Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.704
Filter
1.
Nat Commun ; 15(1): 4756, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834544

ABSTRACT

Given the absence of approved treatments for pathogenic variants in Peripherin-2 (PRPH2), it is imperative to identify a universally effective therapeutic target for PRPH2 pathogenic variants. To test the hypothesis that formation of the elongated discs in presence of PRPH2 pathogenic variants is due to the presence of the full complement of rhodopsin in absence of the required amounts of functional PRPH2. Here we demonstrate the therapeutic potential of reducing rhodopsin levels in ameliorating disease phenotype in knockin models for p.Lys154del (c.458-460del) and p.Tyr141Cys (c.422 A > G) in PRPH2. Reducing rhodopsin levels improves physiological function, mitigates the severity of disc abnormalities, and decreases retinal gliosis. Additionally, intravitreal injections of a rhodopsin-specific antisense oligonucleotide successfully enhance the physiological function of photoreceptors and improves the ultrastructure of discs in mutant mice. Presented findings shows that reducing rhodopsin levels is an effective therapeutic strategy for the treatment of inherited retinal degeneration associated with PRPH2 pathogenic variants.


Subject(s)
Peripherins , Rhodopsin , Peripherins/genetics , Peripherins/metabolism , Animals , Rhodopsin/genetics , Rhodopsin/metabolism , Mice , Humans , Disease Models, Animal , Down-Regulation , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/therapy , Oligonucleotides, Antisense/genetics , Retina/metabolism , Retina/pathology , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Diseases/therapy , Mice, Inbred C57BL , Mutation , Female , Gene Knock-In Techniques , Male
2.
Front Immunol ; 15: 1383612, 2024.
Article in English | MEDLINE | ID: mdl-38742107

ABSTRACT

Introduction: SARS-CoV-2, the cause of the COVID pandemic, is an RNA virus with a high propensity to mutate. Successive virus variants, including variants of concern (VOC), have emerged with increased transmission or immune escape. The original pandemic virus and early variants replicated poorly, if at all, in mice at least partly due to a mismatch between the receptor binding domain on the viral spike protein and the murine angiotensin converting enzyme 2 (ACE2). Omicron VOC emerged in late 2021 harboring > 50 new mutations, 35 of them in the spike protein. This variant resulted in a very large wave of infections, even in the face of prior immunity, albeit being inherently less severe than earlier variants. Reflecting the lower severity reported in humans, Omicron displayed attenuated infection in hamsters and also in the K18-hACE2 mouse model. K18-hACE2 mice express both the human ACE2 as well as the endogenous mouse ACE2. Methods: Here we infected hACE2 knock-in mice that express only human ACE2 and no murine ACE2, or C57BL/6 wildtype mice with SARS-CoV-2 D614G (first-wave isolate), Delta or Omicron BA.1 variants and assessed infectivity and downstream innate immune responses. Results: While replication of SARS-CoV-2 Omicron was lower in the lungs of hACE2 knock-in mice compared with SARS-CoV-2 D614G and VOC Delta, it replicated more efficiently than the earlier variants in C57BL/6 wildtype mice. This opens the opportunity to test the effect of host genetics on SARS-CoV-2 infections in wildtype mice. As a proof of principle, we tested Omicron infection in mice lacking expression of the interferon-alpha receptor-1 (IFNAR1). In these mice we found that loss of type I IFN receptor signaling resulted in higher viral loads in the lungs were detected. Finally, using a chimeric virus of first wave SARS-CoV-2 harboring the Omicron spike protein, we show that Omicron spike increase infection of C57BL/6 wildtype mice, but non-spike genes of Omicron confer attenuation of viral replication. Discussion: Since this chimeric virus efficiently infected C57BL/6 wildtype mice, and replicated in their lungs, our findings illustrate a pathway for genetic mapping of virushost interactions during SARS-CoV-2 infection.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Mice, Inbred C57BL , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virus Replication , Animals , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Mice , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/immunology , COVID-19/virology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Humans , Disease Models, Animal , Gene Knock-In Techniques , Mice, Transgenic
3.
Cell Host Microbe ; 32(5): 632-634, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38723601

ABSTRACT

Inducing HIV-1 broadly neutralizing antibodies (bnAbs) through vaccination poses exceptional challenges. In this issue of Cell Host & Microbe, Wiehe and colleagues report the elicitation of affinity-matured bnAbs in knock-in mice through boosting immunogen vaccination, which selects for key improbable mutations.


Subject(s)
AIDS Vaccines , Antibodies, Neutralizing , HIV Antibodies , HIV Infections , HIV-1 , Vaccine Development , AIDS Vaccines/immunology , AIDS Vaccines/genetics , HIV-1/immunology , HIV-1/genetics , Animals , Mice , HIV Antibodies/immunology , Antibodies, Neutralizing/immunology , HIV Infections/prevention & control , HIV Infections/immunology , Humans , Gene Knock-In Techniques , Immunization, Secondary , Vaccination
4.
Biochem Biophys Res Commun ; 716: 150010, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38704892

ABSTRACT

Calcium (Ca2+) in mitochondria plays crucial roles in neurons including modulating metabolic processes. Moreover, excessive Ca2+ in mitochondria can lead to cell death. Thus, altered mitochondrial Ca2+ regulation has been implicated in several neurodegenerative diseases including Huntington's disease (HD). HD is a progressive hereditary neurodegenerative disorder that results from abnormally expanded cytosine-adenine-guanine trinucleotide repeats in the huntingtin gene. One neuropathological hallmark of HD is neuronal loss in the striatum and cortex. However, mechanisms underlying selective loss of striatal and cortical neurons in HD remain elusive. Here, we measured the basal Ca2+ levels and Ca2+ uptake in single presynaptic mitochondria during 100 external electrical stimuli using highly sensitive mitochondria-targeted Ca2+ indicators in cultured cortical and striatal neurons of a knock-in mouse model of HD (zQ175 mice). We observed elevated presynaptic mitochondrial Ca2+ uptake during 100 electrical stimuli in HD cortical neurons compared with wild-type (WT) cortical neurons. We also found the highly elevated presynaptic mitochondrial basal Ca2+ level and Ca2+ uptake during 100 stimuli in HD striatal neurons. The elevated presynaptic mitochondrial basal Ca2+ level in HD striatal neurons and Ca2+ uptake during stimulation in HD striatal and cortical neurons can disrupt neurotransmission and induce mitochondrial Ca2+ overload, eventually leading to neuronal death in the striatum and cortex of HD.


Subject(s)
Calcium , Cerebral Cortex , Corpus Striatum , Disease Models, Animal , Gene Knock-In Techniques , Huntington Disease , Mitochondria , Presynaptic Terminals , Animals , Huntington Disease/metabolism , Huntington Disease/pathology , Huntington Disease/genetics , Calcium/metabolism , Mitochondria/metabolism , Mice , Corpus Striatum/metabolism , Corpus Striatum/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Presynaptic Terminals/metabolism , Cells, Cultured , Neurons/metabolism , Neurons/pathology , Mice, Transgenic
5.
Genesis ; 62(3): e23601, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703044

ABSTRACT

HAND2 is a basic helix-loop-helix transcription factor with diverse functions during development. To facilitate the investigation of genetic and functional diversity among Hand2-expressing cells in the mouse, we have generated Hand2Dre, a knock-in allele expressing Dre recombinase. To avoid disrupting Hand2 function, the Dre cDNA is inserted at the 3' end of the Hand2 coding sequence following a viral 2A peptide. Hand2Dre homozygotes can therefore be used in complex crosses to increase the proportion of useful genotypes among offspring. Dre expression in mid-gestation Hand2Dre embryos is indistinguishable from wild-type Hand2 expression, and HandDre efficiently recombines rox target sites in vivo. In combination with existing Cre and Flp mouse lines, Hand2Dre will therefore extend the ability to perform genetic intersectional labeling, fate mapping, and functional manipulation of subpopulations of cells characterized by developmental expression of Hand2.


Subject(s)
Alleles , Basic Helix-Loop-Helix Transcription Factors , Gene Knock-In Techniques , Animals , Female , Mice , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Knock-In Techniques/methods , Integrases/genetics , Integrases/metabolism , Male
6.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731886

ABSTRACT

The cerebrovascular endothelial cells with distinct characteristics line cerebrovascular blood vessels and are the fundamental structure of the blood-brain barrier, which is important for the development and homeostatic maintenance of the central nervous system. Cre-LoxP system-based spatial gene manipulation in mice is critical for investigating the physiological functions of key factors or signaling pathways in cerebrovascular endothelial cells. However, there is a lack of Cre recombinase mouse lines that specifically target cerebrovascular endothelial cells. Here, using a publicly available single-cell RNAseq database, we screened the solute carrier organic anion transporter family member 1a4 (Slco1a4) as a candidate marker of cerebrovascular endothelial cells. Then, we generated an inducible Cre mouse line in which a CreERT2-T2A-tdTomato cassette was placed after the initiation codon ATG of the Slco1a4 locus. We found that tdTomato, which can indicate the endogenous Slco1a4 expression, was expressed in almost all cerebrovascular endothelial cells but not in any other non-endothelial cell types in the brain, including neurons, astrocytes, oligodendrocytes, pericytes, smooth muscle cells, and microglial cells, as well as in other organs. Consistently, when crossing the ROSA26LSL-EYFP Cre reporter mouse, EYFP also specifically labeled almost all cerebrovascular endothelial cells upon tamoxifen induction. Overall, we generated a new inducible Cre line that specifically targets cerebrovascular endothelial cells.


Subject(s)
Brain , Endothelial Cells , Integrases , Animals , Mice , Endothelial Cells/metabolism , Integrases/metabolism , Integrases/genetics , Brain/metabolism , Gene Knock-In Techniques , Mice, Transgenic , Blood-Brain Barrier/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Tamoxifen/pharmacology , Red Fluorescent Protein
7.
Nat Commun ; 15(1): 4561, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811575

ABSTRACT

The mammalian SWI/SNF-like BAF complexes play critical roles during animal development and pathological conditions. Previous gene deletion studies and characterization of human gene mutations implicate that the complexes both repress and activate a large number of genes. However, the direct function of the complexes in cells remains largely unclear due to the relatively long-term nature of gene deletion or natural mutation. Here we generate a mouse line by knocking in the auxin-inducible degron tag (AID) to the Smarca4 gene, which encodes BRG1, the essential ATPase subunit of the BAF complexes. We show that the tagged BRG1 can be efficiently depleted by osTIR1 expression and auxin treatment for 6 to 10 h in CD4 + T cells, hepatocytes, and fibroblasts isolated from the knock-in mice. The acute depletion of BRG1 leads to decreases in nascent RNAs and RNA polymerase II binding at a large number of genes, which are positively correlated with the loss of BRG1. Further, these changes are correlated with diminished accessibility at DNase I Hypersensitive Sites (DHSs) and p300 binding. The acute BRG1 depletion results in three major patterns of nucleosome shifts leading to narrower nucleosome spacing surrounding transcription factor motifs and at enhancers and transcription start sites (TSSs), which are correlated with loss of BRG1, decreased chromatin accessibility and decreased nascent RNAs. Acute depletion of BRG1 severely compromises the Trichostatin A (TSA) -induced histone acetylation, suggesting a substantial interplay between the chromatin remodeling activity of BRG1 and histone acetylation. Our data suggest BRG1 mainly plays a direct positive role in chromatin accessibility, RNAPII binding, and nascent RNA production by regulating nucleosome positioning and facilitating transcription factor binding to their target sites.


Subject(s)
DNA Helicases , Nuclear Proteins , Transcription Factors , Animals , Transcription Factors/metabolism , Transcription Factors/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Mice , Nucleosomes/metabolism , Nucleosomes/genetics , Indoleacetic Acids/metabolism , RNA Polymerase II/metabolism , Fibroblasts/metabolism , Gene Knock-In Techniques , Hepatocytes/metabolism , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/genetics , Transcriptional Activation , Transcription, Genetic , Histones/metabolism , Deoxyribonuclease I/metabolism , Chromatin/metabolism , Humans
8.
Sci Rep ; 14(1): 12393, 2024 05 29.
Article in English | MEDLINE | ID: mdl-38811759

ABSTRACT

Parkinson's disease (PD) is a progressive late-onset neurodegenerative disease leading to physical and cognitive decline. Mutations of leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of PD. LRRK2 is a complex scaffolding protein with known regulatory roles in multiple molecular pathways. Two prominent examples of LRRK2-modulated pathways are Wingless/Int (Wnt) and nuclear factor of activated T-cells (NFAT) signaling. Both are well described key regulators of immune and nervous system development as well as maturation. The aim of this study was to establish the physiological and pathogenic role of LRRK2 in Wnt and NFAT signaling in the brain, as well as the potential contribution of the non-canonical Wnt/Calcium pathway. In vivo cerebral Wnt and NFATc1 signaling activity was quantified in LRRK2 G2019S mutant knock-in (KI) and LRRK2 knockout (KO) male and female mice with repeated measures over 28 weeks, employing lentiviral luciferase biosensors, and analyzed using a mixed-effect model. To establish spatial resolution, we investigated tissues, and primary neuronal cell cultures from different brain regions combining luciferase signaling activity, immunohistochemistry, qPCR and western blot assays. Results were analyzed by unpaired t-test with Welch's correction or 2-way ANOVA with post hoc corrections. In vivo Wnt signaling activity in LRRK2 KO and LRRK2 G2019S KI mice was increased significantly ~ threefold, with a more pronounced effect in males (~ fourfold) than females (~ twofold). NFATc1 signaling was reduced ~ 0.5-fold in LRRK2 G2019S KI mice. Brain tissue analysis showed region-specific expression changes in Wnt and NFAT signaling components. These effects were predominantly observed at the protein level in the striatum and cerebral cortex of LRRK2 KI mice. Primary neuronal cell culture analysis showed significant genotype-dependent alterations in Wnt and NFATc1 signaling under basal and stimulated conditions. Wnt and NFATc1 signaling was primarily dysregulated in cortical and hippocampal neurons respectively. Our study further built on knowledge of LRRK2 as a Wnt and NFAT signaling protein. We identified complex changes in neuronal models of LRRK2 PD, suggesting a role for mutant LRRK2 in the dysregulation of NFAT, and canonical and non-canonical Wnt signaling.


Subject(s)
Disease Models, Animal , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , NFATC Transcription Factors , Parkinson Disease , Wnt Signaling Pathway , Animals , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Male , Mice , Female , Gene Knock-In Techniques , Mice, Knockout , Neurons/metabolism , Brain/metabolism , Brain/pathology , Mutation , Humans
9.
Sci Adv ; 10(20): eadj5942, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758779

ABSTRACT

Acetyl-CoA synthetase short-chain family member 1 (ACSS1) uses acetate to generate mitochondrial acetyl-CoA and is regulated by deacetylation by sirtuin 3. We generated an ACSS1-acetylation (Ac) mimic mouse, where lysine-635 was mutated to glutamine (K635Q). Male Acss1K635Q/K635Q mice were smaller with higher metabolic rate and blood acetate and decreased liver/serum ATP and lactate levels. After a 48-hour fast, Acss1K635Q/K635Q mice presented hypothermia and liver aberrations, including enlargement, discoloration, lipid droplet accumulation, and microsteatosis, consistent with nonalcoholic fatty liver disease (NAFLD). RNA sequencing analysis suggested dysregulation of fatty acid metabolism, cellular senescence, and hepatic steatosis networks, consistent with NAFLD. Fasted Acss1K635Q/K635Q mouse livers showed increased fatty acid synthase (FASN) and stearoyl-CoA desaturase 1 (SCD1), both associated with NAFLD, and increased carbohydrate response element-binding protein binding to Fasn and Scd1 enhancer regions. Last, liver lipidomics showed elevated ceramide, lysophosphatidylethanolamine, and lysophosphatidylcholine, all associated with NAFLD. Thus, we propose that ACSS1-K635-Ac dysregulation leads to aberrant lipid metabolism, cellular senescence, and NAFLD.


Subject(s)
Cellular Senescence , Mitochondria , Non-alcoholic Fatty Liver Disease , Stearoyl-CoA Desaturase , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Mice , Cellular Senescence/genetics , Acetylation , Mitochondria/metabolism , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/genetics , Male , Acetate-CoA Ligase/metabolism , Acetate-CoA Ligase/genetics , Gene Knock-In Techniques , Liver/metabolism , Liver/pathology , Lipid Metabolism , Sirtuin 3/metabolism , Sirtuin 3/genetics , Disease Models, Animal , Coenzyme A Ligases , Fatty Acid Synthase, Type I
10.
Proc Natl Acad Sci U S A ; 121(21): e2404763121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38743626

ABSTRACT

Congenital stationary night blindness (CSNB) is an inherited retinal disease that causes a profound loss of rod sensitivity without severe retinal degeneration. One well-studied rhodopsin point mutant, G90D-Rho, is thought to cause CSNB because of its constitutive activity in darkness causing rod desensitization. However, the nature of this constitutive activity and its precise molecular source have not been resolved for almost 30 y. In this study, we made a knock-in (KI) mouse line with a very low expression of G90D-Rho (equal in amount to ~0.1% of normal rhodopsin, WT-Rho, in WT rods), with the remaining WT-Rho replaced by REY-Rho, a mutant with a very low efficiency of activating transducin due to a charge reversal of the highly conserved ERY motif to REY. We observed two kinds of constitutive noise: one being spontaneous isomerization (R*) of G90D-Rho at a molecular rate (R* s-1) 175-fold higher than WT-Rho and the other being G90D-Rho-generated dark continuous noise comprising low-amplitude unitary events occurring at a very high molecular rate equivalent in effect to ~40,000-fold of R* s-1 from WT-Rho. Neither noise type originated from G90D-Opsin because exogenous 11-cis-retinal had no effect. Extrapolating the above observations at low (0.1%) expression of G90D-Rho to normal disease exhibited by a KI mouse model with RhoG90D/WTand RhoG90D/G90D genotypes predicts the disease condition very well quantitatively. Overall, the continuous noise from G90D-Rho therefore predominates, constituting the major equivalent background light causing rod desensitization in CSNB.


Subject(s)
Eye Diseases, Hereditary , Genetic Diseases, X-Linked , Myopia , Night Blindness , Rhodopsin , Animals , Night Blindness/genetics , Night Blindness/metabolism , Eye Diseases, Hereditary/genetics , Eye Diseases, Hereditary/metabolism , Mice , Rhodopsin/genetics , Rhodopsin/metabolism , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Myopia/genetics , Myopia/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Darkness , Transducin/genetics , Transducin/metabolism , Gene Knock-In Techniques , Disease Models, Animal
11.
Emerg Microbes Infect ; 13(1): 2353302, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38753462

ABSTRACT

Animal models of COVID-19 facilitate the development of vaccines and antivirals against SARS-CoV-2. The efficacy of antivirals or vaccines may differ in different animal models with varied degrees of disease. Here, we introduce a mouse model expressing human angiotensin-converting enzyme 2 (ACE2). In this model, ACE2 with the human cytokeratin 18 promoter was knocked into the Hipp11 locus of C57BL/6J mouse by CRISPR - Cas9 (K18-hACE2 KI). Upon intranasal inoculation with high (3 × 105 PFU) or low (2.5 × 102 PFU) dose of SARS-CoV-2 wildtype (WT), Delta, Omicron BA.1, or Omicron BA.2 variants, all mice showed obvious infection symptoms, including weight loss, high viral loads in the lung, and interstitial pneumonia. 100% lethality was observed in K18-hACE2 KI mice infected by variants with a delay of endpoint for Delta and BA.1, and a significantly attenuated pathogenicity was observed for BA.2. The pneumonia of infected mice was accompanied by the infiltration of neutrophils and pulmonary fibrosis in the lung. Compared with K18-hACE2 Tg mice and HFH4-hACE2 Tg mice, K18-hACE2 KI mice are more susceptible to SARS-CoV-2. In the antivirals test, REGN10933 and Remdesivir had limited antiviral efficacies in K18-hACE2 KI mice upon the challenge of SARS-CoV-2 infections, while Nirmatrelvir, monoclonal antibody 4G4, and mRNA vaccines potently protected the mice from death. Our results suggest that the K18-hACE2 KI mouse model is lethal and stable for SARS-CoV-2 infection, and is practicable and stringent to antiviral development.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antiviral Agents , COVID-19 , Disease Models, Animal , Mice, Inbred C57BL , SARS-CoV-2 , Animals , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/virology , Mice , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , Humans , Lung/virology , Lung/pathology , COVID-19 Drug Treatment , Keratin-18/genetics , Viral Load , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adenosine Monophosphate/therapeutic use , Alanine/analogs & derivatives , Alanine/pharmacology , Gene Knock-In Techniques , Antibodies, Viral/immunology , Antibodies, Viral/blood , Female
12.
Science ; 384(6697): eadk0582, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38753770

ABSTRACT

Germline-targeting (GT) HIV vaccine strategies are predicated on deriving broadly neutralizing antibodies (bnAbs) through multiple boost immunogens. However, as the recruitment of memory B cells (MBCs) to germinal centers (GCs) is inefficient and may be derailed by serum antibody-induced epitope masking, driving further B cell receptor (BCR) modification in GC-experienced B cells after boosting poses a challenge. Using humanized immunoglobulin knockin mice, we found that GT protein trimer immunogen N332-GT5 could prime inferred-germline precursors to the V3-glycan-targeted bnAb BG18 and that B cells primed by N332-GT5 were effectively boosted by either of two novel protein immunogens designed to have minimum cross-reactivity with the off-target V1-binding responses. The delivery of the prime and boost immunogens as messenger RNA lipid nanoparticles (mRNA-LNPs) generated long-lasting GCs, somatic hypermutation, and affinity maturation and may be an effective tool in HIV vaccine development.


Subject(s)
AIDS Vaccines , Broadly Neutralizing Antibodies , Germinal Center , HIV Antibodies , HIV-1 , Immunization, Secondary , Nanoparticles , mRNA Vaccines , Animals , Humans , Mice , AIDS Vaccines/immunology , B-Lymphocytes/immunology , Broadly Neutralizing Antibodies/immunology , Cross Reactions , Gene Knock-In Techniques , Germinal Center/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , HIV-1/genetics , Liposomes , Memory B Cells/immunology , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/genetics , Somatic Hypermutation, Immunoglobulin , mRNA Vaccines/immunology , Female , Mice, Inbred C57BL
13.
Nat Immunol ; 25(6): 1083-1096, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38816616

ABSTRACT

Current prophylactic human immunodeficiency virus 1 (HIV-1) vaccine research aims to elicit broadly neutralizing antibodies (bnAbs). Membrane-proximal external region (MPER)-targeting bnAbs, such as 10E8, provide exceptionally broad neutralization, but some are autoreactive. Here, we generated humanized B cell antigen receptor knock-in mouse models to test whether a series of germline-targeting immunogens could drive MPER-specific precursors toward bnAbs. We found that recruitment of 10E8 precursors to germinal centers (GCs) required a minimum affinity for germline-targeting immunogens, but the GC residency of MPER precursors was brief due to displacement by higher-affinity endogenous B cell competitors. Higher-affinity germline-targeting immunogens extended the GC residency of MPER precursors, but robust long-term GC residency and maturation were only observed for MPER-HuGL18, an MPER precursor clonotype able to close the affinity gap with endogenous B cell competitors in the GC. Thus, germline-targeting immunogens could induce MPER-targeting antibodies, and B cell residency in the GC may be regulated by a precursor-competitor affinity gap.


Subject(s)
Antibody Affinity , B-Lymphocytes , Germinal Center , HIV Antibodies , HIV-1 , Germinal Center/immunology , Animals , Mice , Humans , B-Lymphocytes/immunology , HIV-1/immunology , HIV Antibodies/immunology , Antibody Affinity/immunology , Antibodies, Neutralizing/immunology , HIV Infections/immunology , AIDS Vaccines/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/immunology , Gene Knock-In Techniques , Mice, Transgenic , Broadly Neutralizing Antibodies/immunology , Mice, Inbred C57BL
14.
Yi Chuan ; 46(3): 219-231, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38632100

ABSTRACT

CRISPR/Cas9 gene editing technology, as a highly efficient genome editing method, has been extensively employed in the realm of animal husbandry for genetic improvement. With its remarkable efficiency and precision, this technology has revolutionized the field of animal husbandry. Currently, CRISPR/Cas9-based gene knockout, gene knock-in and gene modification techniques are widely employed to achieve precise enhancements in crucial production traits of livestock and poultry species. In this review, we summarize the operational principle and development history of CRISPR/Cas9 technology. Additionally, we highlight the research advancements utilizing this technology in muscle growth and development, fiber growth, milk quality composition, disease resistance breeding, and animal welfare within the livestock and poultry sectors. Our aim is to provide a more comprehensive understanding of the application of CRISPR/Cas9 technology in gene editing for livestock and poultry.


Subject(s)
CRISPR-Cas Systems , Livestock , Animals , Livestock/genetics , Poultry/genetics , Gene Editing/methods , Gene Knock-In Techniques
15.
Sci Adv ; 10(17): eadl1088, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38669339

ABSTRACT

A sharp drop in lenticular glutathione (GSH) plays a pivotal role in age-related cataract (ARC) formation. Despite recognizing GSH's importance in lens defense for decades, its decline with age remains puzzling. Our recent study revealed an age-related truncation affecting the essential GSH biosynthesis enzyme, the γ-glutamylcysteine ligase catalytic subunit (GCLC), at aspartate residue 499. Intriguingly, these truncated GCLC fragments compete with full-length GCLC in forming a heterocomplex with the modifier subunit (GCLM) but exhibit markedly reduced enzymatic activity. Crucially, using an aspartate-to-glutamate mutation knock-in (D499E-KI) mouse model that blocks GCLC truncation, we observed a notable delay in ARC formation compared to WT mice: Nearly 50% of D499E-KI mice remained cataract-free versus ~20% of the WT mice at their age of 20 months. Our findings concerning age-related GCLC truncation might be the key to understanding the profound reduction in lens GSH with age. By halting GCLC truncation, we can rejuvenate lens GSH levels and considerably postpone cataract onset.


Subject(s)
Aging , Catalytic Domain , Cataract , Glutamate-Cysteine Ligase , Glutathione , Lens, Crystalline , Cataract/pathology , Cataract/genetics , Cataract/metabolism , Animals , Glutamate-Cysteine Ligase/metabolism , Glutamate-Cysteine Ligase/genetics , Mice , Glutathione/metabolism , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Aging/metabolism , Humans , Disease Models, Animal , Mutation , Gene Knock-In Techniques
16.
Stem Cell Reports ; 19(5): 744-757, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38579711

ABSTRACT

Precise insertion of fluorescent proteins into lineage-specific genes in human pluripotent stem cells (hPSCs) presents challenges due to low knockin efficiency and difficulties in isolating targeted cells. To overcome these hurdles, we present the modified mRNA (ModRNA)-based Activation for Gene Insertion and Knockin (MAGIK) method. MAGIK operates in two steps: first, it uses a Cas9-2A-p53DD modRNA with a mini-donor plasmid (without a drug selection cassette) to significantly enhance efficiency. Second, a deactivated Cas9 activator modRNA and a 'dead' guide RNA are used to temporarily activate the targeted gene, allowing for live cell sorting of targeted cells. Consequently, MAGIK eliminates the need for drug selection cassettes or labor-intensive single-cell colony screening, expediting precise gene editing. We showed MAGIK can be utilized to insert fluorescent proteins into various genes, including SOX17, NKX6.1, NKX2.5, and PDX1, across multiple hPSC lines. This underscores its robust performance and offers a promising solution for achieving knockin in hPSCs within a significantly shortened time frame.


Subject(s)
Cell Lineage , Gene Knock-In Techniques , Pluripotent Stem Cells , Humans , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Cell Lineage/genetics , Gene Knock-In Techniques/methods , Genes, Reporter , CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems/genetics , Cell Line , Gene Editing/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
J Exp Med ; 221(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38661718

ABSTRACT

Chemokines guide immune cells during their response against pathogens and tumors. Various techniques exist to determine chemokine production, but none to identify cells that directly sense chemokines in vivo. We have generated CCL3-EASER (ErAse, SEnd, Receive) mice that simultaneously report for Ccl3 transcription and translation, allow identifying Ccl3-sensing cells, and permit inducible deletion of Ccl3-producing cells. We infected these mice with murine cytomegalovirus (mCMV), where Ccl3 and NK cells are critical defense mediators. We found that NK cells transcribed Ccl3 already in homeostasis, but Ccl3 translation required type I interferon signaling in infected organs during early infection. NK cells were both the principal Ccl3 producers and sensors of Ccl3, indicating auto/paracrine communication that amplified NK cell response, and this was essential for the early defense against mCMV. CCL3-EASER mice represent the prototype of a new class of dual fluorescence reporter mice for analyzing cellular communication via chemokines, which may be applied also to other chemokines and disease models.


Subject(s)
Cell Communication , Chemokine CCL3 , Models, Animal , Protein Biosynthesis , Transcription, Genetic , Animals , Mice , Cell Communication/immunology , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Gene Knock-In Techniques , Mice, Transgenic , Muromegalovirus , Protein Biosynthesis/drug effects , Protein Biosynthesis/immunology , Transcription, Genetic/immunology , Killer Cells, Natural/immunology , Interferon-beta/pharmacology , Herpesviridae Infections/immunology
18.
FASEB J ; 38(8): e23606, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38648465

ABSTRACT

Rhodopsin mislocalization encompasses various blind conditions. Rhodopsin mislocalization is the primary factor leading to rod photoreceptor dysfunction and degeneration in autosomal dominant retinitis pigmentosa (adRP) caused by class I mutations. In this study, we report a new knock-in mouse model that harbors a class I Q344X mutation in the endogenous rhodopsin gene, which causes rod photoreceptor degeneration in an autosomal dominant pattern. In RhoQ344X/+ mice, mRNA transcripts from the wild-type (Rho) and RhoQ344X mutant rhodopsin alleles are expressed at equal levels. However, the amount of RHOQ344X mutant protein is 2.7 times lower than that of wild-type rhodopsin, a finding consistent with the rapid degradation of the mutant protein. Immunofluorescence microscopy indicates that RHOQ344X is mislocalized to the inner segment and outer nuclear layers of rod photoreceptors in both RhoQ344X/+ and RhoQ344X/Q344X mice, confirming the essential role of the C-terminal VxPx motif in promoting OS delivery of rhodopsin. The mislocalization of RHOQ344X is associated with the concurrent mislocalization of wild-type rhodopsin in RhoQ344X/+ mice. To understand the global changes in proteostasis, we conducted quantitative proteomics analysis and found attenuated expression of rod-specific OS membrane proteins accompanying reduced expression of ciliopathy causative gene products, including constituents of BBSome and axonemal dynein subunit. Those studies unveil a novel negative feedback regulation involving ciliopathy-associated proteins. In this process, a defect in the trafficking signal leads to a reduced quantity of the trafficking apparatus, culminating in a widespread reduction in the transport of ciliary proteins.


Subject(s)
Disease Models, Animal , Gene Knock-In Techniques , Retinal Rod Photoreceptor Cells , Retinitis Pigmentosa , Rhodopsin , Animals , Rhodopsin/metabolism , Rhodopsin/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Mice , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Cilia/metabolism , Cilia/pathology
19.
Nature ; 628(8009): 844-853, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38570685

ABSTRACT

Mitochondria are critical modulators of antiviral tolerance through the release of mitochondrial RNA and DNA (mtDNA and mtRNA) fragments into the cytoplasm after infection, activating virus sensors and type-I interferon (IFN-I) response1-4. The relevance of these mechanisms for mitochondrial diseases remains understudied. Here we investigated mitochondrial recessive ataxia syndrome (MIRAS), which is caused by a common European founder mutation in DNA polymerase gamma (POLG1)5. Patients homozygous for the MIRAS variant p.W748S show exceptionally variable ages of onset and symptoms5, indicating that unknown modifying factors contribute to disease manifestation. We report that the mtDNA replicase POLG1 has a role in antiviral defence mechanisms to double-stranded DNA and positive-strand RNA virus infections (HSV-1, TBEV and SARS-CoV-2), and its p.W748S variant dampens innate immune responses. Our patient and knock-in mouse data show that p.W748S compromises mtDNA replisome stability, causing mtDNA depletion, aggravated by virus infection. Low mtDNA and mtRNA release into the cytoplasm and a slow IFN response in MIRAS offer viruses an early replicative advantage, leading to an augmented pro-inflammatory response, a subacute loss of GABAergic neurons and liver inflammation and necrosis. A population databank of around 300,000 Finnish individuals6 demonstrates enrichment of immunodeficient traits in carriers of the POLG1 p.W748S mutation. Our evidence suggests that POLG1 defects compromise antiviral tolerance, triggering epilepsy and liver disease. The finding has important implications for the mitochondrial disease spectrum, including epilepsy, ataxia and parkinsonism.


Subject(s)
Alleles , DNA Polymerase gamma , Encephalitis Viruses, Tick-Borne , Herpesvirus 1, Human , Immune Tolerance , SARS-CoV-2 , Animals , Female , Humans , Male , Mice , Age of Onset , COVID-19/immunology , COVID-19/virology , COVID-19/genetics , DNA Polymerase gamma/genetics , DNA Polymerase gamma/immunology , DNA Polymerase gamma/metabolism , DNA, Mitochondrial/immunology , DNA, Mitochondrial/metabolism , Encephalitis Viruses, Tick-Borne/immunology , Encephalitis, Tick-Borne/genetics , Encephalitis, Tick-Borne/immunology , Encephalitis, Tick-Borne/virology , Founder Effect , Gene Knock-In Techniques , Herpes Simplex/genetics , Herpes Simplex/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Immune Tolerance/genetics , Immune Tolerance/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon Type I/immunology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/immunology , Mutation , RNA, Mitochondrial/immunology , RNA, Mitochondrial/metabolism , SARS-CoV-2/immunology
20.
Science ; 384(6694): 438-446, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38662831

ABSTRACT

Liver mitochondria play a central role in metabolic adaptations to changing nutritional states, yet their dynamic regulation upon anticipated changes in nutrient availability has remained unaddressed. Here, we found that sensory food perception rapidly induced mitochondrial fragmentation in the liver through protein kinase B/AKT (AKT)-dependent phosphorylation of serine 131 of the mitochondrial fission factor (MFFS131). This response was mediated by activation of hypothalamic pro-opiomelanocortin (POMC)-expressing neurons. A nonphosphorylatable MFFS131G knock-in mutation abrogated AKT-induced mitochondrial fragmentation in vitro. In vivo, MFFS131G knock-in mice displayed altered liver mitochondrial dynamics and impaired insulin-stimulated suppression of hepatic glucose production. Thus, rapid activation of a hypothalamus-liver axis can adapt mitochondrial function to anticipated changes of nutritional state in control of hepatic glucose metabolism.


Subject(s)
Food , Gluconeogenesis , Glucose , Liver , Membrane Proteins , Mitochondria, Liver , Mitochondrial Dynamics , Mitochondrial Proteins , Perception , Animals , Male , Mice , Gene Knock-In Techniques , Glucose/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Liver/metabolism , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Neurons/metabolism , Phosphorylation , Pro-Opiomelanocortin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...