1
|
Sandhu A, Rawat K, Gautam V, Kumar A, Sharma A, Bhatia A, Grover S, Saini L, Saha L. Neuroprotective effect of PPAR gamma agonist in rat model of autism spectrum disorder: Role of Wnt/β-catenin pathway. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111126. [PMID: 39179196 DOI: 10.1016/j.pnpbp.2024.111126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND The clinical manifestation of autism spectrum disorder (ASD) is linked to the disruption of fundamental neurodevelopmental pathways. Emerging evidences claim to have an upregulation of canonical Wnt/β-catenin pathway while downregulation of PPARγ pathway in ASD. This study aims to investigate the therapeutic potential of pioglitazone, a PPARγ agonist, in rat model of ASD. The study further explores the possible role of PPARγ and Wnt/β-catenin pathway and their interaction in ASD by using their modulators. MATERIAL AND METHODS Pregnant female Wistar rats received 600 mg/kg of valproic acid (VPA) to induce autistic symptoms in pups. Pioglitazone (10 mg/kg) was used to evaluate neurobehaviors, relative mRNA expression of inflammatory (IL-1β, IL-6, IL-10, TNF-α), apoptotic markers (Bcl-2, Bax, & Caspase-3) and histopathology (H&E, Nissl stain, Immunohistochemistry). Effect of pioglitazone was evaluated on Wnt pathway and 4 μg/kg dose of 6-BIO (Wnt modulator) was used to study the PPARγ pathway. RESULTS ASD model was established in pups as indicated by core autistic symptoms, increased neuroinflammation, apoptosis and histopathological neurodegeneration in cerebellum, hippocampus and amygdala. Pioglitazone significantly attenuated these alterations in VPA-exposed rats. The expression study results indicated an increase in key transcription factor, β-catenin in VPA-rats suggesting an upregulation of canonical Wnt pathway in them. Pioglitazone significantly downregulated the Wnt signaling by suppressing the expression of Wnt signaling-associated proteins. The inhibiting effect of Wnt pathway on PPARγ activity was indicated by downregulation of PPARγ-associated protein in VPA-exposed rats and those administered with 6-BIO. CONCLUSION In the present study, upregulation of canonical Wnt/β-catenin pathway was demonstrated in ASD rat model. Pioglitazone administration significantly ameliorated these symptoms potentially through its neuroprotective effect and its ability to downregulate the Wnt/β-catenin pathway. The antagonism between the PPARγ and Wnt pathway offers a promising therapeutic approach for addressing ASD.
Collapse
Affiliation(s)
- Arushi Sandhu
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India
| | - Kajal Rawat
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India
| | - Vipasha Gautam
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India
| | - Anil Kumar
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India
| | - Antika Sharma
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Sandeep Grover
- Department of Psychiatry, Post Graduate Institute ofMedical Education and Research (PGIMER), Chandigarh 160012, India
| | - Lokesh Saini
- Department of Paediatrics, All India Institute of Medical Sciences (AIIMS), Jodhpur, Rajasthan 342001, India
| | - Lekha Saha
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh 160012, India.
| |
Collapse
|
2
|
Fu Y, Gu Z, Cao H, Zuo C, Huang Y, Song Y, Miao J, Jiang Y, Wang F. Proteomic characterization of the medial prefrontal cortex in chronic restraint stress mice. J Proteomics 2024; 307:105278. [PMID: 39142625 DOI: 10.1016/j.jprot.2024.105278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/30/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
Depression is a prominent contributor to global disability. A growing body of data suggests that depression is associated with the pathophysiology of the medial prefrontal cortex (mPFC), but the underlying mechanisms remain poorly understood. Mice were subjected to chronic restraint stress (CRS) for 3 weeks to create depression models during this investigation. Protein tandem mass tag (TMT) quantification and LC-MS/MS analysis were conducted to examine proteome patterns. Afterwards, to further explore the enrichment of differential proteins and the signaling pathways involved, we annotated these differentially expressed proteins. We confirmed that CRS mice developed depression-like and anxiety-like behaviors. Among the 8081 measured proteins, a total of 15 proteins were found to be differentially expressed. These proteins exhibited functional enrichment in a variety of biological functions, and among these pathways, alterations in synaptic function and autophagy are noteworthy. In addition, we identified a differentially expressed protein called Wnt2b and found that CRS may disrupt synaptic plasticity by affecting the activation of the Wnt2b/β-catenin pathway. Our findings showed depression-like behaviors in the CRS mouse model and molecular alterations in the mPFC, which may help explain the pathogenesis of depression and identify novel antidepressant medication targets. SIGNIFICANCE: Depression is a prevalent and frequent chronic mental illness and is now a significant contributor to global disability. In this study, we used chronic restraint stress to establish a mouse model of depression, and differentially expressed proteins in the medial prefrontal cortex of depressed model mice were detected by TMT proteomics. Our study verified the presence of altered synaptic function and excessive autophagy in the mPFC of CRS-induced mice from a proteomic perspective. Furthermore, we demonstrated that CRS may disrupt synaptic plasticity by affecting the activation of the Wnt2b/β-catenin pathway, which may be a key link in the pathogenesis of depression and may provide new insights for identifying new antidepressant drug targets.
Collapse
Affiliation(s)
- Yufeng Fu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Zhongya Gu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Huan Cao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Chengchao Zuo
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yaqi Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yu Song
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Jinfeng Miao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yongsheng Jiang
- Cancer Center of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China.
| | - Furong Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China; Key Laboratory of Vascular Aging (HUST), Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China.
| |
Collapse
|
3
|
Zhang N, Westerhaus A, Wilson M, Wang E, Goff L, Sockanathan S. Physiological regulation of neuronal Wnt activity is essential for TDP-43 localization and function. EMBO J 2024; 43:3388-3413. [PMID: 38918634 PMCID: PMC11329687 DOI: 10.1038/s44318-024-00156-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/31/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Nuclear exclusion of the RNA- and DNA-binding protein TDP-43 can induce neurodegeneration in different diseases. Diverse processes have been implicated to influence TDP-43 mislocalization, including disrupted nucleocytoplasmic transport (NCT); however, the physiological pathways that normally ensure TDP-43 nuclear localization are unclear. The six-transmembrane enzyme glycerophosphodiester phosphodiesterase 2 (GDE2 or GDPD5) cleaves the glycosylphosphatidylinositol (GPI) anchor that tethers some proteins to the membrane. Here we show that GDE2 maintains TDP-43 nuclear localization by regulating the dynamics of canonical Wnt signaling. Ablation of GDE2 causes aberrantly sustained Wnt activation in adult neurons, which is sufficient to cause NCT deficits, nuclear pore abnormalities, and TDP-43 nuclear exclusion. Disruption of GDE2 coincides with TDP-43 abnormalities in postmortem tissue from patients with amyotrophic lateral sclerosis (ALS). Further, GDE2 deficits are evident in human neural cell models of ALS, which display erroneous Wnt activation that, when inhibited, increases mRNA levels of genes regulated by TDP-43. Our study identifies GDE2 as a critical physiological regulator of Wnt signaling in adult neurons and highlights Wnt pathway activation as an unappreciated mechanism contributing to nucleocytoplasmic transport and TDP-43 abnormalities in disease.
Collapse
Affiliation(s)
- Nan Zhang
- The Solomon Snyder Department of Neuroscience, The Johns Hopkins School of Medicine, 725 N Wolfe Street, Baltimore, MD, 21205, USA
| | - Anna Westerhaus
- The Solomon Snyder Department of Neuroscience, The Johns Hopkins School of Medicine, 725 N Wolfe Street, Baltimore, MD, 21205, USA
| | - Macey Wilson
- The Solomon Snyder Department of Neuroscience, The Johns Hopkins School of Medicine, 725 N Wolfe Street, Baltimore, MD, 21205, USA
- Department of Cellular Biology, University of Georgia, Biological Sciences 302, 120 Cedar St., Athens, GA, 30602, USA
| | - Ethan Wang
- The Solomon Snyder Department of Neuroscience, The Johns Hopkins School of Medicine, 725 N Wolfe Street, Baltimore, MD, 21205, USA
| | - Loyal Goff
- The Solomon Snyder Department of Neuroscience, The Johns Hopkins School of Medicine, 725 N Wolfe Street, Baltimore, MD, 21205, USA
- McKusick-Nathans Department of Genetic Medicine, Kavli Neurodiscovery Institute, The Johns Hopkins School of Medicine, 725 N Wolfe Street, Baltimore, MD, 21205, USA
| | - Shanthini Sockanathan
- The Solomon Snyder Department of Neuroscience, The Johns Hopkins School of Medicine, 725 N Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
4
|
Su L, Zhang M, Ji F, Zhao J, Wang Y, Wang W, Zhang S, Ma H, Wang Y, Jiao J. Microglia homeostasis mediated by epigenetic ARID1A regulates neural progenitor cells response and leads to autism-like behaviors. Mol Psychiatry 2024; 29:1595-1609. [PMID: 35858990 DOI: 10.1038/s41380-022-01703-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 01/26/2023]
Abstract
Microglia are resident macrophages of the central nervous system that selectively emerge in embryonic cortical proliferative zones and regulate neurogenesis by altering molecular and phenotypic states. Despite their important roles in inflammatory phagocytosis and neurodegenerative diseases, microglial homeostasis during early brain development has not been fully elucidated. Here, we demonstrate a notable interplay between microglial homeostasis and neural progenitor cell signal transduction during embryonic neurogenesis. ARID1A, an epigenetic subunit of the SWI/SNF chromatin-remodeling complex, disrupts genome-wide H3K9me3 occupancy in microglia and changes the epigenetic chromatin landscape of regulatory elements that influence the switching of microglial states. Perturbation of microglial homeostasis impairs the release of PRG3, which regulates neural progenitor cell self-renewal and differentiation during embryonic development. Furthermore, the loss of microglia-driven PRG3 alters the downstream cascade of the Wnt/β-catenin signaling pathway through its interaction with the neural progenitor receptor LRP6, which leads to misplaced regulation in neuronal development and causes autism-like behaviors at later stages. Thus, during early fetal brain development, microglia progress toward a more homeostatic competent phenotype, which might render neural progenitor cells respond to environmental cross-talk perturbations.
Collapse
Affiliation(s)
- Libo Su
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Mengtian Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Fen Ji
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Jinyue Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yuanyuan Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Wenwen Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, 230026, China
| | - Shukui Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- College of Life Sciences, Yantai University, Yantai, 264005, Shandong, China
| | - Hongyan Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yanyan Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
| |
Collapse
|
5
|
Fetsko AR, Sebo DJ, Budzynski LB, Scharbarth A, Taylor MR. IL-1β disrupts the initiation of blood-brain barrier development by inhibiting endothelial Wnt/β-catenin signaling. iScience 2024; 27:109651. [PMID: 38638574 PMCID: PMC11025013 DOI: 10.1016/j.isci.2024.109651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/06/2024] [Accepted: 03/29/2024] [Indexed: 04/20/2024] Open
Abstract
During neuroinflammation, the proinflammatory cytokine interleukin-1β (IL-1β) impacts blood-brain barrier (BBB) function by disrupting brain endothelial tight junctions, promoting vascular permeability, and increasing transmigration of immune cells. Here, we examined the effects of Il-1β on the in vivo initiation of BBB development. We generated doxycycline-inducible transgenic zebrafish to secrete Il-1β in the CNS. To validate the utility of our model, we showed Il-1β dose-dependent mortality, recruitment of neutrophils, and expansion of microglia. Using live imaging, we discovered that Il-1β causes a significant reduction in CNS angiogenesis and barriergenesis. To demonstrate specificity, we rescued the Il-1β induced phenotypes by targeting the zebrafish il1r1 gene using CRISPR-Cas9. Mechanistically, we determined that Il-1β disrupts the initiation of BBB development by decreasing Wnt/β-catenin transcriptional activation in brain endothelial cells. Given that several neurodevelopmental disorders are associated with inflammation, our findings support further investigation into the connections between proinflammatory cytokines, neuroinflammation, and neurovascular development.
Collapse
Affiliation(s)
- Audrey R. Fetsko
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dylan J. Sebo
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Lilyana B. Budzynski
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Alli Scharbarth
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Michael R. Taylor
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
6
|
Fetsko AR, Sebo DJ, Budzynski LB, Scharbarth A, Taylor MR. IL-1β disrupts blood-brain barrier development by inhibiting endothelial Wnt/β-catenin signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.04.569943. [PMID: 38106202 PMCID: PMC10723338 DOI: 10.1101/2023.12.04.569943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
During neuroinflammation, the proinflammatory cytokine Interleukin-1β (IL-1β) impacts blood-brain barrier (BBB) function by disrupting brain endothelial tight junctions, promoting vascular permeability, and increasing transmigration of immune cells. Here, we examined the effects of Il-1β on the in vivo development of the BBB. We generated a doxycycline-inducible transgenic zebrafish model that drives secretion of Il-1β in the CNS. To validate the utility of our model, we showed Il-1β dose-dependent mortality, recruitment of neutrophils, and expansion of microglia. Using live imaging, we discovered that Il-1β causes a significant reduction in CNS angiogenesis and barriergenesis. To demonstrate specificity, we rescued the Il-1β induced phenotypes by targeting the zebrafish il1r1 gene using CRISPR/Cas9. Mechanistically, we determined that Il-1β disrupts BBB development by decreasing Wnt/β-catenin transcriptional activation in brain endothelial cells. Given that several neurodevelopmental disorders are associated with inflammation, our findings support further investigation into the connections between proinflammatory cytokines, neuroinflammation, and neurovascular development.
Collapse
Affiliation(s)
- Audrey R. Fetsko
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Dylan J. Sebo
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Lilyana B. Budzynski
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Alli Scharbarth
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael R. Taylor
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
7
|
Ding Y, Li L, Wang S, Cao Y, Yang M, Dai Y, Lin H, Li J, Liu Y, Wang Z, Liu W, Tao J. Electroacupuncture promotes neurogenesis in the dentate gyrus and improves pattern separation in an early Alzheimer's disease mouse model. Biol Res 2023; 56:65. [PMID: 38041203 PMCID: PMC10693055 DOI: 10.1186/s40659-023-00472-z] [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: 11/08/2022] [Accepted: 11/03/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Impaired pattern separation occurs in the early stage of Alzheimer's disease (AD), and hippocampal dentate gyrus (DG) neurogenesis participates in pattern separation. Here, we investigated whether spatial memory discrimination impairment can be improved by promoting the hippocampal DG granule cell neogenesis-mediated pattern separation in the early stage of AD by electroacupuncture (EA). METHODS Five familial AD mutations (5 × FAD) mice received EA treatment at Baihui and Shenting points for 4 weeks. During EA, mice were intraperitoneally injected with BrdU (50 mg/kg) twice a day. rAAV containing Wnt5a shRNA was injected into the bilateral DG region, and the viral efficiency was evaluated by detecting Wnt5a mRNA levels. Cognitive behavior tests were conducted to assess the impact of EA treatment on cognitive function. The hippocampal DG area Aβ deposition level was detected by immunohistochemistry after the intervention; The number of BrdU+/CaR+ cells and the gene expression level of calretinin (CaR) and prospero homeobox 1(Prox1) in the DG area of the hippocampus was detected to assess neurogenesis by immunofluorescence and western blotting after the intervention; The gene expression levels of FZD2, Wnt5a, DVL2, p-DVL2, CaMKII, and p-CaMKII in the Wnt signaling pathway were detected by Western blotting after the intervention. RESULTS Cognitive behavioral tests showed that 5 × FAD mice had impaired pattern separation (P < 0.001), which could be improved by EA (P < 0.01). Immunofluorescence and Western blot showed that the expression of Wnt5a in the hippocampus was decreased (P < 0.001), and the neurogenesis in the DG was impaired (P < 0.001) in 5 × FAD mice. EA could increase the expression level of Wnt5a (P < 0.05) and promote the neurogenesis of immature granule cells (P < 0.05) and the development of neuronal dendritic spines (P < 0.05). Interference of Wnt5a expression aggravated the damage of neurogenesis (P < 0.05), weakened the memory discrimination ability (P < 0.05), and inhibited the beneficial effect of EA (P < 0.05) in AD mice. The expression level of Wnt pathway related proteins such as FZD2, DVL2, p-DVL2, CAMKII, p-CAMKII increased after EA, but the effect of EA was inhibited after Wnt5a was knocked down. In addition, EA could reduce the deposition of Aβ plaques in the DG without any impact on Wnt5a. CONCLUSION EA can promote hippocampal DG immature granule cell neogenesis-mediated pattern separation to improve spatial memory discrimination impairment by regulating Wnt5a in 5 × FAD mice.
Collapse
Affiliation(s)
- Yanyi Ding
- The Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Long Li
- The Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Sinuo Wang
- The Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yajun Cao
- The Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Minguang Yang
- The Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yaling Dai
- The Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Huawei Lin
- The Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Jianhong Li
- The Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yulu Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Zhifu Wang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fuzhou, Fujian, 350122, China
- Fujian Key Laboratory of Cognitive Rehabilitation, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, 350001, China
| | - Weilin Liu
- The Institute of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China.
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China.
- Fujian Key Laboratory of Rehabilitation Technology, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, 350001, China.
- Provincial and Ministerial Co-founded Collaborative Innovation Center of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| |
Collapse
|
8
|
Meng Z, Liu Q, Liu Y, Yang Y, Shao C, Zhang S. Frizzled-3 suppression overcomes multidrug chemoresistance by Wnt/β-catenin signaling pathway inhibition in hepatocellular carcinoma cells. J Chemother 2023; 35:653-661. [PMID: 36843499 DOI: 10.1080/1120009x.2023.2182573] [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: 07/19/2022] [Revised: 02/04/2023] [Accepted: 02/15/2023] [Indexed: 02/28/2023]
Abstract
Multidrug resistance (MDR) is a major obstacle to the efficacy of hepatocellular carcinoma (HCC) chemotherapy. Previous studies have identified that low FZD3 predicted decreased survival after intraperitoneal versus intravenous-only chemotherapy in ovarian cancer. This study aimed to identify a potential target in HCC chemotherapy. The FZD3 expression variant in HCC cell lines was detected by RT-qPCR and western blotting. The FZD3 expression in the early recurrent HCC group (RE group) and the non-early recurrent HCC group (non-RE group) was measured by RT-qPCR. Then, the 50% inhibitory concentrations (IC50) in HCC cell lines were studied by MTT assay. TOP/FOP FLASH luciferase assay was performed to measure TCF-binding activities. We found that FZD3 was upregulated in three HCC cell lines, and the FZD3 expression was significantly higher in the RE group than in the non-RE group (P = 0.0344). A positive correlation between FZD3 and MDR1 was observed in HCC tissues (R2 = 0.6368, P = 0.0001). Then, we found that FZD3 knockdown significantly altered Huh-7 cell chemotherapeutic sensitivity to cisplatin [50.43 µM in the FZD3 siRNA (siFZD3) group vs 98.59 µM in the siRNA negative control (siNC) group; P = 0.007] or doxorubicin (7.43 µM in the siFZD3 group vs 14.93 µM in the siNC group; P = 0.017). TOP/FOP FLASH luciferase assay showed FZD3 could inhibit Wnt/β-catenin signaling in HCC cells. Moreover, FZD3 expression knockdown in SNU-449 and Huh-7 cells markedly reduced β-catenin and phosho-β-catenin (S37) protein expression, and Cyclin D1, c-myc and MDR1 were significantly decreased. This is the first study to describe the significantly increased FZD3 expression in patients with early recurrent HCC. FZD3 knockdown led to increased sensitivity to chemotherapy by Wnt/β-catenin signaling inhibition in HCC cell lines. Our study suggests FZD3 as a potential target for reversing chemoresistance in HCC.
Collapse
Affiliation(s)
- Zifan Meng
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qing Liu
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanfei Liu
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuanming Yang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changfeng Shao
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shaoqiang Zhang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Wang Z, Li XN, Yang SN, Wang Y, Gao KJ, Han B, Ma AJ. Exosomal miR-320e through wnt2targeted inhibition of the Wnt/β-catenin pathway allevisate cerebral small vessel disease and cognitive impairment. World J Psychiatry 2023; 13:630-644. [PMID: 37771642 PMCID: PMC10523201 DOI: 10.5498/wjp.v13.i9.630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/20/2023] [Accepted: 07/14/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Exosomal miRNAs play crucial roles in many central nervous system diseases. Cerebral small vessel disease (CVSD) is a small vessel disease that is affected by various factors. This study aimed to investigate the role of exosomal miR-320e in the Wnt/β-catenin pathway stimulated by oxidative stress and assess its clinical correlation with psychiatric symptoms in patients with CVSD. AIM To explore whether exosomal miR-320e could suppress the Wnt/β-catenin pathway and play a protective role in CVSD progression, as well as examine its potential correlation with cognitive impairment and depression in patients with CVSD. METHODS Differentially expressed exosomal miRNAs were filtered by sequencing plasma exosomes from patients with CVSD and healthy controls. Bioinformatics and dual luciferase analyses were used to confirm the binding of miR-320e to Wnt2, and the mRNA and protein levels of downstream components in the Wnt/β-catenin pathway were evaluated when overexpressed or with knockdown of miR-320e under H2O2-induced oxidative stress. In addition, Wnt2-targeting siRNA was used to confirm the role of miR-320e in the Wnt2-mediated inhibition of the Wnt/β-catenin pathway. A retrospective analysis was conducted among patients with CVSD to confirm the correlation between miR-320e expression and the severity of cognitive impairment and depression, which were quantified using the Montreal Cognitive Assessment (MoCA)/Executive Function Assessment (EFA), and the Hamilton Depression Scale (HAMD)/Beck Depression Inventory (BDI), respectively. RESULTS High-throughput sequencing revealed that exosomal miR-320e was downregulated in patients with CVSD. Bioinformatics analysis and dual-luciferase reporter gene experiments showed that exosomal miR-320e inhibited the Wnt/β-catenin pathway in response to oxidative stress by targeting the 3' noncoding region of Wnt2. Uptake of exosomes carrying miR-320e into endothelial cells could also target Wnt2 and inhibit the Wnt2/β-catenin pathway. Elevated miR-320e expression may protect patients with CVSD from relatively severe cognitive impairment and depression, as it was found to have a positive correlation with the MoCA/EFA and HAMD/BDI scores. CONCLUSION Our results suggest that exosomal miR-320e suppresses the Wnt/β-catenin pathway and may play a protective role in CVSD progression.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Internal Medicine-Neurology, Affiliated Hospital of Qingdao University, Qingdao 266001, Shandong Province, China
| | - Xue-Ning Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266001, Shandong Province, China
| | - Shao-Nan Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266001, Shandong Province, China
| | - Yuan Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266001, Shandong Province, China
| | - Ke-Jin Gao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266001, Shandong Province, China
| | - Bin Han
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266001, Shandong Province, China
| | - Ai-Jun Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266001, Shandong Province, China
| |
Collapse
|
10
|
Stajano D, Lombino FL, Schweizer M, Glatzel M, Saftig P, Gromova KV, Kneussel M. Tetraspanin 15 depletion impairs extracellular vesicle docking at target neurons. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e113. [PMID: 38938373 PMCID: PMC11080857 DOI: 10.1002/jex2.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 08/11/2023] [Accepted: 08/30/2023] [Indexed: 06/29/2024]
Abstract
Neurons in the central nervous system release extracellular vesicles (EVs) and exosomes in response to synaptic activity to regulate physiological processes at target neurons. The intercellular transfer of proteins, mRNAs, lipids or metabolites through EVs potentially modulates the structure and function of neurons and circuits. Whereas the biogenesis of EVs, their release from donor cells, and their molecular composition have been studied extensively, the critical factors and mechanisms regulating EV interactions with target cells are incompletely understood. Here, we identified tetraspanin 15 (Tspan15) as a component of tumor susceptibility gene 101 protein (TSG101)- and CD81-positive EV fractions. Tspan15 fluorescent fusion proteins were released from donor cells and interacted with target cells together with the exosomal marker CD63. EVs collected from wildtype cortical neurons (WT-EVs) underwent similar association with target neurons derived from either wildtype (+/+) or Tspan15 knockout (-/-) mice. In contrast, target cell interactions of EVs collected from Tspan15 (-/-) cortical donor neurons (KO-EVs) were significantly impaired, as compared to WT-EVs. Our data suggest that Tspan15 is dispensable at target neuron plasma membranes, but is required at the EV surface to promote EV docking at target neurons.
Collapse
Affiliation(s)
- Daniele Stajano
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Franco L. Lombino
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Michaela Schweizer
- Core Facility Morphology, Center for Molecular Neurobiology, ZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Markus Glatzel
- Institute of NeuropathologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Paul Saftig
- Biochemical InstituteChristian‐Albrechts‐University KielKielGermany
| | - Kira V. Gromova
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Matthias Kneussel
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
11
|
Park G, Jang WE, Kim S, Gonzales EL, Ji J, Choi S, Kim Y, Park JH, Mohammad HB, Bang G, Kang M, Kim S, Jeon SJ, Kim JY, Kim KP, Shin CY, An JY, Kim MS, Lee YS. Dysregulation of the Wnt/β-catenin signaling pathway via Rnf146 upregulation in a VPA-induced mouse model of autism spectrum disorder. Exp Mol Med 2023; 55:1783-1794. [PMID: 37524878 PMCID: PMC10474298 DOI: 10.1038/s12276-023-01065-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/15/2023] [Accepted: 05/29/2023] [Indexed: 08/02/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder associated with impaired social behavior and communication, repetitive behaviors, and restricted interests. In addition to genetic factors, environmental factors such as prenatal drug exposure contribute to the development of ASD. However, how those prenatal factors induce behavioral deficits in the adult stage is not clear. To elucidate ASD pathogenesis at the molecular level, we performed a high-resolution mass spectrometry-based quantitative proteomic analysis on the prefrontal cortex (PFC) of mice exposed to valproic acid (VPA) in utero, a widely used animal model of ASD. Differentially expressed proteins (DEPs) in VPA-exposed mice showed significant overlap with ASD risk genes, including differentially expressed genes from the postmortem cortex of ASD patients. Functional annotations of the DEPs revealed significant enrichment in the Wnt/β-catenin signaling pathway, which is dysregulated by the upregulation of Rnf146 in VPA-exposed mice. Consistently, overexpressing Rnf146 in the PFC impaired social behaviors and altered the Wnt signaling pathway in adult mice. Furthermore, Rnf146-overexpressing PFC neurons showed increased excitatory synaptic transmission, which may underlie impaired social behavior. These results demonstrate that Rnf146 is critical for social behavior and that dysregulation of Rnf146 underlies social deficits in VPA-exposed mice.
Collapse
Affiliation(s)
- Gaeun Park
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Wooyoung Eric Jang
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Seoyeon Kim
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, Republic of Korea
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841, Republic of Korea
| | - Edson Luck Gonzales
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jungeun Ji
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, Republic of Korea
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841, Republic of Korea
| | - Seunghwan Choi
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, 02841, Republic of Korea
| | - Yujin Kim
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, Republic of Korea
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841, Republic of Korea
| | - Ji Hwan Park
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | | | - Geul Bang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Minkyung Kang
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Soobin Kim
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Se Jin Jeon
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, 17104, Republic of Korea
- Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Chan Young Shin
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea.
| | - Joon-Yong An
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, Republic of Korea.
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841, Republic of Korea.
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, 02841, Republic of Korea.
| | - Min-Sik Kim
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea.
- New Biology Research Center, DGIST, Daegu, 42988, Republic of Korea.
- Center for Cell Fate Reprogramming and Control, DGIST, Daegu, 42988, Republic of Korea.
| | - Yong-Seok Lee
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Wide River Institute of Immunology, Seoul National University, Hongcheon, 25159, Republic of Korea.
| |
Collapse
|
12
|
Gu X, Jia C, Wang J. Advances in Understanding the Molecular Mechanisms of Neuronal Polarity. Mol Neurobiol 2023; 60:2851-2870. [PMID: 36738353 DOI: 10.1007/s12035-023-03242-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
The establishment and maintenance of neuronal polarity are important for neural development and function. Abnormal neuronal polarity establishment commonly leads to a variety of neurodevelopmental disorders. Over the past three decades, with the continuous development and improvement of biological research methods and techniques, we have made tremendous progress in the understanding of the molecular mechanisms of neuronal polarity establishment. The activity of positive and negative feedback signals and actin waves are both essential in this process. They drive the directional transport and aggregation of key molecules of neuronal polarity, promote the spatiotemporal regulation of ordered and coordinated interactions of actin filaments and microtubules, stimulate the specialization and growth of axons, and inhibit the formation of multiple axons. In this review, we focus on recent advances in these areas, in particular the important findings about neuronal polarity in two classical models, in vitro primary hippocampal/cortical neurons and in vivo cortical pyramidal neurons, and discuss our current understanding of neuronal polarity..
Collapse
Affiliation(s)
- Xi Gu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Chunhong Jia
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Junhao Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
13
|
Mechanotransduction in tumor dynamics modeling. Phys Life Rev 2023; 44:279-301. [PMID: 36841159 DOI: 10.1016/j.plrev.2023.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023]
Abstract
Mechanotherapy is a groundbreaking approach to impact carcinogenesis. Cells sense and respond to mechanical stimuli, translating them into biochemical signals in a process known as mechanotransduction. The impact of stress on tumor growth has been studied in the last three decades, and many papers highlight the role of mechanics as a critical self-inducer of tumor fate at the in vitro and in vivo biological levels. Meanwhile, mathematical models attempt to determine laws to reproduce tumor dynamics. This review discusses biological mechanotransduction mechanisms and mathematical-biomechanical models together. The aim is to provide a common framework for the different approaches that have emerged in the literature from the perspective of tumor avascularity and to provide insight into emerging mechanotherapies that have attracted interest in recent years.
Collapse
|
14
|
Haws W, England S, Grieb G, Susana G, Hernandez S, Mirer H, Lewis K. Analyses of binding partners and functional domains for the developmentally essential protein Hmx3a/HMX3. Sci Rep 2023; 13:1151. [PMID: 36670152 PMCID: PMC9859826 DOI: 10.1038/s41598-023-27878-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 01/09/2023] [Indexed: 01/22/2023] Open
Abstract
HMX3 is a homeodomain protein with essential roles in CNS and ear development. Homeodomains are DNA-binding domains and hence homeodomain-containing proteins are usually assumed to be transcription factors. However, intriguingly, our recent data suggest that zebrafish Hmx3a may not require its homeodomain to function, raising the important question of what molecular interactions mediate its effects. To investigate this, we performed a yeast two-hybrid screen and identified 539 potential binding partners of mouse HMX3. Using co-immunoprecipitation, we tested whether a prioritized subset of these interactions are conserved in zebrafish and found that Tle3b, Azin1b, Prmt2, Hmgb1a, and Hmgn3 bind Hmx3a. Next, we tested whether these proteins bind the products of four distinct hmx3a mutant alleles that all lack the homeodomain. Embryos homozygous for two of these alleles develop abnormally and die, whereas zebrafish homozygous for the other two alleles are viable. We found that all four mutations abrogate binding to Prmt2 and Tle3b, whereas Azin1b binding was preserved in all cases. Interestingly, Hmgb1a and Hmgn3 had more affinity for products of the viable mutant alleles. These data shed light on how HMX3/Hmx3a might function at a molecular level and identify new targets for future study in these vital developmental processes.
Collapse
Affiliation(s)
- William Haws
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Samantha England
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Ginny Grieb
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Gabriela Susana
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Sophie Hernandez
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Hunter Mirer
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Katharine Lewis
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA.
| |
Collapse
|
15
|
Geng S, Paul F, Kowalczyk I, Raimundo S, Sporbert A, Mamo TM, Hammes A. Balancing WNT signalling in early forebrain development: The role of LRP4 as a modulator of LRP6 function. Front Cell Dev Biol 2023; 11:1173688. [PMID: 37091972 PMCID: PMC10119419 DOI: 10.3389/fcell.2023.1173688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
The specification of the forebrain relies on the precise regulation of WNT/ß-catenin signalling to support neuronal progenitor cell expansion, patterning, and morphogenesis. Imbalances in WNT signalling activity in the early neuroepithelium lead to congenital disorders, such as neural tube defects (NTDs). LDL receptor-related protein (LRP) family members, including the well-studied receptors LRP5 and LRP6, play critical roles in modulating WNT signalling capacity through tightly regulated interactions with their co-receptor Frizzled, WNT ligands, inhibitors and intracellular WNT pathway components. However, little is known about the function of LRP4 as a potential modulator of WNT signalling in the central nervous system. In this study, we investigated the role of LRP4 in the regulation of WNT signalling during early mouse forebrain development. Our results demonstrate that LRP4 can modulate LRP5- and LRP6-mediated WNT signalling in the developing forebrain prior to the onset of neurogenesis at embryonic stage 9.5 and is therefore essential for accurate neural tube morphogenesis. Specifically, LRP4 functions as a genetic modifier for impaired mitotic activity and forebrain hypoplasia, but not for NTDs in LRP6-deficient mutants. In vivo and in vitro data provide evidence that LRP4 is a key player in fine-tuning WNT signalling capacity and mitotic activity of mouse neuronal progenitors and of human retinal pigment epithelial (hTERT RPE-1) cells. Our data demonstrate the crucial roles of LRP4 and LRP6 in regulating WNT signalling and forebrain development and highlight the need to consider the interaction between different signalling pathways to understand the underlying mechanisms of disease. The findings have significant implications for our mechanistic understanding of how LRPs participate in controlling WNT signalling.
Collapse
Affiliation(s)
- Shuang Geng
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Fabian Paul
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Izabela Kowalczyk
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Sandra Raimundo
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Tamrat Meshka Mamo
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- *Correspondence: Tamrat Meshka Mamo, ; Annette Hammes,
| | - Annette Hammes
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- *Correspondence: Tamrat Meshka Mamo, ; Annette Hammes,
| |
Collapse
|
16
|
Bone Tissue and the Nervous System: What Do They Have in Common? Cells 2022; 12:cells12010051. [PMID: 36611845 PMCID: PMC9818711 DOI: 10.3390/cells12010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022] Open
Abstract
Degenerative diseases affecting bone tissues and the brain represent important problems with high socio-economic impact. Certain bone diseases, such as osteoporosis, are considered risk factors for the progression of neurological disorders. Often, patients with neurodegenerative diseases have bone fractures or reduced mobility linked to osteoarthritis. The bone is a dynamic tissue involved not only in movement but also in the maintenance of mineral metabolism. Bone is also associated with the generation of both hematopoietic stem cells (HSCs), and thus the generation of the immune system, and mesenchymal stem cells (MSCs). Bone marrow is a lymphoid organ and contains MSCs and HSCs, both of which are involved in brain health via the production of cytokines with endocrine functions. Hence, it seems clear that bone is involved in the regulation of the neuronal system and vice versa. This review summarizes the recent knowledge on the interactions between the nervous system and bone and highlights the importance of the interaction between nerve and bone cells. In addition, experimental models that study the interaction between nerve and skeletal cells are discussed, and innovative models are suggested to better evaluate the molecular interactions between these two cell types.
Collapse
|
17
|
Sun H, Wu M, Wang M, Zhang X, Zhu J. The regulatory role of endoplasmic reticulum chaperone proteins in neurodevelopment. Front Neurosci 2022; 16:1032607. [DOI: 10.3389/fnins.2022.1032607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
The endoplasmic reticulum (ER) is the largest tubular reticular organelle spanning the cell. As the main site of protein synthesis, Ca2+ homeostasis maintenance and lipid metabolism, the ER plays a variety of essential roles in eukaryotic cells, with ER molecular chaperones participate in all these processes. In recent years, it has been reported that the abnormal expression of ER chaperones often leads to a variety of neurodevelopmental disorders (NDDs), including abnormal neuronal migration, neuronal morphogenesis, and synaptic function. Neuronal development is a complex and precisely regulated process. Currently, the mechanism by which neural development is regulated at the ER level remains under investigation. Therefore, in this work, we reviewed the recent advances in the roles of ER chaperones in neural development and developmental disorders caused by the deficiency of these molecular chaperones.
Collapse
|
18
|
Sanabria-de la Torre R, García-Fontana C, González-Salvatierra S, Andújar-Vera F, Martínez-Heredia L, García-Fontana B, Muñoz-Torres M. The Contribution of Wnt Signaling to Vascular Complications in Type 2 Diabetes Mellitus. Int J Mol Sci 2022; 23:6995. [PMID: 35805996 PMCID: PMC9266892 DOI: 10.3390/ijms23136995] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 02/01/2023] Open
Abstract
Vascular complications are the leading cause of morbidity and mortality among patients with type 2 diabetes mellitus (T2DM). These vascular abnormalities result in a chronic hyperglycemic state, which influences many signaling molecular pathways that initially lead to increased oxidative stress, increased inflammation, and endothelial dysfunction, leading to both microvascular and macrovascular complications. Endothelial dysfunction represents the initial stage in both types of vascular complications; it represents "mandatory damage" in the development of microvascular complications and only "introductory damage" in the development of macrovascular complications. Increasing scientific evidence has revealed an important role of the Wnt pathway in the pathophysiology of the vascular wall. It is well known that the Wnt pathway is altered in patients with T2DM. This review aims to be an update of the current literature related to the Wnt pathway molecules that are altered in patients with T2DM, which may also be the cause of damage to the vasculature. Both microvascular complications (retinopathy, nephropathy, and neuropathy) and macrovascular complications (coronary artery disease, cerebrovascular disease, and peripheral arterial disease) are analyzed. This review aims to concisely concentrate all the evidence to facilitate the view on the vascular involvement of the Wnt pathway and its components by highlighting the importance of exploring possible therapeutic strategy for patients with T2DM who develop vascular pathologies.
Collapse
Affiliation(s)
- Raquel Sanabria-de la Torre
- Department of Medicine, University of Granada, 18016 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (M.M.-T.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
| | - Cristina García-Fontana
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sheila González-Salvatierra
- Department of Medicine, University of Granada, 18016 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (M.M.-T.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
| | - Francisco Andújar-Vera
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Department of Computer Science and Artificial Intelligence, University of Granada, 18071 Granada, Spain
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI Institute), 18014 Granada, Spain
| | - Luis Martínez-Heredia
- Department of Medicine, University of Granada, 18016 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (M.M.-T.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
| | - Beatriz García-Fontana
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Manuel Muñoz-Torres
- Department of Medicine, University of Granada, 18016 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (M.M.-T.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
19
|
Abou-Fadel J, Bhalli M, Grajeda B, Zhang J. CmP Signaling Network Leads to Identification of Prognostic Biomarkers for Triple-Negative Breast Cancer in Caucasian Women. Genet Test Mol Biomarkers 2022; 26:198-219. [PMID: 35481969 DOI: 10.1089/gtmb.2021.0221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective: Triple-negative breast cancer (TNBC) constitutes ∼15% of all diagnosed invasive breast cancer cases with limited options for treatment since immunotherapies that target ER, PR, and HER2 receptors are ineffective. Progesterone (PRG) can induce its effects through either classic, nonclassic, or combined responses by binding to classic nuclear PRG receptors (nPRs) or nonclassic membrane PRG receptors (mPRs). Under PRG-induced actions, we previously demonstrated that the CCM signaling complex (CSC) can couple both nPRs and mPRs into a CmPn signaling network, which plays an important role during nPR(+) breast cancer tumorigenesis. We recently defined the novel CmP signaling network in African American women (AAW)-derived TNBC cells, which overlapped with our previously defined CmPn network in nPR(+) breast cancer cells. Methods: Under mPR-specific steroid actions, we measured alterations to key tumorigenic pathways in Caucasian American women (CAW)- derived TNBC cells, with RNAseq/proteomic and systems biology approaches. Exemption from ethics approval from IRB: This study only utilized cultured NBC cell lines with publicly available TNBC clinical data sets. Results: Our results demonstrated that TNBCs in CAW share similar altered signaling pathways, as TNBCs in AAW, under mPR-specific steroid actions, demonstrating the overall aggressive nature of TNBCs, regardless of racial differences. Furthermore, in this report, we have deconvoluted the CmP signalosome, using systems biology approaches and CAW-TNBC clinical data, to identify 21 new CAW-TNBC-specific prognostic biomarkers that reinforce the definitive role of CSC and mPR signaling during CAW-TNBC tumorigenesis. Conclusion: This new set of potential prognostic biomarkers may revolutionize molecular mechanisms and currently known concepts of tumorigenesis in CAW-TNBCs, leading to hopeful new therapeutic strategies.
Collapse
Affiliation(s)
- Johnathan Abou-Fadel
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, Texas, USA
| | - Muaz Bhalli
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, Texas, USA
| | - Brian Grajeda
- Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas, USA
| | - Jun Zhang
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, Texas, USA
| |
Collapse
|
20
|
Shen L, Pan L, Ju C, Wu X. The role of Wnt/β-catenin pathway for skin-derived precursors differentiating into corneal endothelial cell-like cells. Exp Eye Res 2022; 218:109008. [DOI: 10.1016/j.exer.2022.109008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/04/2022] [Accepted: 02/22/2022] [Indexed: 11/04/2022]
|
21
|
Nagu P, Sharma V, Behl T, Pathan AKA, Mehta V. Molecular Insights to the Wnt Signaling During Alzheimer's Disorder: a Potential Target for Therapeutic Interventions. J Mol Neurosci 2022; 72:679-690. [PMID: 34997460 DOI: 10.1007/s12031-021-01940-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/30/2021] [Indexed: 11/25/2022]
Abstract
In the adult brain, Wnt signaling is crucial for neurogenesis, and it also regulates neuronal development, neuronal maturation, neuronal differential, and proliferation. Impaired Wnt signaling pathways are associated with enhanced levels of amyloid-β, reduced β-catenin levels, and increased expression of GSK-3β enzyme, suggesting its direct association with the pathogenesis of Alzheimer's disorder (AD). These findings are consolidated by reports where activation of Wnt signaling by genetic factors and pharmacological intervention has improved the cognitive functions in animals and restored neurogenesis in the adult brain. Various natural and synthetic molecules have been identified that modulate Wnt signaling in the adult brain and promote neurogenesis and alleviate behavioral dysfunction. These molecules include lithium, valproic acid, ethosuximide, selenomethionine, curcumin, andrographolide, xanthoceraside, huperzine A, pyridostigmine, ginkgolide-B, ricinine, cannabidiol, and resveratrol. These molecules are associated with the DKK1 and GSK-3β inhibition and β-catenin stabilization along with their effects on neurogenesis, neuronal proliferation, and differentiation in the hippocampus through modulation of Wnt signaling and thereby could prove beneficial in the management of AD pathogenesis. Although modulation of the Wnt signaling seems to suggest to be promising in the management of AD, unfortunately, most of the literature available for the association of Wnt signaling and AD pathogenesis is either from preclinical studies or post-mortem brain. Therefore, it will be interesting to understand the role of Wnt signaling in AD patients, and a rigorous investigation could provide us with a better understanding of AD pathogenesis and the identification of novel targets for therapeutic interventions.
Collapse
Affiliation(s)
- Priyanka Nagu
- Department of Pharmacy, Shri Jagdishprasad Jhabarmal Tibrewala University, Jhunjhunu, Rajasthan, India.,Department of Pharmaceutics, Government College of Pharmacy, Rohru, Himachal Pradesh, India
| | - Vivek Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.,Department of Pharmacology, Government College of Pharmacy, Himachal Pradesh 171207, Rohru, District Shimla, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Amjad Khan A Pathan
- Department of Pharmacy, Shri Jagdishprasad Jhabarmal Tibrewala University, Jhunjhunu, Rajasthan, India
| | - Vineet Mehta
- Department of Pharmacology, Government College of Pharmacy, Himachal Pradesh 171207, Rohru, District Shimla, India.
| |
Collapse
|
22
|
Scher MS. Neurologic Sequelae Associated with Hypertensive Disorders of Pregnancy. CHILDREN (BASEL, SWITZERLAND) 2021; 8:945. [PMID: 34828658 PMCID: PMC8617864 DOI: 10.3390/children8110945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 11/23/2022]
Abstract
Hypertensive disorders of pregnancy (HDP) contribute to adverse gene-environment interactions prior to conception and continue throughout pregnancy. Embryonic/fetal brain disorders occur from interactions between genetic susceptibilities interacting with acquired diseases or conditions affecting the maternal/placental fetal (MPF) triad. Trimester-specific pathophysiological mechanisms, such as maternal immune activation and ischemic placental syndrome, contribute to adverse peripartum, neonatal and childhood outcomes. Two diagnostic approaches provide timelier diagnoses over the first 1000 days from conception until two years of age. Horizontal analyses assess the maturation of the triad, neonate and child. Vertical analyses consider systems-biology from genetic, molecular, cellular, tissue through organ networks during each developmental niche. Disease expressions associated with HDP have cumulative adverse effects across the lifespan when subjected to subsequent adverse events. Critical/sensitive periods of developmental neuroplasticity over the first 1000 days are more likely to result in permanent sequelae. Novel diagnostic approaches, beginning during pre-conception, will facilitate the development of effective preventive, rescue and reparative neurotherapeutic strategies in response to HDP-related trimester-specific disease pathways. Public health policies require the inclusion of women's health advocacy during and beyond their reproductive years to reduce sequelae experienced by mothers and their offspring. A lower global burden of neurologic disease from HDP will benefit future generations.
Collapse
Affiliation(s)
- Mark S. Scher
- Pediatrics and Neurology, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
- Department of Pediatrics, Division of Pediatric Neurology Fetal/Neonatal Neurology Program, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
23
|
Lombardo MV, Eyler L, Pramparo T, Gazestani VH, Hagler DJ, Chen CH, Dale AM, Seidlitz J, Bethlehem RAI, Bertelsen N, Barnes CC, Lopez L, Campbell K, Lewis NE, Pierce K, Courchesne E. Atypical genomic cortical patterning in autism with poor early language outcome. SCIENCE ADVANCES 2021; 7:eabh1663. [PMID: 34516910 PMCID: PMC8442861 DOI: 10.1126/sciadv.abh1663] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/15/2021] [Indexed: 05/21/2023]
Abstract
Cortical regionalization develops via genomic patterning along anterior-posterior (A-P) and dorsal-ventral (D-V) gradients. Here, we find that normative A-P and D-V genomic patterning of cortical surface area (SA) and thickness (CT), present in typically developing and autistic toddlers with good early language outcome, is absent in autistic toddlers with poor early language outcome. Autistic toddlers with poor early language outcome are instead specifically characterized by a secondary and independent genomic patterning effect on CT. Genes involved in these effects can be traced back to midgestational A-P and D-V gene expression gradients and different prenatal cell types (e.g., progenitor cells and excitatory neurons), are functionally important for vocal learning and human-specific evolution, and are prominent in prenatal coexpression networks enriched for high-penetrance autism risk genes. Autism with poor early language outcome may be explained by atypical genomic cortical patterning starting in prenatal development, which may detrimentally affect later regional functional specialization and circuit formation.
Collapse
Affiliation(s)
- Michael V. Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Lisa Eyler
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- VISN 22 Mental Illness Research, Education, and Clinical Center, VA San Diego Healthcare System, San Diego, CA, USA
| | - Tiziano Pramparo
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Vahid H. Gazestani
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Donald J. Hagler
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | - Chi-Hua Chen
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Anders M. Dale
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Jakob Seidlitz
- Department of Child and Adolescent Psychiatry and Behavioral Science, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard A. I. Bethlehem
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Natasha Bertelsen
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
- Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - Cynthia Carter Barnes
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Linda Lopez
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Kathleen Campbell
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Nathan E. Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, La Jolla, CA, USA
| | - Karen Pierce
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Eric Courchesne
- Autism Center of Excellence, Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
24
|
Giacomini E, Minetto S, Li Piani L, Pagliardini L, Somigliana E, Viganò P. Genetics and Inflammation in Endometriosis: Improving Knowledge for Development of New Pharmacological Strategies. Int J Mol Sci 2021; 22:ijms22169033. [PMID: 34445738 PMCID: PMC8396487 DOI: 10.3390/ijms22169033] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
According to a rich body of literature, immune cell dysfunctions, both locally and systemically, and an inflammatory environment characterize all forms of endometriosis. Alterations in transcripts and proteins involved in the recruitment of immune cells, in the interaction between cytokines and their receptors, cellular adhesion and apoptosis have been demonstrated in endometriotic lesions. The objective of this narrative review is to provide an overview of the components and mechanisms at the intersection between inflammation and genetics that may constitute vanguard therapeutic approaches in endometriosis. The GWAS technology and pathway-based analysis highlighted the role of the MAPK and the WNT/β-catenin cascades in the pathogenesis of endometriosis. These signaling pathways have been suggested to interfere with the disease establishment via several mechanisms, including apoptosis, migration and angiogenesis. Extracellular vesicle-associated molecules may be not only interesting to explain some aspects of endometriosis progression, but they may also serve as therapeutic regimens per se. Immune/inflammatory dysfunctions have always represented attractive therapeutic targets in endometriosis. These would be even more interesting if genetic evidence supported the involvement of functional pathways at the basis of these alterations. Targeting these dysfunctions through next-generation inhibitors can constitute a therapeutic alternative for endometriosis.
Collapse
Affiliation(s)
- Elisa Giacomini
- Reproductive Sciences Laboratory, Obstetrics and Gynecology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (E.G.); (L.P.)
| | - Sabrina Minetto
- Obstetrics and Gynecology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
| | - Letizia Li Piani
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (L.L.P.); (E.S.)
| | - Luca Pagliardini
- Reproductive Sciences Laboratory, Obstetrics and Gynecology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (E.G.); (L.P.)
| | - Edgardo Somigliana
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (L.L.P.); (E.S.)
| | - Paola Viganò
- Infertility Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-02-5503-4302
| |
Collapse
|
25
|
Wang C, Ruan L, Shi H, Lin W, Liu L, Li S. Phosphorylation of Shrimp Tcf by a Viral Protein Kinase WSV083 Suppresses Its Antiviral Effect. Front Immunol 2021; 12:698697. [PMID: 34408747 PMCID: PMC8365339 DOI: 10.3389/fimmu.2021.698697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022] Open
Abstract
Nuclear DNA-binding TCF proteins, which act as the main downstream effectors of Wnt signaling, are essential for the regulation of cell fate and innate immunity. However, their role during viral infection in shrimp remains unknown. Herein, we demonstrated that Litopenaeus vannamei TCF (LvTcf) acts independently of Lvβ-catenin to promote interferon-like protein LvVago1 production, thus mounting the response to WSSV infection. Further, we observed that WSV083, a WSSV serine/threonine protein kinase, bound to LvTcf and phosphorylated it. Phosphorylated LvTcf was then recognized and degraded via the ubiquitin-proteasome pathway. Moreover, mass spectrometry analyses indicated that the T39 and T104 residues of LvTcf were target sites phosphorylated by WSV083. Point mutation analyses suggested that additional sites of LvTcf may undergo phosphorylation via WSV083. Taken together, the current work provides valuable insights into host immunity and viral pathogenesis. LvTcf is not only a modulator of shrimp innate immunity but is also an important target for WSSV immune evasion. Thus, the current findings will help improve disease control in shrimps.
Collapse
Affiliation(s)
- Chuanqi Wang
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China.,School of Life Science, Xiamen University, Xiamen, China
| | - Lingwei Ruan
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| | - Hong Shi
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| | - Wenyang Lin
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China.,School of Life Science, Xiamen University, Xiamen, China
| | - Linmin Liu
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| | - Sujie Li
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| |
Collapse
|
26
|
Kirolos SA, Rijal R, Consalvo KM, Gomer RH. Using Dictyostelium to Develop Therapeutics for Acute Respiratory Distress Syndrome. Front Cell Dev Biol 2021; 9:710005. [PMID: 34350188 PMCID: PMC8326840 DOI: 10.3389/fcell.2021.710005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) involves damage to lungs causing an influx of neutrophils from the blood into the lung airspaces, and the neutrophils causing further damage, which attracts more neutrophils in a vicious cycle. There are ∼190,000 cases of ARDS per year in the US, and because of the lack of therapeutics, the mortality rate is ∼40%. Repelling neutrophils out of the lung airspaces, or simply preventing neutrophil entry, is a potential therapeutic. In this minireview, we discuss how our lab noticed that a protein called AprA secreted by growing Dictyostelium cells functions as a repellent for Dictyostelium cells, causing cells to move away from a source of AprA. We then found that AprA has structural similarity to a human secreted protein called dipeptidyl peptidase IV (DPPIV), and that DPPIV is a repellent for human neutrophils. In animal models of ARDS, inhalation of DPPIV or DPPIV mimetics blocks neutrophil influx into the lungs. To move DPPIV or DPPIV mimetics into the clinic, we need to know how this repulsion works to understand possible drug interactions and side effects. Combining biochemistry and genetics in Dictyostelium to elucidate the AprA signal transduction pathway, followed by drug studies in human neutrophils to determine similarities and differences between neutrophil and Dictyostelium chemorepulsion, will hopefully lead to the safe use of DPPIV or DPPIV mimetics in the clinic.
Collapse
Affiliation(s)
| | | | | | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
27
|
Hu L, Chen Y, Yang CP, Huang Y, Song NN, Chen JY, Sun YL, Ding YQ, Lang B. Ulk4, a Newly Discovered Susceptibility Gene for Schizophrenia, Regulates Corticogenesis in Mice. Front Cell Dev Biol 2021; 9:645368. [PMID: 34235142 PMCID: PMC8255617 DOI: 10.3389/fcell.2021.645368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/23/2021] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia (SCZ) is a chronic and severe mental disease that affects around 1% of the population. The precise etiology of SCZ still remains largely unknown, and no conclusive mechanisms are firmly established. Recent advances in epidemiological and clinical investigation support an overwhelmingly strong neurodevelopmental origin for SCZ. Here, we demonstrated that Unc-51-like kinase 4 (Ulk4), a novel risk factor for major mental disorders including schizophrenia, is involved in the corticogenesis. Deletion of Ulk4 in mice led to significantly thinner layers of II–III, and V in the cerebral cortex, which was confirmed in conditional Ulk4 deletion mice achieved by Cre-loxp strategy. This abnormality might be caused by decreased intermediate neural progenitors and increased apoptosis. Thus, our data suggest that Ulk4 manipulates the behaviors of neural progenitors during brain development and, when functionally defective, leads to the reduction of specific cortical layers. This anomaly may increase predisposition to a range of neurodevelopmental disorders, including SCZ.
Collapse
Affiliation(s)
- Ling Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yi Chen
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Cui-Ping Yang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Ying Huang
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Ning-Ning Song
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jia-Yin Chen
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yu-Ling Sun
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Yu-Qiang Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.,Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China.,Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Bing Lang
- National Clinical Research Centre for Mental Health, Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
28
|
Mahmoud DE, Kaabachi W, Sassi N, Mokhtar A, Moalla M, Ammar LB, Jemmali S, Rekik S, Tarhouni L, Kallel-Sellami M, Cheour E, Laadhar L. SFRP5 Enhances Wnt5a Induced-Inflammation in Rheumatoid Arthritis Fibroblast-Like Synoviocytes. Front Immunol 2021; 12:663683. [PMID: 34211463 PMCID: PMC8239419 DOI: 10.3389/fimmu.2021.663683] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/01/2021] [Indexed: 12/24/2022] Open
Abstract
Background Tissue derived fibroblast-like synoviocytes (td-FLS) are key actors in pannus formation and contribute to joint destruction and inflammation during rheumatoid arthritis (RA). Several members of the Wnt family, including Wnt5a, may contribute to RA td-FLS activation and can potentially serve as therapeutic targets. Objective The present work aimed to investigate the expression of Wnt5a signaling elements in RA td-FLS and their potential precursors (fluid derived (fd) FLS and fibrocytes). We also studied the role of Wnt5a in RA td-FLS pro-inflammatory activity and whether the inhibitor SFRP5 could restore Wnt5a-induced synovial dysfunction in vitro. Materials and Methods The levels of Wnt5a, SFRP5, Wnt5a receptors/coreceptors and Wnt5a pro-inflammatory targets were determined in cultured RA td-FLS, fd-FLS and fibrocytes using qPCR under basal conditions. The expression of pro-inflammatory molecules was assessed after RA td-FLS stimulation with Wnt5a and SFRP5 at different time points. Results Our data showed that td-FLS, fd-FLS and fibrocytes from patients with RA expressed similar levels of Wnt5a and a set of Wnt5a receptors/coreceptors. We also demonstrated that Wnt5a stimulated the expression of the pro-inflammatory targets, especially IL1β, IL8 and IL6 in RA td-FLS. Wnt5a-induced inflammation was enhanced in the presence of SFRP5. Furthermore, Wnt5a alone and in conjunction with SFRP5 inhibited the gene expression of TCF4 and the protein levels of the canonical coreceptor LRP5. Conclusion Wnt5a pro-inflammatory effect is not inhibited but enhanced by SFRP5 in RA td-FLS. This research highlights the importance of carefully evaluating changes in Wnt5a response in the presence of SFRP5.
Collapse
Affiliation(s)
- Dorra Elhaj Mahmoud
- Immuno-Rheumatology Research Laboratory, Rheumatology Department, La Rabta Hospital, University of Tunis-El Manar, Tunis, Tunisia
| | - Wajih Kaabachi
- Medicine School of Tunis, University of Tunis-El Manar, Tunis, Tunisia
| | - Nadia Sassi
- Immuno-Rheumatology Research Laboratory, Rheumatology Department, La Rabta Hospital, University of Tunis-El Manar, Tunis, Tunisia
| | - Amel Mokhtar
- Immuno-Rheumatology Research Laboratory, Rheumatology Department, La Rabta Hospital, University of Tunis-El Manar, Tunis, Tunisia
| | - Myriam Moalla
- Rheumatology Department, La Rabta Hospital, Tunis, Tunisia
| | | | - Samia Jemmali
- Rheumatology Department, La Rabta Hospital, Tunis, Tunisia
| | - Sonia Rekik
- Rheumatology Department, La Rabta Hospital, Tunis, Tunisia
| | - Lamjed Tarhouni
- Department of Hand and Reconstructive Surgery, Kassab Institute of Traumatic and Orthopedic Surgery, Tunis, Tunisia
| | - Maryam Kallel-Sellami
- Immuno-Rheumatology Research Laboratory, Rheumatology Department, La Rabta Hospital, University of Tunis-El Manar, Tunis, Tunisia
| | - Elhem Cheour
- Immuno-Rheumatology Research Laboratory, Rheumatology Department, La Rabta Hospital, University of Tunis-El Manar, Tunis, Tunisia
| | - Lilia Laadhar
- Immuno-Rheumatology Research Laboratory, Rheumatology Department, La Rabta Hospital, University of Tunis-El Manar, Tunis, Tunisia
| |
Collapse
|
29
|
Downregulation of miR-383 reduces depression-like behavior through targeting Wnt family member 2 (Wnt2) in rats. Sci Rep 2021; 11:9223. [PMID: 33927285 PMCID: PMC8085118 DOI: 10.1038/s41598-021-88560-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 03/22/2021] [Indexed: 01/04/2023] Open
Abstract
This study aimed to evaluate the role of miR-383 in the regulation of Wnt-2 signaling in the rat model of chronic stress. The male SD rats with depressive-like behaviors were stimulated with chronic unpredictable mild stress (CUMS) including ice-water swimming for 5 min, food deprivation for 24 h, water deprivation for 24 h, stimulating tail for 1 min, turning night into day, shaking for 15 min (once/s), and wrap restraint (5 min/time) every day for 21 days. The expression levels of miRNAs were detected by qRT-PCR, and the expression levels of Wnt2, depression-impacted proteins (GFAP, BDNF, CREB), brain neurotransmitters (5-HT, NE, DA) and apoptosis-related proteins (Bax and Bcl-2) were evaluated by qRT-PCR and western blot. Bioinformatic analysis and luciferase reporter assay were performed to determine the relationship between miR-383 and Wnt2. Ethological analysis was evaluated by sugar preference test, refuge island test and open field tests. Rescue experiments including knockdown of miR-383, overexpression and silencing of Wnt2 were performed to determine the role of miR-383. High expression levels of miR-383 were observed in the hippocampus of rats submitted to CUMS model. Downregulation of miR-383 significantly inhibited the apoptosis and inflammatory response of hippocampal neurons, and increased the expression levels of GFAP, BDNF and CREB which were impacted in depression, as well as neurotransmitters, then attenuated neural injury in rats induced by CUMS. Furthermore, Wnt family member 2 (Wnt2) was identified as a target of miR-383, and silencing of Wnt2 obviously attenuated the protective effect of miR-383 inhibitor on the apoptosis and inflammatory response in hippocampal neurons, as well as neural injury in CUMS-induced rats. Downregulation of miR-383 ameliorated the behavioral and neurochemical changes induced by chronic stress in rats by directly targeting Wnt2, indicating that the miR-383/Wnt2 axis might be a potential therapeutic target for MDD.
Collapse
|
30
|
Zhang H, Chen R, Shao J. MicroRNA-96-5p Facilitates the Viability, Migration, and Invasion and Suppresses the Apoptosis of Cervical Cancer Cells byNegatively Modulating SFRP4. Technol Cancer Res Treat 2021; 19:1533033820934132. [PMID: 32527205 PMCID: PMC7294480 DOI: 10.1177/1533033820934132] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The current study was intended to research the functional role and regulatory mechanism of microRNA-96-5p in the progression of cervical cancer. METHODS MicroRNA-96-5p expression in cervical cancer tissues was assessed by quantitative real-time polymerase chain reaction. The association between microRNA-96-5p expression and clinicopathological features of patients with cervical cancer was analyzed. MTT, flow cytometry, wound healing, and transwell assay were performed to evaluate the viability, apoptosis, migration, and invasion of Hela and SiHa cells. Targetscan, dual-luciferase reporter gene assay, and RNA pull-down analysis were constructed to evaluate the target relationship between microRNA-96-5p and secreted frizzled-related protein 4. RESULTS MicroRNA-96-5p was overexpressed in cervical cancer tissues, and microRNA-96-5p expression was markedly associated with the clinical stage and lymph node metastasis of patients with cervical cancer. Overexpressed microRNA-96-5p facilitated the viability, migration, invasion, and inhibited the apoptosis of Hela and SiHa cells, whereas suppression of microRNA-96-5p exerted the opposite trend. Secreted frizzled-related protein 4 was proved to be a target of microRNA-96-5p. Silencing of secreted frizzled-related protein 4 eliminated the anti-tumor effect of microRNA-96-5p on cervical cancer cells. CONCLUSIONS MicroRNA-96-5p facilitated the viability, migration, and invasion and inhibited the apoptosis of cervical cancer cells via negatively regulating secreted frizzled-related protein 4.
Collapse
Affiliation(s)
- Huiling Zhang
- Department of Clinical Laboratory, Huai'an Maternity and Child Health Hospital, Qingjiangpu District, Huaian City, Jiangsu Province, China
| | - Ruxin Chen
- Department of Obstetrics and Gynecology, Jinan Maternal and Child Health Hospital, Jinan City, Shandong Province, China
| | - Jinyan Shao
- Department of Obstetrics and Gynecology, Laishan Branch of Yantai Yuhuangding Hospital, Laishan District, Yantai City, Shandong Province, China
| |
Collapse
|
31
|
Abstract
Pancreatic cancer (PC) is one of the deadliest malignancies. The high mortality rate of PC largely results from delayed diagnosis and early metastasis. Therefore, identifying novel treatment targets for patients with PC is urgently required to improve survival rates. A major barrier to successful treatment of PC is the presence of a hypoxic tumor microenvironment, which is associated with poor prognosis, treatment resistance, increased invasion and metastasis. Recent studies have identified a number of novel molecules and pathways in PC cells that promote cancer cells progression under hypoxic conditions, which may provide new therapy strategies to inhibit the development and metastasis of PC. This review summarizes the latest research of hypoxia in PC and provides an overview of how the current therapies have the capacity to overcome hypoxia and improve PC patient treatment. These findings will eventually provide guidance for future PC management and clinical trials and hopefully improve the survival of patients with PC.
Collapse
Affiliation(s)
- Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
32
|
Urresti J, Zhang P, Moran-Losada P, Yu NK, Negraes PD, Trujillo CA, Antaki D, Amar M, Chau K, Pramod AB, Diedrich J, Tejwani L, Romero S, Sebat J, Yates III JR, Muotri AR, Iakoucheva LM. Cortical organoids model early brain development disrupted by 16p11.2 copy number variants in autism. Mol Psychiatry 2021; 26:7560-7580. [PMID: 34433918 PMCID: PMC8873019 DOI: 10.1038/s41380-021-01243-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 11/09/2022]
Abstract
Reciprocal deletion and duplication of the 16p11.2 region is the most common copy number variation (CNV) associated with autism spectrum disorders. We generated cortical organoids from skin fibroblasts of patients with 16p11.2 CNV to investigate impacted neurodevelopmental processes. We show that organoid size recapitulates macrocephaly and microcephaly phenotypes observed in the patients with 16p11.2 deletions and duplications. The CNV dosage affects neuronal maturation, proliferation, and synapse number, in addition to its effect on organoid size. We demonstrate that 16p11.2 CNV alters the ratio of neurons to neural progenitors in organoids during early neurogenesis, with a significant excess of neurons and depletion of neural progenitors observed in deletions. Transcriptomic and proteomic profiling revealed multiple pathways dysregulated by the 16p11.2 CNV, including neuron migration, actin cytoskeleton, ion channel activity, synaptic-related functions, and Wnt signaling. The level of the active form of small GTPase RhoA was increased in both, deletions and duplications. Inhibition of RhoA activity rescued migration deficits, but not neurite outgrowth. This study provides insights into potential neurobiological mechanisms behind the 16p11.2 CNV during neocortical development.
Collapse
Affiliation(s)
- Jorge Urresti
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Pan Zhang
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Patricia Moran-Losada
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Nam-Kyung Yu
- grid.214007.00000000122199231Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA USA
| | - Priscilla D. Negraes
- grid.266100.30000 0001 2107 4242Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Department of Pediatrics/Rady Children’s Hospital San Diego, University of California, San Diego, La Jolla, CA USA
| | - Cleber A. Trujillo
- grid.266100.30000 0001 2107 4242Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Department of Pediatrics/Rady Children’s Hospital San Diego, University of California, San Diego, La Jolla, CA USA
| | - Danny Antaki
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA USA
| | - Megha Amar
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Kevin Chau
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Akula Bala Pramod
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| | - Jolene Diedrich
- grid.214007.00000000122199231Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA USA
| | - Leon Tejwani
- grid.266100.30000 0001 2107 4242Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Department of Pediatrics/Rady Children’s Hospital San Diego, University of California, San Diego, La Jolla, CA USA
| | - Sarah Romero
- grid.266100.30000 0001 2107 4242Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Department of Pediatrics/Rady Children’s Hospital San Diego, University of California, San Diego, La Jolla, CA USA
| | - Jonathan Sebat
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242University of California San Diego, Beyster Center for Psychiatric Genomics, La Jolla, CA USA
| | - John R. Yates III
- grid.214007.00000000122199231Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA USA
| | - Alysson R. Muotri
- grid.266100.30000 0001 2107 4242Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Department of Pediatrics/Rady Children’s Hospital San Diego, University of California, San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242University of California San Diego, Kavli Institute for Brain and Mind, La Jolla, CA USA ,Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, CA USA
| | - Lilia M. Iakoucheva
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA
| |
Collapse
|
33
|
Scher MS. "The First Thousand Days" Define a Fetal/Neonatal Neurology Program. Front Pediatr 2021; 9:683138. [PMID: 34408995 PMCID: PMC8365757 DOI: 10.3389/fped.2021.683138] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/27/2021] [Indexed: 01/11/2023] Open
Abstract
Gene-environment interactions begin at conception to influence maternal/placental/fetal triads, neonates, and children with short- and long-term effects on brain development. Life-long developmental neuroplasticity more likely results during critical/sensitive periods of brain maturation over these first 1,000 days. A fetal/neonatal program (FNNP) applying this perspective better identifies trimester-specific mechanisms affecting the maternal/placental/fetal (MPF) triad, expressed as brain malformations and destructive lesions. Maladaptive MPF triad interactions impair progenitor neuronal/glial populations within transient embryonic/fetal brain structures by processes such as maternal immune activation. Destructive fetal brain lesions later in pregnancy result from ischemic placental syndromes associated with the great obstetrical syndromes. Trimester-specific MPF triad diseases may negatively impact labor and delivery outcomes. Neonatal neurocritical care addresses the symptomatic minority who express the great neonatal neurological syndromes: encephalopathy, seizures, stroke, and encephalopathy of prematurity. The asymptomatic majority present with neurologic disorders before 2 years of age without prior detection. The developmental principle of ontogenetic adaptation helps guide the diagnostic process during the first 1,000 days to identify more phenotypes using systems-biology analyses. This strategy will foster innovative interdisciplinary diagnostic/therapeutic pathways, educational curricula, and research agenda among multiple FNNP. Effective early-life diagnostic/therapeutic programs will help reduce neurologic disease burden across the lifespan and successive generations.
Collapse
Affiliation(s)
- Mark S Scher
- Division of Pediatric Neurology, Department of Pediatrics, Fetal/Neonatal Neurology Program, Emeritus Scholar Tenured Full Professor in Pediatrics and Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
34
|
Li B, Niswander LA. TMEM132A, a Novel Wnt Signaling Pathway Regulator Through Wntless (WLS) Interaction. Front Cell Dev Biol 2020; 8:599890. [PMID: 33324648 PMCID: PMC7726220 DOI: 10.3389/fcell.2020.599890] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/04/2020] [Indexed: 11/24/2022] Open
Abstract
Wnt signaling pathway plays indispensable roles in embryonic development and adult tissue homeostasis. However, the regulatory mechanisms involved in Wnt ligand trafficking within and secretion from the signal sending cells is still relatively uncharacterized. Here, we discover a novel regulator of Wnt signaling pathway called transmembrane protein 132A (TMEM132A). Our evidence shows a physical and functional interaction of TMEM132A with the Wnt ligand transporting protein Wntless (WLS). We show that TMEM132A stabilizes Wnt ligand, enhances WLS–Wnt ligand interaction, and activates the Wnt signaling pathway. Our results shed new light on the cellular mechanism underlying the fundamental aspect of WNT secretion from Wnt signal sending cells.
Collapse
Affiliation(s)
- Binbin Li
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, United States
| | - Lee A Niswander
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, United States
| |
Collapse
|
35
|
Arredondo SB, Valenzuela-Bezanilla D, Mardones MD, Varela-Nallar L. Role of Wnt Signaling in Adult Hippocampal Neurogenesis in Health and Disease. Front Cell Dev Biol 2020; 8:860. [PMID: 33042988 PMCID: PMC7525004 DOI: 10.3389/fcell.2020.00860] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/10/2020] [Indexed: 12/11/2022] Open
Abstract
Neurogenesis persists during adulthood in the dentate gyrus of the hippocampus. Signals provided by the local hippocampal microenvironment support neural stem cell proliferation, differentiation, and maturation of newborn neurons into functional dentate granule cells, that integrate into the neural circuit and contribute to hippocampal function. Increasing evidence indicates that Wnt signaling regulates multiple aspects of adult hippocampal neurogenesis. Wnt ligands bind to Frizzled receptors and co-receptors to activate the canonical Wnt/β-catenin signaling pathway, or the non-canonical β-catenin-independent signaling cascades Wnt/Ca2+ and Wnt/planar cell polarity. Here, we summarize current knowledge on the roles of Wnt signaling components including ligands, receptors/co-receptors and soluble modulators in adult hippocampal neurogenesis. Also, we review the data suggesting distinctive roles for canonical and non-canonical Wnt signaling cascades in regulating different stages of neurogenesis. Finally, we discuss the evidence linking the dysfunction of Wnt signaling to the decline of neurogenesis observed in aging and Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | - Lorena Varela-Nallar
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
36
|
Wallace J, Narasipura SD, Sha BE, French AL, Al-Harthi L. Canonical Wnts Mediate CD8 + T Cell Noncytolytic Anti-HIV-1 Activity and Correlate with HIV-1 Clinical Status. THE JOURNAL OF IMMUNOLOGY 2020; 205:2046-2055. [PMID: 32887752 DOI: 10.4049/jimmunol.1801379] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 08/04/2020] [Indexed: 11/19/2022]
Abstract
CD8+ T cells do not rely solely on cytotoxic functions for significant HIV control. Moreover, the noncytotoxic CD8+ T cell antiviral response is a primary mediator of natural HIV control such as that seen in HIV elite controllers and long-term nonprogressors that does not require combined antiretroviral therapy. In this study, we investigated the biological factors contributing to the noncytotoxic control of HIV replication mediated by primary human CD8+ T cells. We report that canonical Wnt signaling inhibits HIV transcription in an MHC-independent, noncytotoxic manner and that mediators of this pathway correlate with HIV controller clinical status. We show that CD8+ T cells express all 19 Wnts and CD8+ T cell-conditioned medium (CM) induced canonical Wnt signaling in infected recipient cells while simultaneously inhibiting HIV transcription. Antagonizing canonical Wnt activity in CD8+ T cell CM resulted in increased HIV transcription in infected cells. Further, Wnt2b expression was upregulated in HIV controllers versus viremic patients, and in vitro depletion of Wnt2b and/or Wnt9b from CD8+ CM reversed HIV inhibitory activity. Finally, plasma concentration of Dkk-1, an antagonist of canonical Wnt signaling, was higher in viremic patients with lower CD4 counts. This study demonstrates that canonical Wnt signaling inhibits HIV and significantly correlates with HIV controller status.
Collapse
Affiliation(s)
- Jennillee Wallace
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612
| | - Srinivas D Narasipura
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612
| | - Beverly E Sha
- Division of Infectious Diseases, Rush University Medical Center, Chicago, IL 60612
| | - Audrey L French
- Division of Infectious Diseases, Rush University Medical Center, Chicago, IL 60612.,Stroger Hospital of Cook County, Cook County Health and Hospitals System, Chicago, IL 60612; and.,Ruth M. Rothstein CORE Center, Cook County Health and Hospitals System, Chicago, IL 60612
| | - Lena Al-Harthi
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612;
| |
Collapse
|
37
|
The COMPASS Family Protein ASH2L Mediates Corticogenesis via Transcriptional Regulation of Wnt Signaling. Cell Rep 2020; 28:698-711.e5. [PMID: 31315048 DOI: 10.1016/j.celrep.2019.06.055] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 05/12/2019] [Accepted: 06/14/2019] [Indexed: 12/19/2022] Open
Abstract
Histone methylation is essential for regulating gene expression during organogenesis to maintain stem cells and execute a proper differentiation program for their descendants. Here we show that the COMPASS family histone methyltransferase co-factor ASH2L is required for maintaining neural progenitor cells (NPCs) and the production and positioning of projection neurons during neocortex development. Specifically, loss of Ash2l in NPCs results in malformation of the neocortex; the mutant neocortex has fewer neurons, which are also abnormal in composition and laminar position. Moreover, ASH2L loss impairs trimethylation of H3K4 and the transcriptional machinery specific for Wnt-β-catenin signaling, inhibiting the proliferation ability of NPCs at late stages of neurogenesis by disrupting S phase entry to inhibit cell cycle progression. Overexpressing β-catenin after ASH2L elimination rescues the proliferation deficiency. Therefore, our findings demonstrate that ASH2L is crucial for modulating Wnt signaling to maintain NPCs and generate a full complement of neurons during mammalian neocortex development.
Collapse
|
38
|
Mancini A, Howard SR, Marelli F, Cabrera CP, Barnes MR, Sternberg MJ, Leprovots M, Hadjidemetriou I, Monti E, David A, Wehkalampi K, Oleari R, Lettieri A, Vezzoli V, Vassart G, Cariboni A, Bonomi M, Garcia MI, Guasti L, Dunkel L. LGR4 deficiency results in delayed puberty through impaired Wnt/β-catenin signaling. JCI Insight 2020; 5:133434. [PMID: 32493844 PMCID: PMC7308048 DOI: 10.1172/jci.insight.133434] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 04/23/2020] [Indexed: 12/13/2022] Open
Abstract
The initiation of puberty is driven by an upsurge in hypothalamic gonadotropin-releasing hormone (GnRH) secretion. In turn, GnRH secretion upsurge depends on the development of a complex GnRH neuroendocrine network during embryonic life. Although delayed puberty (DP) affects up to 2% of the population, is highly heritable, and is associated with adverse health outcomes, the genes underlying DP remain largely unknown. We aimed to discover regulators by whole-exome sequencing of 160 individuals of 67 multigenerational families in our large, accurately phenotyped DP cohort. LGR4 was the only gene remaining after analysis that was significantly enriched for potentially pathogenic, rare variants in 6 probands. Expression analysis identified specific Lgr4 expression at the site of GnRH neuron development. LGR4 mutant proteins showed impaired Wnt/β-catenin signaling, owing to defective protein expression, trafficking, and degradation. Mice deficient in Lgr4 had significantly delayed onset of puberty and fewer GnRH neurons compared with WT, whereas lgr4 knockdown in zebrafish embryos prevented formation and migration of GnRH neurons. Further, genetic lineage tracing showed strong Lgr4-mediated Wnt/β-catenin signaling pathway activation during GnRH neuron development. In conclusion, our results show that LGR4 deficiency impairs Wnt/β-catenin signaling with observed defects in GnRH neuron development, resulting in a DP phenotype. Defects of LGR4/Wnt-β-catenin activity compromise the development of the GnRH neuroendocrine network, resulting in delayed onset of puberty in humans and mice.
Collapse
Affiliation(s)
- Alessandra Mancini
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sasha R Howard
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Federica Marelli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,IRCCS Istituto Auxologico Italiano, Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Milan, Italy
| | - Claudia P Cabrera
- Centre for Translational Bioinformatics, William Harvey Research Institute, and.,NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Michael R Barnes
- Centre for Translational Bioinformatics, William Harvey Research Institute, and.,NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Michael Je Sternberg
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London, United Kingdom
| | | | - Irene Hadjidemetriou
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Elena Monti
- St George's NHS Foundation Trust, London, United Kingdom
| | - Alessia David
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Karoliina Wehkalampi
- Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Antonella Lettieri
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Valeria Vezzoli
- IRCCS Istituto Auxologico Italiano, Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Milan, Italy
| | | | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Marco Bonomi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,IRCCS Istituto Auxologico Italiano, Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Milan, Italy
| | | | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Leo Dunkel
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
39
|
Suzuki N, Ochi H. Regeneration enhancers: A clue to reactivation of developmental genes. Dev Growth Differ 2020; 62:343-354. [PMID: 32096563 PMCID: PMC7383998 DOI: 10.1111/dgd.12654] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 12/18/2022]
Abstract
During tissue and organ regeneration, cells initially detect damage and then alter nuclear transcription in favor of tissue/organ reconstruction. Until recently, studies of tissue regeneration have focused on the identification of relevant genes. These studies show that many developmental genes are reused during regeneration. Concurrently, comparative genomics studies have shown that the total number of genes does not vastly differ among vertebrate taxa. Moreover, functional analyses of developmental genes using various knockout/knockdown techniques demonstrated that the functions of these genes are conserved among vertebrates. Despite these data, the ability to regenerate damaged body parts varies widely between animals. Thus, it is important to determine how regenerative transcriptional programs are triggered and why animals with low regenerative potential fail to express developmental genes after injury. Recently, we discovered relevant enhancers and named them regeneration signal-response enhancers (RSREs) after identifying their activation mechanisms in a Xenopus laevis transgenic system. In this review, we summarize recent studies of injury/regeneration-associated enhancers and then discuss their mechanisms of activation.
Collapse
Affiliation(s)
- Nanoka Suzuki
- Amphibian Research CenterHiroshima UniversityHigashi‐HiroshimaJapan
| | - Haruki Ochi
- Institute for Promotion of Medical Science ResearchFaculty of MedicineYamagata UniversityYamagataJapan
| |
Collapse
|
40
|
Alvarez-Zavala M, Barreto-Vargas C, Torres-Reyes LA, De la Peña-Castro RF, Aguilar-Lemarroy A, Jave-Suarez LF. Exogenous Expression of WNT7A in Leukemia-Derived Cell Lines Induces Resistance to Chemotherapeutic Agents. Anticancer Agents Med Chem 2020; 20:1504-1514. [PMID: 32436833 DOI: 10.2174/1871520620666200521114100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/08/2020] [Accepted: 02/28/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Dysregulations of the WNT pathway are implicated in the malignant transformation of different types of neoplasia. WNT7A is expressed in normal peripheral lymphocytes, but is decreased in the tumoral counterpart. Furthermore, the treatment of leukemic cells with recombinant WNT7A decreases proliferation, suggesting its possible use as a therapeutic biomolecule. This study aimed to evaluate the concomitant action of WNT7A and different chemotherapeutic agents over proliferation and cell death of leukemia/ lymphoma derived cell lines. METHODS Ectopic expression of WNT7A was induced in CEM and BJAB cell lines by using a lentiviral system. RNA expression was analyzed by microarrays and qPCR, and protein expression was determined by Western Blot. Cell proliferation was measured by cell counting, metabolic activity by WST-1 assay, cell death and DNA content by flow cytometry. RESULTS WNT7A ectopic expression was shown to decrease cell proliferation, but the apoptosis rate of leukemic cells was not altered. Moreover, these cells acquired resistance to doxorubicin, vincristine and MG-132. Cell cycle analysis reveals a decrease in G1 and an increase in S and G2 phases with a further upregulation of senescence- associated genes. Microarray analysis reveals that most gene expression changes were related to cancer and metabolic associated pathways. All those changes appear to be independent of the WNT canonical pathway regulation. CONCLUSION WNT7A negatively regulates cell proliferation in leukemic cell lines and promotes resistance to chemotherapeutic agents by inducing a senescence-like phenotype independently of the WNT canonical pathway.
Collapse
Affiliation(s)
- Monserrat Alvarez-Zavala
- Instituto de Investigacion en Inmunodeficiencias y VIH, Departamento de Clínicas Medicas, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Christian Barreto-Vargas
- Division de Inmunologia, Centro de Investigacion Biomedica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Luis A Torres-Reyes
- Departamento de Biologia Molecular y Genomica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Roberto F De la Peña-Castro
- Division de Inmunologia, Centro de Investigacion Biomedica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Adriana Aguilar-Lemarroy
- Division de Inmunologia, Centro de Investigacion Biomedica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Luis F Jave-Suarez
- Division de Inmunologia, Centro de Investigacion Biomedica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| |
Collapse
|
41
|
Gels for Live Analysis of Compartmentalized Environments (GLAnCE): A tissue model to probe tumour phenotypes at tumour-stroma interfaces. Biomaterials 2020; 228:119572. [DOI: 10.1016/j.biomaterials.2019.119572] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 09/19/2019] [Accepted: 10/18/2019] [Indexed: 12/15/2022]
|
42
|
Kozielewicz P, Turku A, Schulte G. Molecular Pharmacology of Class F Receptor Activation. Mol Pharmacol 2019; 97:62-71. [PMID: 31591260 DOI: 10.1124/mol.119.117986] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 09/25/2019] [Indexed: 12/29/2022] Open
Abstract
The class Frizzled (FZD) or class F of G protein-coupled receptors consists of 10 FZD paralogues and Smoothened (SMO). FZDs coordinate wingless/Int-1 signaling and SMO mediates Hedgehog signaling. Class F receptor signaling is intrinsically important for embryonic development and its dysregulation leads to diseases, including diverse forms of tumors. With regard to the importance of class F signaling in human disease, these receptors provide an attractive target for therapeutics, exemplified by the use of SMO antagonists for the treatment of basal cell carcinoma. Here, we review recent structural insights in combination with a more detailed functional understanding of class F receptor activation, G protein coupling, conformation-based functional selectivity, and mechanistic details of activating cancer mutations, which will lay the basis for further development of class F-targeting small molecules for human therapy. SIGNIFICANCE STATEMENT: Stimulated by recent insights into the activation mechanisms of class F receptors from structural and functional analysis of Frizzled and Smoothened, we aim to summarize what we know about the molecular details of ligand binding, agonist-driven conformational changes, and class F receptor activation. A better understanding of receptor activation mechanisms will allow us to engage in structure- and mechanism-driven drug discovery with the potential to develop more isoform-selective and potentially pathway-selective drugs for human therapy.
Collapse
Affiliation(s)
- Pawel Kozielewicz
- Section of Receptor Biology and Signaling, Department Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ainoleena Turku
- Section of Receptor Biology and Signaling, Department Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Schulte
- Section of Receptor Biology and Signaling, Department Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
43
|
Semaan C, Henderson BR, Molloy MP. Proteomic screen with the proto-oncogene beta-catenin identifies interaction with Golgi coatomer complex I. Biochem Biophys Rep 2019; 19:100662. [PMID: 31338436 PMCID: PMC6626114 DOI: 10.1016/j.bbrep.2019.100662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/17/2019] [Accepted: 06/22/2019] [Indexed: 11/28/2022] Open
Abstract
Beta-catenin is well-known as a key effector of Wnt signalling and aberrant expression is associated with several human cancers. Stabilisation of and atypical subcellular localisation of beta-catenin, regulated in part through specific protein-protein interactions has been linked to cancer development, however the mechanisms behind these pathologies is yet to be fully elucidated. Affinity purification and mass spectrometry were used to identify potential β-catenin interacting proteins in SW480 colon cancer cells. Recombinant β-catenin constructs were used to co-isolate interacting proteins from stable isotope labelled cells followed by detection using mass spectrometry. Several known and new putative interactors were observed. In particular, we identified interaction with a set of coatomer complex I subunits implicated in retrograde transport at the Golgi, and confirmed endogenous interaction of β-catenin with coatomer subunit COPB using immunoprecipitation assays and immunofluorescence microscopy. These observations suggest a hitherto unrecognised role for β-catenin in the secretory pathway and warrant further functional studies to unravel its activity at this cellular location.
Collapse
Affiliation(s)
- Crystal Semaan
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, Australia.,Gene and Stem Cell Therapy Program Centenary Institute, The University of Sydney, Camperdown, 2050, NSW, Australia
| | - Beric R Henderson
- Westmead Institute for Medical Research, University of Sydney, Australia
| | - Mark P Molloy
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, Australia.,Australian Proteome Analysis Facility, Macquarie University, Sydney, Australia.,Bowel Cancer and Biomarker Laboratory, Kolling Institute, The University of Sydney, Australia
| |
Collapse
|
44
|
Gossypol Promotes Wnt/ β-Catenin Signaling through WIF1 in Ovariectomy-Induced Osteoporosis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8745487. [PMID: 31139657 PMCID: PMC6500658 DOI: 10.1155/2019/8745487] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 03/24/2019] [Accepted: 03/27/2019] [Indexed: 01/23/2023]
Abstract
Osteoporosis is one of the most frequent diseases related with age. Previously, we have reported a novel potential drug, gossypol, for the treatment of osteoporosis through its regulation of Wnt/β-catenin signaling. This study aims to identify the detailed mechanism of gossypol in human osteoporosis. Mice injected with gossypol were subjected for RNA-seq analysis and the transcription level of WIF1 was shown to be decreased dramatically in gossypol-treated mice, which was further confirmed by qRT-PCR and western blot analysis. Luciferase reporter assay showed gossypol inhibited the activity of WIF1 and the methylation of WIF1 was significantly upregulated, evidenced by ChIP assay. Cell viability assays demonstrated that gossypol promoted cell proliferation while cotreatment with WIF1 expressing plasmid reversed the effect in a dose- and time-dependent manner. Similarly, cell apoptotic assays and TUNEL assays showed gossypol suppressed cell apoptosis, which was revised by WIF1 overexpression. The mouse model suggested gossypol injection ameliorated osteoporosis, while coinjection of AAV5-WIF1 eliminated the protection effects of gossypol, as evidenced by H&E staining, serum osteocalcin level, serum OPG level, serum RANKL level, bone density, ultimate strength, and postyield displacement. This study is a supplement to the former publication, which reinforced the protection effect of gossypol in human osteoporosis.
Collapse
|
45
|
Skaper SD, Barbierato M, Facci L, Borri M, Contarini G, Zusso M, Giusti P. Co-Ultramicronized Palmitoylethanolamide/Luteolin Facilitates the Development of Differentiating and Undifferentiated Rat Oligodendrocyte Progenitor Cells. Mol Neurobiol 2019; 55:103-114. [PMID: 28822061 DOI: 10.1007/s12035-017-0722-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oligodendrocytes, the myelin-producing cells of the central nervous system (CNS), have limited capability to bring about repair in chronic CNS neuroinflammatory demyelinating disorders such as multiple sclerosis (MS). MS lesions are characterized by a compromised pool of undifferentiated oligodendrocyte progenitor cells (OPCs) unable to mature into myelin-producing oligodendrocytes. An attractive strategy may be to replace lost OLs and/or promote their maturation. N-palmitoylethanolamine (PEA) is an endogenous fatty acid amide signaling molecule with anti-inflammatory and neuroprotective actions. Recent studies show a co-ultramicronized composite of PEA and the flavonoid luteolin (co-ultraPEALut) to be more efficacious than PEA in improving outcome in CNS injury models. Here, we examined the effects of co-ultraPEALut on development of OPCs from newborn rat cortex cultured under conditions favoring either differentiation (Sato medium) or proliferation (fibroblast growth factor-2 and platelet-derived growth factor (PDGF)-AA-supplemented serum-free medium ("SFM")). OPCs in SFM displayed high expression of PDGF receptor alpha gene and the proliferation marker Ki-67. In Sato medium, in contrast, OPCs showed rapid decreases in PDGF receptor alpha and Ki-67 expression with a concomitant rise in myelin basic protein (MBP) expression. In these conditions, co-ultraPEALut (10 μM) enhanced OPC morphological complexity and expression of MBP and the transcription factor TCF7l2. Surprisingly, co-ultraPEALut also up-regulated MBP mRNA expression in OPCs in SFM. MBP expression in all cases was sensitive to inhibition of mammalian target of rapamycin. Within the context of strategies to promote endogenous remyelination in MS which focus on enhancing long-term survival of OPCs and stimulating their differentiation into remyelinating oligodendrocytes, co-ultraPEALut may represent a novel pharmacological approach.
Collapse
Affiliation(s)
- Stephen D Skaper
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "Egidio Meneghetti" 2, 35131, Padua, Italy.
| | - Massimo Barbierato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "Egidio Meneghetti" 2, 35131, Padua, Italy
| | - Laura Facci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "Egidio Meneghetti" 2, 35131, Padua, Italy
| | - Mila Borri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "Egidio Meneghetti" 2, 35131, Padua, Italy
| | - Gabriella Contarini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "Egidio Meneghetti" 2, 35131, Padua, Italy
| | - Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "Egidio Meneghetti" 2, 35131, Padua, Italy
| | - Pietro Giusti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "Egidio Meneghetti" 2, 35131, Padua, Italy
| |
Collapse
|
46
|
Lee JM, Kim TW, Park SS, Kim CJ, Shin MS, Lee SJ, Kim SH, Baek SS. Wnt signaling pathway is implicated in the alleviating effect of treadmill exercise on maternal separation-induced depression. J Exerc Rehabil 2019; 15:200-205. [PMID: 31111001 PMCID: PMC6509450 DOI: 10.12965/jer.1938148.074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/21/2019] [Indexed: 01/31/2023] Open
Abstract
Maternal separation in the developmental stage has a negative influence on brain development and causes depression. The extracellular ligand, Wnt, and its receptors play an important role in axis formation and neural development. Exercise inhibits apoptosis, increases cell proliferation, and exerts antidepressive effect. In this study, the effect of treadmill exercise on the maternal separation-induced depression was investigated in the aspect of Wnt signaling pathway. The maternal separation started on the postnatal day 14. The rat pups in the exercise groups were forced to run on a treadmill for 30 min once a day from postnatal day 21 to postnatal day 34. The rat pups in the maternal separation and fluoxetine-treated group were intraperitoneally injected with 5-mg/kg fluoxetine once a day from postnatal day 21 to postnatal day 34. Forced swimming test was performed to evaluate the depression level. Western blotting was performed for the expressions of Wnt signaling ligands, Wnt2 and Wnt3a, and Wnt signaling inhibitors, Dkk1, and sFRP3. Maternal separation showed depressive behaviors in the forced swimming test. Treadmill exercise alleviated depressive behaviors in the maternal separation rat pups. Expressions of Wnt2 and Wnt3a were decreased by maternal separation. Treadmill exercise alleviated maternal separation-induced reduction of Wnt2 and Wnt3a expressions. Expressions of Dkk1 and sFRP3 in the hippocampus were increased by maternal separation. Treadmill exercise alleviated maternal separation-induced reduction of Dkk1 and sFRP3 expressions. Our study demonstrated that treadmill exercise activates Wnt signaling pathway, and then exerted antidepressive effect.
Collapse
Affiliation(s)
- Jae-Min Lee
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Tae-Woon Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sang-Seo Park
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Mal-Soon Shin
- School of Global Sport Studies, Korea University, Sejong, Korea
| | - Sam-Jun Lee
- Department of Physical Education, College of Health, Welfare, and Education, Tong Myong University, Busan, Korea
| | - Sang-Hoon Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea.,Department of Sport & Health Care, College of Art & Culture, Sangmyung University, Seoul, Korea
| | - Seung-Soo Baek
- Department of Sport & Health Care, College of Art & Culture, Sangmyung University, Seoul, Korea
| |
Collapse
|
47
|
Gou X, Tang Y, Qu Y, Xiao D, Ying J, Mu D. Could the inhibitor of DNA binding 2 and 4 play a role in white matter injury? Rev Neurosci 2019; 30:625-638. [PMID: 30738015 DOI: 10.1515/revneuro-2018-0090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/02/2018] [Indexed: 01/12/2023]
Abstract
Abstract
White matter injury (WMI) prevents the normal development of myelination, leading to central nervous system myelination disorders and the production of chronic sequelae associated with WMI, such as chronic dyskinesia, cognitive impairment and cerebral palsy. This results in a large emotional and socioeconomic burden. Decreased myelination in preterm infant WMI is associated with the delayed development or destruction of oligodendrocyte (OL) lineage cells, particularly oligodendrocyte precursor cells (OPCs). The development of cells from the OL lineage involves the migration, proliferation and different stages of OL differentiation, finally leading to myelination. A series of complex intrinsic, extrinsic and epigenetic factors regulate the OPC cell cycle withdrawal, OL lineage progression and myelination. We focus on the inhibitor of DNA binding 2 (ID2), because it is widely involved in the different stages of OL differentiation and genesis. ID2 is a key transcription factor for the normal development of OL lineage cells, and the pathogenesis of WMI is closely linked with OL developmental disorders. ID4, another family member of the IDs protein, also plays a similar role in OL differentiation and genesis. ID2 and ID4 belong to the helix-loop-helix family; they lack the DNA-binding sequences and inhibit oligodendrogenesis and OPC differentiation. In this review, we mainly discuss the roles of ID2 in OL development, especially during OPC differentiation, and summarize the ID2-mediated intracellular and extracellular signaling pathways that regulate these processes. We also discuss ID4 in relation to bone morphogenetic protein signaling and oligodendrogenesis. It is likely that these developmental mechanisms are also involved in the myelin repair or remyelination in human neurological diseases.
Collapse
Affiliation(s)
- Xiaoyun Gou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Ying Tang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Dongqiong Xiao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| |
Collapse
|
48
|
Lian N, Niu Q, Lei Y, Li X, Li Y, Song X. MiR-221 is involved in depression by regulating Wnt2/CREB/BDNF axis in hippocampal neurons. Cell Cycle 2018; 17:2745-2755. [PMID: 30589396 DOI: 10.1080/15384101.2018.1556060] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE The aim of this study was to investigate the mechanism of miR-221 in depression. METHODS The molecules expressions were measured by qRT-PCR and western blot. The sucrose preference test (SPT), forced swimming test (FST) and tail suspension test (TST) were used to detect depressive-like symptoms. MTT assay and flow cytometric was used to measure the proliferation and apoptosis of hippocampal neuronal. RESULTS MiR-221 expression in the cerebrospinal fluid and serum of major depressive disorder patients and the hippocampus of chronic unpredictable mild stress (CUMS) mice were increased, while the expression of Wnt2, p-CREB and BDNF were decreased. Additionally, silence of miR-221 increased sucrose preference of CUMS mice and shortened the immobility time of CUMS mice in SPT and FST. MiR-221 could targeted regulate Wnt2, and knockdown of Wnt2 reversed the effect of miR-221 inhibitor on the proliferation and apoptosis of hippocampal neurons and countered the promoting effect of miR-221 inhibitor on the expression of Wnt2, p-CREB and BDNF. CONCLUSION MiR-221 could promote the development of depression by regulating Wnt2/CREB/BDNF axis.
Collapse
Affiliation(s)
- Nan Lian
- a Department of Psychiatry , The First Affiliated Hospital of Zhengzhou University , Zhengzhou Henan , China.,b Biological Psychiatry International Joint Laboratory of Henan , Zhengzhou University , Zhengzhou Henan , China.,c Henan Psychiatric Transformation Research Key Laboratory , Zhengzhou University , Zhengzhou Henan , China
| | - Qihui Niu
- a Department of Psychiatry , The First Affiliated Hospital of Zhengzhou University , Zhengzhou Henan , China.,b Biological Psychiatry International Joint Laboratory of Henan , Zhengzhou University , Zhengzhou Henan , China.,c Henan Psychiatric Transformation Research Key Laboratory , Zhengzhou University , Zhengzhou Henan , China
| | - Yang Lei
- a Department of Psychiatry , The First Affiliated Hospital of Zhengzhou University , Zhengzhou Henan , China.,b Biological Psychiatry International Joint Laboratory of Henan , Zhengzhou University , Zhengzhou Henan , China.,c Henan Psychiatric Transformation Research Key Laboratory , Zhengzhou University , Zhengzhou Henan , China
| | - Xue Li
- a Department of Psychiatry , The First Affiliated Hospital of Zhengzhou University , Zhengzhou Henan , China.,b Biological Psychiatry International Joint Laboratory of Henan , Zhengzhou University , Zhengzhou Henan , China.,c Henan Psychiatric Transformation Research Key Laboratory , Zhengzhou University , Zhengzhou Henan , China
| | - Youhui Li
- a Department of Psychiatry , The First Affiliated Hospital of Zhengzhou University , Zhengzhou Henan , China.,b Biological Psychiatry International Joint Laboratory of Henan , Zhengzhou University , Zhengzhou Henan , China.,c Henan Psychiatric Transformation Research Key Laboratory , Zhengzhou University , Zhengzhou Henan , China
| | - Xueqin Song
- a Department of Psychiatry , The First Affiliated Hospital of Zhengzhou University , Zhengzhou Henan , China.,b Biological Psychiatry International Joint Laboratory of Henan , Zhengzhou University , Zhengzhou Henan , China.,c Henan Psychiatric Transformation Research Key Laboratory , Zhengzhou University , Zhengzhou Henan , China
| |
Collapse
|
49
|
Kuptanon C, Srichomthong C, Sangsin A, Kovitvanitcha D, Suphapeetiporn K, Shotelersuk V. The most 5' truncating homozygous mutation of WNT1 in siblings with osteogenesis imperfecta with a variable degree of brain anomalies: a case report. BMC MEDICAL GENETICS 2018; 19:117. [PMID: 30012084 PMCID: PMC6048891 DOI: 10.1186/s12881-018-0639-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 07/03/2018] [Indexed: 01/22/2023]
Abstract
Background WNT1 mutations cause bone fragility as well as brain anomalies. There are some reported cases of WNT1 mutations with normal cognition. Genotype and phenotype correlation of WNT1 mutations has not been established. Case presentation Here we present two female siblings with osteogenesis imperfecta (OI) born to a consanguineous couple. Both sustained severe bone deformities. However, only the younger had severe brain anomalies resulting in an early death from pneumonia, while the older had normal intellectual development. Next generation sequencing showed a homozygous mutation, c.6delG, p.Leu3Serfs*36 in WNT1. To our knowledge, it is the most 5′ truncating mutation to date. Conclusion This report emphasizes the intrafamilial variability of brain anomalies found in this OI type and suggests that WNT1 may not be necessary for normal human cognitive development.
Collapse
Affiliation(s)
- Chulaluck Kuptanon
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, Bangkok, 10400, Thailand
| | - Chalurmpon Srichomthong
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.,Excellence Center for Medical Genetics, the Thai Red Cross Society, King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand
| | - Apiruk Sangsin
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.,Excellence Center for Medical Genetics, the Thai Red Cross Society, King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand.,Department of Orthopaedics, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Dool Kovitvanitcha
- Department of Orthopedics, Queen Sirikit National Institute of Child Health, Bangkok, 10400, Thailand
| | - Kanya Suphapeetiporn
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand. .,Excellence Center for Medical Genetics, the Thai Red Cross Society, King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand. .,Division of Medical Genetics and Metabolism, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Sor Kor Building 11th floor, Bangkok, 10330, Thailand.
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.,Excellence Center for Medical Genetics, the Thai Red Cross Society, King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand
| |
Collapse
|
50
|
Sarubbo F, Moranta D, Pani G. Dietary polyphenols and neurogenesis: Molecular interactions and implication for brain ageing and cognition. Neurosci Biobehav Rev 2018; 90:456-470. [DOI: 10.1016/j.neubiorev.2018.05.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 04/05/2018] [Accepted: 05/07/2018] [Indexed: 12/17/2022]
|