1
|
Kadam R, Gupta M, Lazarov O, Prabhakar BS. Brain-immune interactions: implication for cognitive impairments in Alzheimer's disease and autoimmune disorders. J Leukoc Biol 2024; 116:1269-1290. [PMID: 38869088 DOI: 10.1093/jleuko/qiae134] [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: 11/13/2023] [Revised: 05/09/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024] Open
Abstract
Progressive memory loss and cognitive dysfunction, encompassing deficits in learning, memory, problem solving, spatial reasoning, and verbal expression, are characteristics of Alzheimer's disease and related dementia. A wealth of studies has described multiple roles of the immune system in the development or exacerbation of dementia. Individuals with autoimmune disorders can also develop cognitive dysfunction, a phenomenon termed "autoimmune dementia." Together, these findings underscore the pivotal role of the neuroimmune axis in both Alzheimer's disease and related dementia and autoimmune dementia. The dynamic interplay between adaptive and innate immunity, both in and outside the brain, significantly affects the etiology and progression of these conditions. Multidisciplinary research shows that cognitive dysfunction arises from a bidirectional relationship between the nervous and immune systems, though the specific mechanisms that drive cognitive impairments are not fully understood. Intriguingly, this reciprocal regulation occurs at multiple levels, where neuronal signals can modulate immune responses, and immune system-related processes can influence neuronal viability and function. In this review, we consider the implications of autoimmune responses in various autoimmune disorders and Alzheimer's disease and explore their effects on brain function. We also discuss the diverse cellular and molecular crosstalk between the brain and the immune system, as they may shed light on potential triggers of peripheral inflammation, their effect on the integrity of the blood-brain barrier, and brain function. Additionally, we assess challenges and possibilities associated with developing immune-based therapies for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Rashmi Kadam
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| |
Collapse
|
2
|
Stankovic S, Lazic A, Parezanovic M, Stevanovic M, Pavlovic S, Stojiljkovic M, Klaassen K. Transcriptome Profiling of Phenylalanine-Treated Human Neuronal Model: Spotlight on Neurite Impairment and Synaptic Connectivity. Int J Mol Sci 2024; 25:10019. [PMID: 39337507 PMCID: PMC11431966 DOI: 10.3390/ijms251810019] [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: 07/30/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Phenylketonuria (PKU) is the most common inherited disorder of amino acid metabolism, characterized by high levels of phenylalanine (Phe) in the blood and brain, leading to cognitive impairment without treatment. Nevertheless, Phe-mediated brain dysfunction is not fully understood. The objective of this study was to address gene expression alterations due to excessive Phe exposure in the human neuronal model and provide molecular advances in PKU pathophysiology. Hence, we performed NT2/D1 differentiation in culture, and, for the first time, we used Phe-treated NT2-derived neurons (NT2/N) as a novel model for Phe-mediated neuronal impairment. NT2/N were treated with 1.25 mM, 2.5 mM, 5 mM, 10 mM, and 30 mM Phe and subjected to whole-mRNA short-read sequencing. Differentially expressed genes (DEGs) were analyzed and enrichment analysis was performed. Under three different Phe concentrations (2.5 mM, 5 mM, and 10 mM), DEGs pointed to the PREX1, LRP4, CDC42BPG, GPR50, PRMT8, RASGRF2, and CDH6 genes, placing them in the context of PKU for the first time. Enriched processes included dendrite and axon impairment, synaptic transmission, and membrane assembly. In contrast to these groups, the 30 mM Phe treatment group clearly represented the neurotoxicity of Phe, exhibiting enrichment in apoptotic pathways. In conclusion, we established NT2/N as a novel model for Phe-mediated neuronal dysfunction and outlined the Phe-induced gene expression changes resulting in neurite impairment and altered synaptic connectivity.
Collapse
Affiliation(s)
- Sara Stankovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.); (A.L.); (M.P.); (M.S.); (S.P.); (M.S.)
| | - Andrijana Lazic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.); (A.L.); (M.P.); (M.S.); (S.P.); (M.S.)
| | - Marina Parezanovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.); (A.L.); (M.P.); (M.S.); (S.P.); (M.S.)
| | - Milena Stevanovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.); (A.L.); (M.P.); (M.S.); (S.P.); (M.S.)
- Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Studentski trg 16, 11158 Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Kneza Mihaila 35, 11001 Belgrade, Serbia
| | - Sonja Pavlovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.); (A.L.); (M.P.); (M.S.); (S.P.); (M.S.)
| | - Maja Stojiljkovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.); (A.L.); (M.P.); (M.S.); (S.P.); (M.S.)
| | - Kristel Klaassen
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (S.S.); (A.L.); (M.P.); (M.S.); (S.P.); (M.S.)
| |
Collapse
|
3
|
DePew AT, Bruckner JJ, O'Connor-Giles KM, Mosca TJ. Neuronal LRP4 directs the development, maturation and cytoskeletal organization of Drosophila peripheral synapses. Development 2024; 151:dev202517. [PMID: 38738619 PMCID: PMC11190576 DOI: 10.1242/dev.202517] [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: 11/09/2023] [Accepted: 05/02/2024] [Indexed: 05/14/2024]
Abstract
Synaptic development requires multiple signaling pathways to ensure successful connections. Transmembrane receptors are optimally positioned to connect the synapse and the rest of the neuron, often acting as synaptic organizers to synchronize downstream events. One such organizer, the LDL receptor-related protein LRP4, is a cell surface receptor that has been most well-studied postsynaptically at mammalian neuromuscular junctions. Recent work, however, identified emerging roles, but how LRP4 acts as a presynaptic organizer and the downstream mechanisms of LRP4 are not well understood. Here, we show that LRP4 functions presynaptically at Drosophila neuromuscular synapses, acting in motoneurons to instruct pre- and postsynaptic development. Loss of presynaptic LRP4 results in multiple defects, impairing active zone organization, synapse growth, physiological function, microtubule organization, synaptic ultrastructure and synapse maturation. We further demonstrate that LRP4 promotes most aspects of presynaptic development via a downstream SR-protein kinase, SRPK79D. These data demonstrate a function for presynaptic LRP4 as a peripheral synaptic organizer, highlight a downstream mechanism conserved with its CNS function in Drosophila, and underscore previously unappreciated but important developmental roles for LRP4 in cytoskeletal organization, synapse maturation and active zone organization.
Collapse
Affiliation(s)
- Alison T. DePew
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Joseph J. Bruckner
- Cell and Molecular Biology Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kate M. O'Connor-Giles
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Timothy J. Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
4
|
Liu ZY, Li YQ, Wang DL, Wang Y, Qiu WT, Qiu YY, Zhang HL, You QL, Liu SM, Liang QN, Wu EJ, Hu BJ, Sun XD. Agrin-Lrp4 pathway in hippocampal astrocytes restrains development of temporal lobe epilepsy through adenosine signaling. Cell Biosci 2024; 14:66. [PMID: 38783336 PMCID: PMC11112884 DOI: 10.1186/s13578-024-01241-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Human patients often experience an episode of serious seizure activity, such as status epilepticus (SE), prior to the onset of temporal lobe epilepsy (TLE), suggesting that SE can trigger the development of epilepsy. Yet, the underlying mechanisms are not fully understood. The low-density lipoprotein receptor related protein (Lrp4), a receptor for proteoglycan-agrin, has been indicated to modulate seizure susceptibility. However, whether agrin-Lrp4 pathway also plays a role in the development of SE-induced TLE is not clear. METHODS Lrp4f/f mice were crossed with hGFAP-Cre and Nex-Cre mice to generate brain conditional Lrp4 knockout mice (hGFAP-Lrp4-/-) and pyramidal neuron specific knockout mice (Nex-Lrp4-/-). Lrp4 was specifically knocked down in hippocampal astrocytes by injecting AAV virus carrying hGFAP-Cre into the hippocampus. The effects of agrin-Lrp4 pathway on the development of SE-induced TLE were evaluated on the chronic seizure model generated by injecting kainic acid (KA) into the amygdala. The spontaneous recurrent seizures (SRS) in mice were video monitored. RESULTS We found that Lrp4 deletion from the brain but not from the pyramidal neurons elevated the seizure threshold and reduced SRS numbers, with no change in the stage or duration of SRS. More importantly, knockdown of Lrp4 in the hippocampal astrocytes after SE induction decreased SRS numbers. In accord, direct injection of agrin into the lateral ventricle of control mice but not mice with Lrp4 deletion in hippocampal astrocytes also increased the SRS numbers. These results indicate a promoting effect of agrin-Lrp4 signaling in hippocampal astrocytes on the development of SE-induced TLE. Last, we observed that knockdown of Lrp4 in hippocampal astrocytes increased the extracellular adenosine levels in the hippocampus 2 weeks after SE induction. Blockade of adenosine A1 receptor in the hippocampus by DPCPX after SE induction diminished the effects of Lrp4 on the development of SE-induced TLE. CONCLUSION These results demonstrate a promoting role of agrin-Lrp4 signaling in hippocampal astrocytes in the development of SE-induced development of epilepsy through elevating adenosine levels. Targeting agrin-Lrp4 signaling may serve as a potential therapeutic intervention strategy to treat TLE.
Collapse
Affiliation(s)
- Zi-Yang Liu
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuan-Quan Li
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology of the Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Die-Lin Wang
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ying Wang
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wan-Ting Qiu
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yu-Yang Qiu
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - He-Lin Zhang
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiang-Long You
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shi-Min Liu
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiu-Ni Liang
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Er-Jian Wu
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bing-Jie Hu
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Xiang-Dong Sun
- School of Basic Medical Sciences, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and Department of GFNeurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Chen BH, Lin ZY, Zeng XX, Jiang YH, Geng F. LRP4-related signalling pathways and their regulatory role in neurological diseases. Brain Res 2024; 1825:148705. [PMID: 38065285 DOI: 10.1016/j.brainres.2023.148705] [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: 10/04/2023] [Revised: 11/17/2023] [Accepted: 12/03/2023] [Indexed: 01/28/2024]
Abstract
The mechanism of action of low-density lipoprotein receptor related protein 4 (LRP4) is mediated largely via the Agrin-LRP4-MuSK signalling pathway in the nervous system. LRP4 contributes to the development of synapses in the peripheral nervous system (PNS). It interacts with signalling molecules such as the amyloid beta-protein precursor (APP) and the wingless type protein (Wnt). Its mechanisms of action are complex and mediated via interaction between the pre-synaptic motor neuron and post-synaptic muscle cell in the PNS, which enhances the development of the neuromuscular junction (NMJ). LRP4 may function differently in the central nervous system (CNS) than in the PNS, where it regulates ATP and glutamate release via astrocytes. It mayaffect the growth and development of the CNS by controlling the energy metabolism. LRP4 interacts with Agrin to maintain dendrite growth and density in the CNS. The goal of this article is to review the current studies involving relevant LRP4 signaling pathways in the nervous system. The review also discusses the clinical and etiological roles of LRP4 in neurological illnesses, such as myasthenia gravis, Alzheimer's disease and epilepsy. In this review, we provide a theoretical foundation for the pathogenesis and therapeutic application of LRP4 in neurologic diseases.
Collapse
Affiliation(s)
- Bai-Hui Chen
- Department of Physiology, Shantou University Medical College, Shantou 515041, China
| | - Ze-Yu Lin
- Department of Physiology, Shantou University Medical College, Shantou 515041, China
| | - Xiao-Xue Zeng
- Department of Physiology, Shantou University Medical College, Shantou 515041, China
| | - Yi-Han Jiang
- Department of Physiology, Shantou University Medical College, Shantou 515041, China
| | - Fei Geng
- Department of Physiology, Shantou University Medical College, Shantou 515041, China; Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
6
|
DePew AT, Bruckner JJ, O’Connor-Giles KM, Mosca TJ. Neuronal LRP4 directs the development, maturation, and cytoskeletal organization of peripheral synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.03.564481. [PMID: 37961323 PMCID: PMC10635100 DOI: 10.1101/2023.11.03.564481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Synapse development requires multiple signaling pathways to accomplish the myriad of steps needed to ensure a successful connection. Transmembrane receptors on the cell surface are optimally positioned to facilitate communication between the synapse and the rest of the neuron and often function as synaptic organizers to synchronize downstream signaling events. One such organizer, the LDL receptor-related protein LRP4, is a cell surface receptor most well-studied postsynaptically at mammalian neuromuscular junctions. Recent work, however, has identified emerging roles for LRP4 as a presynaptic molecule, but how LRP4 acts as a presynaptic organizer, what roles LRP4 plays in organizing presynaptic biology, and the downstream mechanisms of LRP4 are not well understood. Here we show that LRP4 functions presynaptically at Drosophila neuromuscular synapses, acting in motor neurons to instruct multiple aspects of pre- and postsynaptic development. Loss of presynaptic LRP4 results in a range of developmental defects, impairing active zone organization, synapse growth, physiological function, microtubule organization, synaptic ultrastructure, and synapse maturation. We further demonstrate that LRP4 promotes most aspects of presynaptic development via a downstream SR-protein kinase, SRPK79D. SRPK79D overexpression suppresses synaptic defects associated with loss of lrp4. These data demonstrate a function for LRP4 as a peripheral synaptic organizer acting presynaptically, highlight a downstream mechanism conserved with its CNS function, and indicate previously unappreciated roles for LRP4 in cytoskeletal organization, synapse maturation, and active zone organization, underscoring its developmental importance.
Collapse
Affiliation(s)
- Alison T. DePew
- Dept. of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Joseph J. Bruckner
- Cell and Molecular Biology Training Program, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Kate M. O’Connor-Giles
- Department of Neuroscience, Brown University, Providence, RI 02912 USA
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Timothy J. Mosca
- Dept. of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107 USA
- Lead Contact
| |
Collapse
|
7
|
Naomi R, Yazid MD, Teoh SH, Balan SS, Shariff H, Kumar J, Bahari H, Embong H. Dietary Polyphenols as a Protection against Cognitive Decline: Evidence from Animal Experiments; Mechanisms and Limitations. Antioxidants (Basel) 2023; 12:antiox12051054. [PMID: 37237920 DOI: 10.3390/antiox12051054] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/27/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Emerging evidence suggests that cognitive impairments may result from various factors, such as neuroinflammation, oxidative stress, mitochondrial damage, impaired neurogenesis, synaptic plasticity, blood-brain barrier (BBB) disruption, amyloid β protein (Aβ) deposition, and gut dysbiosis. Meanwhile, dietary polyphenol intake in a recommended dosage has been suggested to reverse cognitive dysfunction via various pathways. However, excessive intake of polyphenols could trigger unwanted adverse effects. Thus, this review aims to outline possible causes of cognitive impairments and how polyphenols alleviate memory loss via various pathways based on in vivo experimental studies. Thus, to identify potentially relevant articles, the keywords (1) nutritional polyphenol intervention NOT medicine AND neuron growth OR (2) dietary polyphenol AND neurogenesis AND memory impairment OR (3) polyphenol AND neuron regeneration AND memory deterioration (Boolean operators) were used in the Nature, PubMed, Scopus, and Wiley online libraries. Based on the inclusion and exclusion criteria, 36 research papers were selected to be further reviewed. The outcome of all the studies included supports the statement of appropriate dosage by taking into consideration gender differences, underlying conditions, lifestyle, and causative factors for cognitive decline, which will significantly boost memory power. Therefore, this review recapitulates the possible causes of cognitive decline, the mechanism of polyphenols involving various signaling pathways in modulating the memory, gut dysbiosis, endogenous antioxidants, bioavailability, dosage, and safety efficacy of polyphenols. Hence, this review is expected to provide a basic understanding of therapeutic development for cognitive impairments in the future.
Collapse
Affiliation(s)
- Ruth Naomi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Soo Huat Teoh
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia
| | - Santhra Segaran Balan
- Department of Diagnostic and Allied Health Sciences, Faculty of Health and Life Sciences, Management and Science University, Shah Alam 40100, Malaysia
| | - Halim Shariff
- Faculty of Health Sciences, University Technology Mara (UITM) Pulau Pinang, Bertam Campus, Kepala Batas 13200, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Hasnah Bahari
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Hashim Embong
- Department of Emergency Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
8
|
Prömer J, Barresi C, Herbst R. From phosphorylation to phenotype - Recent key findings on kinase regulation, downstream signaling and disease surrounding the receptor tyrosine kinase MuSK. Cell Signal 2023; 104:110584. [PMID: 36608736 DOI: 10.1016/j.cellsig.2022.110584] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/31/2022] [Indexed: 01/04/2023]
Abstract
Muscle-specific kinase (MuSK) is the key regulator of neuromuscular junction development. MuSK acts via several distinct pathways and is responsible for pre- and postsynaptic differentiation. MuSK is unique among receptor tyrosine kinases as activation and signaling are particularly tightly regulated. Initiation of kinase activity requires Agrin, a heparan sulphate proteoglycan derived from motor neurons, the low-density lipoprotein receptor-related protein-4 (Lrp4) and the intracellular adaptor protein Dok-7. There is a great knowledge gap between MuSK activation and downstream signaling. Recent studies using omics techniques have addressed this knowledge gap, thereby greatly contributing to a better understanding of MuSK signaling. Impaired MuSK signaling causes severe muscle weakness as described in congenital myasthenic syndromes or myasthenia gravis but the underlying pathophysiology is often unclear. This review focuses on recent advances in deciphering MuSK activation and downstream signaling. We further highlight latest break-throughs in understanding and treatment of MuSK-related disorders and discuss the role of MuSK in non-muscle tissue.
Collapse
Affiliation(s)
- Jakob Prömer
- Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Cinzia Barresi
- Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ruth Herbst
- Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Geng S, Paul F, Kowalczyk I, Raimundo S, Sporbert A, Mamo TM, Hammes A. Balancing WNT signalling in early forebrain development: The role of LRP4 as a modulator of LRP6 function. Front Cell Dev Biol 2023; 11:1173688. [PMID: 37091972 PMCID: PMC10119419 DOI: 10.3389/fcell.2023.1173688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
The specification of the forebrain relies on the precise regulation of WNT/ß-catenin signalling to support neuronal progenitor cell expansion, patterning, and morphogenesis. Imbalances in WNT signalling activity in the early neuroepithelium lead to congenital disorders, such as neural tube defects (NTDs). LDL receptor-related protein (LRP) family members, including the well-studied receptors LRP5 and LRP6, play critical roles in modulating WNT signalling capacity through tightly regulated interactions with their co-receptor Frizzled, WNT ligands, inhibitors and intracellular WNT pathway components. However, little is known about the function of LRP4 as a potential modulator of WNT signalling in the central nervous system. In this study, we investigated the role of LRP4 in the regulation of WNT signalling during early mouse forebrain development. Our results demonstrate that LRP4 can modulate LRP5- and LRP6-mediated WNT signalling in the developing forebrain prior to the onset of neurogenesis at embryonic stage 9.5 and is therefore essential for accurate neural tube morphogenesis. Specifically, LRP4 functions as a genetic modifier for impaired mitotic activity and forebrain hypoplasia, but not for NTDs in LRP6-deficient mutants. In vivo and in vitro data provide evidence that LRP4 is a key player in fine-tuning WNT signalling capacity and mitotic activity of mouse neuronal progenitors and of human retinal pigment epithelial (hTERT RPE-1) cells. Our data demonstrate the crucial roles of LRP4 and LRP6 in regulating WNT signalling and forebrain development and highlight the need to consider the interaction between different signalling pathways to understand the underlying mechanisms of disease. The findings have significant implications for our mechanistic understanding of how LRPs participate in controlling WNT signalling.
Collapse
Affiliation(s)
- Shuang Geng
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Fabian Paul
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Izabela Kowalczyk
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Sandra Raimundo
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Tamrat Meshka Mamo
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- *Correspondence: Tamrat Meshka Mamo, ; Annette Hammes,
| | - Annette Hammes
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- *Correspondence: Tamrat Meshka Mamo, ; Annette Hammes,
| |
Collapse
|
10
|
Yan M, Xiong M, Wu Y, Lin D, Chen P, Chen J, Liu Z, Zhang H, Ren D, Fei E, Lai X, Zou S, Wang S. LRP4 is required for the olfactory association task in the piriform cortex. Cell Biosci 2022; 12:54. [PMID: 35526070 PMCID: PMC9080164 DOI: 10.1186/s13578-022-00792-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 04/18/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Low-density lipoprotein receptor-related protein 4 (LRP4) plays a critical role in the central nervous system (CNS), including hippocampal synaptic plasticity, maintenance of excitatory synaptic transmission, fear regulation, as well as long-term potentiation (LTP).
Results
In this study, we found that Lrp4 was highly expressed in layer II of the piriform cortex. Both body weight and brain weight decreased in Lrp4ECD/ECD mice without TMD (Transmembrane domain) and ICD (intracellular domain) of LRP4. However, in the piriform cortical neurons of Lrp4ECD/ECD mice, the spine density increased, and the frequency of both mEPSC (miniature excitatory postsynaptic current) and sEPSC (spontaneous excitatory postsynaptic current) was enhanced. Intriguingly, finding food in the buried food-seeking test was prolonged in both Lrp4ECD/ECD mice and Lrp4 cKO (conditional knockout of Lrp4 in the piriform cortex) mice.
Conclusions
This study indicated that the full length of LRP4 in the piriform cortex was necessary for maintaining synaptic plasticity and the integrity of olfactory function.
Collapse
|
11
|
Passarella D, Ciampi S, Di Liberto V, Zuccarini M, Ronci M, Medoro A, Foderà E, Frinchi M, Mignogna D, Russo C, Porcile C. Low-Density Lipoprotein Receptor-Related Protein 8 at the Crossroad between Cancer and Neurodegeneration. Int J Mol Sci 2022; 23:ijms23168921. [PMID: 36012187 PMCID: PMC9408729 DOI: 10.3390/ijms23168921] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
The low-density-lipoprotein receptors represent a family of pleiotropic cell surface receptors involved in lipid homeostasis, cell migration, proliferation and differentiation. The family shares common structural features but also has significant differences mainly due to tissue-specific interactors and to peculiar proteolytic processing. Among the receptors in the family, recent studies place low-density lipoprotein receptor-related protein 8 (LRP8) at the center of both neurodegenerative and cancer-related pathways. From one side, its overexpression has been highlighted in many types of cancer including breast, gastric, prostate, lung and melanoma; from the other side, LRP8 has a potential role in neurodegeneration as apolipoprotein E (ApoE) and reelin receptor, which are, respectively, the major risk factor for developing Alzheimer’s disease (AD) and the main driver of neuronal migration, and as a γ-secretase substrate, the main enzyme responsible for amyloid formation in AD. The present review analyzes the contributions of LDL receptors, specifically of LRP8, in both cancer and neurodegeneration, pointing out that depending on various interactions and peculiar processing, the receptor can contribute to both proliferative and neurodegenerative processes.
Collapse
Affiliation(s)
- Daniela Passarella
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Silvia Ciampi
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Valentina Di Liberto
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, 90133 Palermo, Italy
| | - Mariachiara Zuccarini
- Department of Medical Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100 Chieti, Italy
| | - Maurizio Ronci
- Department of Pharmacy, University of Chieti-Pescara, 66100 Chieti, Italy
| | - Alessandro Medoro
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Emanuele Foderà
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Monica Frinchi
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, 90133 Palermo, Italy
| | - Donatella Mignogna
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Claudio Russo
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
- Correspondence: ; Tel.: +39-0874404897
| | - Carola Porcile
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| |
Collapse
|
12
|
Chin EW, Ma Q, Ruan H, Chin C, Somasundaram A, Zhang C, Liu C, Lewis MD, White M, Smith TL, Battersby M, Yao WD, Lu XY, Arap W, Licinio J, Wong ML. The epigenetic reader PHF21B modulates murine social memory and synaptic plasticity-related genes. JCI Insight 2022; 7:e158081. [PMID: 35866480 PMCID: PMC9431697 DOI: 10.1172/jci.insight.158081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Synaptic dysfunction is a manifestation of several neurobehavioral and neurological disorders. A major therapeutic challenge lies in uncovering the upstream regulatory factors controlling synaptic processes. Plant homeodomain (PHD) finger proteins are epigenetic readers whose dysfunctions are implicated in neurological disorders. However, the molecular mechanisms linking PHD protein deficits to disease remain unclear. Here, we generated a PHD finger protein 21B-depleted (Phf21b-depleted) mutant CRISPR mouse model (hereafter called Phf21bΔ4/Δ4) to examine Phf21b's roles in the brain. Phf21bΔ4/Δ4 animals exhibited impaired social memory. In addition, reduced expression of synaptic proteins and impaired long-term potentiation were observed in the Phf21bΔ4/Δ4 hippocampi. Transcriptome profiling revealed differential expression of genes involved in synaptic plasticity processes. Furthermore, we characterized a potentially novel interaction of PHF21B with histone H3 trimethylated lysine 36 (H3K36me3), a histone modification associated with transcriptional activation, and the transcriptional factor CREB. These results establish PHF21B as an important upstream regulator of synaptic plasticity-related genes and a candidate therapeutic target for neurobehavioral dysfunction in mice, with potential applications in human neurological and psychiatric disorders.
Collapse
Affiliation(s)
| | - Qi Ma
- Department of Psychiatry and Behavioral Sciences
| | - Hongyu Ruan
- Department of Psychiatry and Behavioral Sciences
| | | | | | - Chunling Zhang
- Department of Neuroscience & Physiology, Norton College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Chunyu Liu
- Department of Psychiatry and Behavioral Sciences
- Department of Neuroscience & Physiology, Norton College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Martin D. Lewis
- Neuropsychiatric Laboratory, Lifelong Health Research Unit, and
| | - Melissa White
- Gene Editing Research Unit, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- SA Genome Editing Facility, University of Adelaide, Adelaide, South Australia, Australia
| | - Tracey L. Smith
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Malcolm Battersby
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Wei-Dong Yao
- Department of Psychiatry and Behavioral Sciences
- Department of Neuroscience & Physiology, Norton College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Xin-Yun Lu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Julio Licinio
- Department of Psychiatry and Behavioral Sciences
- Department of Neuroscience & Physiology, Norton College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Ma-Li Wong
- Department of Psychiatry and Behavioral Sciences
- Department of Neuroscience & Physiology, Norton College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| |
Collapse
|
13
|
van Kessel E, Berendsen S, Baumfalk AE, Venugopal H, Krijnen EA, Spliet WGM, van Hecke W, Giuliani F, Seute T, van Zandvoort MJE, Snijders TJ, Robe PA. Tumor-related molecular determinants of neurocognitive deficits in patients with diffuse glioma. Neuro Oncol 2022; 24:1660-1670. [PMID: 35148403 PMCID: PMC9527514 DOI: 10.1093/neuonc/noac036] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Cognitive impairment is a common and debilitating symptom in patients with diffuse glioma, and is the result of multiple factors. We hypothesized that molecular tumor characteristics influence neurocognitive functioning (NCF), and aimed to identify tumor-related markers of NCF in diffuse glioma patients. METHODS We examined the relation between cognitive performance (executive function, memory, and psychomotor speed) and intratumoral expression levels of molecular markers in treatment-naive patients with diffuse glioma. We performed a single-center study in a consecutive cohort, through a two-step design: (1) hypothesis-free differential expression and gene set enrichment analysis to identify candidate oncogenetic markers for cognitive impairment. Nineteen molecular markers of interest were derived from this set of genes, as well as from prior knowledge; (2) correlation of cognitive performance to intratumoral expression levels of these nineteen molecular markers, measured with immunohistochemistry. RESULTS From 708 included patients with immunohistochemical data, we performed an in-depth analysis of neuropsychological data in 197, and differential expression analysis in 65 patients. After correcting for tumor volume and location, we found significant associations between expression levels of CD3 and IDH-1 and psychomotor speed; between IDH-1, ATRX, NLGN3, BDNF, CK2Beta, EAAT1, GAT-3, SRF, and memory performance; and between IDH-1, P-STAT5b, NLGN3, CK2Beta, and executive functioning. P-STAT5b, CD163, CD3, and Semaphorin-3A were independently associated after further correction for histopathological grade. CONCLUSION Molecular characteristics of glioma can be independent determinants of patients' cognitive functioning. This suggests that besides tumor volume, location, and histological grade, variations in glioma biology influence cognitive performance through mechanisms that include perturbation of neuronal communication. These results pave the way towards targeted cognition improving therapies in neuro-oncology.
Collapse
Affiliation(s)
- Emma van Kessel
- Corresponding Author: Emma van Kesssel, MD, University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, internal address G03.232, PO Box 85500, 3508 XC Utrecht, The Netherlands ()
| | - Sharon Berendsen
- University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Neurology & Neurosurgery, Utrecht, The Netherlands
| | - Anniek E Baumfalk
- University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Neurology & Neurosurgery, Utrecht, The Netherlands
| | - Hema Venugopal
- University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Neurology & Neurosurgery, Utrecht, The Netherlands
| | - Eva A Krijnen
- University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Neurology & Neurosurgery, Utrecht, The Netherlands
| | - Wim G M Spliet
- University Medical Center Utrecht, Department of Pathology, Utrecht, The Netherlands
| | - Wim van Hecke
- University Medical Center Utrecht, Department of Pathology, Utrecht, The Netherlands
| | - Fabrizio Giuliani
- University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Neurology & Neurosurgery, Utrecht, The Netherlands
| | - Tatjana Seute
- University Medical Center Utrecht, UMC Utrecht Brain Center, Department of Neurology & Neurosurgery, Utrecht, The Netherlands
| | | | | | | |
Collapse
|
14
|
Hu D, Guo Y, Wu M, Ma Y, Jing W. Manuscript Title: GDAP2 overexpression affects the development of neurons and dysregulates neuronal excitatory synaptic transmission. Neuroscience 2022; 488:32-43. [DOI: 10.1016/j.neuroscience.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 11/28/2022]
|
15
|
Benvenga S, Antonelli A, Fallahi P, Bonanno C, Rodolico C, Guarneri F. Amino acid sequence homology between thyroid autoantigens and central nervous system proteins: Implications for the steroid-responsive encephalopathy associated with autoimmune thyroiditis. J Clin Transl Endocrinol 2021; 26:100274. [PMID: 34849350 PMCID: PMC8609095 DOI: 10.1016/j.jcte.2021.100274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 10/15/2021] [Accepted: 10/27/2021] [Indexed: 11/13/2022] Open
Abstract
Alpha-enolase, aldehyde reductase-I and dimethylargininase-I are SREAT autoantigens. Molecular mimicry between thyroid and CNS autoantigens is hypothesized in SREAT. Homology with TSH-R, Tg and TPO exists for 6, 27 and 47 of 46,809 CNS-proteins. The above homologies are often in epitope-containing parts of thyroid autoantigens. Most of the above proteins are expressed in CNS regions which are altered in SREAT.
A few patients with Hashimoto’s thyroiditis or Graves’ disease develop a multiform syndrome of the central nervous system (CNS) termed Hashimoto’s encephalopathy or steroid-responsive encephalopathy associated with autoimmune thyroid disease (HE/SREAT). They have high levels of thyroid autoantibodies (TgAb, TPOAb and/or TSH-R-Ab) in blood and cerebrospinal fluid. Autoantibodies against alpha-enolase, aldehyde reductase-I (AKRIA) and/or dimethylargininase-I (DDAHI), proteins expressed in the CNS among other tissues, were detected in the blood and, when searched, in the cerebrospinal fluid of HE/SREAT patients. Recently, we reported that alpha-enolase, AKRIA and DDAHI share local sequence homology with each of the three autoantigens (TgAb, TPOAb, TSH-R-Ab), often in epitope-containing segments of the thyroid autoantigens. We hypothesized that there might be additional CNS-expressed proteins homologous to thyroid autoantigens, possibly overlapping known epitopes of the thyroid autoantigens. We used bioinformatic methods to address this hypothesis. Six, 27 and 47 of 46,809 CNS-expressed proteins share homology with TSH-R, Tg and TPO, respectively. The homologous regions often contain epitopes, and some match regions of thyroid autoantigens which have homology with alpha-enolase, AKRIA and/or DDAHI. Several of the aforementioned proteins are present in CNS areas that show abnormalities at neuroimaging in HE/SREAT patients. Furthermore, autoantibodies against some of the said six, 27 and 47 proteins were reported to be associated with a number of autoimmune diseases. Not only we validated our hypothesis, but we think that such a variety of potential CNS targets for thyroid Ab against epitopes contained in regions that have local homology with CNS proteins may explain the polymorphic phenotypes of HE/SREAT. Only when elevated amounts of these Ab are synthesized and trespass the blood-brain barrier, HE/SREAT appears. This might explain why HE/SREAT is so relatively rare.
Collapse
|
16
|
Wang K, Wu J, Wang J, Jiang K. miR-485's anti-drug resistant epilepsy effects by regulating SV2A/PSD-95 and targeting ABCC1 and neuronal signaling-transduction proteins in hippocampus of rats. Brain Behav 2021; 11:e2247. [PMID: 34291586 PMCID: PMC8413801 DOI: 10.1002/brb3.2247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/23/2021] [Accepted: 05/25/2021] [Indexed: 12/14/2022] Open
Abstract
AIM Drug-resistant epilepsy (DRE), most subsequently developing refractory epilepsy, causes a significant burden to the society. microRNAs have been demonstrated as key regulators and therapeutic targets in epilepsy. Accordingly, the aim of the present study was to test whether miR-485 could be a potential target for DRE. METHODS AND RESULTS An in vivo DRE model was developed in Sprague-Dawley rats by lithium chloride-pilocarpine and screened by antiepileptic drugs. We found that miR-485-5p in hippocampus was significant downregulated at early stage and recovered to normal level at late stage of DRE. Overexpression of miR-485-5p in dentate gyrus (DG) of hippocampus in DRE rats could significantly decrease the frequency of seizures and the numbers of epileptiform spikes of hippocampal DG neuron, and could specifically decrease SV2A expression without affecting PSD-95 expression in DG. Furthermore, miR-485-5p overexpression could significantly downregulate the expression of efflux transporter related to multidrug resistance (ABCC1) in hippocampus at late stage of DRE. Finally, a specific expression pattern of neuronal signaling-transduction proteins (LRP4, MDM4, p53, and TMBIM1) for DRE was observed, and miR-485-5p overexpression could modulate these proteins' expression levels toward normal in hippocampus both at early and late stage of DRE. CONCLUSION Collectively, these results suggest that miR-485 was a potential target for anti-DRE, and this effects might be partially via miR-485-5p/homeostatic-synaptic plasticity-molecule axis and/or targeting efflux transporter (ABCC1) and other neuronal signaling-transduction proteins (LRP4, MDM4, p53, and TMBIM1).
Collapse
Affiliation(s)
- Kaixuan Wang
- Department of Pediatrics, Jinhua Central Hospital, Jinhua, China
| | - Jing Wu
- Department of Child Psychology, National Clinical Research Center For Child Health, The Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Jiangping Wang
- Department of Rehabilitation, National Clinical Research Center For Child Health, The Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Kewen Jiang
- Department of Child Psychology, National Clinical Research Center For Child Health, The Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Vergoossen DLE, Keo A, Mahfouz A, Huijbers MG. Timing and localization of myasthenia gravis-related gene expression. Eur J Neurosci 2021; 54:5574-5585. [PMID: 34228850 PMCID: PMC8457065 DOI: 10.1111/ejn.15382] [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: 03/26/2021] [Revised: 06/01/2021] [Accepted: 07/01/2021] [Indexed: 11/29/2022]
Abstract
Myasthenia gravis (MG) is an acquired autoimmune disorder caused by autoantibodies binding acetylcholine receptors (AChR), muscle‐specific kinase (MuSK), agrin or low‐density lipoprotein receptor‐related protein 4 (Lrp4). These autoantibodies inhibit neuromuscular transmission by blocking the function of these proteins and thereby cause fluctuating skeletal muscle weakness. Several reports suggest that these autoantibodies might also affect the central nervous system (CNS) in MG patients. A comprehensive overview of the timing and localization of the expression of MG‐related antigens in other organs is currently lacking. To investigate the spatio‐temporal expression of MG‐related genes outside skeletal muscle, we used in silico tools to assess public expression databases. Acetylcholine esterase, nicotinic AChR α1 subunit, agrin, collagen Q, downstream of kinase‐7, Lrp4, MuSK and rapsyn were included as MG‐related genes because of their well‐known involvement in either congenital or autoimmune MG. We investigated expression of MG‐related genes in (1) all human tissues using GTEx data, (2) specific brain regions, (3) neurodevelopmental stages, and (4) cell types using datasets from the Allen Institute for Brain Sciences. MG‐related genes show heterogenous spatio‐temporal expression patterns in the human body as well as in the CNS. For each of these genes, several (new) tissues, brain areas and cortical cell types with (relatively) high expression were identified suggesting a potential role for these genes outside skeletal muscle. The possible presence of MG‐related antigens outside skeletal muscle suggests that autoimmune MG, congenital MG or treatments targeting the same proteins may affect MG‐related protein function in other organs.
Collapse
Affiliation(s)
- Dana L E Vergoossen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Arlin Keo
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, The Netherlands.,Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
| | - Ahmed Mahfouz
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Computational Biology Center, Leiden University Medical Center, Leiden, The Netherlands.,Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
| | - Maartje G Huijbers
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.,Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
18
|
Hui TK, Lai XS, Dong X, Jing H, Liu Z, Fei E, Chen WB, Wang S, Ren D, Zou S, Wu HT, Pan BX. Ablation of Lrp4 in Schwann Cells Promotes Peripheral Nerve Regeneration in Mice. BIOLOGY 2021; 10:biology10060452. [PMID: 34063992 PMCID: PMC8223976 DOI: 10.3390/biology10060452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/16/2021] [Accepted: 05/18/2021] [Indexed: 11/16/2022]
Abstract
Low-density lipoprotein receptor-related protein 4 (Lrp4) is a critical protein involved in the Agrin-Lrp4-MuSK signaling pathway that drives the clustering of acetylcholine receptors (AChRs) at the neuromuscular junction (NMJ). Many studies have shown that Lrp4 also functions in kidney development, bone formation, nervous system development, etc. However, whether Lrp4 participates in nerve regeneration in mammals remains unknown. Herein, we show that Lrp4 is expressed in SCs and that conditional knockout (cKO) of Lrp4 in SCs promotes peripheral nerve regeneration. In Lrp4 cKO mice, the demyelination of SCs was accelerated, and the proliferation of SCs was increased in the injured nerve. Furthermore, we identified that two myelination-related genes, Krox-20 and Mpz, were downregulated more dramatically in the cKO group than in the control group. Our results elucidate a novel role of Lrp4 in peripheral nerve regeneration and thereby provide a potential therapeutic target for peripheral nerve recovery.
Collapse
Affiliation(s)
- Tian-Kun Hui
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Xin-Sheng Lai
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Xia Dong
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Hongyang Jing
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Ziyang Liu
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Erkang Fei
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Wen-Bing Chen
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Shunqi Wang
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Dongyan Ren
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Suqi Zou
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
- Correspondence: (S.Z.); (H.-T.W.); (B.-X.P.)
| | - Hai-Tao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
- Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
- Correspondence: (S.Z.); (H.-T.W.); (B.-X.P.)
| | - Bing-Xing Pan
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
- Correspondence: (S.Z.); (H.-T.W.); (B.-X.P.)
| |
Collapse
|
19
|
DePew AT, Mosca TJ. Conservation and Innovation: Versatile Roles for LRP4 in Nervous System Development. J Dev Biol 2021; 9:9. [PMID: 33799485 PMCID: PMC8006230 DOI: 10.3390/jdb9010009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023] Open
Abstract
As the nervous system develops, connections between neurons must form to enable efficient communication. This complex process of synaptic development requires the coordination of a series of intricate mechanisms between partner neurons to ensure pre- and postsynaptic differentiation. Many of these mechanisms employ transsynaptic signaling via essential secreted factors and cell surface receptors to promote each step of synaptic development. One such cell surface receptor, LRP4, has emerged as a synaptic organizer, playing a critical role in conveying extracellular signals to initiate diverse intracellular events during development. To date, LRP4 is largely known for its role in development of the mammalian neuromuscular junction, where it functions as a receptor for the synaptogenic signal Agrin to regulate synapse development. Recently however, LRP4 has emerged as a synapse organizer in the brain, where new functions for the protein continue to arise, adding further complexity to its already versatile roles. Additional findings indicate that LRP4 plays a role in disorders of the nervous system, including myasthenia gravis, amyotrophic lateral sclerosis, and Alzheimer's disease, demonstrating the need for further study to understand disease etiology. This review will highlight our current knowledge of how LRP4 functions in the nervous system, focusing on the diverse developmental roles and different modes this essential cell surface protein uses to ensure the formation of robust synaptic connections.
Collapse
Affiliation(s)
| | - Timothy J. Mosca
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| |
Collapse
|
20
|
Pharmacological upregulation of GLT-1 alleviates the cognitive impairments in the animal model of temporal lobe epilepsy. PLoS One 2021; 16:e0246068. [PMID: 33507976 PMCID: PMC7842975 DOI: 10.1371/journal.pone.0246068] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
It is known that hippocampal epileptogenesis is accompanied by hyperexcitability, glutamate-related neuronal dysfunctions and consequently cognitive deficits. However, the neuroprotective role of astrocytic glutamate uptake through the Glutamate Transporter-1 (GLT-1) remains to be unknown in these processes. Therefore, to assess the effect of glutamate uptake, pharmacological upregulation of GLT-1 using ceftriaxone administration (200 mg/kg/day, i.p, 5 days) was utilized in Li-PIL animal models of temporal lobe epilepsy (TLE). Glutamate concentration and glutamine synthetase activity were analyzed using biochemical assays. In addition, GLT-1 gene expression was assessed by RT-qPCR. Finally, cognitive function was studied using Morris water maze (MWM) test and novel object recognition task (NORT). Our results demonstrated that the acute phase of epileptogenesis (first 72 hours after Status Epilepticus) was accompanied by an increase in the hippocampal glutamate and downregulation of GLT-1 mRNA expression compared to controls. Ceftriaxone administration in epileptic animals led to a reduction of glutamate along with elevation of the level of glutamine synthetase activity and GLT-1 expression in the acute phase. In the chronic phase of epileptogenesis (4 weeks after Status Epilepticus), glutamate levels and GLT-1 expression were decreased compared to controls. Ceftriaxone treatment increased the levels of GLT-1 expression. Furthermore, impaired learning and memory ability in the chronic phase of epileptogenesis was rescued by Ceftriaxone administration. This study shows that astrocytic glutamate uptake can profoundly impact the processes of hippocampal epileptogenesis through the reduction of glutamate-induced excitotoxicity and consequently rescuing of cognitive deficits caused by epilepsy.
Collapse
|
21
|
Yan M, Guo A, Chen P, Jing H, Ren D, Zhong Y, Wu Y, Fei E, Lai X, Zou S, Wang S. LRP4 LDLα repeats of astrocyte enhance dendrite arborization of the neuron. Mol Brain 2020; 13:166. [PMID: 33302985 PMCID: PMC7730773 DOI: 10.1186/s13041-020-00708-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
The low-density lipoprotein receptor-related protein 4 (LRP4) is essential for inducing the neuromuscular junction (NMJ) formation in muscle fibers, and LRP4 plays a critical role in dendritic development and synaptogenesis in the central nervous system (CNS). As a single transmembrane protein, LRP4 contains an enormously sizeable extracellular domain (ECD), containing multiple LDLα repeats in the N-terminal of ECD. LRP4 only with extracellular domain acts as a similar mechanism of full-length LRP4 in muscles to stimulate acetylcholine receptor clustering. In this study, we elucidated that LDLα repeats of LRP4 maintained the body weight and survival rate. Dendritic branches of the pyramidal neurons in Lrp4-null mice with LRP4 LDLα repeats residue were more than in Lrp4-null mice without residual LRP4 domain. Supplement with conditioned medium from LRP4 LDLα overexpression cells, the primary culture pyramidal neurons achieved strong dendritic arborization ability. Besides, astrocytes with LRP4 LDLα repeats residue could promote pyramidal neuronal dendrite arborization in the primary co-cultured system. These observations signify that LRP4 LDLα repeats play a prominent underlying role in dendrite arborization.
Collapse
Affiliation(s)
- Min Yan
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,School of Basic Medical Sciences, Nanchang University, Nanchang, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Amin Guo
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Peng Chen
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Hongyang Jing
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Dongyan Ren
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Yanzi Zhong
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Yongqiang Wu
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Erkang Fei
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Xinsheng Lai
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Suqi Zou
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Shunqi Wang
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China. .,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
22
|
A Role of Low-Density Lipoprotein Receptor-Related Protein 4 (LRP4) in Astrocytic Aβ Clearance. J Neurosci 2020; 40:5347-5361. [PMID: 32457076 DOI: 10.1523/jneurosci.0250-20.2020] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/10/2020] [Accepted: 05/16/2020] [Indexed: 01/28/2023] Open
Abstract
Amyloid-β (Aβ) deposition occurs years before cognitive symptoms appear and is considered a cause of Alzheimer's disease (AD). The imbalance of Aβ production and clearance leads to Aβ accumulation and Aβ deposition. Increasing evidence indicates an important role of astrocytes, the most abundant cell type among glial cells in the brain, in Aβ clearance. We explored the role of low-density lipoprotein receptor-related protein 4 (LRP4), a member of the LDLR family, in AD pathology. We show that Lrp4 is specifically expressed in astrocytes and its levels in astrocytes were higher than those of Ldlr and Lrp1, both of which have been implicated in Aβ uptake. LRP4 was reduced in postmortem brain tissues of AD patients. Genetic deletion of the Lrp4 gene augmented Aβ plaques in 5xFAD male mice, an AD mouse model, and exacerbated the deficits in neurotransmission, synchrony between the hippocampus and PFC, and cognition. Mechanistically, LRP4 promotes Aβ uptake by astrocytes likely by interacting with ApoE. Together, our study demonstrates that astrocytic LRP4 plays an important role in Aβ pathology and cognitive function.SIGNIFICANCE STATEMENT This study investigates how astrocytes, a type of non-nerve cells in the brain, may contribute to Alzheimer's disease (AD) development. We demonstrate that the low-density lipoprotein receptor-related protein 4 (LRP4) is reduced in the brain of AD patients. Mimicking the reduced levels in an AD mouse model exacerbates cognitive impairment and increases amyloid aggregates that are known to damage the brain. We show that LRP4 could promote the clearance of amyloid protein by astrocytes. Our results reveal a previously unappreciated role of LRP4 in AD development.
Collapse
|
23
|
Zhou F, Yin G, Gao Y, Liu D, Xie J, Ouyang L, Fan Y, Yu H, Zha Z, Wang K, Shao L, Feng C, Fan G. Toxicity assessment due to prenatal and lactational exposure to lead, cadmium and mercury mixtures. ENVIRONMENT INTERNATIONAL 2019; 133:105192. [PMID: 31639605 DOI: 10.1016/j.envint.2019.105192] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/25/2019] [Accepted: 09/16/2019] [Indexed: 06/10/2023]
Abstract
The heavy metals lead (Pb), cadmium (Cd) and mercury (Hg) are common environmental pollutants that can be detected simultaneously in blood, serum, and urine samples from the general human population. However, there is limited information regarding toxicity of low-level exposure to Pb, Cd, and Hg mixtures. Our previous research showed the interaction of these three elements at low concentrations in vitro. In this study, we further evaluate early effects of low dose exposure to Pb, Cd, and Hg mixtures on the brain, heart, liver, kidney, and testicle in rats. Pregnant rats were exposed to various concentrations of heavy metal mixtures (MM) in drinking water, during gestation and lactation, and the impacts on offspring were measured at postnatal day 23. Our results showed that the concentrations of Pb, Cd, and Hg in the blood of rat pups were similar to those in the blood of the general human population. Additionally, the MM concentrations in their blood and brain significantly increased in a dose-dependent manner. MM exposure caused histopathological changes in the brain, liver, kidney and testicle. Statistically significant increases in liver CYP450 and PON1, kidney KIM1, and decrease in testicle SDH were observed. In the brain, significant increases were detected in oxidative stress, intracellular free calcium, and cell apoptosis. Further neurobehavioral testing revealed that MM exposure caused dose-dependent impairments in learning and memory as well as sensory perception. MM exposure also disrupted synapse remodeling, which may be associated with pathways involved in dendritic spine growth, maintenance, and elimination. These results suggested that exposure to Pb, Cd, and Hg mixtures, at human environmental exposure related levels, caused damage to multiple organs as well as impairments in neurobehavioral functions of rats. Our findings emphasize the need to control and regulate potential sources of heavy metal contamination.
Collapse
Affiliation(s)
- Fankun Zhou
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Guangming Yin
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Yanyan Gao
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Dong Liu
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Jie Xie
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Lu Ouyang
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Ying Fan
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Han Yu
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Zhipeng Zha
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Kai Wang
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Lijian Shao
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Chang Feng
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China
| | - Guangqin Fan
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
24
|
Cognitive dysfunction in mice with passively induced MuSK antibody seropositive myasthenia gravis. J Neurol Sci 2019; 399:15-21. [DOI: 10.1016/j.jns.2019.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/23/2019] [Accepted: 02/01/2019] [Indexed: 11/19/2022]
|
25
|
The role of agrin, Lrp4 and MuSK during dendritic arborization and synaptogenesis in cultured embryonic CNS neurons. Dev Biol 2019; 445:54-67. [DOI: 10.1016/j.ydbio.2018.10.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 01/06/2023]
|
26
|
Cardoso AL, Fernandes A, Aguilar-Pimentel JA, de Angelis MH, Guedes JR, Brito MA, Ortolano S, Pani G, Athanasopoulou S, Gonos ES, Schosserer M, Grillari J, Peterson P, Tuna BG, Dogan S, Meyer A, van Os R, Trendelenburg AU. Towards frailty biomarkers: Candidates from genes and pathways regulated in aging and age-related diseases. Ageing Res Rev 2018; 47:214-277. [PMID: 30071357 DOI: 10.1016/j.arr.2018.07.004] [Citation(s) in RCA: 324] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Use of the frailty index to measure an accumulation of deficits has been proven a valuable method for identifying elderly people at risk for increased vulnerability, disease, injury, and mortality. However, complementary molecular frailty biomarkers or ideally biomarker panels have not yet been identified. We conducted a systematic search to identify biomarker candidates for a frailty biomarker panel. METHODS Gene expression databases were searched (http://genomics.senescence.info/genes including GenAge, AnAge, LongevityMap, CellAge, DrugAge, Digital Aging Atlas) to identify genes regulated in aging, longevity, and age-related diseases with a focus on secreted factors or molecules detectable in body fluids as potential frailty biomarkers. Factors broadly expressed, related to several "hallmark of aging" pathways as well as used or predicted as biomarkers in other disease settings, particularly age-related pathologies, were identified. This set of biomarkers was further expanded according to the expertise and experience of the authors. In the next step, biomarkers were assigned to six "hallmark of aging" pathways, namely (1) inflammation, (2) mitochondria and apoptosis, (3) calcium homeostasis, (4) fibrosis, (5) NMJ (neuromuscular junction) and neurons, (6) cytoskeleton and hormones, or (7) other principles and an extensive literature search was performed for each candidate to explore their potential and priority as frailty biomarkers. RESULTS A total of 44 markers were evaluated in the seven categories listed above, and 19 were awarded a high priority score, 22 identified as medium priority and three were low priority. In each category high and medium priority markers were identified. CONCLUSION Biomarker panels for frailty would be of high value and better than single markers. Based on our search we would propose a core panel of frailty biomarkers consisting of (1) CXCL10 (C-X-C motif chemokine ligand 10), IL-6 (interleukin 6), CX3CL1 (C-X3-C motif chemokine ligand 1), (2) GDF15 (growth differentiation factor 15), FNDC5 (fibronectin type III domain containing 5), vimentin (VIM), (3) regucalcin (RGN/SMP30), calreticulin, (4) PLAU (plasminogen activator, urokinase), AGT (angiotensinogen), (5) BDNF (brain derived neurotrophic factor), progranulin (PGRN), (6) α-klotho (KL), FGF23 (fibroblast growth factor 23), FGF21, leptin (LEP), (7) miRNA (micro Ribonucleic acid) panel (to be further defined), AHCY (adenosylhomocysteinase) and KRT18 (keratin 18). An expanded panel would also include (1) pentraxin (PTX3), sVCAM/ICAM (soluble vascular cell adhesion molecule 1/Intercellular adhesion molecule 1), defensin α, (2) APP (amyloid beta precursor protein), LDH (lactate dehydrogenase), (3) S100B (S100 calcium binding protein B), (4) TGFβ (transforming growth factor beta), PAI-1 (plasminogen activator inhibitor 1), TGM2 (transglutaminase 2), (5) sRAGE (soluble receptor for advanced glycosylation end products), HMGB1 (high mobility group box 1), C3/C1Q (complement factor 3/1Q), ST2 (Interleukin 1 receptor like 1), agrin (AGRN), (6) IGF-1 (insulin-like growth factor 1), resistin (RETN), adiponectin (ADIPOQ), ghrelin (GHRL), growth hormone (GH), (7) microparticle panel (to be further defined), GpnmB (glycoprotein nonmetastatic melanoma protein B) and lactoferrin (LTF). We believe that these predicted panels need to be experimentally explored in animal models and frail cohorts in order to ascertain their diagnostic, prognostic and therapeutic potential.
Collapse
|
27
|
Yang Y, Tian X, Xu D, Zheng F, Lu X, Zhang Y, Ma Y, Li Y, Xu X, Zhu B, Wang X. GPR40 modulates epileptic seizure and NMDA receptor function. SCIENCE ADVANCES 2018; 4:eaau2357. [PMID: 30345361 PMCID: PMC6192686 DOI: 10.1126/sciadv.aau2357] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 09/12/2018] [Indexed: 05/21/2023]
Abstract
Epilepsy is a common neurological disease, and approximately 30% of patients do not respond adequately to antiepileptic drug treatment. Recent studies suggest that G protein-coupled receptor 40 (GPR40) is expressed in the central nervous system and is involved in the regulation of neurological function. However, the impact of GPR40 on epileptic seizures remains unclear. In this study, we first reported that GPR40 expression was increased in epileptic brains. In the kainic acid-induced epilepsy model, GPR40 activation after status epilepticus alleviated epileptic activity, whereas GPR40 inhibition showed the opposite effect. In the pentylenetetrazole-induced kindling model, susceptibility to epilepsy was reduced with GPR40 activation and increased with GPR40 inhibition. Whole-cell patch-clamp recordings demonstrated that GPR40 affected N-methyl-d-aspartate (NMDA) receptor-mediated synaptic transmission. Moreover, GPR40 regulated NR2A and NR2B expression on the surface of neurons. In addition, endocytosis of NMDA receptors and binding of GPR40 with NR2A and NR2B can be regulated by GPR40. Together, our findings indicate that GPR40 modulates epileptic seizures, providing a novel antiepileptic target.
Collapse
Affiliation(s)
- Yong Yang
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Department of Neurology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266003 Shandong, China
| | - Xin Tian
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Demei Xu
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fangshuo Zheng
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xi Lu
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yanke Zhang
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuanlin Ma
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yun Li
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xin Xu
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Binglin Zhu
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Corresponding author. (B.Z.); (X.W.)
| | - Xuefeng Wang
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing 100101, China
- Corresponding author. (B.Z.); (X.W.)
| |
Collapse
|
28
|
Fundamental Molecules and Mechanisms for Forming and Maintaining Neuromuscular Synapses. Int J Mol Sci 2018; 19:ijms19020490. [PMID: 29415504 PMCID: PMC5855712 DOI: 10.3390/ijms19020490] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 02/08/2023] Open
Abstract
The neuromuscular synapse is a relatively large synapse with hundreds of active zones in presynaptic motor nerve terminals and more than ten million acetylcholine receptors (AChRs) in the postsynaptic membrane. The enrichment of proteins in presynaptic and postsynaptic membranes ensures a rapid, robust, and reliable synaptic transmission. Over fifty years ago, classic studies of the neuromuscular synapse led to a comprehensive understanding of how a synapse looks and works, but these landmark studies did not reveal the molecular mechanisms responsible for building and maintaining a synapse. During the past two-dozen years, the critical molecular players, responsible for assembling the specialized postsynaptic membrane and regulating nerve terminal differentiation, have begun to be identified and their mechanism of action better understood. Here, we describe and discuss five of these key molecular players, paying heed to their discovery as well as describing their currently understood mechanisms of action. In addition, we discuss the important gaps that remain to better understand how these proteins act to control synaptic differentiation and maintenance.
Collapse
|
29
|
Ce O, Rs P, Ab W, S D, Cj W, Qm M, D L. Potential Link Between Proprotein Convertase Subtilisin/Kexin Type 9 and Alzheimer's Disease. ACTA ACUST UNITED AC 2018; 1. [PMID: 32352077 DOI: 10.31531/2581-4745.1000106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Alzheimer's disease [AD] is not only the most common neurodegenerative disease but is also currently incurable. Proprotein convertase subtilisin/kexin-9 [PCSK9] is an indirect regulator of plasma low density lipoprotein [LDL] levels controlling LDL receptor expression at the plasma membrane. PCSK9 also appears to regulate the development of glucose intolerance, insulin resistance, abdominal obesity, inflammation, and hypertension, conditions that have been identified as risk factors for AD. PCSK9 levels also depend on age, sex, and ethnic background, factors associated with AD. Herein, we will review indirect evidence that suggests a link between PCSK9 levels and AD.
Collapse
Affiliation(s)
- Oldham Ce
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Powell Rs
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Williams Ab
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Dixon S
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Wooten Cj
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Melendez Qm
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| | - Lopez D
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise [BRITE], College of Arts and Sciences, North Carolina Central University, Durham, USA
| |
Collapse
|
30
|
Ye XC, Hu JX, Li L, Li Q, Tang FL, Lin S, Sun D, Sun XD, Cui GY, Mei L, Xiong WC. Astrocytic Lrp4 (Low-Density Lipoprotein Receptor-Related Protein 4) Contributes to Ischemia-Induced Brain Injury by Regulating ATP Release and Adenosine-A 2AR (Adenosine A2A Receptor) Signaling. Stroke 2018; 49:165-174. [PMID: 29212737 PMCID: PMC5742060 DOI: 10.1161/strokeaha.117.018115] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 10/27/2017] [Accepted: 11/03/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND PURPOSE Lrp4 (low-density lipoprotein receptor-related protein 4) is predominantly expressed in astrocytes, where it regulates glutamatergic neurotransmission by suppressing ATP release. Here, we investigated Lrp4's function in ischemia/stroke-induced brain injury response, which includes glutamate-induced neuronal death and reactive astrogliosis. METHODS The brain-specific Lrp4 conditional knockout mice (Lrp4GFAP-Cre), astrocytic-specific Lrp4 conditional knockout mice (Lrp4GFAP-creER), and their control mice (Lrp4f/f) were subjected to photothrombotic ischemia and the transient middle cerebral artery occlusion. After ischemia/stroke, mice or their brain samples were subjected to behavior tests, brain histology, immunofluorescence staining, Western blot, and quantitative real-time polymerase chain reaction. In addition, primary astrocytes and neurons were cocultured with or without oxygen and glucose deprivation and in the presence or absence of the antagonist for adenosine-A2AR (adenosine A2A receptor) or ATP-P2X7R (P2X purinoceptor 7) signaling. Gliotransmitters, such as glutamate, d-serine, ATP, and adenosine, in the condition medium of cultured astrocytes were also measured. RESULTS Lrp4, largely expressed in astrocytes, was increased in response to ischemia/stroke. Both Lrp4GFAP-Cre and Lrp4GFAP-creER mice showed less brain injury, including reduced neuronal death, and impaired reactive astrogliosis. Mechanistically, Lrp4 conditional knockout in astrocytes increased ATP release and the production of ATP derivative, adenosine, which were further elevated by oxygen and glucose deprivation. Pharmacological inhibition of ATP-P2X7R or adenosine-A2AR signaling diminished Lrp4GFAP-creER's protective effect. CONCLUSIONS The astrocytic Lrp4 plays an important role in ischemic brain injury response. Lrp4 deficiency in astrocytes seems to be protective in response to ischemic brain injury, likely because of the increased ATP release and adenosine-A2AR signaling.
Collapse
Affiliation(s)
- Xin-Chun Ye
- From the Department of Neuroscience and Regenerative Medicine (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.) and Department of Neurology (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.), Medical College of Georgia, Augusta University; Institute of Nervous System Diseases (X.-C.Y., J.-X.H.) and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University (X.-C.Y., J.-X.H., G.-Y.C.), Xuzhou Medical University, Jiangsu, China; and Department of Hand Surgery, China Japan Union Hospital, Jilin University, Changchun (Q.L.)
| | - Jin-Xia Hu
- From the Department of Neuroscience and Regenerative Medicine (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.) and Department of Neurology (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.), Medical College of Georgia, Augusta University; Institute of Nervous System Diseases (X.-C.Y., J.-X.H.) and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University (X.-C.Y., J.-X.H., G.-Y.C.), Xuzhou Medical University, Jiangsu, China; and Department of Hand Surgery, China Japan Union Hospital, Jilin University, Changchun (Q.L.)
| | - Lei Li
- From the Department of Neuroscience and Regenerative Medicine (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.) and Department of Neurology (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.), Medical College of Georgia, Augusta University; Institute of Nervous System Diseases (X.-C.Y., J.-X.H.) and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University (X.-C.Y., J.-X.H., G.-Y.C.), Xuzhou Medical University, Jiangsu, China; and Department of Hand Surgery, China Japan Union Hospital, Jilin University, Changchun (Q.L.)
| | - Qiang Li
- From the Department of Neuroscience and Regenerative Medicine (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.) and Department of Neurology (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.), Medical College of Georgia, Augusta University; Institute of Nervous System Diseases (X.-C.Y., J.-X.H.) and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University (X.-C.Y., J.-X.H., G.-Y.C.), Xuzhou Medical University, Jiangsu, China; and Department of Hand Surgery, China Japan Union Hospital, Jilin University, Changchun (Q.L.)
| | - Fu-Lei Tang
- From the Department of Neuroscience and Regenerative Medicine (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.) and Department of Neurology (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.), Medical College of Georgia, Augusta University; Institute of Nervous System Diseases (X.-C.Y., J.-X.H.) and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University (X.-C.Y., J.-X.H., G.-Y.C.), Xuzhou Medical University, Jiangsu, China; and Department of Hand Surgery, China Japan Union Hospital, Jilin University, Changchun (Q.L.)
| | - Sen Lin
- From the Department of Neuroscience and Regenerative Medicine (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.) and Department of Neurology (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.), Medical College of Georgia, Augusta University; Institute of Nervous System Diseases (X.-C.Y., J.-X.H.) and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University (X.-C.Y., J.-X.H., G.-Y.C.), Xuzhou Medical University, Jiangsu, China; and Department of Hand Surgery, China Japan Union Hospital, Jilin University, Changchun (Q.L.)
| | - Dong Sun
- From the Department of Neuroscience and Regenerative Medicine (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.) and Department of Neurology (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.), Medical College of Georgia, Augusta University; Institute of Nervous System Diseases (X.-C.Y., J.-X.H.) and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University (X.-C.Y., J.-X.H., G.-Y.C.), Xuzhou Medical University, Jiangsu, China; and Department of Hand Surgery, China Japan Union Hospital, Jilin University, Changchun (Q.L.)
| | - Xiang-Dong Sun
- From the Department of Neuroscience and Regenerative Medicine (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.) and Department of Neurology (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.), Medical College of Georgia, Augusta University; Institute of Nervous System Diseases (X.-C.Y., J.-X.H.) and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University (X.-C.Y., J.-X.H., G.-Y.C.), Xuzhou Medical University, Jiangsu, China; and Department of Hand Surgery, China Japan Union Hospital, Jilin University, Changchun (Q.L.)
| | - Gui-Yun Cui
- From the Department of Neuroscience and Regenerative Medicine (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.) and Department of Neurology (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.), Medical College of Georgia, Augusta University; Institute of Nervous System Diseases (X.-C.Y., J.-X.H.) and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University (X.-C.Y., J.-X.H., G.-Y.C.), Xuzhou Medical University, Jiangsu, China; and Department of Hand Surgery, China Japan Union Hospital, Jilin University, Changchun (Q.L.)
| | - Lin Mei
- From the Department of Neuroscience and Regenerative Medicine (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.) and Department of Neurology (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.), Medical College of Georgia, Augusta University; Institute of Nervous System Diseases (X.-C.Y., J.-X.H.) and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University (X.-C.Y., J.-X.H., G.-Y.C.), Xuzhou Medical University, Jiangsu, China; and Department of Hand Surgery, China Japan Union Hospital, Jilin University, Changchun (Q.L.)
| | - Wen-Cheng Xiong
- From the Department of Neuroscience and Regenerative Medicine (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.) and Department of Neurology (X.-C.Y., J.-X.H., L.L., Q.L., F.-L.T., S.L., D.S., X.-D.S., L.M., W.-C.X.), Medical College of Georgia, Augusta University; Institute of Nervous System Diseases (X.-C.Y., J.-X.H.) and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University (X.-C.Y., J.-X.H., G.-Y.C.), Xuzhou Medical University, Jiangsu, China; and Department of Hand Surgery, China Japan Union Hospital, Jilin University, Changchun (Q.L.).
| |
Collapse
|
31
|
Karakatsani A, Marichal N, Urban S, Kalamakis G, Ghanem A, Schick A, Zhang Y, Conzelmann KK, Rüegg MA, Berninger B, Ruiz de Almodovar C, Gascón S, Kröger S. Neuronal LRP4 regulates synapse formation in the developing CNS. Development 2017; 144:4604-4615. [PMID: 29061639 DOI: 10.1242/dev.150110] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 10/11/2017] [Indexed: 01/19/2023]
Abstract
The low-density lipoprotein receptor-related protein 4 (LRP4) is essential in muscle fibers for the establishment of the neuromuscular junction. Here, we show that LRP4 is also expressed by embryonic cortical and hippocampal neurons, and that downregulation of LRP4 in these neurons causes a reduction in density of synapses and number of primary dendrites. Accordingly, overexpression of LRP4 in cultured neurons had the opposite effect inducing more but shorter primary dendrites with an increased number of spines. Transsynaptic tracing mediated by rabies virus revealed a reduced number of neurons presynaptic to the cortical neurons in which LRP4 was knocked down. Moreover, neuron-specific knockdown of LRP4 by in utero electroporation of LRP4 miRNA in vivo also resulted in neurons with fewer primary dendrites and a lower density of spines in the developing cortex and hippocampus. Collectively, our results demonstrate an essential and novel role of neuronal LRP4 in dendritic development and synaptogenesis in the CNS.
Collapse
Affiliation(s)
- Andromachi Karakatsani
- Department of Physiological Genomics, Ludwig-Maximilians-University, Grosshaderner Str. 9, D-82152 Planegg-Martinsried, Germany.,Biochemistry Center (BZH), Heidelberg University, 69120 Heidelberg, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Nicolás Marichal
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch Weg 19, D-55128 Mainz, Germany.,Focus Program Translational Neurosciences Mainz, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, D-55131 Mainz, Germany
| | - Severino Urban
- Biochemistry Center (BZH), Heidelberg University, 69120 Heidelberg, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Georgios Kalamakis
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Alexander Ghanem
- Max von Pettenkofer Institute and Gene Center, Ludwig-Maximilians-University, D-81377, Munich, Germany
| | - Anna Schick
- Department of Physiological Genomics, Ludwig-Maximilians-University, Grosshaderner Str. 9, D-82152 Planegg-Martinsried, Germany
| | - Yina Zhang
- Department of Physiological Genomics, Ludwig-Maximilians-University, Grosshaderner Str. 9, D-82152 Planegg-Martinsried, Germany
| | - Karl-Klaus Conzelmann
- Max von Pettenkofer Institute and Gene Center, Ludwig-Maximilians-University, D-81377, Munich, Germany
| | - Markus A Rüegg
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Benedikt Berninger
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch Weg 19, D-55128 Mainz, Germany.,Focus Program Translational Neurosciences Mainz, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, D-55131 Mainz, Germany
| | - Carmen Ruiz de Almodovar
- Biochemistry Center (BZH), Heidelberg University, 69120 Heidelberg, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Sergio Gascón
- Department of Physiological Genomics, Ludwig-Maximilians-University, Grosshaderner Str. 9, D-82152 Planegg-Martinsried, Germany .,Institute for Stem Cell Research, Helmholtz Center Munich at the Biomedical Center (BMC), Grosshaderner Strasse 9, D-82152 Planegg-Martinsried, Germany.,Toxicology and Pharmacology Department, Faculty of Veterinary Medicine, Complutense University, Ave. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Stephan Kröger
- Department of Physiological Genomics, Ludwig-Maximilians-University, Grosshaderner Str. 9, D-82152 Planegg-Martinsried, Germany
| |
Collapse
|
32
|
Mosca TJ, Luginbuhl DJ, Wang IE, Luo L. Presynaptic LRP4 promotes synapse number and function of excitatory CNS neurons. eLife 2017; 6. [PMID: 28606304 PMCID: PMC5469616 DOI: 10.7554/elife.27347] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/08/2017] [Indexed: 12/24/2022] Open
Abstract
Precise coordination of synaptic connections ensures proper information flow within circuits. The activity of presynaptic organizing molecules signaling to downstream pathways is essential for such coordination, though such entities remain incompletely known. We show that LRP4, a conserved transmembrane protein known for its postsynaptic roles, functions presynaptically as an organizing molecule. In the Drosophila brain, LRP4 localizes to the nerve terminals at or near active zones. Loss of presynaptic LRP4 reduces excitatory (not inhibitory) synapse number, impairs active zone architecture, and abolishes olfactory attraction - the latter of which can be suppressed by reducing presynaptic GABAB receptors. LRP4 overexpression increases synapse number in excitatory and inhibitory neurons, suggesting an instructive role and a common downstream synapse addition pathway. Mechanistically, LRP4 functions via the conserved kinase SRPK79D to ensure normal synapse number and behavior. This highlights a presynaptic function for LRP4, enabling deeper understanding of how synapse organization is coordinated.
Collapse
Affiliation(s)
- Timothy J Mosca
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, United States.,Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - David J Luginbuhl
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Irving E Wang
- Department of Neurobiology, Stanford University, Stanford, United States
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| |
Collapse
|
33
|
Lane-Donovan C, Herz J. ApoE, ApoE Receptors, and the Synapse in Alzheimer's Disease. Trends Endocrinol Metab 2017; 28:273-284. [PMID: 28057414 PMCID: PMC5366078 DOI: 10.1016/j.tem.2016.12.001] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/29/2016] [Accepted: 12/07/2016] [Indexed: 01/24/2023]
Abstract
As the population ages, neurodegenerative diseases such as Alzheimer's disease (AD) are becoming a significant burden on patients, their families, and health-care systems. Neurodegenerative processes may start up to 15 years before outward signs and symptoms of AD, as evidenced by data from AD patients and mouse models. A major genetic risk factor for late-onset AD is the ɛ4 isoform of apolipoprotein E (ApoE4), which is present in almost 20% of the population. In this review we discuss the contribution of ApoE receptor signaling to the function of each component of the tripartite synapse - the axon terminal, the postsynaptic dendritic spine, and the astrocyte - and examine how these systems fail in the context of ApoE4 and AD.
Collapse
Affiliation(s)
- Courtney Lane-Donovan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Neuroscience, Department of Neuroanatomy, Albert Ludwig University, Freiburg, Germany.
| |
Collapse
|
34
|
Pohlkamp T, Wasser CR, Herz J. Functional Roles of the Interaction of APP and Lipoprotein Receptors. Front Mol Neurosci 2017; 10:54. [PMID: 28298885 PMCID: PMC5331069 DOI: 10.3389/fnmol.2017.00054] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/16/2017] [Indexed: 11/24/2022] Open
Abstract
The biological fates of the key initiator of Alzheimer’s disease (AD), the amyloid precursor protein (APP), and a family of lipoprotein receptors, the low-density lipoprotein (LDL) receptor-related proteins (LRPs) and their molecular roles in the neurodegenerative disease process are inseparably interwoven. Not only does APP bind tightly to the extracellular domains (ECDs) of several members of the LRP group, their intracellular portions are also connected through scaffolds like the one established by FE65 proteins and through interactions with adaptor proteins such as X11/Mint and Dab1. Moreover, the ECDs of APP and LRPs share common ligands, most notably Reelin, a regulator of neuronal migration during embryonic development and modulator of synaptic transmission in the adult brain, and Agrin, another signaling protein which is essential for the formation and maintenance of the neuromuscular junction (NMJ) and which likely also has critical, though at this time less well defined, roles for the regulation of central synapses. Furthermore, the major independent risk factors for AD, Apolipoprotein (Apo) E and ApoJ/Clusterin, are lipoprotein ligands for LRPs. Receptors and ligands mutually influence their intracellular trafficking and thereby the functions and abilities of neurons and the blood-brain-barrier to turn over and remove the pathological product of APP, the amyloid-β peptide. This article will review and summarize the molecular mechanisms that are shared by APP and LRPs and discuss their relative contributions to AD.
Collapse
Affiliation(s)
- Theresa Pohlkamp
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA
| | - Catherine R Wasser
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA; Department of Neuroscience, UT Southwestern Medical CenterDallas, TX, USA; Department of Neurology and Neurotherapeutics, UT Southwestern Medical CenterDallas, TX, USA
| |
Collapse
|
35
|
Rivner MH, Liu S, Quarles B, Fleenor B, Shen C, Pan J, Mei L. Agrin and low-density lipoprotein-related receptor protein 4 antibodies in amyotrophic lateral sclerosis patients. Muscle Nerve 2016; 55:430-432. [PMID: 27756107 DOI: 10.1002/mus.25438] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2016] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The prevalence and characteristics of agrin and low-density lipoprotein-related receptor protein 4 (LRP4) antibody-positive amyotrophic lateral sclerosis (ALS) patients were studied. METHODS We tested 82 ALS patients and 59 controls for agrin and LRP4 antibodies using enzyme-linked immunoassay (ELISA). RESULTS We found that 13.8% of ALS patients had agrin antibodies, and 9.8% had LRP4 antibodies. Women with ALS are twice as likely as men to have antibodies. Agrin-positive ALS patients are younger than agrin-negative ALS patients. CONCLUSIONS Antibodies to agrin and LRP4 are found in ALS patients. It must be determined whether these antibodies are pathogenic. Because antibody-positive patients have upper as well as lower motor neuron findings, the antibodies' effects cannot be explained solely by their actions at the neuromuscular junction. A breakdown in interneuronal signaling may be the cause of ALS. Further research is needed to resolve this question. Muscle Nerve, 2016 Muscle Nerve 55: 430-432, 2017.
Collapse
Affiliation(s)
- Michael H Rivner
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Siyang Liu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Brandy Quarles
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Brandi Fleenor
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Chengyong Shen
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Jinxiu Pan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Lin Mei
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
36
|
Borralleras C, Mato S, Amédée T, Matute C, Mulle C, Pérez-Jurado LA, Campuzano V. Synaptic plasticity and spatial working memory are impaired in the CD mouse model of Williams-Beuren syndrome. Mol Brain 2016; 9:76. [PMID: 27485321 PMCID: PMC4971717 DOI: 10.1186/s13041-016-0258-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 07/29/2016] [Indexed: 01/22/2023] Open
Abstract
Mice heterozygous for a complete deletion (CD) equivalent to the most common deletion found in individuals with Williams-Beuren syndrome (WBS) recapitulate relevant features of the neurocognitive phenotype, such as hypersociability, along with some neuroanatomical alterations in specific brain areas. However, the pathophysiological mechanisms underlying these phenotypes still remain largely unknown. We have studied the synaptic function and cognition in CD mice using hippocampal slices and a behavioral test sensitive to hippocampal function. We have found that long-term potentiation (LTP) elicited by theta burst stimulation (TBS) was significantly impaired in hippocampal field CA1 of CD animals. This deficit might be associated with the observed alterations in spatial working memory. However, we did not detect changes in presynaptic function, LTP induction mechanisms or AMPA and NMDA receptor function. Reduced levels of Brain-derived neurotrophic factor (BDNF) were present in the CA1-CA3 hippocampal region of CD mice, which could account for LTP deficits in these mice. Taken together, these results suggest a defect of CA1 synapses in CD mice to sustain synaptic strength after stimulation. These data represent the first description of synaptic functional deficits in CD mice and further highlights the utility of the CD model to study the mechanisms underlying the WBS neurocognitive profile.
Collapse
Affiliation(s)
- Cristina Borralleras
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain.,Neurosciences Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Susana Mato
- Department of Neuroscience, Neurotek-University of the Basque Country, Leioa, Spain.,Achucarro Basque Center for Neuroscience, Zamudio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Thierry Amédée
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 - University of Bordeaux, F-33000, Bordeaux, France
| | - Carlos Matute
- Department of Neuroscience, Neurotek-University of the Basque Country, Leioa, Spain.,Achucarro Basque Center for Neuroscience, Zamudio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Christophe Mulle
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 - University of Bordeaux, F-33000, Bordeaux, France
| | - Luis A Pérez-Jurado
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain.,Neurosciences Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Victoria Campuzano
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain. .,Neurosciences Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain.
| |
Collapse
|
37
|
Sun XD, Li L, Liu F, Huang ZH, Bean JC, Jiao HF, Barik A, Kim SM, Wu H, Shen C, Tian Y, Lin TW, Bates R, Sathyamurthy A, Chen YJ, Yin DM, Xiong L, Lin HP, Hu JX, Li BM, Gao TM, Xiong WC, Mei L. Lrp4 in astrocytes modulates glutamatergic transmission. Nat Neurosci 2016; 19:1010-8. [PMID: 27294513 PMCID: PMC4961622 DOI: 10.1038/nn.4326] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 05/08/2016] [Indexed: 02/06/2023]
Abstract
Neurotransmission requires precise control of neurotransmitter release from axon terminals. This process is regulated by glial cells; however, underlying mechanisms are not fully understood. Here we report that glutamate release in the brain is impaired in mice lacking low density lipoprotein receptor-related protein 4 (Lrp4), a protein critical for neuromuscular junction formation. Electrophysiological studies indicate compromised release probability in astrocyte-specific Lrp4 knockout mice. Lrp4 mutant astrocytes suppress glutamate transmission by enhancing the release of ATP, whose levels are elevated in the hippocampus of Lrp4 mutant mice. Consequently, the mutant mice are impaired in locomotor activity and spatial memory and are resistant to seizure induction. These impairments could be ameliorated by adenosine A1 receptor antagonist. The results reveal a critical role of Lrp4, in response to agrin, in modulating astrocytic ATP release and synaptic transmission. Our study provides insight into the interaction between neurons and astrocytes for synaptic homeostasis and/or plasticity.
Collapse
Affiliation(s)
- Xiang-Dong Sun
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Lei Li
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Fang Liu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Zhi-Hui Huang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Jonathan C Bean
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Hui-Feng Jiao
- Center for Neuropsychiatric Diseases, Institute of Life Science, Nanchang University, Nanchang, China
| | - Arnab Barik
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Seon-Myung Kim
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Haitao Wu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Chengyong Shen
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Yun Tian
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Thiri W Lin
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Ryan Bates
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Anupama Sathyamurthy
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Yong-Jun Chen
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Dong-Min Yin
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Lei Xiong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Hui-Ping Lin
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Jin-Xia Hu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA
| | - Bao-Ming Li
- Center for Neuropsychiatric Diseases, Institute of Life Science, Nanchang University, Nanchang, China.,Jiangxi Medical School, Nanchang University, Nanchang, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, Southern Medical University, Guangzhou, China
| | - Wen-Cheng Xiong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA.,Charlie Norwood Virginia Medical Center, Augusta, Georgia, USA
| | - Lin Mei
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Georgia, USA.,Center for Neuropsychiatric Diseases, Institute of Life Science, Nanchang University, Nanchang, China.,Jiangxi Medical School, Nanchang University, Nanchang, China.,Charlie Norwood Virginia Medical Center, Augusta, Georgia, USA
| |
Collapse
|
38
|
Tanahashi H, Tian QB, Hara Y, Sakagami H, Endo S, Suzuki T. Polyhydramnios in Lrp4 knockout mice with bilateral kidney agenesis: Defects in the pathways of amniotic fluid clearance. Sci Rep 2016; 6:20241. [PMID: 26847765 PMCID: PMC4742865 DOI: 10.1038/srep20241] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/29/2015] [Indexed: 12/26/2022] Open
Abstract
Amniotic fluid volume during mid-to-late gestation depends mainly on the urine excretion from the foetal kidneys and partly on the fluid secretion from the foetal lungs during foetal breathing-like movements. Urine is necessary for foetal breathing-like movements, which is critical for foetal lung development. Bilateral renal agenesis and/or obstruction of the urinary tract lead to oligohydramnios, which causes infant death within a short period after birth due to pulmonary hypoplasia. Lrp4, which functions as an agrin receptor, is essential for the formation of neuromuscular junctions. Herein, we report novel phenotypes of Lrp4 knockout (Lrp4(-/-)) mice. Most Lrp4(-/-) foetuses showed unilateral or bilateral kidney agenesis, and Lrp4 knockout resulted in polyhydramnios. The loss of Lrp4 compromised foetal swallowing and breathing-like movements and downregulated the expression of aquaporin-9 in the foetal membrane and aquaporin-1 in the placenta, which possibly affected the amniotic fluid clearance. These results suggest that amniotic fluid removal was compromised in Lrp4(-/-) foetuses, resulting in polyhydramnios despite the impairment of urine production. Our findings indicate that amniotic fluid removal plays an essential role in regulating the amniotic fluid volume.
Collapse
Affiliation(s)
- Hiroshi Tanahashi
- Department of Neuroplasticity, Institute of Pathogenesis and Disease Prevention, Graduate School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Japan.,Department of Biological Sciences for Intractable Neurological Diseases, Institute for Biomedical Sciences, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Qing-Bao Tian
- Department of Neuroplasticity, Institute of Pathogenesis and Disease Prevention, Graduate School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Yoshinobu Hara
- Department of Anatomy, Kitasato University School of Medicine, 1-15-1, Kitasato, Sagamihara, Kanagawa 252-0374, Japan
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, 1-15-1, Kitasato, Sagamihara, Kanagawa 252-0374, Japan
| | - Shogo Endo
- Research Team for Aging Neuroscience, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Sakae-cho, Itabashi, Tokyo 173-0015, Japan
| | - Tatsuo Suzuki
- Department of Neuroplasticity, Institute of Pathogenesis and Disease Prevention, Graduate School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Japan.,Department of Biological Sciences for Intractable Neurological Diseases, Institute for Biomedical Sciences, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| |
Collapse
|
39
|
Smith PD, Coulson-Thomas VJ, Foscarin S, Kwok JCF, Fawcett JW. "GAG-ing with the neuron": The role of glycosaminoglycan patterning in the central nervous system. Exp Neurol 2015; 274:100-14. [PMID: 26277685 DOI: 10.1016/j.expneurol.2015.08.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/17/2015] [Accepted: 08/06/2015] [Indexed: 01/17/2023]
Abstract
Proteoglycans (PGs) are a diverse family of proteins that consist of one or more glycosaminoglycan (GAG) chains, covalently linked to a core protein. PGs are major components of the extracellular matrix (ECM) and play critical roles in development, normal function and damage-response of the central nervous system (CNS). GAGs are classified based on their disaccharide subunits, into the following major groups: chondroitin sulfate (CS), heparan sulfate (HS), heparin (HEP), dermatan sulfate (DS), keratan sulfate (KS) and hyaluronic acid (HA). All except HA are modified by sulfation, giving GAG chains specific charged structures and binding properties. While significant neuroscience research has focused on the role of one PG family member, chondroitin sulfate proteoglycan (CSPG), there is ample evidence in support of a role for the other PGs in regulating CNS function in normal and pathological conditions. This review discusses the role of all the identified PG family members (CS, HS, HEP, DS, KS and HA) in normal CNS function and in the context of pathology. Understanding the pleiotropic roles of these molecules in the CNS may open the door to novel therapeutic strategies for a number of neurological conditions.
Collapse
Affiliation(s)
- Patrice D Smith
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK; Department of Neuroscience, Carleton University, Ottawa, ON, Canada.
| | - Vivien J Coulson-Thomas
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - Simona Foscarin
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - Jessica C F Kwok
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - James W Fawcett
- John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK.
| |
Collapse
|
40
|
Pohlkamp T, Durakoglugil M, Lane-Donovan C, Xian X, Johnson EB, Hammer RE, Herz J. Lrp4 domains differentially regulate limb/brain development and synaptic plasticity. PLoS One 2015; 10:e0116701. [PMID: 25688974 PMCID: PMC4331535 DOI: 10.1371/journal.pone.0116701] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 12/14/2014] [Indexed: 01/01/2023] Open
Abstract
Apolipoprotein E (ApoE) genotype is the strongest predictor of Alzheimer’s Disease (AD) risk. ApoE is a cholesterol transport protein that binds to members of the Low-Density Lipoprotein (LDL) Receptor family, which includes LDL Receptor Related Protein 4 (Lrp4). Lrp4, together with one of its ligands Agrin and its co-receptors Muscle Specific Kinase (MuSK) and Amyloid Precursor Protein (APP), regulates neuromuscular junction (NMJ) formation. All four proteins are also expressed in the adult brain, and APP, MuSK, and Agrin are required for normal synapse function in the CNS. Here, we show that Lrp4 is also required for normal hippocampal plasticity. In contrast to the closely related Lrp8/Apoer2, the intracellular domain of Lrp4 does not appear to be necessary for normal expression and maintenance of long-term potentiation at central synapses or for the formation and maintenance of peripheral NMJs. However, it does play a role in limb development.
Collapse
Affiliation(s)
- Theresa Pohlkamp
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
- * E-mail: (TP); (JH)
| | - Murat Durakoglugil
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
| | - Courtney Lane-Donovan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
| | - Xunde Xian
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
| | - Eric B. Johnson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
| | - Robert E. Hammer
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, United States of America
- * E-mail: (TP); (JH)
| |
Collapse
|