1
|
Schibalski RS, Shulha AS, Tsao BP, Palygin O, Ilatovskaya DV. The role of polyamine metabolism in cellular function and physiology. Am J Physiol Cell Physiol 2024; 327:C341-C356. [PMID: 38881422 PMCID: PMC11427016 DOI: 10.1152/ajpcell.00074.2024] [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: 01/31/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Polyamines are molecules with multiple amino groups that are essential for cellular function. The major polyamines are putrescine, spermidine, spermine, and cadaverine. Polyamines are important for posttranscriptional regulation, autophagy, programmed cell death, proliferation, redox homeostasis, and ion channel function. Their levels are tightly controlled. High levels of polyamines are associated with proliferative pathologies such as cancer, whereas low polyamine levels are observed in aging, and elevated polyamine turnover enhances oxidative stress. Polyamine metabolism is implicated in several pathophysiological processes in the nervous, immune, and cardiovascular systems. Currently, manipulating polyamine levels is under investigation as a potential preventive treatment for several pathologies, including aging, ischemia/reperfusion injury, pulmonary hypertension, and cancer. Although polyamines have been implicated in many intracellular mechanisms, our understanding of these processes remains incomplete and is a topic of ongoing investigation. Here, we discuss the regulation and cellular functions of polyamines, their role in physiology and pathology, and emphasize the current gaps in knowledge and potential future research directions.
Collapse
Affiliation(s)
- Ryan S Schibalski
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Anastasia S Shulha
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Betty P Tsao
- Division of Rheumatology & Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
2
|
Zhu X, Wang B, Yu H, Li C, Zhao Y, Zhong Y, Tang W, Zhou Y, Huang X, Zhu H, Wu Y, Yang K, Wei Y, Gao Z, Dong J. Icariin attenuates asthmatic airway inflammation via modulating alveolar macrophage activation based on network pharmacology and in vivo experiments. J Gene Med 2024; 26:e3718. [PMID: 38979822 DOI: 10.1002/jgm.3718] [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: 09/07/2023] [Revised: 03/23/2024] [Accepted: 06/19/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Icariin (ICA) inhibits inflammatory response in various diseases, but the mechanism underlying ICA treating airway inflammation in asthma needs further understood. We aimed to predict and validate the potential targets of ICA against asthma-associated airway inflammation using network pharmacology and experiments. METHODS The ovalbumin-induced asthma-associated airway inflammation mice model was established. The effects of ICA were evaluated by behavioral, airway hyperresponsiveness, lung pathological changes, inflammatory cell and cytokines counts. Next, the corresponding targets of ICA were mined via the SEA, CTD, HERB, PharmMapper, Symmap database and the literature. Pubmed-Gene and GeneCards databases were used to screen asthma and airway inflammation-related targets. The overlapping targets were used to build an interaction network, analyze gene ontology and enrich pathways. Subsequently, flow cytometry, quantitative real-time PCR and western blotting were employed for validation. RESULTS ICA alleviated the airway inflammation of asthma; 402 targets of ICA, 5136 targets of asthma and 4531 targets of airway inflammation were screened; 216 overlapping targets were matched and predicted ICA possesses the potential to modulate asthmatic airway inflammation by macrophage activation/polarization. Additionally, ICA decreased M1 but elevated M2. Potential targets that were disrupted by asthma inflammation were restored by ICA treatment. CONCLUSIONS ICA alleviates airway inflammation in asthma by inhibiting the M1 polarization of alveolar macrophages, which is related to metabolic reprogramming. Jun, Jak2, Syk, Tnf, Aldh2, Aldh9a1, Nos1, Nos2 and Nos3 represent potential targets of therapeutic intervention. The present study enhances understanding of the anti-airway inflammation effects of ICA, especially in asthma.
Collapse
Affiliation(s)
- Xiaofei Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Bin Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Congcong Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yuhang Zhao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yuanyuan Zhong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yaolong Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Xi Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Huahe Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yueren Wu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Kai Yang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhen Gao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Zhang L, Buonfiglio F, Fieß A, Pfeiffer N, Gericke A. Retinopathy of Prematurity-Targeting Hypoxic and Redox Signaling Pathways. Antioxidants (Basel) 2024; 13:148. [PMID: 38397746 PMCID: PMC10885953 DOI: 10.3390/antiox13020148] [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: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a proliferative vascular ailment affecting the retina. It is the main risk factor for visual impairment and blindness in infants and young children worldwide. If left undiagnosed and untreated, it can progress to retinal detachment and severe visual impairment. Geographical variations in ROP epidemiology have emerged over recent decades, attributable to differing levels of care provided to preterm infants across countries and regions. Our understanding of the causes of ROP, screening, diagnosis, treatment, and associated risk factors continues to advance. This review article aims to present the pathophysiological mechanisms of ROP, including its treatment. Specifically, it delves into the latest cutting-edge treatment approaches targeting hypoxia and redox signaling pathways for this condition.
Collapse
Affiliation(s)
| | | | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (L.Z.); (F.B.); (A.F.); (N.P.)
| |
Collapse
|
4
|
Shosha E, Shahror RA, Morris CA, Xu Z, Lucas R, McGee-Lawrence ME, Rusch NJ, Caldwell RB, Fouda AY. The arginase 1/ornithine decarboxylase pathway suppresses HDAC3 to ameliorate the myeloid cell inflammatory response: implications for retinal ischemic injury. Cell Death Dis 2023; 14:621. [PMID: 37735154 PMCID: PMC10514323 DOI: 10.1038/s41419-023-06147-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023]
Abstract
The enzyme arginase 1 (A1) hydrolyzes the amino acid arginine to form L-ornithine and urea. Ornithine is further converted to polyamines by the ornithine decarboxylase (ODC) enzyme. We previously reported that deletion of myeloid A1 in mice exacerbates retinal damage after ischemia/reperfusion (IR) injury. Furthermore, treatment with A1 protects against retinal IR injury in wild-type mice. PEG-A1 also mitigates the exaggerated inflammatory response of A1 knockout (KO) macrophages in vitro. Here, we sought to identify the anti-inflammatory pathway that confers macrophage A1-mediated protection against retinal IR injury. Acute elevation of intraocular pressure was used to induce retinal IR injury in mice. A multiplex cytokine assay revealed a marked increase in the inflammatory cytokines interleukin 1β (IL-1β) and tumor necrosis factor α (TNF-α) in the retina at day 5 after IR injury. In vitro, blocking the A1/ODC pathway augmented IL-1β and TNF-α production in stimulated macrophages. Furthermore, A1 treatment attenuated the stimulated macrophage metabolic switch to a pro-inflammatory glycolytic phenotype, whereas A1 deletion had the opposite effect. Screening for histone deacetylases (HDACs) which play a role in macrophage inflammatory response showed that A1 deletion or ODC inhibition increased the expression of HDAC3. We further showed the involvement of HDAC3 in the upregulation of TNF-α but not IL-1β in stimulated macrophages deficient in the A1/ODC pathway. Investigating HDAC3 KO macrophages showed a reduced inflammatory response and a less glycolytic phenotype upon stimulation. In vivo, HDAC3 co-localized with microglia/macrophages at day 2 after IR in WT retinas and was further increased in A1-deficient retinas. Collectively, our data provide initial evidence that A1 exerts its anti-inflammatory effect in macrophages via ODC-mediated suppression of HDAC3 and IL-1β. Collectively we propose that interventions that augment the A1/ODC pathway and inhibit HDAC3 may confer therapeutic benefits for the treatment of retinal ischemic diseases.
Collapse
Affiliation(s)
- Esraa Shosha
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Rami A Shahror
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Carol A Morris
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Zhimin Xu
- Vascular Biology Center, Augusta University, Augusta, GA, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Rudolf Lucas
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | | | - Nancy J Rusch
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Abdelrahman Y Fouda
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
5
|
Fevereiro-Martins M, Marques-Neves C, Guimarães H, Bicho M. Retinopathy of prematurity: A review of pathophysiology and signaling pathways. Surv Ophthalmol 2023; 68:175-210. [PMID: 36427559 DOI: 10.1016/j.survophthal.2022.11.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022]
Abstract
Retinopathy of prematurity (ROP) is a vasoproliferative disorder of the retina and a leading cause of visual impairment and childhood blindness worldwide. The disease is characterized by an early stage of retinal microvascular degeneration, followed by neovascularization that can lead to subsequent retinal detachment and permanent visual loss. Several factors play a key role during the different pathological stages of the disease. Oxidative and nitrosative stress and inflammatory processes are important contributors to the early stage of ROP. Nitric oxide synthase and arginase play important roles in ischemia/reperfusion-induced neurovascular degeneration. Destructive neovascularization is driven by mediators of the hypoxia-inducible factor pathway, such as vascular endothelial growth factor and metabolic factors (succinate). The extracellular matrix is involved in hypoxia-induced retinal neovascularization. Vasorepulsive molecules (semaphorin 3A) intervene preventing the revascularization of the avascular zone. This review focuses on current concepts about signaling pathways and their mediators, involved in the pathogenesis of ROP, highlighting new potentially preventive and therapeutic modalities. A better understanding of the intricate molecular mechanisms underlying the pathogenesis of ROP should allow the development of more effective and targeted therapeutic agents to reduce aberrant vasoproliferation and facilitate physiological retinal vascular development.
Collapse
Affiliation(s)
- Mariza Fevereiro-Martins
- Laboratório de Genética and Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Investigação Científica Bento da Rocha Cabral, Lisboa, Portugal; Departamento de Oftalmologia, Hospital Cuf Descobertas, Lisboa, Portugal.
| | - Carlos Marques-Neves
- Centro de Estudos das Ci.¼ncias da Visão, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| | - Hercília Guimarães
- Departamento de Ginecologia-Obstetrícia e Pediatria, Faculdade de Medicina, Universidade do Porto, Porto, Portugal.
| | - Manuel Bicho
- Laboratório de Genética and Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Investigação Científica Bento da Rocha Cabral, Lisboa, Portugal.
| |
Collapse
|
6
|
Tang K, Zhang H, Deng J, Wang D, Liu S, Lu S, Cui Q, Chen C, Liu J, Yang Z, Li Y, Chen J, Lv J, Ma J, Huang B. Ammonia detoxification promotes CD8 + T cell memory development by urea and citrulline cycles. Nat Immunol 2023; 24:162-173. [PMID: 36471170 DOI: 10.1038/s41590-022-01365-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 10/17/2022] [Indexed: 12/12/2022]
Abstract
Amino acid metabolism is essential for cell survival, while the byproduct ammonia is toxic and can injure cellular longevity. Here we show that CD8+ memory T (TM) cells mobilize the carbamoyl phosphate (CP) metabolic pathway to clear ammonia, thus promoting memory development. CD8+ TM cells use β-hydroxybutyrylation to upregulate CP synthetase 1 and trigger the CP metabolic cascade to form arginine in the cytosol. This cytosolic arginine is then translocated into the mitochondria where it is split by arginase 2 to urea and ornithine. Cytosolic arginine is also converted to nitric oxide and citrulline by nitric oxide synthases. Thus, both the urea and citrulline cycles are employed by CD8+ T cells to clear ammonia and enable memory development. This ammonia clearance machinery might be targeted to improve T cell-based cancer immunotherapies.
Collapse
Affiliation(s)
- Ke Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, China
| | - Huafeng Zhang
- Department of Pathology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinghui Deng
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dianheng Wang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Shichuan Liu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuya Lu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingfa Cui
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jincheng Liu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuoshun Yang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yonggang Li
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Jie Chen
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Jiadi Lv
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Huang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China.
| |
Collapse
|
7
|
Li S, Li J, Fan Y, Huang T, Zhou Y, Fan H, Zhang Q, Qiu R. The mechanism of formononetin/calycosin compound optimizing the effects of temozolomide on C6 malignant glioma based on metabolomics and network pharmacology. Biomed Pharmacother 2022; 153:113418. [PMID: 36076540 DOI: 10.1016/j.biopha.2022.113418] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/28/2022] [Accepted: 07/13/2022] [Indexed: 12/30/2022] Open
Abstract
The complex of formononetin and calycosin (FMN/CAL) shows a synergistic effect on temozolomide in the treatment of malignant glioma, however the mechanism is unclear. We investigated the mechanism through means of metabolomics, network pharmacology and molecular biology. FMN/CAL enhanced the inhibition of TMZ on the growth and infiltration of C6 glioma. The metabolomic results showed that the TMZ sensitization of FMN/CAL mainly involved 5 metabolic pathways and 4 metabolites in cells, 1 metabolic pathway and 2 metabolites in tumor tissues, and 7 metabolic pathways and 8 metabolites in serum. Further network pharmacological analysis revealed that NOS2 was a potential target for FMN/CAL to regulate the metabolism in TMZ-treated C6 glioma cells, serums and tissues, and TNF-α was another potential target identified in tissues. FMN/CAL down-regulated the expression of NOS2 in tumor cells and tissues, and reduced the secretion of TNF-α in tumor region. FMN/CAL promoted TMZ-induced C6 cell apoptosis by inhibiting NOS2, but the inhibition of cell vitality and migration was not through NOS2. Our work revealed that FMN/CAL can increase the sensitivity of malignant glioma to TMZ by inhibiting NOS2-dependent cell survival, which provides a basis for the application of this combination in adjuvant treatment of glioma.
Collapse
Affiliation(s)
- Songya Li
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Jiayi Li
- Medical Insurance Office, SIR RUN RUN Hospital Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Yani Fan
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Tao Huang
- College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu 210000, China
| | - Yanfen Zhou
- College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu 210000, China
| | - Hongwei Fan
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China.
| | - Qi Zhang
- College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu 210000, China.
| | - Runze Qiu
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China.
| |
Collapse
|
8
|
Yao M, Fang J, Li J, Ng ACK, Liu J, Leung GKK, Song F, Zhang J, Chang C. Modulation of the proteoglycan receptor PTPσ promotes white matter integrity and functional recovery after intracerebral hemorrhage stroke in mice. J Neuroinflammation 2022; 19:207. [PMID: 35982473 PMCID: PMC9387079 DOI: 10.1186/s12974-022-02561-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/25/2022] [Indexed: 11/29/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) is associated with high morbidity and mortality rates. However, extant investigations have mainly focused on gray matter injury within the primary injury site after ICH rather than on white matter (WM) injury in the brain and spinal cord. This focus partly accounts for the diminished therapeutic discovery. Recent evidence suggests that chondroitin sulphate proteoglycans (CSPG), which can bind to the neural transmembrane protein tyrosine phosphatase-sigma (PTPσ), may facilitate axonal regrowth and remyelination by ameliorating neuroinflammation. Methods A clinically relevant ICH model was established using adult C57BL/6 mice. The mice were then treated systemically with intracellular sigma peptide (ISP), which specifically targets PTPσ. Sensorimotor function was assessed by various behavioral tests and electrophysiological assessment. Western blot was used to verify the expression levels of Iba-1 and different inflammatory cytokines. The morphology of white matter tracts of brain and spinal cord was evaluated by immunofluorescence staining and transmission electron microscopy (TEM). Adeno-associated virus (AAV) 2/9 injection was used to assess the ipsilateral axonal compensation after injury. Parallel in vitro studies on the effects of CSPG interference on oligodendrocyte–DRG neuron co-culture explored the molecular mechanism through which ISP treatment promoted myelination capability. Results ISP, by targeting PTPσ, improved WM integrity and sensorimotor recovery via immunomodulation. In addition, ISP administration significantly decreased WM injury in the peri-hematomal region as well as cervical spinal cord, enhanced axonal myelination and facilitated neurological restoration, including electrophysiologically assessed sensorimotor functions. Parallel in vitro studies showed that inhibition of PTPσ by ISP fosters myelination by modulating the Erk/CREB signaling pathway. Conclusions Our findings revealed for the first time that manipulation of PTPσ signaling by ISP can promote prolonged neurological recovery by restoration of the integrity of neural circuits in the CNS through modulation of Erk/CREB signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02561-4.
Collapse
Affiliation(s)
- Min Yao
- School of Pharmaceutical Sciences, Health Science Centre, Shenzhen University, Shenzhen, 518060, China.,School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China.,Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jie Fang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Jiewei Li
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Anson Cho Kiu Ng
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jiaxin Liu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Gilberto Ka Kit Leung
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Fanglai Song
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Jian Zhang
- School of Pharmaceutical Sciences, Health Science Centre, Shenzhen University, Shenzhen, 518060, China.
| | - Chunqi Chang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
9
|
Aldosari DI, Malik A, Alhomida AS, Ola MS. Implications of Diabetes-Induced Altered Metabolites on Retinal Neurodegeneration. Front Neurosci 2022; 16:938029. [PMID: 35911994 PMCID: PMC9328693 DOI: 10.3389/fnins.2022.938029] [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: 05/06/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic retinopathy (DR) is one of the major complications of diabetic eye diseases, causing vision loss and blindness worldwide. The concept of diabetic retinopathy has evolved from microvascular disease into more complex neurovascular disorders. Early in the disease progression of diabetes, the neuronal and glial cells are compromised before any microvascular abnormalities clinically detected by the ophthalmoscopic examination. This implies understanding the pathophysiological mechanisms at the early stage of disease progression especially due to diabetes-induced metabolic alterations to damage the neural retina so that early intervention and treatments options can be identified to prevent and inhibit the progression of DR. Hyperglycemia has been widely considered the major contributor to the progression of the retinal damage, even though tight control of glucose does not seem to have a bigger effect on the incidence or progression of retinal damage that leads to DR. Emerging evidence suggests that besides diabetes-induced hyperglycemia, dyslipidemia and amino acid defects might be a major contributor to the progression of early neurovascular retinal damage. In this review, we have discussed recent advances in the alterations of key metabolites of carbohydrate, lipid, and amino acids and their implications for neurovascular damage in DR.
Collapse
Affiliation(s)
| | | | | | - Mohammad S. Ola
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Abstract
The microbiota-gut-brain-axis (MGBA) is a bidirectional communication network between gut microbes and their host. Many environmental and host-related factors affect the gut microbiota. Dysbiosis is defined as compositional and functional alterations of the gut microbiota that contribute to the pathogenesis, progression and treatment responses to disease. Dysbiosis occurs when perturbations of microbiota composition and function exceed the ability of microbiota and its host to restore a symbiotic state. Dysbiosis leads to dysfunctional signaling of the MGBA, which regulates the development and the function of the host's immune, metabolic, and nervous systems. Dysbiosis-induced dysfunction of the MGBA is seen with aging and stroke, and is linked to the development of common stroke risk factors such as obesity, diabetes, and atherosclerosis. Changes in the gut microbiota are also seen in response to stroke, and may impair recovery after injury. This review will begin with an overview of the tools used to study the MGBA with a discussion on limitations and potential experimental confounders. Relevant MGBA components are introduced and summarized for a better understanding of age-related changes in MGBA signaling and its dysfunction after stroke. We will then focus on the relationship between the MGBA and aging, highlighting that all components of the MGBA undergo age-related alterations that can be influenced by or even driven by the gut microbiota. In the final section, the current clinical and preclinical evidence for the role of MGBA signaling in the development of stroke risk factors such as obesity, diabetes, hypertension, and frailty are summarized, as well as microbiota changes with stroke in experimental and clinical populations. We conclude by describing the current understanding of microbiota-based therapies for stroke including the use of pre-/pro-biotics and supplementations with bacterial metabolites. Ongoing progress in this new frontier of biomedical sciences will lead to an improved understanding of the MGBA's impact on human health and disease.
Collapse
Affiliation(s)
- Pedram Honarpisheh
- Department of Neurology, University of Texas McGovern Medical School, Houston (P.H., L.D.M.)
| | - Robert M Bryan
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX (R.M.B.)
| | - Louise D McCullough
- Department of Neurology, University of Texas McGovern Medical School, Houston (P.H., L.D.M.)
| |
Collapse
|
11
|
Molecular regulation of neuroinflammation in glaucoma: Current knowledge and the ongoing search for new treatment targets. Prog Retin Eye Res 2022; 87:100998. [PMID: 34348167 PMCID: PMC8803988 DOI: 10.1016/j.preteyeres.2021.100998] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022]
Abstract
Neuroinflammation relying on the inflammatory responses of glial cells has emerged as an impactful component of the multifactorial etiology of neurodegeneration in glaucoma. It has become increasingly evident that despite early adaptive and reparative features of glial responses, prolonged reactivity of the resident glia, along with the peripheral immune cells, create widespread toxicity to retinal ganglion cell (RGC) axons, somas, and synapses. As much as the synchronized responses of astrocytes and microglia to glaucoma-related stress or neuron injury, their bi-directional interactions are critical to build and amplify neuroinflammation and to dictate the neurodegenerative outcome. Although distinct molecular programs regulate somatic and axonal degeneration in glaucoma, inhibition of neurodegenerative inflammation can provide a broadly beneficial treatment strategy to rescue RGC integrity and function. Since inflammatory toxicity and mitochondrial dysfunction are converging etiological paths that can boost each other and feed into a vicious cycle, anti-inflammatory treatments may also offer a multi-target potential. This review presents an overview of the current knowledge on neuroinflammation in glaucoma with particular emphasis on the cell-intrinsic and cell-extrinsic factors involved in the reciprocal regulation of glial responses, the interdependence between inflammatory and mitochondrial routes of neurodegeneration, and the research aspects inspiring for prospective immunomodulatory treatments. With the advent of powerful technologies, ongoing research on molecular and functional characteristics of glial responses is expected to accumulate more comprehensive and complementary information and to rapidly move the field forward to safe and effective modulation of the glial pro-inflammatory activities, while restoring or augmenting the glial immune-regulatory and neurosupport functions.
Collapse
|
12
|
Bunch KL, Abdelrahman AA, Caldwell RB, Caldwell RW. Novel Therapeutics for Diabetic Retinopathy and Diabetic Macular Edema: A Pathophysiologic Perspective. Front Physiol 2022; 13:831616. [PMID: 35250632 PMCID: PMC8894892 DOI: 10.3389/fphys.2022.831616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) and diabetic macular edema (DME) are retinal complications of diabetes that can lead to loss of vision and impaired quality of life. The current gold standard therapies for treatment of DR and DME focus on advanced disease, are invasive, expensive, and can trigger adverse side-effects, necessitating the development of more effective, affordable, and accessible therapies that can target early stage disease. The pathogenesis and pathophysiology of DR is complex and multifactorial, involving the interplay between the effects of hyperglycemia, hyperlipidemia, hypoxia, and production of reactive oxygen species (ROS) in the promotion of neurovascular dysfunction and immune cell polarization to a proinflammatory state. The pathophysiology of DR provides several therapeutic targets that have the potential to attenuate disease progression. Current novel DR and DME therapies under investigation include erythropoietin-derived peptides, inducers of antioxidant gene expression, activators of nitric oxide/cyclic GMP signaling pathways, and manipulation of arginase activity. This review aims to aid understanding of DR and DME pathophysiology and explore novel therapies that capitalize on our knowledge of these diabetic retinal complications.
Collapse
Affiliation(s)
- Katharine L. Bunch
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Ammar A. Abdelrahman
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Ruth B. Caldwell
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - R. William Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
- *Correspondence: R. William Caldwell,
| |
Collapse
|
13
|
Fouda AY, Eldahshan W, Xu Z, Lemtalsi T, Shosha E, Zaidi SA, Abdelrahman AA, Cheng PNM, Narayanan SP, Caldwell RW, Caldwell RB. Preclinical investigation of Pegylated arginase 1 as a treatment for retina and brain injury. Exp Neurol 2022; 348:113923. [PMID: 34780773 PMCID: PMC9122100 DOI: 10.1016/j.expneurol.2021.113923] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/12/2021] [Accepted: 11/08/2021] [Indexed: 02/03/2023]
Abstract
Arginase 1 (A1) is the enzyme that hydrolyzes the amino acid, L-arginine, to ornithine and urea. We have previously shown that A1 deletion worsens retinal ischemic injury, suggesting a protective role of A1. In this translational study, we aimed to study the utility of systemic pegylated A1 (PEG-A1, recombinant human arginase linked to polyethylene glycol) treatment in mouse models of acute retinal and brain injury. Cohorts of WT mice were subjected to retinal ischemia-reperfusion (IR) injury, traumatic optic neuropathy (TON) or brain cerebral ischemia via middle cerebral artery occlusion (MCAO) and treated with intraperitoneal injections of PEG-A1 or vehicle (PEG only). Drug penetration into retina and brain tissues was measured by western blotting and immunolabeling for PEG. Neuroprotection was measured in a blinded fashion by quantitation of NeuN (neuronal marker) immunolabeling of retina flat-mounts and brain infarct area using triphenyl tetrazolium chloride (TTC) staining. Furthermore, ex vivo retina explants and in vitro retina neuron cultures were subjected to oxygen-glucose deprivation (OGD) followed by reoxygenation (R) and treated with PEG-A1. PEG-A1 given systemically did not cross the intact blood-retina/brain barriers in sham controls but reached the retina and brain after injury. PEG-A1 provided neuroprotection after retinal IR injury, TON and cerebral ischemia. PEG-A1 treatment was also neuroprotective in retina explants subjected to OGD/R but did not improve survival in retinal neuronal cultures exposed to OGD/R. In summary, systemic PEG-A1 administration is neuroprotective and provides an excellent route to deliver the drug to the retina and the brain after acute injury.
Collapse
Affiliation(s)
- Abdelrahman Y Fouda
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Wael Eldahshan
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Zhimin Xu
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Tahira Lemtalsi
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Esraa Shosha
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Syed Ah Zaidi
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Ammar A Abdelrahman
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Paul Ning-Man Cheng
- Bio-cancer Treatment International, 511-513, Bioinformatics Building, Hong Kong Science Park, Tai Po, Hong Kong, China
| | - S Priya Narayanan
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Cellular Biology & Anatomy, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA; Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA, United States
| | - R William Caldwell
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Cellular Biology & Anatomy, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
14
|
Honig MG, Del Mar NA, Henderson DL, O'Neal D, Doty JB, Cox R, Li C, Perry AM, Moore BM, Reiner A. Raloxifene Modulates Microglia and Rescues Visual Deficits and Pathology After Impact Traumatic Brain Injury. Front Neurosci 2021; 15:701317. [PMID: 34776838 PMCID: PMC8585747 DOI: 10.3389/fnins.2021.701317] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/07/2021] [Indexed: 11/29/2022] Open
Abstract
Mild traumatic brain injury (TBI) involves widespread axonal injury and activation of microglia, which initiates secondary processes that worsen the TBI outcome. The upregulation of cannabinoid type-2 receptors (CB2) when microglia become activated allows CB2-binding drugs to selectively target microglia. CB2 inverse agonists modulate activated microglia by shifting them away from the harmful pro-inflammatory M1 state toward the helpful reparative M2 state and thus can stem secondary injury cascades. We previously found that treatment with the CB2 inverse agonist SMM-189 after mild TBI in mice produced by focal cranial blast rescues visual deficits and the optic nerve axon loss that would otherwise result. We have further shown that raloxifene, which is Food and Drug Administration (FDA)-approved as an estrogen receptor modulator to treat osteoporosis, but also possesses CB2 inverse agonism, yields similar benefit in this TBI model through its modulation of microglia. As many different traumatic events produce TBI in humans, it is widely acknowledged that diverse animal models must be used in evaluating possible therapies. Here we examine the consequences of TBI created by blunt impact to the mouse head for visual function and associated pathologies and assess raloxifene benefit. We found that mice subjected to impact TBI exhibited decreases in contrast sensitivity and the B-wave of the electroretinogram, increases in light aversion and resting pupil diameter, and optic nerve axon loss, which were rescued by daily injection of raloxifene at 5 or 10 mg/ml for 2 weeks. Raloxifene treatment was associated with reduced M1 activation and/or enhanced M2 activation in retina, optic nerve, and optic tract after impact TBI. Our results suggest that the higher raloxifene dose, in particular, may be therapeutic for the optic nerve by enhancing the phagocytosis of axonal debris that would otherwise promote inflammation, thereby salvaging less damaged axons. Our current work, together with our prior studies, shows that microglial activation drives secondary injury processes after both impact and cranial blast TBI and raloxifene mitigates microglial activation and visual system injury in both cases. The results thus provide a strong basis for phase 2 human clinical trials evaluating raloxifene as a TBI therapy.
Collapse
Affiliation(s)
- Marcia G Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nobel A Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Desmond L Henderson
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Dylan O'Neal
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - John B Doty
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Rachel Cox
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chunyan Li
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Aaron M Perry
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Bob M Moore
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
15
|
Zhu L, Zhou H, Xu F, Yang H, Li P, Sheng Y, Liu P, Kong W, Liu X, Yang L, Liu L, Liu X. Hepatic Ischemia-Reperfusion Impairs Blood-Brain Barrier Partly Due to Release of Arginase From Injured Liver. Front Pharmacol 2021; 12:724471. [PMID: 34721021 PMCID: PMC8548691 DOI: 10.3389/fphar.2021.724471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 09/13/2021] [Indexed: 11/20/2022] Open
Abstract
Aim: Hepatic ischemia-reperfusion (HIR) induces remote organs injury, including the brain. The homeostasis of the brain is maintained by the blood-brain barrier (BBB); thus, we aimed to investigate whether HIR impaired BBB and attempted to elucidate its underlying mechanism. Methods: Cell viability of human cerebral microvascular endothelial cells (hCMEC/D3) was measured following 24 h incubation with a serum of HIR rat undergoing 1 h ischemia and 4 h reperfusion, liver homogenate, or lysate of primary hepatocytes of the rat. The liver homogenate was precipitated using (NH4)2SO4 followed by separation on three columns and electrophoresis to identify the toxic molecule. Cell activity, apoptosis, proliferation, cell cycle, and expressions of proteins related to cell cycle were measured in hCMEC/D3 cells incubated with identified toxic molecules. HIR rats undergoing 1 h ischemia and 24 h reperfusion were developed to determine the release of an identified toxic molecule. BBB function was indexed as permeability to fluorescein and brain water. Endothelial cell proliferation and expressions of proteins related to the cell cycle in cerebral microvessels were measured by immunofluorescence and western blot. Results: Toxic molecule to BBB in the liver was identified to be arginase. Arginase inhibitor nor-NOHA efficiently attenuated hCMEC/D3 damage caused by liver homogenate and serum of HIR rats. Both arginase and serum of HIR rats significantly lowered arginine (Arg) in the culture medium. Arg addition efficiently attenuated the impairment of hCMEC/D3 caused by arginase or Arg deficiency, demonstrating that arginase impaired hCMEC/D3 via depriving Arg. Both arginase and Arg deficiency damaged hCMEC/D3 cells by inhibiting cell proliferation, retarding the cell cycle to G1 phase, and downregulating expressions of cyclin A, cyclin D, CDK2, and CDK4. HIR notably increased plasma arginase activity and lowered Arg level, increased the BBB permeability accompanied with enhanced brain water, and decreased the proliferative cells (marked by Ki67) in cerebral microvessels (marked by CD31) and protein expressions of cyclin A, cyclin D, CDK2 and CDK4 in isolated brain microvessels. Oral supplement of Arg remarkably attenuated these HIR-induced alterations. Conclusion: HIR leads to substantial release of arginase from the injured liver and then deprives systemic Arg. The Arg deficiency further impairs BBB via inhibiting the proliferation of brain microvascular endothelial cells by cell cycle arrest.
Collapse
Affiliation(s)
- Liang Zhu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Han Zhou
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Feng Xu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hanyu Yang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ping Li
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yun Sheng
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Peihua Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Weimin Kong
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaonan Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lu Yang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
16
|
Tomita Y, Usui-Ouchi A, Nilsson AK, Yang J, Ko M, Hellström A, Fu Z. Metabolism in Retinopathy of Prematurity. Life (Basel) 2021; 11:1119. [PMID: 34832995 PMCID: PMC8620873 DOI: 10.3390/life11111119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Retinopathy of prematurity is defined as retinal abnormalities that occur during development as a consequence of disturbed oxygen conditions and nutrient supply after preterm birth. Both neuronal maturation and retinal vascularization are impaired, leading to the compensatory but uncontrolled retinal neovessel growth. Current therapeutic interventions target the hypoxia-induced neovessels but negatively impact retinal neurons and normal vessels. Emerging evidence suggests that metabolic disturbance is a significant and underexplored risk factor in the disease pathogenesis. Hyperglycemia and dyslipidemia correlate with the retinal neurovascular dysfunction in infants born prematurely. Nutritional and hormonal supplementation relieve metabolic stress and improve retinal maturation. Here we focus on the mechanisms through which metabolism is involved in preterm-birth-related retinal disorder from clinical and experimental investigations. We will review and discuss potential therapeutic targets through the restoration of metabolic responses to prevent disease development and progression.
Collapse
Affiliation(s)
- Yohei Tomita
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.T.); (J.Y.); (M.K.)
| | - Ayumi Usui-Ouchi
- Department of Ophthalmology, Juntendo University Urayasu Hospital, Chiba 279-0021, Japan;
| | - Anders K. Nilsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 19 Gothenburg, Sweden; (A.K.N.); (A.H.)
| | - Jay Yang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.T.); (J.Y.); (M.K.)
| | - Minji Ko
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.T.); (J.Y.); (M.K.)
| | - Ann Hellström
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 19 Gothenburg, Sweden; (A.K.N.); (A.H.)
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.T.); (J.Y.); (M.K.)
| |
Collapse
|
17
|
Lee D, Jeong H, Miwa Y, Shinojima A, Katada Y, Tsubota K, Kurihara T. Retinal dysfunction induced in a mouse model of unilateral common carotid artery occlusion. PeerJ 2021; 9:e11665. [PMID: 34221738 PMCID: PMC8223895 DOI: 10.7717/peerj.11665] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Background Retinal ischemic stresses are associated with the pathogenesis of various retinal vascular diseases. To investigate pathological mechanisms of retinal ischemia, reproducible, robust and clinically significant experimental rodent models are highly needed. Previously, we established a stable murine model of chronic hypoperfusion retinal injuries by permanent unilateral common carotid artery occlusion (UCCAO) and demonstrated chronic pathological processes in the ischemic retina after the occlusion; however, retinal functional deficits and other acute retinal ischemic injuries by UCCAO still remain obscure. In this study, we attempted to examine retinal functional changes as well as acute retinal ischemic alterations such as retinal thinning, gliosis and cell death after UCCAO. Methods Adult mice (male C57BL/6, 6–8 weeks old) were subjected to UCCAO in the right side, and retinal function was primarily measured using electroretinography for 14 days after the surgery. Furthermore, retinal thinning, gliosis and cell death were investigated using optical coherence tomography, immunohistochemistry and TUNEL assay, respectively. Results Functional deficits in the unilateral right retina started to be seen 7 days after the occlusion. Specifically, the amplitude of b-wave dramatically decreased while that of a-wave was slightly affected. 14 days after the occlusion, the amplitudes of both waves and oscillatory potentials were significantly detected decreased in the unilateral right retina. Even though a change in retinal thickness was not dramatically observed among all the eyes, retinal gliosis and cell death in the unilateral right retina were substantially observed after UCCAO. Conclusions Along with previous retinal ischemic results in this model, UCCAO can stimulate retinal ischemia leading to functional, morphological and molecular changes in the retina. This model can be useful for the investigation of pathological mechanisms for human ischemic retinopathies and furthermore can be utilized to test new drugs for various ischemic ocular diseases.
Collapse
Affiliation(s)
- Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Heonuk Jeong
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Yukihiro Miwa
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan.,Animal eye-care, Tokyo Animal Eye Clinic, Tokyo, Japan
| | - Ari Shinojima
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Yusaku Katada
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan.,Tsubota Laboratory, Inc., Tokyo, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
18
|
Shosha E, Fouda AY, Lemtalsi T, Haigh S, Fulton D, Ibrahim A, Al-Shabrawey M, Caldwell RW, Caldwell RB. Endothelial arginase 2 mediates retinal ischemia/reperfusion injury by inducing mitochondrial dysfunction. Mol Metab 2021; 53:101273. [PMID: 34139341 PMCID: PMC8274341 DOI: 10.1016/j.molmet.2021.101273] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/05/2021] [Accepted: 06/11/2021] [Indexed: 12/19/2022] Open
Abstract
Objective Retinal ischemic disease is a major cause of vision loss. Current treatment options are limited to late-stage diseases, and the molecular mechanisms of the initial insult are not fully understood. We have previously shown that the deletion of the mitochondrial arginase isoform, arginase 2 (A2), limits neurovascular injury in models of ischemic retinopathy. Here, we investigated the involvement of A2-mediated alterations in mitochondrial dynamics and function in the pathology. Methods We used wild-type (WT), global A2 knockout (A2KO-) mice, cell-specific A2 knockout mice subjected to retinal ischemia/reperfusion (I/R), and bovine retinal endothelial cells (BRECs) subjected to an oxygen-glucose deprivation/reperfusion (OGD/R) insult. We used western blotting to measure levels of cell stress and death markers and the mitochondrial fragmentation protein, dynamin related protein 1 (Drp1). We also used live cell mitochondrial labeling and Seahorse XF analysis to evaluate mitochondrial fragmentation and function, respectively. Results We found that the global deletion of A2 limited the I/R-induced disruption of retinal layers, fundus abnormalities, and albumin extravasation. The specific deletion of A2 in endothelial cells was protective against I/R-induced neurodegeneration. The OGD/R insult in BRECs increased A2 expression and induced cell stress and cell death, along with decreased mitochondrial respiration, increased Drp1 expression, and mitochondrial fragmentation. The overexpression of A2 in BREC also decreased mitochondrial respiration, promoted increases in the expression of Drp1, mitochondrial fragmentation, and cell stress and resulted in decreased cell survival. In contrast, the overexpression of the cytosolic isoform, arginase 1 (A1), did not affect these parameters. Conclusions This study is the first to show that A2 in endothelial cells mediates retinal ischemic injury through a mechanism involving alterations in mitochondrial dynamics and function. Ischemic retinopathy is a common feature of blinding eye disease. Arginase 2 overexpression in endothelial cells induces mitochondrial dysfunction. Endothelial-specific arginase 2 deletion improves neuronal survival after ischemia. Endothelial cell arginase 2 plays a crucial role in ischemic retinal injury.
Collapse
Affiliation(s)
- Esraa Shosha
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Abdelrahman Y Fouda
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Tahira Lemtalsi
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Stephen Haigh
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - David Fulton
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Ahmed Ibrahim
- Vision Discovery Institute, Augusta University, Augusta, GA, USA; Wayne State University, Department of Ophthalmology, Visual, and Anatomical Sciences, Department of Pharmacology, Detroit, MI, USA; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed Al-Shabrawey
- Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Oral Biology, Dental College of Georgia, Augusta, GA, USA
| | - R William Caldwell
- Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
19
|
Li T, Zhang W, Hu E, Sun Z, Li P, Yu Z, Zhu X, Zheng F, Xing Z, Xia Z, He F, Luo J, Tang T, Wang Y. Integrated metabolomics and network pharmacology to reveal the mechanisms of hydroxysafflor yellow A against acute traumatic brain injury. Comput Struct Biotechnol J 2021; 19:1002-1013. [PMID: 33613866 PMCID: PMC7868816 DOI: 10.1016/j.csbj.2021.01.033] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/02/2021] [Accepted: 01/20/2021] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) has become a leading cause of mortality, morbidity and disability worldwide. Hydroxysafflor yellow A (HSYA) is effective in treating TBI, but the potential mechanisms require further exploration. We aimed to reveal the mechanisms of HSYA against acute TBI by an integrated strategy combining metabolomics with network pharmacology. A controlled cortical impact (CCI) rat model was established, and neurological functions were evaluated. Metabolomics of brain tissues was used to identify differential metabolites, and the metabolic pathways were enriched by MetaboAnalyst. Then, network pharmacology was applied to dig out the potential targets against TBI induced by HSYA. The integrated network of metabolomics and network pharmacology was constructed based on Cytoscape. Finally, the obtained key targets were verified by molecular docking. HSYA alleviated the neurological deficits of TBI. Fifteen potentially significant metabolites were found to be involved in the therapeutic effects of HSYA against acute TBI. Most of these metabolites were regulated to recover after HSYA treatment. We found 10 hub genes according to network pharmacology, which was partly consistent with the metabolomics findings. Further integrated analysis focused on 4 key targets, including NOS1, ACHE, PTGS2 and XDH, as well as their related core metabolites and pathways. Molecular docking showed high affinities between key targets and HSYA. Region-specific metabolic alterations in the cortex and hippocampus were illuminated. This study reveals the complicated mechanisms of HSYA against acute TBI. Our work provides a novel paradigm to identify the potential mechanisms of pharmacological effects derived from a natural compound.
Collapse
Affiliation(s)
- Teng Li
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - En Hu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhengji Sun
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Pengfei Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Zhe Yu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiaofei Zhu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Fei Zheng
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhihua Xing
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zian Xia
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Feng He
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiekun Luo
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Tao Tang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yang Wang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
20
|
Chen J, Wang X, Hu J, Du J, Dordoe C, Zhou Q, Huang W, Guo R, Han F, Guo K, Ye S, Lin L, Li X. FGF20 Protected Against BBB Disruption After Traumatic Brain Injury by Upregulating Junction Protein Expression and Inhibiting the Inflammatory Response. Front Pharmacol 2021; 11:590669. [PMID: 33568994 PMCID: PMC7868342 DOI: 10.3389/fphar.2020.590669] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
Disruption of the blood-brain barrier (BBB) and the cerebral inflammatory response occurring after traumatic brain injury (TBI) facilitate further brain damage, which leads to long-term complications of TBI. Fibroblast growth factor 20 (FGF20), a neurotrophic factor, plays important roles in brain development and neuronal homeostasis. The aim of the current study was to assess the protective effects of FGF20 on TBI via BBB maintenance. In the present study, recombinant human FGF20 (rhFGF20) reduced neurofunctional deficits, brain edema, Evans blue extravasation and neuroinflammation in a TBI mouse model. In an in vitro TNF-α-induced human brain microvascular endothelial cell (HBMEC) model of BBB disruption, rhFGF20 reduced paracellular permeability and increased trans-endothelial electrical resistance (TEER). Both in the TBI mouse model and in vitro, rhFGF20 increased the expression of proteins composing in BBB-associated tight junctions (TJs) and adherens junctions (AJs), and decreased the inflammatory response, which protected the BBB integrity. Notably, rhFGF20 preserved BBB function by activating the AKT/GSK3β pathway and inhibited the inflammatory response by regulating the JNK/NFκB pathway. Thus, FGF20 is a potential candidate treatment for TBI that protects the BBB by upregulating junction protein expression and inhibiting the inflammatory response.
Collapse
Affiliation(s)
- Jun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingting Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Confidence Dordoe
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qiulin Zhou
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenting Huang
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ruili Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fanyi Han
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Kaiming Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shasha Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Wenzhou, China
| |
Collapse
|
21
|
Ma C, Hunt JB, Selenica MLB, Sanneh A, Sandusky-Beltran LA, Watler M, Daas R, Kovalenko A, Liang H, Placides D, Cao C, Lin X, Orr MB, Zhang B, Gensel JC, Feola DJ, Gordon MN, Morgan D, Bickford PC, Lee DC. Arginase 1 Insufficiency Precipitates Amyloid- β Deposition and Hastens Behavioral Impairment in a Mouse Model of Amyloidosis. Front Immunol 2021; 11:582998. [PMID: 33519806 PMCID: PMC7840571 DOI: 10.3389/fimmu.2020.582998] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/23/2020] [Indexed: 12/17/2022] Open
Abstract
Alzheimer’s disease (AD) includes several hallmarks comprised of amyloid-β (Aβ) deposition, tau neuropathology, inflammation, and memory impairment. Brain metabolism becomes uncoupled due to aging and other AD risk factors, which ultimately lead to impaired protein clearance and aggregation. Increasing evidence indicates a role of arginine metabolism in AD, where arginases are key enzymes in neurons and glia capable of depleting arginine and producing ornithine and polyamines. However, currently, it remains unknown if the reduction of arginase 1 (Arg1) in myeloid cell impacts amyloidosis. Herein, we produced haploinsufficiency of Arg1 by the hemizygous deletion in myeloid cells using Arg1fl/fl and LysMcreTg/+ mice crossed with APP Tg2576 mice. Our data indicated that Arg1 haploinsufficiency promoted Aβ deposition, exacerbated some behavioral impairment, and decreased components of Ragulator-Rag complex involved in mechanistic target of rapamycin complex 1 (mTORC1) signaling and autophagy. Additionally, Arg1 repression and arginine supplementation both impaired microglial phagocytosis in vitro. These data suggest that proper function of Arg1 and arginine metabolism in myeloid cells remains essential to restrict amyloidosis.
Collapse
Affiliation(s)
- Chao Ma
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Sanders-Brown Center on Aging, Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Jerry B Hunt
- Sanders-Brown Center on Aging, Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Maj-Linda B Selenica
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States.,Sanders-Brown Center on Aging, Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Awa Sanneh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Leslie A Sandusky-Beltran
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Mallory Watler
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Rana Daas
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Andrii Kovalenko
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Huimin Liang
- Sanders-Brown Center on Aging, Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Devon Placides
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Chuanhai Cao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Xiaoyang Lin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Michael B Orr
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Bei Zhang
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States.,Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - David J Feola
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Marcia N Gordon
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Dave Morgan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Paula C Bickford
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Research Service, James A. Haley Veterans Affairs Hospital, Tampa, FL, United States
| | - Daniel C Lee
- Sanders-Brown Center on Aging, Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| |
Collapse
|
22
|
Oxidative Stress and Vascular Dysfunction in the Retina: Therapeutic Strategies. Antioxidants (Basel) 2020; 9:antiox9080761. [PMID: 32824523 PMCID: PMC7465265 DOI: 10.3390/antiox9080761] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022] Open
Abstract
Many retinal diseases, such as diabetic retinopathy, glaucoma, and age-related macular (AMD) degeneration, are associated with elevated reactive oxygen species (ROS) levels. ROS are important intracellular signaling molecules that regulate numerous physiological actions, including vascular reactivity and neuron function. However, excessive ROS formation has been linked to vascular endothelial dysfunction, neuron degeneration, and inflammation in the retina. ROS can directly modify cellular molecules and impair their function. Moreover, ROS can stimulate the production of inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) causing inflammation and cell death. However, there are various compounds with direct or indirect antioxidant activity that have been used to reduce ROS accumulation in animal models and humans. In this review, we report on the physiological and pathophysiological role of ROS in the retina with a special focus on the vascular system. Moreover, we present therapeutic approaches for individual retinal diseases targeting retinal signaling pathways involving ROS.
Collapse
|
23
|
López-Bernal Á, García-Tejera O, Testi L, Villalobos FJ. Genotypic variability in radial resistance to water flow in olive roots and its response to temperature variations. TREE PHYSIOLOGY 2020; 40:445-453. [PMID: 32031664 DOI: 10.1093/treephys/tpaa010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/30/2019] [Accepted: 01/23/2020] [Indexed: 06/10/2023]
Abstract
As radial root resistance (Rp) represents one of the key components of the soil-plant-atmosphere continuum resistance catena modulating water transport, understanding its control is essential for physiologists, modelers and breeders. Reports of Rp, however, are still scarce and scattered in the scientific literature. In this study, we assessed genetic variability in Rp and its dependence on temperature in five widely used olive cultivars. In a first experiment, cultivar differences in Rp at 25 °C were evaluated from flow-pressure measurements in excised roots and subsequent analysis of root traits. In a second experiment, similar determinations were performed continually over a 5-h period in which temperature was gradually increased from 12 to 32 °C, enabling the assessment of Rp response to changing temperature. Despite some variability, our results did not show statistical differences in Rp among cultivars in the first experiment. In the second, cultivar differences in Rp were not significant at 12 °C, but they became so as temperature increased. Furthermore, the changes in Rp between 12 and 32 °C were higher than those expected by the temperature-driven decrease in water viscosity, with the degree of that change differing among cultivars. Also, Rp at 25 °C reached momentarily in the second experiment was consistently higher than in the first at that same, but fixed, temperature. Overall, our results suggest that there is limited variability in Rp among the studied cultivars when plants have been exposed to a given temperature for sufficient time. Temperature-induced variation in Rp might thus be partly explained by changes in membrane permeability that occur slowly, which explains why our values at 25 °C differed between experiments. The observed cultivar differences in Rp with warming also indicate faster acclimation of Rp to temperature changes in some cultivars than others.
Collapse
Affiliation(s)
- Á López-Bernal
- Departamento de Agronomía, Universidad de Córdoba, Campus de Rabanales, Edificio C4, 14071 Córdoba, Spain
| | - O García-Tejera
- Efficient Use of Water Program, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Parc de Gardeny, Edifici Fruitcentre, 25003 Lleida, Spain
| | - L Testi
- Instituto de Agricultura Sostenible (IAS), Consejo Superior de Investigaciones Científicas (CSIC), Av. Menéndez Pidal s/n, 14080 Córdoba, Spain
| | - F J Villalobos
- Departamento de Agronomía, Universidad de Córdoba, Campus de Rabanales, Edificio C4, 14071 Córdoba, Spain
- Instituto de Agricultura Sostenible (IAS), Consejo Superior de Investigaciones Científicas (CSIC), Av. Menéndez Pidal s/n, 14080 Córdoba, Spain
| |
Collapse
|
24
|
Pillar S, Moisseiev E, Sokolovska J, Grzybowski A. Recent Developments in Diabetic Retinal Neurodegeneration: A Literature Review. J Diabetes Res 2020; 2020:5728674. [PMID: 34151902 PMCID: PMC7787838 DOI: 10.1155/2020/5728674] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/11/2020] [Accepted: 11/24/2020] [Indexed: 02/08/2023] Open
Abstract
Neurodegeneration plays a significant role in the complex pathology of diabetic retinopathy. Evidence suggests the onset of neurodegeneration occurs early on in the disease, and so a greater understanding of the process is essential for prompt detection and targeted therapies. Neurodegeneration is a common pathway of assorted processes, including activation of inflammatory pathways, reduction of neuroprotective factors, DNA damage, and apoptosis. Oxidative stress and formation of advanced glycation end products amplify these processes and are elevated in the setting of hyperglycemia, hyperlipidemia, and glucose variability. These key pathophysiologic mechanisms are discussed, as well as diagnostic modalities and novel therapeutic avenues, with an emphasis on recent discoveries. The aim of this article is to highlight the crucial role of neurodegeneration in diabetic retinopathy and to review the molecular basis for this neuronal dysfunction, its diagnostic features, and the progress currently made in relevant therapeutic interventions.
Collapse
Affiliation(s)
- Shani Pillar
- Department of Ophthalmology, Meir Medical Center, Kfar Saba, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elad Moisseiev
- Department of Ophthalmology, Meir Medical Center, Kfar Saba, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Andrzej Grzybowski
- Department of Ophthalmology, University of Warmia and Mazury, Olsztyn, Poland
- Institute for Research in Ophthalmology, Foundation for Ophthalmology Development, Poznan, Poland
| |
Collapse
|