subsequent barrier permeability. The aim of this study was to investigate the in vitro effects of Wnt7a mediated β-catenin signaling on endothelial barrier integrity. Mouse brain endothelial cells, bEnd.3, were treated with recombinant Wnt7a protein or XAV939, a selective inhibitor of Wnt/β-catenin mediated transcription to modulate the Wnt signaling pathway. The involvement of Wnt/HIF1α signaling was investigated by inhibiting Hif1α signaling with Hif1α siRNA. Wnt7a stimulation led to activation and nuclear translocation of β-catenin, which was inhibited by XAV939. Wnt7a stimulation decreased Claudin-5 expression mediated by β-catenin and decreased endothelial barrier formation. Wnt7a increased Hif1α and Vegfa expression mediated by β-catenin. However, Hif1α signaling pathway did not regulate tight junction proteins Claudin-5 and Occludin. Our data suggest that Wnt7a stimulation leads to a decrease in tight junction proteins mediated by the nuclear translocation of β-catenin, which hampers proper endothelial barrier formation. This process might be crucial in initiating endothelial cell proliferation and angiogenesis. Although HIF1α did not modulate the expression of tight junction proteins, it might play a role in brain angiogenesis and underlie pathogenic mechanisms in Wnt/HIF1α signaling in diseases such as cerebral small vessel disease.
Collapse
Affiliation(s)
- Narek Manukjan
- Department of Pharmacology and Toxicology, Maastricht University, 50 Universiteitssingel, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| | - Steven Chau
- Department of Pharmacology and Toxicology, Maastricht University, 50 Universiteitssingel, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
| | - Florian Caiment
- Department of Toxicogenomics, GROW - School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
| | - Marcel van Herwijnen
- Department of Toxicogenomics, GROW - School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
| | - Hubert J Smeets
- Department of Toxicogenomics, GROW - School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
- MHeNs-School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
| | - Daniel Fulton
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK.
- Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Maastricht University, 50 Universiteitssingel, P.O. Box 616, Maastricht, 6200 MD, The Netherlands.
- CARIM-School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands.
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Maastricht University, 50 Universiteitssingel, P.O. Box 616, Maastricht, 6200 MD, The Netherlands.
- CARIM-School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands.
- MHeNs-School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands.
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, Maastricht, 6202 AZ, The Netherlands.
| |
Collapse
|
24
|
Erramilli SK, Dominik PK, Ogbu CP, Kossiakoff AA, Vecchio AJ. Structural and biophysical insights into targeting of claudin-4 by a synthetic antibody fragment. Commun Biol 2024; 7:733. [PMID: 38886509 PMCID: PMC11183071 DOI: 10.1038/s42003-024-06437-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Claudins are a 27-member family of ~25 kDa membrane proteins that integrate into tight junctions to form molecular barriers at the paracellular spaces between endothelial and epithelial cells. As the backbone of tight junction structure and function, claudins are attractive targets for modulating tissue permeability to deliver drugs or treat disease. However, structures of claudins are limited due to their small sizes and physicochemical properties-these traits also make therapy development a challenge. Here we report the development of a synthetic antibody fragment (sFab) that binds human claudin-4 and the determination of a high-resolution structure of it bound to claudin-4/enterotoxin complexes using cryogenic electron microscopy. Structural and biophysical results reveal this sFabs mechanism of select binding to human claudin-4 over other homologous claudins and establish the ability of sFabs to bind hard-to-target claudins to probe tight junction structure and function. The findings provide a framework for tight junction modulation by sFabs for tissue-selective therapies.
Collapse
Affiliation(s)
- Satchal K Erramilli
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, 60637, USA
| | - Pawel K Dominik
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, 60637, USA
- Pfizer, San Diego, CA, 92121, USA
| | - Chinemerem P Ogbu
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Department of Structural Biology, University at Buffalo, Buffalo, NY, 14203, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, 60637, USA
| | - Alex J Vecchio
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.
- Department of Structural Biology, University at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
25
|
Hladky SB, Barrand MA. Alterations in brain fluid physiology during the early stages of development of ischaemic oedema. Fluids Barriers CNS 2024; 21:51. [PMID: 38858667 PMCID: PMC11163777 DOI: 10.1186/s12987-024-00534-8] [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: 02/01/2024] [Accepted: 03/22/2024] [Indexed: 06/12/2024] Open
Abstract
Oedema occurs when higher than normal amounts of solutes and water accumulate in tissues. In brain parenchymal tissue, vasogenic oedema arises from changes in blood-brain barrier permeability, e.g. in peritumoral oedema. Cytotoxic oedema arises from excess accumulation of solutes within cells, e.g. ischaemic oedema following stroke. This type of oedema is initiated when blood flow in the affected core region falls sufficiently to deprive brain cells of the ATP needed to maintain ion gradients. As a consequence, there is: depolarization of neurons; neural uptake of Na+ and Cl- and loss of K+; neuronal swelling; astrocytic uptake of Na+, K+ and anions; swelling of astrocytes; and reduction in ISF volume by fluid uptake into neurons and astrocytes. There is increased parenchymal solute content due to metabolic osmolyte production and solute influx from CSF and blood. The greatly increased [K+]isf triggers spreading depolarizations into the surrounding penumbra increasing metabolic load leading to increased size of the ischaemic core. Water enters the parenchyma primarily from blood, some passing into astrocyte endfeet via AQP4. In the medium term, e.g. after three hours, NaCl permeability and swelling rate increase with partial opening of tight junctions between blood-brain barrier endothelial cells and opening of SUR1-TPRM4 channels. Swelling is then driven by a Donnan-like effect. Longer term, there is gross failure of the blood-brain barrier. Oedema resolution is slower than its formation. Fluids without colloid, e.g. infused mock CSF, can be reabsorbed across the blood-brain barrier by a Starling-like mechanism whereas infused serum with its colloids must be removed by even slower extravascular means. Large scale oedema can increase intracranial pressure (ICP) sufficiently to cause fatal brain herniation. The potentially lethal increase in ICP can be avoided by craniectomy or by aspiration of the osmotically active infarcted region. However, the only satisfactory treatment resulting in retention of function is restoration of blood flow, providing this can be achieved relatively quickly. One important objective of current research is to find treatments that increase the time during which reperfusion is successful. Questions still to be resolved are discussed.
Collapse
Affiliation(s)
- Stephen B Hladky
- Department of Pharmacology, Tennis Court Rd., Cambridge, CB2 1PD, UK.
| | - Margery A Barrand
- Department of Pharmacology, Tennis Court Rd., Cambridge, CB2 1PD, UK
| |
Collapse
|
26
|
Liao J, Gong L, Xu Q, Wang J, Yang Y, Zhang S, Dong J, Lin K, Liang Z, Sun Y, Mu Y, Chen Z, Lu Y, Zhang Q, Lin Z. Revolutionizing Neurocare: Biomimetic Nanodelivery Via Cell Membranes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402445. [PMID: 38583077 DOI: 10.1002/adma.202402445] [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: 02/16/2024] [Revised: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Brain disorders represent a significant challenge in medical science due to the formidable blood-brain barrier (BBB), which severely limits the penetration of conventional therapeutics, hindering effective treatment strategies. This review delves into the innovative realm of biomimetic nanodelivery systems, including stem cell-derived nanoghosts, tumor cell membrane-coated nanoparticles, and erythrocyte membrane-based carriers, highlighting their potential to circumvent the BBB's restrictions. By mimicking native cell properties, these nanocarriers emerge as a promising solution for enhancing drug delivery to the brain, offering a strategic advantage in overcoming the barrier's selective permeability. The unique benefits of leveraging cell membranes from various sources is evaluated and advanced technologies for fabricating cell membrane-encapsulated nanoparticles capable of masquerading as endogenous cells are examined. This enables the targeted delivery of a broad spectrum of therapeutic agents, ranging from small molecule drugs to proteins, thereby providing an innovative approach to neurocare. Further, the review contrasts the capabilities and limitations of these biomimetic nanocarriers with traditional delivery methods, underlining their potential to enable targeted, sustained, and minimally invasive treatment modalities. This review is concluded with a perspective on the clinical translation of these biomimetic systems, underscoring their transformative impact on the therapeutic landscape for intractable brain diseases.
Collapse
Affiliation(s)
- Jun Liao
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Lidong Gong
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Qingqiang Xu
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Jingya Wang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yuanyuan Yang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Shiming Zhang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Junwei Dong
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Kerui Lin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Zichao Liang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yuhan Sun
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Yongxu Mu
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014040, China
| | - Zhengju Chen
- Pooling Medical Research Institutes of 100Biotech, Beijing, 100006, China
| | - Ying Lu
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Qiang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zhiqiang Lin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
27
|
Begley D, Gabathuler R, Pastores G, Garcia-Cazorla A, Ardigò D, Scarpa M, Tomanin R, Tosi G. Challenges and opportunities in neurometabolic disease treatment with enzyme delivery. Expert Opin Drug Deliv 2024; 21:817-828. [PMID: 38963225 DOI: 10.1080/17425247.2024.2375388] [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: 12/30/2023] [Accepted: 06/28/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Neurometabolic disorders remain challenging to treat, largely due to the limited availability of drugs that can cross the blood-brain barrier (BBB) and effectively target brain impairment. Key reasons for inadequate treatment include a lack of coordinated knowledge, few studies on BBB status in these diseases, and poorly designed therapies. AREAS COVERED This paper provides an overview of current research on neurometabolic disorders and therapeutic options, focusing on the treatment of neurological involvement. It highlights the limitations of existing therapies, describes innovative protocols recently developed, and explores new opportunities for therapy design and testing, some of which are already under investigation. The goal is to guide researchers toward innovative and potentially more effective treatments. EXPERT OPINION Advancing research on neurometabolic diseases is crucial for designing effective treatment strategies. The field suffers from a lack of collaboration, and a strong collective effort is needed to enhance synergy, increase knowledge, and develop a new therapeutic paradigm for neurometabolic disorders.
Collapse
Affiliation(s)
- David Begley
- Blood-Brain Barrier Group, King's College London, Strand, London, UK
| | | | | | - Angeles Garcia-Cazorla
- Neurometabolic Unit. Department of Neurology, Hospital Sant Joan de Déu, CIBERER and MetabERN, Barcelona, Spain
| | | | - Maurizio Scarpa
- Regional Coordinating Center for Rare Diseases, Udine University Hospital, Udine, Italy
| | - Rosella Tomanin
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Dept. of Women's and Children's Health, University of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Giovanni Tosi
- Nanotech Lab, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
28
|
Shuvalova M, Dmitrieva A, Belousov V, Nosov G. The role of reactive oxygen species in the regulation of the blood-brain barrier. Tissue Barriers 2024:2361202. [PMID: 38808582 DOI: 10.1080/21688370.2024.2361202] [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: 02/28/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
The blood-brain barrier (BBB) regulates the exchange of metabolites and cells between the blood and brain, and maintains central nervous system homeostasis. Various factors affect BBB barrier functions, including reactive oxygen species (ROS). ROS can act as stressors, damaging biological molecules, but they also serve as secondary messengers in intracellular signaling cascades during redox signaling. The impact of ROS on the BBB has been observed in multiple sclerosis, stroke, trauma, and other neurological disorders, making blocking ROS generation a promising therapeutic strategy for BBB dysfunction. However, it is important to consider ROS generation during normal BBB functioning for signaling purposes. This review summarizes data on proteins expressed by BBB cells that can be targets of redox signaling or oxidative stress. It also provides examples of signaling molecules whose impact may cause ROS generation in the BBB, as well as discusses the most common diseases associated with BBB dysfunction and excessive ROS generation, open questions that arise in the study of this problem, and possible ways to overcome them.
Collapse
Affiliation(s)
- Margarita Shuvalova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anastasiia Dmitrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vsevolod Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| | - Georgii Nosov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| |
Collapse
|
29
|
Li Y, Yu Q, Peng H, Mingjun X, Xu W, Zheng T, Zhao T, Xia M, Wu J, Stavrinou P, Goldbrunner R, Xie Y, Zhang G, Feng Y, Guan Y, Zheng F, Sun P. Jingfang granules protects against intracerebral hemorrhage by inhibiting neuroinflammation and protecting blood-brain barrier damage. Aging (Albany NY) 2024; 16:9023-9046. [PMID: 38809507 PMCID: PMC11164481 DOI: 10.18632/aging.205854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 04/10/2024] [Indexed: 05/30/2024]
Abstract
Intracerebral hemorrhage (ICH) can induce intensive oxidative stress, neuroinflammation, and brain cell apoptosis. However, conventional methods for ICH treatment have many disadvantages. There is an urgent need for alternative, effective therapies with minimal side effects. Pharmacodynamics experiment, molecular docking, network pharmacology, and metabolomics were adopted to investigate the treatment and its mechanism of Jingfang Granules (JFG) in ICH. In this study, we investigated the therapeutic effects of JFG on ICH using behavioral, brain water content and Magnetic resonance imaging experiments. However, the key active component and targets of JFG remain unknown. Here we verified that JFG was beneficial to improve brain injury after ICH. A network pharmacology analysis revealed that the anti-inflammatory effect of JFG is predominantly mediated by its activation of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway through Luteolin, (+)-Anomalin and Phaseol and their targeting of AKT1, tumor necrosis factorα (TNF-α), and interleukin-1β (IL-1β). Molecular docking analyses revealed an average affinity of -8.633 kcal/mol, indicating a binding strength of less than -5 kcal/mol. Metabolomic analysis showed that JFG exerted its therapeutic effect on ICH by regulating metabolic pathways, such as the metabolism of taurine and hypotaurine, biosynthesis of valine, leucine, and isoleucine. In conclusion, we demonstrated that JFG attenuated neuroinflammation and BBB injury subsequent to ICH by activating the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Yanling Li
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Qingying Yu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huiyuan Peng
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine, Zhong Shan, China
| | - Xie Mingjun
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - WenHua Xu
- Prevention and Treatment Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Tingting Zheng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Tingting Zhao
- Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Mengyao Xia
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Jibiao Wu
- Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Pantelis Stavrinou
- Department of Neurosurgery, Center for Neurosurgery, Faculty of Medicine and University Hospital, University of Cologne, Cologne, Germany
| | - Roland Goldbrunner
- Department of Neurosurgery, Center for Neurosurgery, Faculty of Medicine and University Hospital, University of Cologne, Cologne, Germany
| | - Yicheng Xie
- The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Guimin Zhang
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Yu Feng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Yongxia Guan
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Feng Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Peng Sun
- Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| |
Collapse
|
30
|
Porkoláb G, Mészáros M, Szecskó A, Vigh JP, Walter FR, Figueiredo R, Kálomista I, Hoyk Z, Vizsnyiczai G, Gróf I, Jan JS, Gosselet F, Pirity MK, Vastag M, Hudson N, Campbell M, Veszelka S, Deli MA. Synergistic induction of blood-brain barrier properties. Proc Natl Acad Sci U S A 2024; 121:e2316006121. [PMID: 38748577 PMCID: PMC11126970 DOI: 10.1073/pnas.2316006121] [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: 09/14/2023] [Accepted: 04/05/2024] [Indexed: 05/27/2024] Open
Abstract
Blood-brain barrier (BBB) models derived from human stem cells are powerful tools to improve our understanding of cerebrovascular diseases and to facilitate drug development for the human brain. Yet providing stem cell-derived endothelial cells with the right signaling cues to acquire BBB characteristics while also retaining their vascular identity remains challenging. Here, we show that the simultaneous activation of cyclic AMP and Wnt/β-catenin signaling and inhibition of the TGF-β pathway in endothelial cells robustly induce BBB properties in vitro. To target this interaction, we present a small-molecule cocktail named cARLA, which synergistically enhances barrier tightness in a range of BBB models across species. Mechanistically, we reveal that the three pathways converge on Wnt/β-catenin signaling to mediate the effect of cARLA via the tight junction protein claudin-5. We demonstrate that cARLA shifts the gene expressional profile of human stem cell-derived endothelial cells toward the in vivo brain endothelial signature, with a higher glycocalyx density and efflux pump activity, lower rates of endocytosis, and a characteristic endothelial response to proinflammatory cytokines. Finally, we illustrate how cARLA can improve the predictive value of human BBB models regarding the brain penetration of drugs and targeted nanoparticles. Due to its synergistic effect, high reproducibility, and ease of use, cARLA has the potential to advance drug development for the human brain by improving BBB models across laboratories.
Collapse
Affiliation(s)
- Gergő Porkoláb
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
- Doctoral School of Biology, University of Szeged, SzegedH-6720, Hungary
| | - Mária Mészáros
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Anikó Szecskó
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
- Doctoral School of Biology, University of Szeged, SzegedH-6720, Hungary
| | - Judit P. Vigh
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
- Doctoral School of Biology, University of Szeged, SzegedH-6720, Hungary
| | - Fruzsina R. Walter
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | | | - Ildikó Kálomista
- In Vitro Metabolism Laboratory, Gedeon Richter, BudapestH-1103, Hungary
| | - Zsófia Hoyk
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Gaszton Vizsnyiczai
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Ilona Gróf
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Jeng-Shiung Jan
- Department of Chemical Engineering, National Cheng Kung University, Tainan70101, Taiwan
| | - Fabien Gosselet
- Laboratoire de la Barriére Hémato-Encéphalique, Université d’Artois, Lens62307, France
| | - Melinda K. Pirity
- Institute of Genetics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Monika Vastag
- In Vitro Metabolism Laboratory, Gedeon Richter, BudapestH-1103, Hungary
| | - Natalie Hudson
- Smurfit Institute of Genetics, Trinity College Dublin, DublinD02 VF25, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, DublinD02 VF25, Ireland
| | - Szilvia Veszelka
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Mária A. Deli
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| |
Collapse
|
31
|
Varghese SM, Patel S, Nandan A, Jose A, Ghosh S, Sah RK, Menon B, K V A, Chakravarty S. Unraveling the Role of the Blood-Brain Barrier in the Pathophysiology of Depression: Recent Advances and Future Perspectives. Mol Neurobiol 2024:10.1007/s12035-024-04205-5. [PMID: 38730081 DOI: 10.1007/s12035-024-04205-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 04/19/2024] [Indexed: 05/12/2024]
Abstract
Depression is a highly prevalent psychological disorder characterized by persistent dysphoria, psychomotor retardation, insomnia, anhedonia, suicidal ideation, and a remarkable decrease in overall well-being. Despite the prevalence of accessible antidepressant therapies, many individuals do not achieve substantial improvement. Understanding the multifactorial pathophysiology and the heterogeneous nature of the disorder could lead the way toward better outcomes. Recent findings have elucidated the substantial impact of compromised blood-brain barrier (BBB) integrity on the manifestation of depression. BBB functions as an indispensable defense mechanism, tightly overseeing the transport of molecules from the periphery to preserve the integrity of the brain parenchyma. The dysfunction of the BBB has been implicated in a multitude of neurological disorders, and its disruption and consequent brain alterations could potentially serve as important factors in the pathogenesis and progression of depression. In this review, we extensively examine the pathophysiological relevance of the BBB and delve into the specific modifications of its components that underlie the complexities of depression. A particular focus has been placed on examining the effects of peripheral inflammation on the BBB in depression and elucidating the intricate interactions between the gut, BBB, and brain. Furthermore, this review encompasses significant updates on the assessment of BBB integrity and permeability, providing a comprehensive overview of the topic. Finally, we outline the therapeutic relevance and strategies based on BBB in depression, including COVID-19-associated BBB disruption and neuropsychiatric implications. Understanding the comprehensive pathogenic cascade of depression is crucial for shaping the trajectory of future research endeavors.
Collapse
Affiliation(s)
- Shamili Mariya Varghese
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Shashikant Patel
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad, Telangana, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Amritasree Nandan
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Anju Jose
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Soumya Ghosh
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad, Telangana, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ranjay Kumar Sah
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Bindu Menon
- Department of Psychiatry, Amrita School of Medicine, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Athira K V
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India.
| | - Sumana Chakravarty
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad, Telangana, 500007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
32
|
Sun C, Wang S, Ma Z, Zhou J, Ding Z, Yuan G, Pan Y. Neutrophils in glioma microenvironment: from immune function to immunotherapy. Front Immunol 2024; 15:1393173. [PMID: 38779679 PMCID: PMC11109384 DOI: 10.3389/fimmu.2024.1393173] [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: 02/28/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Glioma is a malignant tumor of the central nervous system (CNS). Currently, effective treatment options for gliomas are still lacking. Neutrophils, as an important member of the tumor microenvironment (TME), are widely distributed in circulation. Recently, the discovery of cranial-meningeal channels and intracranial lymphatic vessels has provided new insights into the origins of neutrophils in the CNS. Neutrophils in the brain may originate more from the skull and adjacent vertebral bone marrow. They cross the blood-brain barrier (BBB) under the action of chemokines and enter the brain parenchyma, subsequently migrating to the glioma TME and undergoing phenotypic changes upon contact with tumor cells. Under glycolytic metabolism model, neutrophils show complex and dual functions in different stages of cancer progression, including participation in the malignant progression, immune suppression, and anti-tumor effects of gliomas. Additionally, neutrophils in the TME interact with other immune cells, playing a crucial role in cancer immunotherapy. Targeting neutrophils may be a novel generation of immunotherapy and improve the efficacy of cancer treatments. This article reviews the molecular mechanisms of neutrophils infiltrating the central nervous system from the external environment, detailing the origin, functions, classifications, and targeted therapies of neutrophils in the context of glioma.
Collapse
Affiliation(s)
- Chao Sun
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Siwen Wang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Zhen Ma
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Jinghuan Zhou
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Zilin Ding
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Guoqiang Yuan
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yawen Pan
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
33
|
Duclos S, Choi SW, Andjelkovic AV, Chaudhary N, Camelo-Piragua S, Pandey A, Xu Z. Characterization of Blood-Brain Barrier Opening Induced by Transcranial Histotripsy in Murine Brains. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:639-646. [PMID: 38302370 DOI: 10.1016/j.ultrasmedbio.2023.12.014] [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: 10/18/2022] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 02/03/2024]
Abstract
OBJECTIVE Transcranial histotripsy has shown promise as a non-invasive neurosurgical tool, as it has the ability to treat a wide range of locations in the brain without overheating the skull. One important effect of histotripsy in the brain is the blood-brain barrier (BBB) opening (BBBO) at the ablation site, but there is a knowledge gap concerning the extent of histotripsy-induced BBBO. Here we describe induction of BBBO by transcranial histotripsy and use of magnetic resonance imaging (MRI) and histology to quantify changes in BBBO at the periphery of the histotripsy ablation zone over time in the healthy mouse brain. METHODS An eight-element, 1 MHz histotripsy transducer with a focal distance of 32.5 mm was used to treat the brains of 23 healthy female BL6 mice. T1-gadolinium (T1-Gd) MR images were acquired immediately following histotripsy treatment and during each of the subsequent 4 wk to quantify the size and intensity of BBB leakage. RESULTS The T1-Gd MRI results revealed that the hyperintense BBBO volume increased over the first week and subsided gradually over the following 3 wk. Histology revealed complete loss of tight junction proteins and blood vessels in the center of the ablation region immediately after histotripsy, partial recovery in the periphery of the ablation zone 1 wk following histotripsy and near-complete recovery of tight junction complex after 4 wk. CONCLUSION These results provide the first evidence of transcranial histotripsy-induced BBBO and repair at the periphery of the ablation zone.
Collapse
Affiliation(s)
- Sarah Duclos
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| | - Sang Won Choi
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Anuska V Andjelkovic
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Neeraj Chaudhary
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA; Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | | | - Aditya Pandey
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA; Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Zhen Xu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
34
|
Ke X, Xia S, Yu W, Mabry S, Fu Q, Menden HL, Sampath V, Lane RH. Delta like 4 regulates cerebrovascular development and endothelial integrity via DLL4-NOTCH-CLDN5 pathway and is vulnerable to neonatal hyperoxia. J Physiol 2024; 602:2265-2285. [PMID: 38632887 DOI: 10.1113/jp285716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
The mechanisms governing brain vascularization during development remain poorly understood. A key regulator of developmental vascularization is delta like 4 (DLL4), a Notch ligand prominently expressed in endothelial cells (EC). Exposure to hyperoxia in premature infants can disrupt the development and functions of cerebral blood vessels and lead to long-term cognitive impairment. However, its role in cerebral vascular development and the impact of postnatal hyperoxia on DLL4 expression in mouse brain EC have not been explored. We determined the DLL4 expression pattern and its downstream signalling gene expression in brain EC using Dll4+/+ and Dll4+/LacZ mice. We also performed in vitro studies using human brain microvascular endothelial cells. Finally, we determined Dll4 and Cldn5 expression in mouse brain EC exposed to postnatal hyperoxia. DLL4 is expressed in various cell types, with EC being the predominant one in immature brains. Moreover, DLL4 deficiency leads to persistent abnormalities in brain microvasculature and increased vascular permeability both in vivo and in vitro. We have identified that DLL4 insufficiency compromises endothelial integrity through the NOTCH-NICD-RBPJ-CLDN5 pathway, resulting in the downregulation of the tight junction protein claudin 5 (CLDN5). Finally, exposure to neonatal hyperoxia reduces DLL4 and CLDN5 expression in developing mouse brain EC. We reveal that DLL4 is indispensable for brain vascular development and maintaining the blood-brain barrier's function and is repressed by neonatal hyperoxia. We speculate that reduced DLL4 signalling in brain EC may contribute to the impaired brain development observed in neonates exposed to hyperoxia. KEY POINTS: The role of delta like 4 (DLL4), a Notch ligand in vascular endothelial cells, in brain vascular development and functions remains unknown. We demonstrate that DLL4 is expressed at a high level during postnatal brain development in immature brains and DLL4 insufficiency leads to abnormal cerebral vasculature and increases vascular permeability both in vivo and in vitro. We identify that DLL4 regulates endothelial integrity through NOTCH-NICD-RBPJ-CLDN5 signalling. Dll4 and Cldn5 expression are decreased in mouse brain endothelial cells exposed to postnatal hyperoxia.
Collapse
Affiliation(s)
- Xingrao Ke
- Department of Pediatrics Division of Neonatology, Children's Mercy, Kansas City, MO, USA
| | - Sheng Xia
- Department of Pediatrics Division of Neonatology, Children's Mercy, Kansas City, MO, USA
| | - Wei Yu
- Department of Pediatrics Division of Neonatology, Children's Mercy, Kansas City, MO, USA
| | - Sherry Mabry
- Department of Pediatrics Division of Neonatology, Children's Mercy, Kansas City, MO, USA
| | - Qi Fu
- Department of Pediatrics Division of Neonatology, Children's Mercy, Kansas City, MO, USA
| | - Heather L Menden
- Department of Pediatrics Division of Neonatology, Children's Mercy, Kansas City, MO, USA
| | - Venkatesh Sampath
- Department of Pediatrics Division of Neonatology, Children's Mercy, Kansas City, MO, USA
| | - Robert H Lane
- Department of Administration, Children Mercy Research Institute, Children's Mercy, Kansas City, MO, USA
| |
Collapse
|
35
|
Hana C, Thaw Dar NN, Galo Venegas M, Vulfovich M. Claudins in Cancer: A Current and Future Therapeutic Target. Int J Mol Sci 2024; 25:4634. [PMID: 38731853 PMCID: PMC11083183 DOI: 10.3390/ijms25094634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/13/2024] Open
Abstract
Claudins are a family of 27 proteins that have an important role in the formation of tight junctions. They also have an important function in ion exchange, cell mobility, and the epithelial-to-mesenchymal transition, the latter being very important in cancer invasion and metastasis. Therapeutic targeting of claudins has been investigated to improve cancer outcomes. Recent evidence shows improved outcomes when combining monoclonal antibodies against claudin 18.2 with chemotherapy for patients with gastroesophageal junction cancer. Currently, chimeric antigen receptor T-cells targeting claudin 18 are under investigation. In this review, we will discuss the major functions of claudins, their distribution in the normal as well as cancerous tissues, and their effect in cancer metastasis, with a special focus on the therapeutic targeting of claudins to improve cancer outcomes.
Collapse
Affiliation(s)
- Caroline Hana
- Hematology/Oncology Department, Memorial Healthcare System, Pembroke Pines, FL 33028, USA; (N.N.T.D.); (M.G.V.)
| | | | | | | |
Collapse
|
36
|
Chen L, Song J, Richter-Stretton G, Lee W, Padmanabhan P, Götz J. Multimodal evaluation of blood-brain barrier opening in mice in response to low-intensity ultrasound and a claudin-5 binder. Nanotheranostics 2024; 8:427-441. [PMID: 38961889 PMCID: PMC11217787 DOI: 10.7150/ntno.95146] [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: 02/07/2024] [Accepted: 04/05/2024] [Indexed: 07/05/2024] Open
Abstract
Background: The blood-brain barrier (BBB) is a major bottleneck in delivering therapeutics to the brain. Treatment strategies to transiently open this barrier include focused ultrasound combined with intravenously injected microbubbles (FUS+MB) and targeting of molecules that regulate BBB permeability. Methods: Here, we investigated BBB opening mediated by the claudin-5 binder cCPEm (a microorganismal toxin in a truncated form) and FUS+MB at a centre frequency of 1 MHz, assessing dextran uptake, broadband emission, and endogenous immunoglobulin G (IgG) extravasation. Results: FUS+MB-induced BBB opening was detectable at a pressure ≥0.35 MPa when assessed for leakage of 10 and 70 kDa dextran, and at ≥0.2 MPa for uptake of endogenous IgG. Treating mice with 20 mg/kg cCPEm failed to open the BBB, and pre-treatment with cCPEm followed by FUS+MB at 0.2 and 0.3 MPa did not overtly increase BBB opening compared to FUS+MB alone. Using passive cavitation detection (PCD), we found that broadband emission correlated with the peak negative pressure (PNP) and dextran leakage, indicating the possibility of using broadband emission for developing a feedback controller to monitor BBB opening. Conclusions: Together, our study highlights the challenges in developing combinatorial approaches to open the BBB and presents an additional IgG-based histological detection method for BBB opening.
Collapse
Affiliation(s)
| | | | | | | | | | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane QLD 4072, Australia
| |
Collapse
|
37
|
Zapata-Acevedo JF, Mantilla-Galindo A, Vargas-Sánchez K, González-Reyes RE. Blood-brain barrier biomarkers. Adv Clin Chem 2024; 121:1-88. [PMID: 38797540 DOI: 10.1016/bs.acc.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The blood-brain barrier (BBB) is a dynamic interface that regulates the exchange of molecules and cells between the brain parenchyma and the peripheral blood. The BBB is mainly composed of endothelial cells, astrocytes and pericytes. The integrity of this structure is essential for maintaining brain and spinal cord homeostasis and protection from injury or disease. However, in various neurological disorders, such as traumatic brain injury, Alzheimer's disease, and multiple sclerosis, the BBB can become compromised thus allowing passage of molecules and cells in and out of the central nervous system parenchyma. These agents, however, can serve as biomarkers of BBB permeability and neuronal damage, and provide valuable information for diagnosis, prognosis and treatment. Herein, we provide an overview of the BBB and changes due to aging, and summarize current knowledge on biomarkers of BBB disruption and neurodegeneration, including permeability, cellular, molecular and imaging biomarkers. We also discuss the challenges and opportunities for developing a biomarker toolkit that can reliably assess the BBB in physiologic and pathophysiologic states.
Collapse
Affiliation(s)
- Juan F Zapata-Acevedo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Alejandra Mantilla-Galindo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Karina Vargas-Sánchez
- Laboratorio de Neurofisiología Celular, Grupo de Neurociencia Traslacional, Facultad de Medicina, Universidad de los Andes, Bogotá, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
38
|
Arkelius K, Wendt TS, Andersson H, Arnou A, Gottschalk M, Gonzales RJ, Ansar S. LOX-1 and MMP-9 Inhibition Attenuates the Detrimental Effects of Delayed rt-PA Therapy and Improves Outcomes After Acute Ischemic Stroke. Circ Res 2024; 134:954-969. [PMID: 38501247 DOI: 10.1161/circresaha.123.323371] [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: 08/08/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Acute ischemic stroke triggers endothelial activation that disrupts vascular integrity and increases hemorrhagic transformation leading to worsened stroke outcomes. rt-PA (recombinant tissue-type plasminogen activator) is an effective treatment; however, its use is limited due to a restricted time window and hemorrhagic transformation risk, which in part may involve activation of MMPs (matrix metalloproteinases) mediated through LOX-1 (lectin-like oxLDL [oxidized low-density lipoprotein] receptor 1). This study's overall aim was to evaluate the therapeutic potential of novel MMP-9 (matrix metalloproteinase 9) ± LOX-1 inhibitors in combination with rt-PA to improve stroke outcomes. METHODS A rat thromboembolic stroke model was utilized to investigate the impact of rt-PA delivered 4 hours poststroke onset as well as selective MMP-9 (JNJ0966) ±LOX-1 (BI-0115) inhibitors given before rt-PA administration. Infarct size, perfusion, and hemorrhagic transformation were evaluated by 9.4-T magnetic resonance imaging, vascular and parenchymal MMP-9 activity via zymography, and neurological function was assessed using sensorimotor function testing. Human brain microvascular endothelial cells were exposed to hypoxia plus glucose deprivation/reperfusion (hypoxia plus glucose deprivation 3 hours/R 24 hours) and treated with ±tPA and ±MMP-9 ±LOX-1 inhibitors. Barrier function was assessed via transendothelial electrical resistance, MMP-9 activity was determined with zymography, and LOX-1 and barrier gene expression/levels were measured using qRT-PCR (quantitative reverse transcription PCR) and Western blot. RESULTS Stroke and subsequent rt-PA treatment increased edema, hemorrhage, MMP-9 activity, LOX-1 expression, and worsened neurological outcomes. LOX-1 inhibition improved neurological function, reduced edema, and improved endothelial barrier integrity. Elevated MMP-9 activity correlated with increased edema, infarct volume, and decreased neurological function. MMP-9 inhibition reduced MMP-9 activity and LOX-1 expression. In human brain microvascular endothelial cells, LOX-1/MMP-9 inhibition differentially attenuated MMP-9 levels, inflammation, and activation following hypoxia plus glucose deprivation/R. CONCLUSIONS Our findings indicate that LOX-1 inhibition and ± MMP-9 inhibition attenuate negative aspects of ischemic stroke with rt-PA therapy, thus resulting in improved neurological function. While no synergistic effect was observed with simultaneous LOX-1 and MMP-9 inhibition, a distinct interaction is evident.
Collapse
Affiliation(s)
- Kajsa Arkelius
- Applied Neurovascular Research, Neurosurgery, Department of Clinical Sciences, Lund University, Sweden (K.A., H.A., A.A., S.A.)
| | - Trevor S Wendt
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ (T.S.W., R.J.G.)
| | - Henrik Andersson
- Applied Neurovascular Research, Neurosurgery, Department of Clinical Sciences, Lund University, Sweden (K.A., H.A., A.A., S.A.)
| | - Anaële Arnou
- Applied Neurovascular Research, Neurosurgery, Department of Clinical Sciences, Lund University, Sweden (K.A., H.A., A.A., S.A.)
| | | | - Rayna J Gonzales
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ (T.S.W., R.J.G.)
| | - Saema Ansar
- Applied Neurovascular Research, Neurosurgery, Department of Clinical Sciences, Lund University, Sweden (K.A., H.A., A.A., S.A.)
| |
Collapse
|
39
|
Crockett AM, Kebir H, Anderson SA, Jyonouchi S, Romberg N, Alvarez JI. 22q11.2 Deletion-Associated Blood-Brain Barrier Permeability Potentiates Systemic Capillary Leak Syndrome Neurologic Features. J Clin Immunol 2024; 44:87. [PMID: 38578402 PMCID: PMC11490314 DOI: 10.1007/s10875-024-01686-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/06/2024] [Indexed: 04/06/2024]
Abstract
We present a case study of a young male with a history of 22q11.2 deletion syndrome (22qDS), diagnosed with systemic capillary leak syndrome (SCLS) who presented with acute onset of diffuse anasarca and sub-comatose obtundation. We hypothesized that his co-presentation of neurological sequelae might be due to blood-brain barrier (BBB) susceptibility conferred by the 22q11.2 deletion, a phenotype that we have previously identified in 22qDS. Using pre- and post-intravenous immunoglobulins (IVIG) patient serum, we studied circulating biomarkers of inflammation and assessed the potential susceptibility of the 22qDS BBB. We employed in vitro cultures of differentiated BBB-like endothelial cells derived from a 22qDS patient and a healthy control. We found evidence of peripheral inflammation and increased serum lipopolysaccharide (LPS) alongside endothelial cells in circulation. We report that the patient's serum significantly impairs barrier function of the 22qDS BBB compared to control. Only two other cases of pediatric SCLS with neurologic symptoms have been reported, and genetic risk factors have been suggested in both instances. As the third case to be reported, our findings are consistent with the hypothesis that genetic susceptibility of the BBB conferred by genes such as claudin-5 deleted in the 22q11.2 region promoted neurologic involvement during SCLS in this patient.
Collapse
Affiliation(s)
- Alexis M Crockett
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, 412 Hill, Philadelphia, PA, 19104-4539, USA
| | - Hania Kebir
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, 412 Hill, Philadelphia, PA, 19104-4539, USA
| | - Stewart A Anderson
- Department of Child and Adolescent Psychiatry, Children's Hospital of Philadelphia, Philadelphia, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Soma Jyonouchi
- Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Neil Romberg
- Division of Immunology and Allergy, Children's Hospital of Philadelphia, Philadelphia, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Jorge I Alvarez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, 412 Hill, Philadelphia, PA, 19104-4539, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
40
|
Wu X, Yuan R, Xu Y, Wang K, Yuan H, Meng T, Hu F. Functionalized lipid nanoparticles modulate the blood-brain barrier and eliminate α-synuclein to repair dopamine neurons. Asian J Pharm Sci 2024; 19:100904. [PMID: 38601010 PMCID: PMC11004078 DOI: 10.1016/j.ajps.2024.100904] [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: 08/09/2023] [Revised: 01/16/2024] [Accepted: 02/17/2024] [Indexed: 04/12/2024] Open
Abstract
The challenge in the clinical treatment of Parkinson's disease lies in the lack of disease-modifying therapies that can halt or slow down the progression. Peptide drugs, such as exenatide (Exe), with potential disease-modifying efficacy, have difficulty in crossing the blood-brain barrier (BBB) due to their large molecular weight. Herein, we fabricate multi-functionalized lipid nanoparticles (LNP) Lpc-BoSA/CSO with BBB targeting, permeability-increasing and responsive release functions. Borneol is chemically bonded with stearic acid and, as one of the components of Lpc-BoSA/CSO, is used to increase BBB permeability. Immunofluorescence results of brain tissue of 15-month-old C57BL/6 mice show that Lpc-BoSA/CSO disperses across the BBB into brain parenchyma, and the amount is 4.21 times greater than that of conventional LNP. Motor symptoms of mice in Lpc-BoSA/CSO-Exe group are significantly improved, and the content of dopamine is 1.85 times (substantia nigra compacta) and 1.49 times (striatum) that of PD mice. α-Synuclein expression and Lewy bodies deposition are reduced to 51.85% and 44.72% of PD mice, respectively. Immunohistochemical mechanism studies show AKT expression in Lpc-BoSA/CSO-Exe is 4.23 times that of PD mice and GSK-3β expression is reduced to 18.41%. Lpc-BoSA/CSO-Exe could reduce the production of α-synuclein and Lewy bodies through AKT/GSK-3β pathway, and effectively prevent the progressive deterioration of Parkinson's disease. In summary, Lpc-BoSA/CSO-Exe increases the entry of exenatide into brain and promotes its clinical application for Parkinson's disease therapy.
Collapse
Affiliation(s)
- Xiaomei Wu
- Department of Pharmacy, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200000, China
| | - Renxiang Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Yichong Xu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Kai Wang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| |
Collapse
|
41
|
Hu Y, Zhang F, Ikonomovic M, Yang T. The Role of NRF2 in Cerebrovascular Protection: Implications for Vascular Cognitive Impairment and Dementia (VCID). Int J Mol Sci 2024; 25:3833. [PMID: 38612642 PMCID: PMC11012233 DOI: 10.3390/ijms25073833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Vascular cognitive impairment and dementia (VCID) represents a broad spectrum of cognitive decline secondary to cerebral vascular aging and injury. It is the second most common type of dementia, and the prevalence continues to increase. Nuclear factor erythroid 2-related factor 2 (NRF2) is enriched in the cerebral vasculature and has diverse roles in metabolic balance, mitochondrial stabilization, redox balance, and anti-inflammation. In this review, we first briefly introduce cerebrovascular aging in VCID and the NRF2 pathway. We then extensively discuss the effects of NRF2 activation in cerebrovascular components such as endothelial cells, vascular smooth muscle cells, pericytes, and perivascular macrophages. Finally, we summarize the clinical potential of NRF2 activators in VCID.
Collapse
Affiliation(s)
- Yizhou Hu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Department of Internal Medicine, University of Pittsburgh Medical Center (UPMC) McKeesport, McKeesport, PA 15132, USA
| | - Feng Zhang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Milos Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Tuo Yang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Department of Internal Medicine, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15216, USA
| |
Collapse
|
42
|
Vázquez-Liébanas E, Mocci G, Li W, Laviña B, Reddy A, O'Connor C, Hudson N, Elbeck Z, Nikoloudis I, Gaengel K, Vanlandewijck M, Campbell M, Betsholtz C, Mäe MA. Mosaic deletion of claudin-5 reveals rapid non-cell-autonomous consequences of blood-brain barrier leakage. Cell Rep 2024; 43:113911. [PMID: 38446668 DOI: 10.1016/j.celrep.2024.113911] [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: 07/14/2023] [Revised: 12/19/2023] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Claudin-5 (CLDN5) is an endothelial tight junction protein essential for blood-brain barrier (BBB) formation. Abnormal CLDN5 expression is common in brain disease, and knockdown of Cldn5 at the BBB has been proposed to facilitate drug delivery to the brain. To study the consequences of CLDN5 loss in the mature brain, we induced mosaic endothelial-specific Cldn5 gene ablation in adult mice (Cldn5iECKO). These mice displayed increased BBB permeability to tracers up to 10 kDa in size from 6 days post induction (dpi) and ensuing lethality from 10 dpi. Single-cell RNA sequencing at 11 dpi revealed profound transcriptomic differences in brain endothelial cells regardless of their Cldn5 status in mosaic mice, suggesting major non-cell-autonomous responses. Reactive microglia and astrocytes suggested rapid cellular responses to BBB leakage. Our study demonstrates a critical role for CLDN5 in the adult BBB and provides molecular insight into the consequences and risks associated with CLDN5 inhibition.
Collapse
Affiliation(s)
- Elisa Vázquez-Liébanas
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Giuseppe Mocci
- Single Cell Core Facility of Flemingsberg Campus (SICOF), Karolinska Institute, 14157 Huddinge, Sweden
| | - Weihan Li
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Bàrbara Laviña
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Avril Reddy
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Claire O'Connor
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Natalie Hudson
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Zaher Elbeck
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Ioannis Nikoloudis
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Konstantin Gaengel
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Michael Vanlandewijck
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden; Single Cell Core Facility of Flemingsberg Campus (SICOF), Karolinska Institute, 14157 Huddinge, Sweden; Department of Medicine, Karolinska Institute, 14157 Huddinge, Sweden
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Christer Betsholtz
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden; Department of Medicine, Karolinska Institute, 14157 Huddinge, Sweden
| | - Maarja Andaloussi Mäe
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden.
| |
Collapse
|
43
|
Roche F, Briançon-Marjollet A, Dematteis M, Baldazza M, Gonthier B, Bertholon F, Perek N, Pépin JL. Early Increase in Blood-Brain Barrier Permeability in a Murine Model Exposed to Fifteen Days of Intermittent Hypoxia. Int J Mol Sci 2024; 25:3065. [PMID: 38474310 DOI: 10.3390/ijms25053065] [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: 12/15/2023] [Revised: 02/08/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Obstructive sleep apnea (OSA) is characterized by intermittent repeated episodes of hypoxia-reoxygenation. OSA is associated with cerebrovascular consequences. An enhanced blood-brain barrier (BBB) permeability has been proposed as a marker of those disorders. We studied in mice the effects of 1 day and 15 days intermittent hypoxia (IH) exposure on BBB function. We focused on the dorsal part of the hippocampus and attempted to identify the molecular mechanisms by combining in vivo BBB permeability (Evans blue tests) and mRNA expression of several junction proteins (zona occludens (ZO-1,2,3), VE-cadherin, claudins (1,5,12), cingulin) and of aquaporins (1,4,9) on hippocampal brain tissues. After 15 days of IH exposure we observed an increase in BBB permeability, associated with increased mRNA expressions of claudins 1 and 12, aquaporins 1 and 9. IH seemed to increase early for claudin-1 mRNA expression as it doubled with 1 day of exposure and returned near to its base level after 15 days. Claudin-1 overexpression may represent an immediate response to IH exposure. Then, after 15 days of exposure, an increase in functional BBB permeability was associated with enhanced expression of aquaporin. These BBB alterations are possibly associated with a vasogenic oedema that may affect brain functions and accelerate neurodegenerative processes.
Collapse
Affiliation(s)
- Frederic Roche
- INSERM, SAINBIOSE U1059, Université Jean Monnet Saint-Étienne, Mines Saint Etienne, F-42023 Saint-Étienne, France
- INSERM U1300, HP2 Laboratory, Université Grenoble Alpes, F-38042 Grenoble, France
| | | | - Maurice Dematteis
- INSERM U1300, HP2 Laboratory, Université Grenoble Alpes, F-38042 Grenoble, France
| | - Marie Baldazza
- INSERM U1300, HP2 Laboratory, Université Grenoble Alpes, F-38042 Grenoble, France
| | - Brigitte Gonthier
- INSERM U1300, HP2 Laboratory, Université Grenoble Alpes, F-38042 Grenoble, France
| | - Frederique Bertholon
- Centre de Ressources Biologiques, CHU de Saint Etienne, F-42055 Saint-Étienne, France
| | - Nathalie Perek
- INSERM, SAINBIOSE U1059, Université Jean Monnet Saint-Étienne, Mines Saint Etienne, F-42023 Saint-Étienne, France
| | - Jean-Louis Pépin
- INSERM U1300, HP2 Laboratory, Université Grenoble Alpes, F-38042 Grenoble, France
| |
Collapse
|
44
|
Greene C, Connolly R, Brennan D, Laffan A, O'Keeffe E, Zaporojan L, O'Callaghan J, Thomson B, Connolly E, Argue R, Meaney JFM, Martin-Loeches I, Long A, Cheallaigh CN, Conlon N, Doherty CP, Campbell M. Blood-brain barrier disruption and sustained systemic inflammation in individuals with long COVID-associated cognitive impairment. Nat Neurosci 2024; 27:421-432. [PMID: 38388736 PMCID: PMC10917679 DOI: 10.1038/s41593-024-01576-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/09/2024] [Indexed: 02/24/2024]
Abstract
Vascular disruption has been implicated in coronavirus disease 2019 (COVID-19) pathogenesis and may predispose to the neurological sequelae associated with long COVID, yet it is unclear how blood-brain barrier (BBB) function is affected in these conditions. Here we show that BBB disruption is evident during acute infection and in patients with long COVID with cognitive impairment, commonly referred to as brain fog. Using dynamic contrast-enhanced magnetic resonance imaging, we show BBB disruption in patients with long COVID-associated brain fog. Transcriptomic analysis of peripheral blood mononuclear cells revealed dysregulation of the coagulation system and a dampened adaptive immune response in individuals with brain fog. Accordingly, peripheral blood mononuclear cells showed increased adhesion to human brain endothelial cells in vitro, while exposure of brain endothelial cells to serum from patients with long COVID induced expression of inflammatory markers. Together, our data suggest that sustained systemic inflammation and persistent localized BBB dysfunction is a key feature of long COVID-associated brain fog.
Collapse
Affiliation(s)
- Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Ruairi Connolly
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | - Declan Brennan
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | - Aoife Laffan
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | - Eoin O'Keeffe
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Lilia Zaporojan
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | | | - Bennett Thomson
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Emma Connolly
- The Irish Longitudinal Study on Ageing, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Ruth Argue
- Clinical Research Facility, St James's Hospital, Dublin, Ireland
| | - James F M Meaney
- Thomas Mitchell Centre for Advanced Medical Imaging (CAMI), St. James's Hospital & Trinity College Dublin, Dublin, Ireland
| | - Ignacio Martin-Loeches
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization, Trinity Centre for Health Sciences, St James's University Hospital, Dublin, Ireland
| | - Aideen Long
- Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin, Ireland
| | - Cliona Ni Cheallaigh
- Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin, Ireland
- Department of Immunology, St James's Hospital, Dublin, Ireland
| | - Niall Conlon
- Department of Immunology, St James's Hospital, Dublin, Ireland
- St James's Hospital, Tallaght University Hospital, Trinity College Dublin Allied Researchers (STTAR) Bioresource, Trinity College Dublin, Dublin, Ireland
| | - Colin P Doherty
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland.
- Academic Unit of Neurology, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland.
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland.
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland.
| |
Collapse
|
45
|
Shukla A, Bhardwaj U, Apoorva, Seth P, Singh SK. Hypoxia-Induced miR-101 Impairs Endothelial Barrier Integrity Through Altering VE-Cadherin and Claudin-5. Mol Neurobiol 2024; 61:1807-1817. [PMID: 37776496 DOI: 10.1007/s12035-023-03662-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/12/2023] [Indexed: 10/02/2023]
Abstract
Stroke is a life-threatening medical condition across the world that adversely affects the integrity of the blood-brain barrier (BBB). The brain microvascular endothelial cells are the important constituent of the BBB. These cells line the blood vessels and form a semipermeable barrier. Disruptions in adherens junction and tight junction proteins of brain microvascular endothelial cells compromise the integrity of BBB. The Vascular Endothelial (VE)-cadherin is an integral adherens junction protein required for the establishment and maintenance of the endothelial barrier integrity. This study aims to investigate the role of miRNA in hypoxia-induced endothelial barrier disruption. In this study, brain endothelial cells were exposed to hypoxic conditions for different time points. Western blotting, overexpression and knockdown of miRNA, real-time PCR, TEER, and sodium fluorescein assay were used to examine the effect of hypoxic conditions on brain endothelial cells. Hypoxic exposure was validated using HIF-1α protein. Exposure to hypoxic conditions resulted to a significant decrease in endothelial barrier resistance and an increase in sodium fluorescein migration across the endothelial barrier. Reduction in endothelial barrier resistance demonstrated compromised barrier integrity, whereas the increase in migration of sodium fluorescein across the barrier indicated the increase in barrier permeability. The present study revealed microRNA-101 decreases the expression of VE-cadherin and claudin-5 in brain endothelial cells exposed to the hypoxic conditions.
Collapse
Affiliation(s)
- Astha Shukla
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, UP, India
| | - Utkarsh Bhardwaj
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, UP, India
| | - Apoorva
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, UP, India
| | - Pankaj Seth
- Molecular and Cellular Neurosciences, National Brain Research Centre, Manesar, 122052, Haryana, India
| | - Sunit K Singh
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, UP, India.
- Dr. B R Ambedkar Center for Biomedical Research, University of Delhi (North Campus), New Delhi, 110007, India.
| |
Collapse
|
46
|
Deli MA, Porkoláb G, Kincses A, Mészáros M, Szecskó A, Kocsis AE, Vigh JP, Valkai S, Veszelka S, Walter FR, Dér A. Lab-on-a-chip models of the blood-brain barrier: evolution, problems, perspectives. LAB ON A CHIP 2024; 24:1030-1063. [PMID: 38353254 DOI: 10.1039/d3lc00996c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
A great progress has been made in the development and use of lab-on-a-chip devices to model and study the blood-brain barrier (BBB) in the last decade. We present the main types of BBB-on-chip models and their use for the investigation of BBB physiology, drug and nanoparticle transport, toxicology and pathology. The selection of the appropriate cell types to be integrated into BBB-on-chip devices is discussed, as this greatly impacts the physiological relevance and translatability of findings. We identify knowledge gaps, neglected engineering and cell biological aspects and point out problems and contradictions in the literature of BBB-on-chip models, and suggest areas for further studies to progress this highly interdisciplinary field. BBB-on-chip models have an exceptional potential as predictive tools and alternatives of animal experiments in basic and preclinical research. To exploit the full potential of this technique expertise from materials science, bioengineering as well as stem cell and vascular/BBB biology is necessary. There is a need for better integration of these diverse disciplines that can only be achieved by setting clear parameters for characterizing both the chip and the BBB model parts technically and functionally.
Collapse
Affiliation(s)
- Mária A Deli
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Gergő Porkoláb
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
- Doctoral School of Biology, University of Szeged, Hungary
| | - András Kincses
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Mária Mészáros
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Anikó Szecskó
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
- Doctoral School of Biology, University of Szeged, Hungary
| | - Anna E Kocsis
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Judit P Vigh
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
- Doctoral School of Biology, University of Szeged, Hungary
| | - Sándor Valkai
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Szilvia Veszelka
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Fruzsina R Walter
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - András Dér
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| |
Collapse
|
47
|
Tanelian A, Nankova B, Miari M, Sabban EL. Microbial composition, functionality, and stress resilience or susceptibility: unraveling sex-specific patterns. Biol Sex Differ 2024; 15:20. [PMID: 38409102 PMCID: PMC10898170 DOI: 10.1186/s13293-024-00590-7] [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: 09/05/2023] [Accepted: 01/31/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Following exposure to traumatic stress, women are twice as likely as men to develop mood disorders. Yet, individual responses to such stress vary, with some people developing stress-induced psychopathologies while others exhibit resilience. The factors influencing sex-related disparities in affective disorders as well as variations in resilience remain unclear; however, emerging evidence suggests differences in the gut microbiota play a role. In this study, using the single prolonged stress (SPS) model of post-traumatic stress disorder, we investigated pre- and post-existing differences in microbial composition, functionality, and metabolites that affect stress susceptibility or resilience in each sex. METHODS Male and female Sprague-Dawley rats were randomly assigned to control or SPS groups. Two weeks following SPS, the animals were exposed to a battery of behavioral tests and decapitated a day later. Based on their anxiety index, they were further categorized as SPS-resilient (SPS-R) or SPS-susceptible (SPS-S). On the day of dissection, cecum, and selected brain tissues were isolated. Stool samples were collected before and after SPS, whereas urine samples were taken before and 30 min into the SPS. RESULTS Before SPS exposure, the sympathoadrenal axis exhibited alterations within male subgroups only. Expression of tight junction protein claudin-5 was lower in brain of SPS-S males, but higher in SPS-R females following SPS. Across the study, alpha diversity remained consistently lower in males compared to females. Beta diversity revealed distinct separations between male and female susceptible groups before SPS, with this separation becoming evident in the resilient groups following SPS. At the genus level, Lactobacillus, Lachnospiraceae_Incertae_Sedis, and Barnesiella exhibited sex-specific alterations, displaying opposing abundances in each sex. Additionally, sex-specific changes were observed in microbial predictive functionality and targeted functional modules both before and after SPS. Alterations in the microbial short-chain fatty acids (SCFAs), were also observed, with major and minor SCFAs being lower in SPS-susceptible males whereas branched-chain SCFAs being higher in SPS-susceptible females. CONCLUSION This study highlights distinct pre- and post-trauma differences in microbial composition, functionality, and metabolites, associated with stress resilience in male and female rats. The findings underscore the importance of developing sex-specific therapeutic strategies to effectively address stress-related disorders. Highlights SPS model induces divergent anxiety and social behavioral responses to traumatic stress in both male and female rodents. SPS-resilient females displayed less anxiety-like behavior and initiated more interactions towards a juvenile rat than SPS-resilient males. Sex-specific pre-existing and SPS-induced differences in the gut microbial composition and predictive functionality were observed in susceptible and resilient rats. SPS-resilient males displayed elevated cecal acetate levels, whereas SPS-susceptible females exhibited heightened branched-chain SCFAs.
Collapse
Affiliation(s)
- Arax Tanelian
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, 10595, USA
| | - Bistra Nankova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, 10595, USA
- Division of Newborn Medicine, Departments of Pediatrics, New York Medical College, Valhalla, NY, 10595, USA
| | - Mariam Miari
- Department of Clinical Sciences in Malmo, Lund University Diabetes Center, Malmo, Sweden
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, 10595, USA.
- Department of Psychiatry and Behavioral Science, New York Medical College, Valhalla, NY, 10595, USA.
| |
Collapse
|
48
|
Lund H, Hunt MA, Kurtović Z, Sandor K, Kägy PB, Fereydouni N, Julien A, Göritz C, Vazquez-Liebanas E, Andaloussi Mäe M, Jurczak A, Han J, Zhu K, Harris RA, Lampa J, Graversen JH, Etzerodt A, Haglund L, Yaksh TL, Svensson CI. CD163+ macrophages monitor enhanced permeability at the blood-dorsal root ganglion barrier. J Exp Med 2024; 221:e20230675. [PMID: 38117255 PMCID: PMC10733632 DOI: 10.1084/jem.20230675] [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: 04/20/2023] [Revised: 10/04/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023] Open
Abstract
In dorsal root ganglia (DRG), macrophages reside close to sensory neurons and have largely been explored in the context of pain, nerve injury, and repair. However, we discovered that most DRG macrophages interact with and monitor the vasculature by sampling macromolecules from the blood. Characterization of the DRG vasculature revealed a specialized endothelial bed that transformed in molecular, structural, and permeability properties along the arteriovenous axis and was covered by macrophage-interacting pericytes and fibroblasts. Macrophage phagocytosis spatially aligned with peak endothelial permeability, a process regulated by enhanced caveolar transcytosis in endothelial cells. Profiling the DRG immune landscape revealed two subsets of perivascular macrophages with distinct transcriptome, turnover, and function. CD163+ macrophages self-maintained locally, specifically participated in vasculature monitoring, displayed distinct responses during peripheral inflammation, and were conserved in mouse and man. Our work provides a molecular explanation for the permeability of the blood-DRG barrier and identifies an unappreciated role of macrophages as integral components of the DRG-neurovascular unit.
Collapse
Affiliation(s)
- Harald Lund
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matthew A. Hunt
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zerina Kurtović
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Kancera AB, Karolinska Institutet Science Park, Stockholm, Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paul B. Kägy
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Noah Fereydouni
- Department of Medicine, Rheumatology Unit, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anais Julien
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Elisa Vazquez-Liebanas
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Maarja Andaloussi Mäe
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Alexandra Jurczak
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jinming Han
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Keying Zhu
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Robert A. Harris
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jon Lampa
- Department of Medicine, Rheumatology Unit, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Lisbet Haglund
- Division of Orthopaedic Surgery, Department of Surgery, McGill University, Montreal, Canada
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, CA, USA
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
49
|
Vita SM, Cruise SC, Gilpin NW, Molina PE. Histological comparison of repeated mild weight drop and lateral fluid percussion injury models of traumatic brain injury (TBI) in female and male rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578177. [PMID: 38352449 PMCID: PMC10862833 DOI: 10.1101/2024.01.31.578177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Traumatic brain injury (TBI) heterogeneity has led to the development of several preclinical models, each modeling a distinct subset of outcomes. Selection of an injury model should be guided by the research question and the specific outcome measures of interest. Consequently, there is a need for conducting direct comparisons of different TBI models. Here, we used immunohistochemistry to directly compare the outcomes from two common models, lateral fluid percussion (LFP) and repeat mild weight drop (rmWD), on neuropathology in adult female and male Wistar rats. Specifically, we used immunohistochemistry to measure the effects of LFP and rmWD on cerebrovascular and tight junction disruption, inflammatory markers, mature neurons and perineuronal nets in the cortical site of injury, cortex adjacent to injury, dentate gyrus, and the CA2/3 area of the hippocampus. Animals were randomized into either LFP or rmWD groups. The LFP group received a craniotomy prior to LFP (or corresponding sham procedure) three days later, while rmWD animals underwent either weight drop or sham (isoflurane only) on each of those four days. After a recovery period of 7 days, animals were euthanized, and brains were harvested for analysis of RECA-1, claudin-5, GFAP, Iba-1, CD-68, NeuN, and wisteria floribunda lectin. Overall, our observations revealed that the most significant disruptions were evident in response to LFP, followed by craniotomy-only, while rmWD animals showed the least residual changes compared to isoflurane-only controls. These findings support consideration of rmWD as a mild, transient injury. LFP leads to longer-lasting disruptions that are more closely associated with a moderate TBI. We further show that both craniotomy and LFP produced greater disruptions in females relative to males at 7 days post-injury. These findings support the inclusion of a time-matched experimentally-naïve or anesthesia-only control group in preclinical TBI research to enhance the validity of data interpretation and conclusions.
Collapse
|
50
|
Fei Y, Li T, Wu R, Xu X, Hu S, Yang Y, Jin C, Tang W, Zhang X, Du Q, Liu C. Se-(Methyl)-selenocysteine ameliorates blood-brain barrier disruption of focal cerebral ischemia mice via ferroptosis inhibition and tight junction upregulation in an Akt/GSK3β-dependent manner. Psychopharmacology (Berl) 2024; 241:379-399. [PMID: 38019326 DOI: 10.1007/s00213-023-06495-4] [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: 07/15/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Ischemic stroke still ranks as the most fatal disease worldwide. Blood-brain barrier (BBB) is a promising therapeutic target for protection. Brain microvascular endothelial cell is a core component of BBB, the barrier function maintenance of which can ameliorate ischemic injury and improve neurological deficit. Se-methyl L-selenocysteine (SeMC) has been shown to exert cardiovascular protection. However, the protection of SeMC against ischemic stroke remains to be elucidated. This research was designed to explore the protection of SeMC from the perspective of BBB protection. METHODS To simulate cerebral ischemic injury, C57BL/6J mice were subjected to middle cerebral artery occlusion/reperfusion (MCAO/R), and bEnd.3 was exposed to oxygen-glucose deprivation/reoxygenation (OGD/R). After the intervention of SeMC, the barrier function and the expression of tight junction and ferroptosis-associated proteins were determined. For mechanism exploration, LY294002 (Akt inhibitor) was introduced both in vivo and in vitro. RESULTS SeMC lessened the brain infarct volume and attenuated the leakage of BBB in mice. In vitro, SeMC improved cell viability and maintained the barrier function of bEnd.3 cells. The protection of SeMC was accompanied with ferroptosis inhibition and tight junction protein upregulation. Mechanism studies revealed that the effect of SeMC was reversed by LY294002, indicating that the protection of SeMC against ischemic stroke was mediated by the Akt signal pathway. CONCLUSION These results suggested that SeMC exerted protection against ischemic stroke, which might be attributed to activating the Akt/GSK3β signaling pathway and increasing the nuclear translocation of Nrf2 and β-catenin, subsequently maintaining the integrity of BBB.
Collapse
Affiliation(s)
- Yuxiang Fei
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Tao Li
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China
| | - Ruoyu Wu
- Jinling High School, 169 Zhongshan Road, Nanjing, 210005, People's Republic of China
| | - Xuejiao Xu
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Sheng Hu
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, People's Republic of China
| | - Ya Yang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, People's Republic of China
| | - Chenchen Jin
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Wenlian Tang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, People's Republic of China
| | - Xu Zhang
- Department of Pharmacy, Chengdu First People's Hospital, Chengdu, People's Republic of China.
- Department of Pharmacy, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine of Chengdu Medical College, Chengdu, 610031, People's Republic of China.
| | - Qianming Du
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China.
| | - Chao Liu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, People's Republic of China.
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|