1
|
Song MK, Jo HS, Kim EJ, Kim JK, Lee SG. Gene Expression of Neurogenesis Related to Exercise Intensity in a Cerebral Infarction Rat Model. Int J Mol Sci 2024; 25:8997. [PMID: 39201683 PMCID: PMC11354542 DOI: 10.3390/ijms25168997] [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: 06/30/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 09/03/2024] Open
Abstract
Regular exercise improves several functions, including cognition, in patients with stroke. However, the effect of regular exercise on neurogenesis related to cognition remains doubtful. We investigated the most effective exercise intensity for functional recovery after stroke using RNA sequencing following regular treadmill exercise. Photothrombotic cerebral infarction was conducted for 10-week-old male Sprague-Dawley rats (n = 36). A Morris water maze (MWM) test was performed before a regular treadmill exercise program (5 days/week, 4 weeks). Rats were randomly divided into four groups: group A (no exercise); group B (low intensity, maximal velocity 18 m/min); group C (moderate intensity, maximal velocity 24 m/min) and group D (high intensity, maximal velocity 30 m/min). After 4 weeks, another MWM test was performed, and all rats were sacrificed. RNA sequencing was performed with ipsilesional hippocampal tissue. On the day after cerebral infarction, no differences in escape latency and velocity were observed among the groups. At 4 weeks after cerebral infarction, the escape latencies in groups B, C, and D were shorter than in group A. The escape latencies in groups B and C were shorter than in group D. The velocity in groups A, B, and C was faster than in group D. Thirty gene symbols related to neurogenesis were detected (p < 0.05, fold change > 1.0, average normalized read count > four times). In the neurotrophin-signaling pathway, the CHK gene was upregulated, and the NF-κB gene was downregulated in the low-intensity group. The CHK and NF-κB genes were both downregulated in the moderate-intensity group. The Raf and IRAK genes were downregulated in the high-intensity group. Western blot analysis showed that NF-κB expression was lowest in the moderate-intensity group, whereas CHK and Raf were elevated, and IRAK was decreased in the high-intensity group. Moderate-intensity exercise may contribute to neuroplasticity. Variation in the expression of neurotrophins in neurogenesis according to exercise intensity may reveal the mechanism of neuroplasticity. Thus, NF-κB is the key neurotrophin for neurogenesis related to exercise intensity.
Collapse
Affiliation(s)
| | | | | | | | - Sam-Gyu Lee
- Department of Physical & Rehabilitation Medicine, Chonnam National University Medical School, #160, Baekseo-ro, Dong-gu, Gwangju 61469, Republic of Korea; (M.-K.S.)
| |
Collapse
|
2
|
Gong R, Qin L, Chen L, Wang N, Bao Y, Lu W. Myosin Va-dependent Transport of NMDA Receptors in Hippocampal Neurons. Neurosci Bull 2024; 40:1053-1075. [PMID: 38291290 PMCID: PMC11306496 DOI: 10.1007/s12264-023-01174-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/03/2023] [Indexed: 02/01/2024] Open
Abstract
N-methyl-D-aspartate receptor (NMDAR) trafficking is a key process in the regulation of synaptic efficacy and brain function. However, the molecular mechanism underlying the surface transport of NMDARs is largely unknown. Here we identified myosin Va (MyoVa) as the specific motor protein that traffics NMDARs in hippocampal neurons. We found that MyoVa associates with NMDARs through its cargo binding domain. This association was increased during NMDAR surface transport. Knockdown of MyoVa suppressed NMDAR transport. We further demonstrated that Ca2+/calmodulin-dependent protein kinase II (CaMKII) regulates NMDAR transport through its direct interaction with MyoVa. Furthermore, MyoVa employed Rab11 family-interacting protein 3 (Rab11/FIP3) as the adaptor proteins to couple themselves with NMDARs during their transport. Accordingly, the knockdown of FIP3 impairs hippocampal memory. Together, we conclude that in hippocampal neurons, MyoVa conducts active transport of NMDARs in a CaMKII-dependent manner.
Collapse
Affiliation(s)
- Ru Gong
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Linwei Qin
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Linlin Chen
- Department of Neurobiology, Nanjing Medical University, Nanjing, 210096, China
| | - Ning Wang
- Department of Neurobiology, Nanjing Medical University, Nanjing, 210096, China
| | - Yifei Bao
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Wei Lu
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China.
- Department of Neurosurgery, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China.
- Department of Neurobiology, Nanjing Medical University, Nanjing, 210096, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
3
|
Aguirre S, Pappa S, Serna-Pujol N, Padilla N, Iacobucci S, Nacht AS, Vicent GP, Jordan A, de la Cruz X, Martínez-Balbás MA. PHF2-mediated H3K9me balance orchestrates heterochromatin stability and neural progenitor proliferation. EMBO Rep 2024; 25:3486-3505. [PMID: 38890452 PMCID: PMC11315909 DOI: 10.1038/s44319-024-00178-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/18/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Heterochromatin stability is crucial for progenitor proliferation during early neurogenesis. It relays on the maintenance of local hubs of H3K9me. However, understanding the formation of efficient localized levels of H3K9me remains limited. To address this question, we used neural stem cells to analyze the function of the H3K9me2 demethylase PHF2, which is crucial for progenitor proliferation. Through mass-spectroscopy and genome-wide assays, we show that PHF2 interacts with heterochromatin components and is enriched at pericentromeric heterochromatin (PcH) boundaries where it maintains transcriptional activity. This binding is essential for silencing the satellite repeats, preventing DNA damage and genome instability. PHF2's depletion increases the transcription of heterochromatic repeats, accompanied by a decrease in H3K9me3 levels and alterations in PcH organization. We further show that PHF2's PHD and catalytic domains are crucial for maintaining PcH stability, thereby safeguarding genome integrity. These results highlight the multifaceted nature of PHF2's functions in maintaining heterochromatin stability and regulating gene expression during neural development. Our study unravels the intricate relationship between heterochromatin stability and progenitor proliferation during mammalian neurogenesis.
Collapse
Affiliation(s)
- Samuel Aguirre
- Department of Structural and Molecular Biology, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, 08028, Spain
| | - Stella Pappa
- Department of Structural and Molecular Biology, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, 08028, Spain
| | - Núria Serna-Pujol
- Department of Structural and Molecular Biology, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, 08028, Spain
| | - Natalia Padilla
- Vall d'Hebron Institute of Research (VHIR), Passeig de la Vall d'Hebron, 119, E-08035, Barcelona, Spain
| | - Simona Iacobucci
- Department of Structural and Molecular Biology, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, 08028, Spain
| | - A Silvina Nacht
- Center for Genomic Regulation (CRG), Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Guillermo P Vicent
- Department of Structural and Molecular Biology, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, 08028, Spain
| | - Albert Jordan
- Department of Structural and Molecular Biology, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, 08028, Spain
| | - Xavier de la Cruz
- Vall d'Hebron Institute of Research (VHIR), Passeig de la Vall d'Hebron, 119, E-08035, Barcelona, Spain
- Institut Català per la Recerca i Estudis Avançats (ICREA), Barcelona, 08018, Spain
| | - Marian A Martínez-Balbás
- Department of Structural and Molecular Biology, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, 08028, Spain.
| |
Collapse
|
4
|
Camilleri M, Jencks K. Pharmacogenetics in IBS: update and impact of GWAS studies in drug targets and metabolism. Expert Opin Drug Metab Toxicol 2024; 20:319-332. [PMID: 38785066 PMCID: PMC11139426 DOI: 10.1080/17425255.2024.2349716] [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: 11/28/2023] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION Medications are frequently prescribed for patients with irritable bowel syndrome (IBS) or disorders of gut brain interaction. The level of drug metabolism and modifications in drug targets determine medication efficacy to modify motor or sensory function as well as patient response outcomes. AREAS COVERED The literature search included PubMed searches with the terms: pharmacokinetics, pharmacogenomics, epigenetics, clinical trials, irritable bowel syndrome, disorders of gut brain interaction, and genome-wide association studies. The main topics covered in relation to irritable bowel syndrome were precision medicine, pharmacogenomics related to drug metabolism, pharmacogenomics related to mechanistic targets, and epigenetics. EXPERT OPINION Pharmacogenomics impacting drug metabolism [CYP 2D6 (cytochrome P450 2D6) or 2C19 (cytochrome P450 2C19)] is the most practical approach to precision medicine in the treatment of IBS. Although there are proof of concept studies that have documented the importance of genetic modification of transmitters or receptors in altering responses to medications in IBS, these principles have rarely been applied in patient response outcomes. Genome-wide association (GWAS) studies have now documented the association of symptoms with genetic variation but not the evaluation of treatment responses. Considerably more research, particularly focused on patient response outcomes and epigenetics, is essential to impact this field in clinical medicine.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Kara Jencks
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
5
|
Deng Y, Dong Y, Wu L, Zhang Q, Yang L. ARID5B promoted the histone demethylation of SORBS2 and hampered the metastasis of ovarian cancer. Pathol Res Pract 2023; 252:154911. [PMID: 37948999 DOI: 10.1016/j.prp.2023.154911] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/18/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023]
Abstract
Ovarian cancer (OVCA) is the 4th most common female tumor after breast cancer, cervical cancer, and endometrial cancer, and now is mainly treated with debulking surgery and postoperative cisplatin and paclitaxel-based combination chemotherapy regimens. However, OVCA is insidious in its development and recurrence occurred in some patients after treatment. It is of great significance to study the pathogenesis of ovarian cancer and identify more biomarkers. Recently, the role of histone methyltransferase (HMT) and histone demethylase (HDM) in oncogenesis and development of malignant tumors has raised attention. Unlike other JMJC demethylases that have both JMJC and ARID domains in a single molecule, PHF2 requires assembly into a complex with a DNA-binding subunit (ARID5B) and exerts its enzymatic activity. Therefore, the aim of this manuscript is to investigate the role of histone demethylases ARID5B-PHF2 complex in the metastasis of OVCA. As result, we found ARID5B and PHF2 are both low expressed in OVCA tumor tissues and cell lines and associated with diagnosis and prognosis. Also, ARID5B suppressed rearrangement of the cytoskeleton in the process of EMT in OVCA cell lines. The role of PHF2 as a tumor suppressor was also confirmed both in vivo and in vitro. SORBS2 is low expressed in OVCA tumor tissues and cell lines and associated with diagnosis and prognosis. The expression of SORBS2 is positively corelated with the expression of ARID5B and PHF2. The promoter of SORBS2 is proved combined with ARID5B. The expression of SORBS2 was increased due to ARID5B-PHF2 complex promoted the histone demethylation by mainly binding in site H3K36me2 and therefore promoting the transcription of SORBS2. In conclusion, ARID5B-PHF2 complex promoted the histone demethylation of SORBS2 by mainly bind in site H3K36me2 and therefore promote the transcription of SORBS2 then hampered the process of EMT and tumor generation of OVCA. These results provided a new perspective on the molecular mechanisms of OVCA development and offered a new target of clinical diagnose and treatment of OVCA.
Collapse
Affiliation(s)
- Yue Deng
- Department of Gynecology,The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Ying Dong
- Department of Gynecology,The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Lu Wu
- Department of Gynecology,The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Qin Zhang
- Department of Gynecology,The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Lihua Yang
- Department of Gynecology,The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China.
| |
Collapse
|
6
|
Jeong DW, Park JW, Kim KS, Kim J, Huh J, Seo J, Kim YL, Cho JY, Lee KW, Fukuda J, Chun YS. Palmitoylation-driven PHF2 ubiquitination remodels lipid metabolism through the SREBP1c axis in hepatocellular carcinoma. Nat Commun 2023; 14:6370. [PMID: 37828054 PMCID: PMC10570296 DOI: 10.1038/s41467-023-42170-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 10/02/2023] [Indexed: 10/14/2023] Open
Abstract
Palmitic acid (PA) is the most common fatty acid in humans and mediates palmitoylation through its conversion into palmitoyl coenzyme A. Although palmitoylation affects many proteins, its pathophysiological functions are only partially understood. Here we demonstrate that PA acts as a molecular checkpoint of lipid reprogramming in HepG2 and Hep3B cells. The zinc finger DHHC-type palmitoyltransferase 23 (ZDHHC23) mediates the palmitoylation of plant homeodomain finger protein 2 (PHF2), subsequently enhancing ubiquitin-dependent degradation of PHF2. This study also reveals that PHF2 functions as a tumor suppressor by acting as an E3 ubiquitin ligase of sterol regulatory element-binding protein 1c (SREBP1c), a master transcription factor of lipogenesis. PHF2 directly destabilizes SREBP1c and reduces SREBP1c-dependent lipogenesis. Notably, SREBP1c increases free fatty acids in hepatocellular carcinoma (HCC) cells, and the consequent PA induction triggers the PHF2/SREBP1c axis. Since PA seems central to activating this axis, we suggest that levels of dietary PA should be carefully monitored in patients with HCC.
Collapse
Affiliation(s)
- Do-Won Jeong
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Jong-Wan Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Kyeong Seog Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, 03080, Korea
| | - Jiyoung Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - June Huh
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Korea
| | - Jieun Seo
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea
- Faculty of Engineering, Yokohama National University, Yokohama, 240-8501, Japan
| | - Ye Lee Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Joo-Youn Cho
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, 03080, Korea
| | - Kwang-Woong Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, Yokohama, 240-8501, Japan
| | - Yang-Sook Chun
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.
| |
Collapse
|
7
|
Whittaker DS, Akhmetova L, Carlin D, Romero H, Welsh DK, Colwell CS, Desplats P. Circadian modulation by time-restricted feeding rescues brain pathology and improves memory in mouse models of Alzheimer's disease. Cell Metab 2023; 35:1704-1721.e6. [PMID: 37607543 PMCID: PMC10591997 DOI: 10.1016/j.cmet.2023.07.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 06/12/2023] [Accepted: 07/27/2023] [Indexed: 08/24/2023]
Abstract
Circadian disruptions impact nearly all people with Alzheimer's disease (AD), emphasizing both their potential role in pathology and the critical need to investigate the therapeutic potential of circadian-modulating interventions. Here, we show that time-restricted feeding (TRF) without caloric restriction improved key disease components including behavioral timing, disease pathology, hippocampal transcription, and memory in two transgenic (TG) mouse models of AD. We found that TRF had the remarkable capability of simultaneously reducing amyloid deposition, increasing Aβ42 clearance, improving sleep and memory, and normalizing daily transcription patterns of multiple genes, including those associated with AD and neuroinflammation. Thus, our study unveils for the first time the pleiotropic nature of timed feeding on AD, which has far-reaching effects beyond metabolism, ameliorating neurodegeneration and the misalignment of circadian rhythmicity. Since TRF can substantially modify disease trajectory, this intervention has immediate translational potential, addressing the urgent demand for accessible approaches to reduce or halt AD progression.
Collapse
Affiliation(s)
- Daniel S Whittaker
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Laila Akhmetova
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Daniel Carlin
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Haylie Romero
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - David K Welsh
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Paula Desplats
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA; Department of Pathology, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Vanrobaeys Y, Mukherjee U, Langmack L, Beyer SE, Bahl E, Lin LC, Michaelson JJ, Abel T, Chatterjee S. Mapping the spatial transcriptomic signature of the hippocampus during memory consolidation. Nat Commun 2023; 14:6100. [PMID: 37773230 PMCID: PMC10541893 DOI: 10.1038/s41467-023-41715-7] [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: 01/17/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023] Open
Abstract
Memory consolidation involves discrete patterns of transcriptional events in the hippocampus. Despite the emergence of single-cell transcriptomic profiling techniques, mapping the transcriptomic signature across subregions of the hippocampus has remained challenging. Here, we utilized unbiased spatial sequencing to delineate transcriptome-wide gene expression changes across subregions of the dorsal hippocampus of male mice following learning. We find that each subregion of the hippocampus exhibits distinct yet overlapping transcriptomic signatures. The CA1 region exhibited increased expression of genes related to transcriptional regulation, while the DG showed upregulation of genes associated with protein folding. Importantly, our approach enabled us to define the transcriptomic signature of learning within two less-defined hippocampal subregions, CA1 stratum radiatum, and oriens. We demonstrated that CA1 subregion-specific expression of a transcription factor subfamily has a critical functional role in the consolidation of long-term memory. This work demonstrates the power of spatial molecular approaches to reveal simultaneous transcriptional events across the hippocampus during memory consolidation.
Collapse
Affiliation(s)
- Yann Vanrobaeys
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52242, USA
| | - Utsav Mukherjee
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, 52242, USA
| | - Lucy Langmack
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Biochemistry and Molecular Biology Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Stacy E Beyer
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Ethan Bahl
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52242, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Li-Chun Lin
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Jacob J Michaelson
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
| | - Snehajyoti Chatterjee
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
9
|
Cabej NR. On the origin and nature of nongenetic information in eumetazoans. Ann N Y Acad Sci 2023. [PMID: 37154677 DOI: 10.1111/nyas.15001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Nongenetic information implies all the forms of biological information not related to genes and DNA in general. Despite the deep scientific relevance of the concept, we currently lack reliable knowledge about its carriers and origins; hence, we still do not understand its true nature. Given that genes are the targets of nongenetic information, it appears that a parsimonious approach to find the ultimate source of that information is to trace back the sequential steps of the causal chain upstream of the target genes up to the ultimate link as the source of the nongenetic information. From this perspective, I examine seven nongenetically determined phenomena: placement of locus-specific epigenetic marks on DNA and histones, changes in snRNA expression patterns, neural induction of gene expression, site-specific alternative gene splicing, predator-induced morphological changes, and cultural inheritance. Based on the available evidence, I propose a general model of the common neural origin of all these forms of nongenetic information in eumetazoans.
Collapse
Affiliation(s)
- Nelson R Cabej
- Department of Biology, University of Tirana, Tirana, Albania
| |
Collapse
|
10
|
Zhen W, Zhen H, Wang Y, Chen L, Niu X, Zhang B, Yang Z, Peng D. Mechanism of ERK/CREB pathway in pain and analgesia. Front Mol Neurosci 2023; 16:1156674. [PMID: 37008781 PMCID: PMC10060514 DOI: 10.3389/fnmol.2023.1156674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/22/2023] [Indexed: 03/18/2023] Open
Abstract
Research has long centered on the pathophysiology of pain. The Transient Receiver Potential (TRP) protein family is well known for its function in the pathophysiology of pain, and extensive study has been done in this area. One of the significant mechanisms of pain etiology and analgesia that lacks a systematic synthesis and review is the ERK/CREB (Extracellular Signal-Regulated Kinase/CAMP Response Element Binding Protein) pathway. The ERK/CREB pathway-targeting analgesics may also cause a variety of adverse effects that call for specialized medical care. In this review, we systematically compiled the mechanism of the ERK/CREB pathway in the process of pain and analgesia, as well as the potential adverse effects on the nervous system brought on by the inhibition of the ERK/CREB pathway in analgesic drugs, and we suggested the corresponding solutions.
Collapse
Affiliation(s)
- Weizhe Zhen
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Hongjun Zhen
- Department of Orthopaedics, Handan Chinese Medicine Hospital, Handan, Hebei Province, China
| | - Yuye Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Leian Chen
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoqian Niu
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Graduate School, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Bin Zhang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziyuan Yang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Graduate School, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Dantao Peng
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Dantao Peng,
| |
Collapse
|
11
|
Zhao J, Huai J. Role of primary aging hallmarks in Alzheimer´s disease. Theranostics 2023; 13:197-230. [PMID: 36593969 PMCID: PMC9800733 DOI: 10.7150/thno.79535] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, which severely threatens the health of the elderly and causes significant economic and social burdens. The causes of AD are complex and include heritable but mostly aging-related factors. The primary aging hallmarks include genomic instability, telomere wear, epigenetic changes, and loss of protein stability, which play a dominant role in the aging process. Although AD is closely associated with the aging process, the underlying mechanisms involved in AD pathogenesis have not been well characterized. This review summarizes the available literature about primary aging hallmarks and their roles in AD pathogenesis. By analyzing published literature, we attempted to uncover the possible mechanisms of aberrant epigenetic markers with related enzymes, transcription factors, and loss of proteostasis in AD. In particular, the importance of oxidative stress-induced DNA methylation and DNA methylation-directed histone modifications and proteostasis are highlighted. A molecular network of gene regulatory elements that undergoes a dynamic change with age may underlie age-dependent AD pathogenesis, and can be used as a new drug target to treat AD.
Collapse
|
12
|
Trafficking of NMDA receptors is essential for hippocampal synaptic plasticity and memory consolidation. Cell Rep 2022; 40:111217. [PMID: 35977502 DOI: 10.1016/j.celrep.2022.111217] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 06/14/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022] Open
Abstract
NMDA receptor (NMDAR) plays a vital role in brain development and normal physiological functions. Surface trafficking of NMDAR contributes to the modulation of synaptic functions and information processing. However, it remains unclear whether NMDAR trafficking is independent of long-term potentiation (LTP) and whether it regulates behavior. Here, we report that LTP of AMPAR and NMDAR can occur concurrently and that NMDAR trafficking can regulate AMPAR trafficking and AMPAR-mediated LTP. By contrast, AMPAR trafficking does not impact NMDAR-mediated LTP. Using SAP97-interfering peptide and SAP97 knockin (KI) rat, we show that the effect is mediated by GluN2A-subunit-containing NMDARs. At the behavior level, impaired NMDAR trafficking results in deficits in consolidation, but not acquisition, of fear memory. Collectively, our results suggest the essential role of NMDAR trafficking in LTP and memory consolidation.
Collapse
|
13
|
Wendt FR, Pathak GA, Deak JD, De Angelis F, Koller D, Cabrera-Mendoza B, Lebovitch DS, Levey DF, Stein MB, Kranzler HR, Koenen KC, Gelernter J, Huckins LM, Polimanti R. Using phenotype risk scores to enhance gene discovery for generalized anxiety disorder and posttraumatic stress disorder. Mol Psychiatry 2022; 27:2206-2215. [PMID: 35181757 PMCID: PMC9133008 DOI: 10.1038/s41380-022-01469-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/18/2022] [Accepted: 02/02/2022] [Indexed: 11/09/2022]
Abstract
UK Biobank (UKB) is a key contributor in mental health genome-wide association studies (GWAS) but only ~31% of participants completed the Mental Health Questionnaire ("MHQ responders"). We predicted generalized anxiety disorder (GAD), posttraumatic stress disorder (PTSD), and major depression symptoms using elastic net regression in the ~69% of UKB participants lacking MHQ data ("MHQ non-responders"; NTraining = 50%; NTest = 50%), maximizing the informative sample for these traits. MHQ responders were more likely to be female, from higher socioeconomic positions, and less anxious than non-responders. Genetic correlation of GAD and PTSD between MHQ responders and non-responders ranged from 0.636 to 1.08; both were predicted by polygenic scores generated from independent cohorts. In meta-analyses of GAD (N = 489,579) and PTSD (N = 497,803), we discovered many novel genomic risk loci (13 for GAD and 40 for PTSD). Transcriptomic analyses converged on altered regulation of prenatal dorsolateral prefrontal cortex in these disorders. Our results provide one roadmap by which sample size and statistical power may be improved for gene discovery of incompletely ascertained traits in the UKB and other biobanks with limited mental health assessment.
Collapse
Affiliation(s)
- Frank R Wendt
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- VA CT Healthcare System, West Haven, CT, USA.
| | - Gita A Pathak
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- VA CT Healthcare System, West Haven, CT, USA
| | - Joseph D Deak
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- VA CT Healthcare System, West Haven, CT, USA
| | - Flavio De Angelis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- VA CT Healthcare System, West Haven, CT, USA
| | - Dora Koller
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- VA CT Healthcare System, West Haven, CT, USA
| | - Brenda Cabrera-Mendoza
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- VA CT Healthcare System, West Haven, CT, USA
| | - Dannielle S Lebovitch
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Daniel F Levey
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- VA CT Healthcare System, West Haven, CT, USA
| | - Murray B Stein
- VA San Diego Healthcare System, Psychiatry Service, San Diego, CA, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, USA
| | - Henry R Kranzler
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Mental Illness Research, Education, and Clinical Center, Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA
| | - Karestan C Koenen
- Broad Institute of MIT and Harvard, Stanley Center for Psychiatry Research, Cambridge, MA, USA
- Massachusettes General Hospital, Psychiatry and Neurodevelopmental Genetics Unit (PNGU), Boston, MA, USA
- Harvard School of Public Health, Department of Epidemiology, Boston, MA, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- VA CT Healthcare System, West Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Laura M Huckins
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mental Illness Research, Education and Clinical Center, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, 10468, USA
| | - Renato Polimanti
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- VA CT Healthcare System, West Haven, CT, USA.
| |
Collapse
|
14
|
Chou YJ, Ma YK, Lu YH, King JT, Tasi WS, Yang SB, Kuo TH. Potential cross-species correlations in social hierarchy and memory between mice and young children. Commun Biol 2022; 5:230. [PMID: 35288641 PMCID: PMC8921227 DOI: 10.1038/s42003-022-03173-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 02/15/2022] [Indexed: 11/12/2022] Open
Abstract
Social hierarchy is associated with various phenotypes. Although memory is known to be important for hierarchy formation, the difference in memory abilities between dominant and subordinate individuals remains unclear. In this study, we examined memory performance in mice with different social ranks and found better memory abilities in dominant mice, along with greater long-term potentiation and higher memory-related gene expression in the hippocampus. Daily injection of memory-improving drugs could also enhance dominance. To validate this correlation across species, through inventory, behavioral and event-related potential studies, we identified better memory abilities in preschool children with higher social dominance. Better memory potentially helped children process dominance facial cues and learn social strategies to acquire higher positions. Our study shows a remarkable similarity between humans and mice in the association between memory and social hierarchy and provides valuable insight into social interactions in young animals, with potential implications for preschool education. Memory performance and hippocampal memory-related gene expression are shown to both be increased in more dominant mice, with memory-improving drugs enhancing dominant behaviour. The data also suggests that children with better memory can recognise dominance more easily, demonstrating a potential cross-species correlation in the association between memory and social hierarchy.
Collapse
Affiliation(s)
- Yu-Ju Chou
- Department of Early Childhood Education, National Tsing Hua University, Hsinchu, 300, Taiwan, Republic of China.
| | - Yu-Kai Ma
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, 300, Taiwan, Republic of China
| | - Yi-Han Lu
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, 300, Taiwan, Republic of China
| | - Jung-Tai King
- Institute of Neurosciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan, Republic of China
| | - Wen-Sheng Tasi
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, 300, Taiwan, Republic of China.,Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan, Republic of China
| | - Shi-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan, Republic of China.
| | - Tsung-Han Kuo
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, 300, Taiwan, Republic of China. .,Department of Life Science, National Tsing Hua University, Hsinchu, 300, Taiwan, Republic of China.
| |
Collapse
|
15
|
Calcium-/Calmodulin-Dependent Protein Kinase II (CaMKII) Inhibition Induces Learning and Memory Impairment and Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:4635054. [PMID: 34976299 PMCID: PMC8718318 DOI: 10.1155/2021/4635054] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 11/18/2021] [Indexed: 12/20/2022]
Abstract
Objectives Inhibition of calcium-/calmodulin- (CaM-) dependent kinase II (CaMKII) is correlated with epilepsy. However, the specific mechanism that underlies learning and memory impairment and neuronal death by CaMKII inhibition remains unclear. Materials and Methods In this study, KN93, a CaMKII inhibitor, was used to investigate the role of CaMKII during epileptogenesis. We first identified differentially expressed genes (DEGs) in primary cultured hippocampal neurons with or without KN93 treatment using RNA-sequencing. Then, the impairment of learning and memory by KN93-induced CaMKII inhibition was assessed using the Morris water maze test. In addition, Western blotting, immunohistochemistry, and TUNEL staining were performed to determine neuronal death, apoptosis, and the relative signaling pathway. Results KN93-induced CaMKII inhibition decreased cAMP response element-binding (CREB) protein activity and impaired learning and memory in Wistar and tremor (TRM) rats, an animal model of genetic epilepsy. CaMKII inhibition also induced neuronal death and reactive astrocyte activation in both the Wistar and TRM hippocampi, deregulating mitogen-activated protein kinases. Meanwhile, neuronal death and neuron apoptosis were observed in PC12 and primary cultured hippocampal neurons after exposure to KN93, which was reversed by SP600125, an inhibitor of c-Jun N-terminal kinase (JNK). Conclusions CaMKII inhibition caused learning and memory impairment and apoptosis, which might be related to dysregulated JNK signaling.
Collapse
|
16
|
Maher A, El Sayed N, Nafea H, Gad M. Rolipram rescues memory consolidation deficits caused by sleep deprivation: Implication of the cAMP/PKA and cAMP/Epac pathways. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:631-639. [PMID: 34397335 DOI: 10.2174/1871527320666210816105144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Over the last few years, the number of people suffering from sleeping disorders has increased significantly despite negative effects on cognition and an association with brain inflammation. OBJECTIVES We assessed memory deficits caused by sleep deprivation (SD) to determine the therapeutic effect of phosphodiesterase 4 (PDE4) inhibitors on SD-induced memory deficits and to investigate whether the modulation of memory deficits by PDE4 inhibitors is mediated by a protein kinase A (PKA)-independent pathway in conjunction with a PKA-dependent pathway. METHODS Adult male mice were divided into four groups. Three SD groups were deprived of Rapid eye movement (REM) sleep for 12 h a day for six consecutive days. They were tested daily in the Morris water maze to evaluate learning and memory. One of the SD groups was injected with a PDE4 inhibitor, rolipram (1 mg/kg ip), whereas another had rolipram co-administered with chlorogenic acid (CHA, 20 mg/kg ip), an inhibitor of PKA. After 6 days, the mice were sacrificed, and the hippocampi were evaluated for cyclic AMP (cAMP) and nuclear factor Nrf-2 levels. The hippocampal expression of PKA, phosphorylated cAMP response element-binding protein (CREB), and phosphorylated glycogen synthase 3β (Ser389) were also evaluated. RESULTS SD caused a significant decrease in cAMP levels in the brain and had a detrimental effect on learning and memory. The administration of rolipram or rolipram+CHA resulted in an improvement in cognitive function. CONCLUSION The present study provides evidence that restoration of memory with PDE4 inhibitors occurs through a dual mechanism involving the PKA and Epac pathways.
Collapse
Affiliation(s)
- Ahmed Maher
- Biochemistry Department, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Cairo. Egypt
| | - Nesrine El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University. Egypt
| | - Heba Nafea
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo. Egypt
| | - Mohamed Gad
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo. Egypt
| |
Collapse
|
17
|
Kim YG, Bak MS, Kim A, Kim Y, Chae YC, Kim YL, Chun YS, An JY, Seo SB, Kim SJ, Lee YS. Kdm3b haploinsufficiency impairs the consolidation of cerebellum-dependent motor memory in mice. Mol Brain 2021; 14:106. [PMID: 34217333 PMCID: PMC8254933 DOI: 10.1186/s13041-021-00815-5] [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: 04/30/2021] [Accepted: 06/16/2021] [Indexed: 11/10/2022] Open
Abstract
Histone modifications are a key mechanism underlying the epigenetic regulation of gene expression, which is critically involved in the consolidation of multiple forms of memory. However, the roles of histone modifications in cerebellum-dependent motor learning and memory are not well understood. To test whether changes in histone methylation are involved in cerebellar learning, we used heterozygous Kdm3b knockout (Kdm3b+/-) mice, which show reduced lysine 9 on histone 3 (H3K9) demethylase activity. H3K9 di-methylation is significantly increased selectively in the granule cell layer of the cerebellum of Kdm3b+/- mice. In the cerebellum-dependent optokinetic response (OKR) learning, Kdm3b+/- mice show deficits in memory consolidation, whereas they are normal in basal oculomotor performance and OKR acquisition. In addition, RNA-seq analyses revealed that the expression levels of several plasticity-related genes were altered in the mutant cerebellum. Our study suggests that active regulation of histone methylation is critical for the consolidation of cerebellar motor memory.
Collapse
Affiliation(s)
- Yong Gyu Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Myeong Seong Bak
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Ahbin Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Yujin Kim
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, Korea
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul, Korea
| | - Yun-Cheol Chae
- Department of Life Science, College of Natural Science, Chung-Ang University, Seoul, 06974, Korea
| | - Ye Lee Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Yang-Sook Chun
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Joon-Yong An
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, Korea
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul, Korea
| | - Sang-Beom Seo
- Department of Life Science, College of Natural Science, Chung-Ang University, Seoul, 06974, Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.
| |
Collapse
|
18
|
Yang X, Li F, Liu Y, Li D, Li J. Study on the Correlation Between NF-κB and Central Fatigue. J Mol Neurosci 2021; 71:1975-1986. [PMID: 33586033 PMCID: PMC8500872 DOI: 10.1007/s12031-021-01803-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/26/2021] [Indexed: 12/25/2022]
Abstract
In recent years, the World Health Organization (WHO) has included fatigue as a major risk factor for human life and health. The incidence rate of fatigue is high. In Europe and America, nearly 1/3 of the population is suffering from fatigue. Due to the acceleration of modern people’s life rhythm and the increase of work pressure, more and more attention has been paid to central fatigue. The activation of NF-κB is related to central fatigue, which has been paid little attention by previous studies. At the same time, previous studies have mostly focused on the immune regulation function of NF-κB, while the NF-κB pathway plays an equally important role in regulating nerve function. NF-κB can participate in the occurrence and development of central fatigue by mediating immune inflammatory response, regulating central excitability and inhibitory transmitters, regulating synaptic plasticity and regulating central nervous system (CNS) functional genes. In addition to neuroprotective effects, NF-κB also has nerve damage effects, which is also closely related to the occurrence and development of central fatigue. In this review, we focus on the relationship between NF-κB pathway and central fatigue and further explore the biological mechanism of central fatigue. At the same time, the clinical application and potential of typical NF-κB inhibitors in the treatment of fatigue were analyzed to provide reference for the clinical treatment of central fatigue.
Collapse
Affiliation(s)
- Xingzhe Yang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Feng Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Yan Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Danxi Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
19
|
Sapio L, Salzillo A, Ragone A, Illiano M, Spina A, Naviglio S. Targeting CREB in Cancer Therapy: A Key Candidate or One of Many? An Update. Cancers (Basel) 2020; 12:cancers12113166. [PMID: 33126560 PMCID: PMC7693618 DOI: 10.3390/cancers12113166] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Only 5% of all drug-related targets currently move from preclinical to clinical in cancer, and just some of them achieve patient’s bedside. Among others, intratumor heterogeneity and preclinical cancer model limitations actually represent the main reasons for this failure. Cyclic-AMP response element-binding protein (CREB) has been defined as a proto-oncogene in different tumor types, being involved in maintenance and progression. Due to its relevance in tumor pathophysiology, many CREB inhibitor compounds have been developed and tested over the years. Herein, we examine the current state-of-the-art of both CREB and CREB inhibitors in cancer, retracing some of the most significant findings of the last years. While the scientific statement confers on CREB a proactive role in cancer, its therapeutic potential is still stuck at laboratory bench. Therefore, pursuing every concrete result to achieve CREB inhibition in clinical might give chance and future to cancer patients worldwide. Abstract Intratumor heterogeneity (ITH) is considered the major disorienting factor in cancer treatment. As a result of stochastic genetic and epigenetic alterations, the appearance of a branched evolutionary shape confers tumor plasticity, causing relapse and unfavorable clinical prognosis. The growing evidence in cancer discovery presents to us “the great paradox” consisting of countless potential targets constantly discovered and a small number of candidates being effective in human patients. Among these, cyclic-AMP response element-binding protein (CREB) has been proposed as proto-oncogene supporting tumor initiation, progression and metastasis. Overexpression and hyperactivation of CREB are frequently observed in cancer, whereas genetic and pharmacological CREB downregulation affects proliferation and apoptosis. Notably, the present review is designed to investigate the feasibility of targeting CREB in cancer therapy. In particular, starting with the latest CREB evidence in cancer pathophysiology, we evaluate the advancement state of CREB inhibitor design, including the histone lysine demethylases JMJD3/UTX inhibitor GSKJ4 that we newly identified as a promising CREB modulator in leukemia cells. Moreover, an accurate analysis of strengths and weaknesses is also conducted to figure out whether CREB can actually represent a therapeutic candidate or just one of the innumerable preclinical cancer targets.
Collapse
|
20
|
Wang L, Nie Q, Gao M, Yang L, Xiang JW, Xiao Y, Liu FY, Gong XD, Fu JL, Wang Y, Nguyen QD, Liu Y, Liu M, Li DWC. The transcription factor CREB acts as an important regulator mediating oxidative stress-induced apoptosis by suppressing αB-crystallin expression. Aging (Albany NY) 2020; 12:13594-13617. [PMID: 32554860 PMCID: PMC7377838 DOI: 10.18632/aging.103474] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/03/2020] [Indexed: 12/17/2022]
Abstract
The general transcription factor, CREB has been shown to play an essential role in promoting cell proliferation, neuronal survival and synaptic plasticity in the nervous system. However, its function in stress response remains to be elusive. In the present study, we demonstrated that CREB plays a major role in mediating stress response. In both rat lens organ culture and mouse lens epithelial cells (MLECs), CREB promotes oxidative stress-induced apoptosis. To confirm that CREB is a major player mediating the above stress response, we established stable lines of MLECs stably expressing CREB and found that they are also very sensitive to oxidative stress-induced apoptosis. To define the underlying mechanism, RNAseq analysis was conducted. It was found that CREB significantly suppressed expression of the αB-crystallin gene to sensitize CREB-expressing cells undergoing oxidative stress-induced apoptosis. CREB knockdown via CRISPR/CAS9 technology led to upregulation of αB-crystallin and enhanced resistance against oxidative stress-induced apoptosis. Moreover, overexpression of exogenous human αB-crystallin can restore the resistance against oxidative stress-induced apoptosis. Finally, we provided first evidence that CREB directly regulates αB-crystallin gene. Together, our results demonstrate that CREB is an important transcription factor mediating stress response, and it promotes oxidative stress-induced apoptosis by suppressing αB-crystallin expression.
Collapse
Affiliation(s)
- Ling Wang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Qian Nie
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Meng Gao
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
- Medical College, Henan University of Science and Technology, Luoyang 471000, Henan, China
| | - Lan Yang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Jia-Wen Xiang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
- Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Yuan Xiao
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Fang-Yuan Liu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Xiao-Dong Gong
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Jia-Ling Fu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Yan Wang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Quan Dong Nguyen
- Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Yizhi Liu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - David Wan-Cheng Li
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| |
Collapse
|