1
|
Arutyunov A, Durán-Laforet V, Ai S, Ferrari L, Murphy R, Schafer DP, Klein RS. West Nile Virus-Induced Expression of Senescent Gene Lgals3bp Regulates Microglial Phenotype within Cerebral Cortex. Biomolecules 2024; 14:808. [PMID: 39062523 PMCID: PMC11274721 DOI: 10.3390/biom14070808] [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: 05/29/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Microglia, the resident macrophages of the central nervous system, exhibit altered gene expression in response to various neurological conditions. This study investigates the relationship between West Nile Virus infection and microglial senescence, focusing on the role of LGALS3BP, a protein implicated in both antiviral responses and aging. Using spatial transcriptomics, RNA sequencing and flow cytometry, we characterized changes in microglial gene signatures in adult and aged mice following recovery from WNV encephalitis. Additionally, we analyzed Lgals3bp expression and generated Lgals3bp-deficient mice to assess the impact on neuroinflammation and microglial phenotypes. Our results show that WNV-activated microglia share transcriptional signatures with aged microglia, including upregulation of genes involved in interferon response and inflammation. Lgals3bp was broadly expressed in the CNS and robustly upregulated during WNV infection and aging. Lgals3bp-deficient mice exhibited reduced neuroinflammation, increased homeostatic microglial numbers, and altered T cell populations without differences in virologic control or survival. These data indicate that LGALS3BP has a role in regulating neuroinflammation and microglial activation and suggest that targeting LGALS3BP might provide a potential route for mitigating neuroinflammation-related cognitive decline in aging and post-viral infections.
Collapse
Affiliation(s)
- Artem Arutyunov
- Center for Neuroimmunology & Neuroinfectious Diseases, St. Louis, MO 63110, USA; (A.A.); (S.A.)
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Violeta Durán-Laforet
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (V.D.-L.); (L.F.); (R.M.); (D.P.S.)
| | - Shenjian Ai
- Center for Neuroimmunology & Neuroinfectious Diseases, St. Louis, MO 63110, USA; (A.A.); (S.A.)
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Loris Ferrari
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (V.D.-L.); (L.F.); (R.M.); (D.P.S.)
| | - Robert Murphy
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (V.D.-L.); (L.F.); (R.M.); (D.P.S.)
| | - Dorothy P. Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (V.D.-L.); (L.F.); (R.M.); (D.P.S.)
| | - Robyn S. Klein
- Department of Microbiology & Immunology, Western Institute of Neuroscience, Schulich School of Medicine & Dentistry, University of Western Ontario, 100 Perth Dr, London, ON N6A 5K8, Canada
| |
Collapse
|
2
|
Muralidharan A, Boukany PE. Electrotransfer for nucleic acid and protein delivery. Trends Biotechnol 2024; 42:780-798. [PMID: 38102019 DOI: 10.1016/j.tibtech.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/15/2023] [Accepted: 11/15/2023] [Indexed: 12/17/2023]
Abstract
Electrotransfer of nucleic acids and proteins has become crucial in biotechnology for gene augmentation and genome editing. This review explores the applications of electrotransfer in both ex vivo and in vivo scenarios, emphasizing biomedical uses. We provide insights into completed clinical trials and successful instances of nucleic acid and protein electrotransfer into therapeutically relevant cells such as immune cells and stem and progenitor cells. In addition, we delve into emerging areas of electrotransfer where nanotechnology and deep learning techniques overcome the limitations of traditional electroporation.
Collapse
Affiliation(s)
- Aswin Muralidharan
- Department of Bionanoscience, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, The Netherlands; Kavli Institute of Nanoscience, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, The Netherlands.
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, The Netherlands.
| |
Collapse
|
3
|
Liu L, Wei J, Chen C, Liang Q, Wang B, Wu W, Li G, Zheng X. Electroporation-based Easi-CRISPR yields biallelic insertions of EGFP-HiBiT cassette in immortalized chicken oviduct epithelial cells. Poult Sci 2023; 102:103112. [PMID: 37806084 PMCID: PMC10568294 DOI: 10.1016/j.psj.2023.103112] [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: 05/25/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023] Open
Abstract
Laying hens are an excellent experimental oviduct model for studying reproduction biology. Because chicken oviduct epithelial cells (cOECs) have a crucial role in synthesizing and secreting ovalbumin, laying hens have been regarded an ideal bioreactor for producing pharmaceuticals in egg white through transgene or gene editing of the ovalbumin (OVA) gene. However, related studies in cOECs are largely limited because of the lack of immortalized model cells. In addition, the editing efficiency of conventional CRISPR-HDR knock-in in chicken cells is suboptimal (ranging from 1 to 10%) and remains elevated. Here, primary cOECs were isolated from young laying hens, then infected with a retrovirus vector of human telomerase reverse transcriptase (hTERT), and immortalized cOECs were established. Subsequently, an electroporation-based Easi-CRISPR (Efficient additions with ssDNA inserts-CRISPR) method was adopted to integrate an EGFP-HiBiT cassette into the chicken OVA locus (immediately upstream of the stop codon). The immortalized cOECs reflected the self-renewal capability and phenotype of oviduct epithelial cells. This is because these cells not only maintained stable proliferation and normal karyotype and had no potential for malignant transformation, but also expressed oviduct markers and an epithelial marker and had a morphology similar to that of primary cOECs. EGFP expression was detected in the edited cells through microscopy, flow cytometry, and HiBiT/Western blotting. The EGFP-HiBiT knock-in efficiency reached 27.9% after a single round of electroporation, which was determined through genotyping and DNA sequencing. Two single cell clones contained biallelic insertions of EGFP-HiBiT donor cassettes. In conclusion, our established immortalized cOECs could act as an in vitro cell model for gene editing in chicken, and this electroporation-based Easi-CRISPR strategy will contribute to the generation of avian bioreactors and other gene-edited (GE) birds.
Collapse
Affiliation(s)
- Lingkang Liu
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Jinyu Wei
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; Buffalo Research Institute, Chinese Academy of Agricultural Sciences and Guangxi Zhuang Nationality Autonomous Region, Nanning 530004, China
| | - Chen Chen
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Qianxue Liang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Boyong Wang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Wende Wu
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Gonghe Li
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Xibang Zheng
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China.
| |
Collapse
|
4
|
Schwarz MM, Ganaie SS, Feng A, Brown G, Yangdon T, White JM, Hoehl RM, McMillen CM, Rush RE, Connors KA, Cui X, Leung DW, Egawa T, Amarasinghe GK, Hartman AL. Lrp1 is essential for lethal Rift Valley fever hepatic disease in mice. SCIENCE ADVANCES 2023; 9:eadh2264. [PMID: 37450601 PMCID: PMC10348670 DOI: 10.1126/sciadv.adh2264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
Rift Valley fever virus (RVFV) is an emerging arbovirus found in Africa. While RVFV is pantropic and infects many cells and tissues, viral replication and necrosis within the liver play a critical role in mediating severe disease. The low-density lipoprotein receptor-related protein 1 (Lrp1) is a recently identified host factor for cellular entry and infection by RVFV. The biological significance of Lrp1, including its role in hepatic disease in vivo, however, remains to be determined. Because Lrp1 has a high expression level in hepatocytes, we developed a mouse model in which Lrp1 is specifically deleted in hepatocytes to test how the absence of liver Lrp1 expression affects RVF pathogenesis. Mice lacking Lrp1 expression in hepatocytes showed minimal RVFV replication in the liver, longer time to death, and altered clinical signs toward neurological disease. In contrast, RVFV infection levels in other tissues showed no difference between the two genotypes. Therefore, Lrp1 is essential for RVF hepatic disease in mice.
Collapse
Affiliation(s)
- Madeline M. Schwarz
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Safder S. Ganaie
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Annie Feng
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Griffin Brown
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Tenzin Yangdon
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - J. Michael White
- Transgenic, Knockout and Micro-Injection Core, Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Ryan M. Hoehl
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cynthia M. McMillen
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rachael E. Rush
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kaleigh A. Connors
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoxia Cui
- Genome Engineering & Stem Cell Center, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Daisy W. Leung
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Takeshi Egawa
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Gaya K. Amarasinghe
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Angsutararux P, Dutta AK, Marras M, Abella C, Mellor RL, Shi J, Nerbonne JM, Silva JR. Differential regulation of cardiac sodium channels by intracellular fibroblast growth factors. J Gen Physiol 2023; 155:e202213300. [PMID: 36944081 PMCID: PMC10038838 DOI: 10.1085/jgp.202213300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/17/2023] [Accepted: 02/09/2023] [Indexed: 03/23/2023] Open
Abstract
Voltage-gated sodium (NaV) channels are responsible for the initiation and propagation of action potentials. In the heart, the predominant NaV1.5 α subunit is composed of four homologous repeats (I-IV) and forms a macromolecular complex with multiple accessory proteins, including intracellular fibroblast growth factors (iFGF). In spite of high homology, each of the iFGFs, iFGF11-iFGF14, as well as the individual iFGF splice variants, differentially regulates NaV channel gating, and the mechanisms underlying these differential effects remain elusive. Much of the work exploring iFGF regulation of NaV1.5 has been performed in mouse and rat ventricular myocytes in which iFGF13VY is the predominant iFGF expressed, whereas investigation into NaV1.5 regulation by the human heart-dominant iFGF12B is lacking. In this study, we used a mouse model with cardiac-specific Fgf13 deletion to study the consequences of iFGF13VY and iFGF12B expression. We observed distinct effects on the voltage-dependences of activation and inactivation of the sodium currents (INa), as well as on the kinetics of peak INa decay. Results in native myocytes were recapitulated with human NaV1.5 heterologously expressed in Xenopus oocytes, and additional experiments using voltage-clamp fluorometry (VCF) revealed iFGF-specific effects on the activation of the NaV1.5 voltage sensor domain in repeat IV (VSD-IV). iFGF chimeras further unveiled roles for all three iFGF domains (i.e., the N-terminus, core, and C-terminus) on the regulation of VSD-IV, and a slower time domain of inactivation. We present here a novel mechanism of iFGF regulation that is specific to individual iFGF isoforms and that leads to distinct functional effects on NaV channel/current kinetics.
Collapse
Affiliation(s)
- Paweorn Angsutararux
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Amal K. Dutta
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Martina Marras
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Carlota Abella
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Rebecca L. Mellor
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Jingyi Shi
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Jeanne M. Nerbonne
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jonathan R. Silva
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
6
|
Wallace M, White JM, Kouranova E, Wang ZT, Cui X. Floxing by Electroporating Single-Cell Embryos with Two CRISPR RNPs and Two ssODNs. Methods Mol Biol 2023; 2631:231-252. [PMID: 36995670 DOI: 10.1007/978-1-0716-2990-1_9] [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] [Indexed: 04/27/2023]
Abstract
Floxed alleles and Cre drivers are two components of most conditional knockout mouse models, which are not only important for studying a given gene in a tissue-specific manner, but also useful for functional analysis of various sized genomic regions. With the increased demand for floxed mouse models in biomedical research, reliable and economical creation of floxed alleles is clearly highly valuable yet remains challenging. Here we provide technical details on the method consisting of electroporating single-cell embryos with CRISPR RNPs and ssODNs, next-generation sequencing (NGS)-based genotyping, an in vitro Cre assay (recombination followed by PCR) for loxP phasing determination, and optional second round targeting of an indel in cis with one loxP insertion in embryos obtained via in vitro fertilization (IVF). As importantly, we present protocols for validation of gRNAs and ssODNs before electroporation of embryos, to confirm phasing of loxP and the indel to be retargeted in individual blastocysts and an alternative strategy to insert loxP sites sequentially. Together, we hope to help researchers reliably obtain floxed alleles in a predictable and timely manner.
Collapse
Affiliation(s)
- Mia Wallace
- Mouse Genetics Core, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - J Michael White
- Transgenic, Knockout and Microinjection Core, Department of Pathology & Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Evgenea Kouranova
- Genome Engineering & Stem Cell Center, Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Zi Teng Wang
- Genome Engineering & Stem Cell Center, Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Xiaoxia Cui
- Genome Engineering & Stem Cell Center, Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
7
|
Krus KL, Strickland A, Yamada Y, Devault L, Schmidt RE, Bloom AJ, Milbrandt J, DiAntonio A. Loss of Stathmin-2, a hallmark of TDP-43-associated ALS, causes motor neuropathy. Cell Rep 2022; 39:111001. [PMID: 35767949 PMCID: PMC9327139 DOI: 10.1016/j.celrep.2022.111001] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/18/2022] [Accepted: 06/02/2022] [Indexed: 12/03/2022] Open
Abstract
TDP-43 mediates proper Stathmin-2 (STMN2) mRNA splicing, and STMN2 protein is reduced in the spinal cord of most patients with amyotrophic lateral sclerosis (ALS). To test the hypothesis that STMN2 loss contributes to ALS pathogenesis, we generated constitutive and conditional STMN2 knockout mice. Constitutive STMN2 loss results in early-onset sensory and motor neuropathy featuring impaired motor behavior and dramatic distal neuromuscular junction (NMJ) denervation of fast-fatigable motor units, which are selectively vulnerable in ALS, without axon or motoneuron degeneration. Selective excision of STMN2 in motoneurons leads to similar NMJ pathology. STMN2 knockout heterozygous mice, which better model the partial loss of STMN2 protein found in patients with ALS, display a slowly progressive, motor-selective neuropathy with functional deficits and NMJ denervation. Thus, our findings strongly support the hypothesis that STMN2 reduction owing to TDP-43 pathology contributes to ALS pathogenesis.
Collapse
Affiliation(s)
- Kelsey L Krus
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Amy Strickland
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yurie Yamada
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laura Devault
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robert E Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - A Joseph Bloom
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO 63110, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO 63110, USA.
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO 63110, USA.
| |
Collapse
|