1
|
Kaipa BR, Kasetti R, Sundaresan Y, Li L, Yacoub S, Millar C, Cho W, Skowronska-Krawczyk D, Maddineni P, Palczewski K, Zode G. Impaired axonal transport at the optic nerve head contributes to neurodegeneration in a novel Cre-inducible mouse model of myocilin glaucoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613712. [PMID: 39345520 PMCID: PMC11429981 DOI: 10.1101/2024.09.18.613712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Elevation of intraocular pressure (IOP) due to trabecular meshwork (TM) dysfunction, leading to neurodegeneration, is the pathological hallmark of primary open-angle glaucoma (POAG). Impaired axonal transport is an early and critical feature of glaucomatous neurodegeneration. However, a robust mouse model that replicates these human POAG features accurately has been lacking. We report the development and characterization of a novel Cre-inducible mouse model expressing a DsRed-tagged Y437H mutant of human myocilin (Tg.CreMYOCY437H). A single intravitreal injection of HAd5-Cre induced selective MYOC expression in the TM, causing TM dysfunction, reducing outflow facility, and progressively elevating IOP in Tg.CreMYOCY437H mice. Sustained IOP elevation resulted in significant retinal ganglion cell (RGC) loss and progressive axonal degeneration in Cre-induced Tg.CreMYOCY437H mice. Notably, impaired anterograde axonal transport was observed at the optic nerve head before RGC degeneration, independent of age, indicating that impaired axonal transport contributes to RGC degeneration in Tg.CreMYOCY437H mice. In contrast, axonal transport remained intact in ocular hypertensive mice injected with microbeads, despite significant RGC loss. Our findings indicate that Cre-inducible Tg.CreMYOCY437H mice replicate all glaucoma phenotypes, providing an ideal model for studying early events of TM dysfunction and neuronal loss in POAG.
Collapse
|
2
|
Meng L, Jin H, Yulug B, Altay O, Li X, Hanoglu L, Cankaya S, Coskun E, Idil E, Nogaylar R, Ozsimsek A, Shoaie S, Turkez H, Nielsen J, Zhang C, Borén J, Uhlén M, Mardinoglu A. Multi-omics analysis reveals the key factors involved in the severity of the Alzheimer's disease. Alzheimers Res Ther 2024; 16:213. [PMID: 39358810 PMCID: PMC11448018 DOI: 10.1186/s13195-024-01578-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder with a global impact, yet its pathogenesis remains poorly understood. While age, metabolic abnormalities, and accumulation of neurotoxic substances are potential risk factors for AD, their effects are confounded by other factors. To address this challenge, we first utilized multi-omics data from 87 well phenotyped AD patients and generated plasma proteomics and metabolomics data, as well as gut and saliva metagenomics data to investigate the molecular-level alterations accounting the host-microbiome interactions. Second, we analyzed individual omics data and identified the key parameters involved in the severity of the dementia in AD patients. Next, we employed Artificial Intelligence (AI) based models to predict AD severity based on the significantly altered features identified in each omics analysis. Based on our integrative analysis, we found the clinical relevance of plasma proteins, including SKAP1 and NEFL, plasma metabolites including homovanillate and glutamate, and Paraprevotella clara in gut microbiome in predicting the AD severity. Finally, we validated the predictive power of our AI based models by generating additional multi-omics data from the same group of AD patients by following up for 3 months. Hence, we observed that these results may have important implications for the development of potential diagnostic and therapeutic approaches for AD patients.
Collapse
Affiliation(s)
- Lingqi Meng
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Han Jin
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Burak Yulug
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Ozlem Altay
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Xiangyu Li
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Lutfu Hanoglu
- Department of Neurology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Seyda Cankaya
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Ebru Coskun
- Department of Neurology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ezgi Idil
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Rahim Nogaylar
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Ahmet Ozsimsek
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Saeed Shoaie
- Centre for Host-Microbiome Interaction's, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden.
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden.
- Centre for Host-Microbiome Interaction's, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK.
| |
Collapse
|
3
|
Luo J, Lin S. Association between cadmium exposure and serum neurofilament light chain levels: A nationwide population-based survey. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 282:116771. [PMID: 39047369 DOI: 10.1016/j.ecoenv.2024.116771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/06/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Although cadmium exposure had been demonstrated to be toxic to the nervous system, little was known about the link between cadmium exposure and axonal injury. Therefore, the present study aimed to reveal whether there was any correlation between blood cadmium and serum neurofilament light chain (NfL) levels in the general population. METHODS This study included 1040 participants with a median (IQR) age of 47 (35-60) years from the 2013-2014 National Health and Nutrition Examination Survey. Serum NfL levels were measured through immunoassay, and whole blood cadmium concentrations were detected by means of inductively coupled plasma mass spectrometry. Linear regression and restricted cubic spline model was applied to analyze the significance of relationship between blood cadmium and serum NfL levels. RESULTS In the full adjusted model, blood cadmium levels were found to be positively associated with serum NfL levels (Q4 vs Q1, β = 3.35, 95 %CI: 0.41, 6.30, p for trend = 0.014). A potential linear positive dose-effect relationship was discovered between blood cadmium and serum NfL levels (p for non-linearity = 0.15). According to the result of stratified analysis, the significant positive relationship between blood cadmium and serum NfL levels was present only in the population of middle-aged and older adults. CONCLUSION The present study suggested a positive association between blood cadmium and serum NfL levels in the general US population.
Collapse
Affiliation(s)
- Jing Luo
- School of Rehabilitation, Jiangsu College of Nursing, Huaian, Jiangsu 223003, China
| | - Song Lin
- Department of Clinical Nutrition, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu 223300, China.
| |
Collapse
|
4
|
Ding EA, Kumar S. Neurofilament Biophysics: From Structure to Biomechanics. Mol Biol Cell 2024; 35:re1. [PMID: 38598299 PMCID: PMC11151108 DOI: 10.1091/mbc.e23-11-0438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/25/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
Neurofilaments (NFs) are multisubunit, neuron-specific intermediate filaments consisting of a 10-nm diameter filament "core" surrounded by a layer of long intrinsically disordered protein (IDP) "tails." NFs are thought to regulate axonal caliber during development and then stabilize the mature axon, with NF subunit misregulation, mutation, and aggregation featuring prominently in multiple neurological diseases. The field's understanding of NF structure, mechanics, and function has been deeply informed by a rich variety of biochemical, cell biological, and mouse genetic studies spanning more than four decades. These studies have contributed much to our collective understanding of NF function in axonal physiology and disease. In recent years, however, there has been a resurgence of interest in NF subunit proteins in two new contexts: as potential blood- and cerebrospinal fluid-based biomarkers of neuronal damage, and as model IDPs with intriguing properties. Here, we review established principles and more recent discoveries in NF structure and function. Where possible, we place these findings in the context of biophysics of NF assembly, interaction, and contributions to axonal mechanics.
Collapse
Affiliation(s)
- Erika A. Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720
| | - Sanjay Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
5
|
Yun Q, Ma SF, Zhang WN, Gu M, Wang J. FoxG1 as a Potential Therapeutic Target for Alzheimer's Disease: Modulating NLRP3 Inflammasome via AMPK/mTOR Autophagy Pathway. Cell Mol Neurobiol 2024; 44:35. [PMID: 38630150 PMCID: PMC11023968 DOI: 10.1007/s10571-024-01467-4] [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: 02/27/2024] [Indexed: 04/19/2024]
Abstract
An increasing body of research suggests that promoting microglial autophagy hinders the neuroinflammation initiated though the NLRP3 inflammasome activation in Alzheimer's disease (AD). The function of FoxG1, a crucial transcription factor involved in cell survival by regulating mitochondrial function, remains unknown during the AD process and neuroinflammation occurs. In the present study, we firstly found that Aβ peptides induced AD-like neuroinflammation upregulation and downregulated the level of autophagy. Following low-dose Aβ25-35 stimulation, FoxG1 expression and autophagy exhibited a gradual increase. Nevertheless, with high-concentration Aβ25-35 treatment, progressive decrease in FoxG1 expression and autophagy levels as the concentration of Aβ25-35 escalated. In addition, FoxG1 has a positive effect on cell viability and autophagy in the nervous system. In parallel with the Aβ25-35 stimulation, we employed siRNA to decrease the expression of FoxG1 in N2A cells. A substantial reduction in autophagy level (Beclin1, LC3II, SQSTM1/P62) and a notable growth in inflammatory response (NLRP3, TNF-α, and IL-6) were observed. In addition, we found FoxG1 overexpression owned the effect on the activation of AMPK/mTOR autophagy pathway and siRNA-FoxG1 successfully abolished this effect. Lastly, FoxG1 suppressed the NLRP3 inflammasome and enhanced the cognitive function in AD-like mouse model induced by Aβ25-35. Confirmed by cellular and animal experiments, FoxG1 suppressed NLRP3-mediated neuroinflammation, which was strongly linked to autophagy regulated by AMPK/mTOR. Taken together, FoxG1 may be a critical node in the pathologic progression of AD and has the potential to serve as therapeutic target.
Collapse
Affiliation(s)
- Qi Yun
- Changzhou Children's Hospital Affiliated to Nantong University, 958 Zhongwu Avenue, Changzhou, 213000, Jiangsu Province, China
| | - Si-Fei Ma
- Changzhou Blood Center, 118 Canal Road, Changzhou, 213000, Jiangsu Province, China
| | - Wei-Ning Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 213000, Jiangsu Province, China
| | - Meng Gu
- Changzhou Children's Hospital Affiliated to Nantong University, 958 Zhongwu Avenue, Changzhou, 213000, Jiangsu Province, China.
| | - Jia Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 213000, Jiangsu Province, China.
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu Province, PR China.
| |
Collapse
|
6
|
van Asperen JV, Kotaich F, Caillol D, Bomont P. Neurofilaments: Novel findings and future challenges. Curr Opin Cell Biol 2024; 87:102326. [PMID: 38401181 DOI: 10.1016/j.ceb.2024.102326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/07/2024] [Indexed: 02/26/2024]
Abstract
Neurofilaments (NFs) are abundant cytoskeletal proteins that emerge as a critical hub for cell signalling within neurons. As we start to uncover essential roles of NFs in regulating microtubule and organelle dynamics, nerve conduction and neurotransmission, novel discoveries are expected to arise in genetics, with NFs identified as causal genes for various neurodegenerative diseases. This review will discuss how the latest advances in fundamental and translational research illuminate our understanding of NF biology, particularly their assembly, organisation, transport and degradation. We will emphasise the notion that filaments are not one entity and that future challenges will be to apprehend their diverse composition and structural heterogeneity and to scrutinize how this regulates signalling, sustains neuronal physiology and drives pathophysiology in disease.
Collapse
Affiliation(s)
- Jessy V van Asperen
- ERC Team, NeuroMyoGene Insitute, INMG-PGNM, Inserm U1315, CNRS UMR5261, University of Lyon 1, Lyon, France
| | - Farah Kotaich
- ERC Team, NeuroMyoGene Insitute, INMG-PGNM, Inserm U1315, CNRS UMR5261, University of Lyon 1, Lyon, France
| | - Damien Caillol
- ERC Team, NeuroMyoGene Insitute, INMG-PGNM, Inserm U1315, CNRS UMR5261, University of Lyon 1, Lyon, France
| | - Pascale Bomont
- ERC Team, NeuroMyoGene Insitute, INMG-PGNM, Inserm U1315, CNRS UMR5261, University of Lyon 1, Lyon, France.
| |
Collapse
|
7
|
Kotaich F, Caillol D, Bomont P. Neurofilaments in health and Charcot-Marie-Tooth disease. Front Cell Dev Biol 2023; 11:1275155. [PMID: 38164457 PMCID: PMC10758125 DOI: 10.3389/fcell.2023.1275155] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/02/2023] [Indexed: 01/03/2024] Open
Abstract
Neurofilaments (NFs) are the most abundant component of mature neurons, that interconnect with actin and microtubules to form the cytoskeleton. Specifically expressed in the nervous system, NFs present the particularity within the Intermediate Filament family of being formed by four subunits, the neurofilament light (NF-L), medium (NF-M), heavy (NF-H) proteins and α-internexin or peripherin. Here, we review the current knowledge on NF proteins and neurofilaments, from their domain structures and their model of assembly to the dynamics of their transport and degradation along the axon. The formation of the filament and its behaviour are regulated by various determinants, including post-transcriptional (miRNA and RBP proteins) and post-translational (phosphorylation and ubiquitination) modifiers. Altogether, the complex set of modifications enable the neuron to establish a stable but elastic NF array constituting the structural scaffold of the axon, while permitting the local expression of NF proteins and providing the dynamics necessary to fulfil local demands and respond to stimuli and injury. Thus, in addition to their roles in mechano-resistance, radial axonal outgrowth and nerve conduction, NFs control microtubule dynamics, organelle distribution and neurotransmission at the synapse. We discuss how the studies of neurodegenerative diseases with NF aggregation shed light on the biology of NFs. In particular, the NEFL and NEFH genes are mutated in Charcot-Marie-Tooth (CMT) disease, the most common inherited neurological disorder of the peripheral nervous system. The clinical features of the CMT forms (axonal CMT2E, CMT2CC; demyelinating CMT1F; intermediate I-CMT) with symptoms affecting the central nervous system (CNS) will allow us to further investigate the physiological roles of NFs in the brain. Thus, NF-CMT mouse models exhibit various degrees of sensory-motor deficits associated with CNS symptoms. Cellular systems brought findings regarding the dominant effect of NF-L mutants on NF aggregation and transport, although these have been recently challenged. Neurofilament detection without NF-L in recessive CMT is puzzling, calling for a re-examination of the current model in which NF-L is indispensable for NF assembly. Overall, we discuss how the fundamental and translational fields are feeding each-other to increase but also challenge our knowledge of NF biology, and to develop therapeutic avenues for CMT and neurodegenerative diseases with NF aggregation.
Collapse
Affiliation(s)
| | | | - Pascale Bomont
- ERC team, NeuroMyoGene Institute-Pathophysiology and Genetics of Neuron and Muscle, Inserm U1315, CNRS UMR5261, University of Lyon 1, Lyon, France
| |
Collapse
|
8
|
Donini L, Tanel R, Zuccarino R, Basso M. Protein biomarkers for the diagnosis and prognosis of Amyotrophic Lateral Sclerosis. Neurosci Res 2023; 197:31-41. [PMID: 37689321 DOI: 10.1016/j.neures.2023.09.002] [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: 07/01/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/11/2023]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is the most common motor neuron disease, still incurable. The disease is highly heterogenous both genetically and phenotypically. Therefore, developing efficacious treatments is challenging in many aspects because it is difficult to predict the rate of disease progression and stratify the patients to minimize statistical variability in clinical studies. Moreover, there is a lack of sensitive measures of therapeutic effect to assess whether a pharmacological intervention ameliorates the disease. There is also urgency of markers that reflect a molecular mechanism dysregulated by ALS pathology and can be rescued when a treatment relieves the condition. Here, we summarize and discuss biomarkers tested in multicentered studies and across different laboratories like neurofilaments, the most used marker in ALS clinical studies, neuroinflammatory-related proteins, p75ECD, p-Tau/t-Tau, and UCHL1. We also explore the applicability of muscle proteins and extracellular vesicles as potential biomarkers.
Collapse
Affiliation(s)
- Luisa Donini
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Italy.
| | - Raffaella Tanel
- Clinical Center NeMO, APSS Ospedale Riabilitativo Villa Rosa, Pergine 38057, TN, Italy.
| | - Riccardo Zuccarino
- Clinical Center NeMO, APSS Ospedale Riabilitativo Villa Rosa, Pergine 38057, TN, Italy
| | - Manuela Basso
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Italy.
| |
Collapse
|
9
|
Han X, Yu S, Cui Y, Li J, Fan J, Wang L, Wang M, Pan Y, Xu G. MiR-23a promotes autophagy of yak cumulus cells to alleviate apoptosis via the apoptosis signal-regulating kinase 1/c-Jun N-terminal kinase pathway. Theriogenology 2023; 212:50-63. [PMID: 37690377 DOI: 10.1016/j.theriogenology.2023.08.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/04/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023]
Abstract
The ultimate fate of Graafian follicles is ovulation or atresia which relies on the highly coordinated processes of apoptosis and autophagy in ovarian cells. Long non-coding RNA maternally expressed gene 3 (LncRNA MEG3), miR-23a, and apoptosis signal-regulating kinase 1 (ASK1) are factors associated with autophagy. However, whether these factors can regulate autophagy in cumulus cells (CCs) of yak is unclear. Here, miR-23a overexpression upregulated the LC3-II/LC3-I ratio and Beclin1 abundance while reducing p62 accumulation (p < 0.05). The monodansylcadaverine assay exhibited a marked increase in punctate green fluorescence, and the GFP-LC3B displayed increased yellow fluorescence (p < 0.05). The opposite effect was observed for miR-23a inhibitors. Furthermore, miR-23a overexpression downregulated the abundance of ASK1 mRNA and total ASK1 protein (t-ASK1), whereas miR-23a inhibitors up-regulated them (p < 0.05). The effects of miR-23a overexpression on ASK1 phosphorylated protein at serine 845 (P-845), total JNK (c-Jun N-terminal kinase) (t-JNK) and the JNK phosphorylated protein (p-JNK) were similar to those of t-ASK1 but elicited the opposite effect on ASK1 phosphorylated protein at serine 967 (P-967) (p < 0.05). We further demonstrated that ASK1 expression can be silenced by small-interfering RNA (siRNA), which had no significant effect on t-JNK abundance (p > 0.05) but significantly suppressed the p-JNK expression (p < 0.05). Silencing ASK1 significantly improved Beclin1 abundance and the LC3-II/LC3-I ratio, but decreased p62 abundance (p < 0.05). An increase in yellow GFP-LC3B puncta and green MDC staining puncta were observed (p < 0.05). Overexpression of LncRNA MEG3 significantly increased the expression of t-ASK1, P-845, and JNK and decreased the abundance of P-967 and miR-23a (p < 0.05). In addition, miR-23a upregulation reduced the number of the TUNEL-positive cells, and the addition of 8 mM 3-methyladenine (3-MA) reversed this downregulation (p < 0.05). Similar trends were observed for the Bax/Bcl2 ratio and cleaved-caspase3 abundance. In summary, miR-23a promotes autophagy by inhibiting ASK1 abundance, which reduces apoptosis of yak CCs. This effect can be inhibited by LncRNA MEG3, which has implications for decreasing abnormal Graafian follicular atresia and maintaining development.
Collapse
Affiliation(s)
- Xiaohong Han
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Sijiu Yu
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Yan Cui
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jingjing Li
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jiangfeng Fan
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Libin Wang
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Meng Wang
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Yangyang Pan
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Gengquan Xu
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| |
Collapse
|
10
|
Zang L, Yang X, Chen Y, Huang F, Yuan Y, Chen X, Zuo Y, Miao Y, Gu J, Guo H, Xia W, Peng Y, Tang M, Huang Z, Wang Y, Ma J, Jiang J, Zhou W, Zheng H, Shi W. Ubiquitin E3 ligase SPOP is a host negative regulator of enterovirus 71-encoded 2A protease. J Virol 2023; 97:e0078623. [PMID: 37796126 PMCID: PMC10617436 DOI: 10.1128/jvi.00786-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/25/2023] [Indexed: 10/06/2023] Open
Abstract
IMPORTANCE EV71 poses a significant health threat to children aged 5 and below. The process of EV71 infection and replication is predominantly influenced by ubiquitination modifications. Our previous findings indicate that EV71 prompts the activation of host deubiquitinating enzymes, thereby impeding the host interferon signaling pathway as a means of evading the immune response. Nevertheless, the precise mechanisms by which the host employs ubiquitination modifications to hinder EV71 infection remain unclear. The present study demonstrated that the nonstructural protein 2Apro, which is encoded by EV71, exhibits ubiquitination and degradation mediated by the host E3 ubiquitin ligase SPOP. In addition, it is the first report, to our knowledge, that SPOP is involved in the host antiviral response.
Collapse
Affiliation(s)
- Lichao Zang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Xinyu Yang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yan Chen
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Fan Huang
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Yukang Yuan
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Xiangjie Chen
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Yibo Zuo
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Ying Miao
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Jin Gu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Hui Guo
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Wenxin Xia
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yang Peng
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Mengyuan Tang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Ziwei Huang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yangyang Wang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jinhong Ma
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Wei Zhou
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Hui Zheng
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Weifeng Shi
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
11
|
Cheah M, Cheng Y, Petrova V, Cimpean A, Jendelova P, Swarup V, Woolf CJ, Geschwind DH, Fawcett JW. Integrin-Driven Axon Regeneration in the Spinal Cord Activates a Distinctive CNS Regeneration Program. J Neurosci 2023; 43:4775-4794. [PMID: 37277179 PMCID: PMC10312060 DOI: 10.1523/jneurosci.2076-22.2023] [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: 11/06/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 06/07/2023] Open
Abstract
The peripheral branch of sensory dorsal root ganglion (DRG) neurons regenerates readily after injury unlike their central branch in the spinal cord. However, extensive regeneration and reconnection of sensory axons in the spinal cord can be driven by the expression of α9 integrin and its activator kindlin-1 (α9k1), which enable axons to interact with tenascin-C. To elucidate the mechanisms and downstream pathways affected by activated integrin expression and central regeneration, we conducted transcriptomic analyses of adult male rat DRG sensory neurons transduced with α9k1, and controls, with and without axotomy of the central branch. Expression of α9k1 without the central axotomy led to upregulation of a known PNS regeneration program, including many genes associated with peripheral nerve regeneration. Coupling α9k1 treatment with dorsal root axotomy led to extensive central axonal regeneration. In addition to the program upregulated by α9k1 expression, regeneration in the spinal cord led to expression of a distinctive CNS regeneration program, including genes associated with ubiquitination, autophagy, endoplasmic reticulum (ER), trafficking, and signaling. Pharmacological inhibition of these processes blocked the regeneration of axons from DRGs and human iPSC-derived sensory neurons, validating their causal contributions to sensory regeneration. This CNS regeneration-associated program showed little correlation with either embryonic development or PNS regeneration programs. Potential transcriptional drivers of this CNS program coupled to regeneration include Mef2a, Runx3, E2f4, and Yy1. Signaling from integrins primes sensory neurons for regeneration, but their axon growth in the CNS is associated with an additional distinctive program that differs from that involved in PNS regeneration.SIGNIFICANCE STATEMENT Restoration of neurologic function after spinal cord injury has yet to be achieved in human patients. To accomplish this, severed nerve fibers must be made to regenerate. Reconstruction of nerve pathways has not been possible, but recently, a method for stimulating long-distance axon regeneration of sensory fibers in rodents has been developed. This research uses profiling of messenger RNAs in the regenerating sensory neurons to discover which mechanisms are activated. This study shows that the regenerating neurons initiate a novel CNS regeneration program which includes molecular transport, autophagy, ubiquitination, and modulation of the endoplasmic reticulum (ER). The study identifies mechanisms that neurons need to activate to regenerate their nerve fibers.
Collapse
Affiliation(s)
- Menghon Cheah
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, United Kingdom
| | - Yuyan Cheng
- Program in Neurogenetics, Department of Neurology, and Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Veselina Petrova
- Department of Neurobiology, Harvard Medical School; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115
| | - Anda Cimpean
- Centre for Reconstructive Neuroscience, Institute of Experimental Medicine Czech Academy of Science, Prague, Czech Republic
| | - Pavla Jendelova
- Centre for Reconstructive Neuroscience, Institute of Experimental Medicine Czech Academy of Science, Prague, Czech Republic
| | - Vivek Swarup
- Program in Neurogenetics, Department of Neurology, and Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California 90095
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697
| | - Clifford J Woolf
- Department of Neurobiology, Harvard Medical School; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, and Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - James W Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, United Kingdom
- Centre for Reconstructive Neuroscience, Institute of Experimental Medicine Czech Academy of Science, Prague, Czech Republic
| |
Collapse
|
12
|
Rooney TC, Aldred GG, Boffey HK, Willems HG, Edwards S, Chawner SJ, Scott DE, Green C, Winpenny D, Skidmore J, Clarke JH, Andrews SP. The Identification of Potent, Selective, and Brain Penetrant PI5P4Kγ Inhibitors as In Vivo-Ready Tool Molecules. J Med Chem 2022; 66:804-821. [PMID: 36516442 PMCID: PMC9841522 DOI: 10.1021/acs.jmedchem.2c01693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Owing to their central role in regulating cell signaling pathways, the phosphatidylinositol 5-phosphate 4-kinases (PI5P4Ks) are attractive therapeutic targets in diseases such as cancer, neurodegeneration, and immunological disorders. Until now, tool molecules for these kinases have been either limited in potency or isoform selectivity, which has hampered further investigation of biology and drug development. Herein we describe the virtual screening workflow which identified a series of thienylpyrimidines as PI5P4Kγ-selective inhibitors, as well as the medicinal chemistry optimization of this chemotype, to provide potent and selective tool molecules for further use. In vivo pharmacokinetics data are presented for exemplar tool molecules, along with an X-ray structure for ARUK2001607 (15) in complex with PI5P4Kγ, along with its selectivity data against >150 kinases and a Cerep safety panel.
Collapse
|