Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.771
Filter
Add more filters

Coleção CLAP
Publication year range
1.
Cell ; 186(18): 3793-3809.e26, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37562401

ABSTRACT

Hepatocytes, the major metabolic hub of the body, execute functions that are human-specific, altered in human disease, and currently thought to be regulated through endocrine and cell-autonomous mechanisms. Here, we show that key metabolic functions of human hepatocytes are controlled by non-parenchymal cells (NPCs) in their microenvironment. We developed mice bearing human hepatic tissue composed of human hepatocytes and NPCs, including human immune, endothelial, and stellate cells. Humanized livers reproduce human liver architecture, perform vital human-specific metabolic/homeostatic processes, and model human pathologies, including fibrosis and non-alcoholic fatty liver disease (NAFLD). Leveraging species mismatch and lipidomics, we demonstrate that human NPCs control metabolic functions of human hepatocytes in a paracrine manner. Mechanistically, we uncover a species-specific interaction whereby WNT2 secreted by sinusoidal endothelial cells controls cholesterol uptake and bile acid conjugation in hepatocytes through receptor FZD5. These results reveal the essential microenvironmental regulation of hepatic metabolism and its human-specific aspects.


Subject(s)
Endothelial Cells , Liver , Animals , Humans , Mice , Endothelial Cells/metabolism , Hepatocytes/metabolism , Kupffer Cells/metabolism , Liver/cytology , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Fibrosis/metabolism
2.
Cell ; 180(2): 221-232, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31978342

ABSTRACT

Human diseases are increasingly linked with an altered or "dysbiotic" gut microbiota, but whether such changes are causal, consequential, or bystanders to disease is, for the most part, unresolved. Human microbiota-associated (HMA) rodents have become a cornerstone of microbiome science for addressing causal relationships between altered microbiomes and host pathology. In a systematic review, we found that 95% of published studies (36/38) on HMA rodents reported a transfer of pathological phenotypes to recipient animals, and many extrapolated the findings to make causal inferences to human diseases. We posit that this exceedingly high rate of inter-species transferable pathologies is implausible and overstates the role of the gut microbiome in human disease. We advocate for a more rigorous and critical approach for inferring causality to avoid false concepts and prevent unrealistic expectations that may undermine the credibility of microbiome science and delay its translation.


Subject(s)
Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Rodentia/microbiology , Animals , Disease/etiology , Fecal Microbiota Transplantation/methods , Humans , Mice , Microbiota/physiology , Models, Animal , Rats
3.
Cell ; 180(3): 471-489.e22, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32004464

ABSTRACT

Broadly neutralizing antibodies (bNAbs) represent a promising approach to prevent and treat HIV-1 infection. However, viral escape through mutation of the HIV-1 envelope glycoprotein (Env) limits clinical applications. Here we describe 1-18, a new VH1-46-encoded CD4 binding site (CD4bs) bNAb with outstanding breadth (97%) and potency (GeoMean IC50 = 0.048 µg/mL). Notably, 1-18 is not susceptible to typical CD4bs escape mutations and effectively overcomes HIV-1 resistance to other CD4bs bNAbs. Moreover, mutational antigenic profiling uncovered restricted pathways of HIV-1 escape. Of most promise for therapeutic use, even 1-18 alone fully suppressed viremia in HIV-1-infected humanized mice without selecting for resistant viral variants. A 2.5-Å cryo-EM structure of a 1-18-BG505SOSIP.664 Env complex revealed that these characteristics are likely facilitated by a heavy-chain insertion and increased inter-protomer contacts. The ability of 1-18 to effectively restrict HIV-1 escape pathways provides a new option to successfully prevent and treat HIV-1 infection.


Subject(s)
Broadly Neutralizing Antibodies/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Animals , Antibodies, Monoclonal/immunology , Binding Sites , CD4 Antigens/metabolism , CHO Cells , Cohort Studies , Cricetulus , Epitopes/immunology , Female , HEK293 Cells , HIV Infections/prevention & control , HIV Infections/virology , Heterografts , Humans , Male , Mice , Mice, Inbred NOD , Middle Aged , Mutation , Protein Binding/immunology , env Gene Products, Human Immunodeficiency Virus/genetics
4.
Cell ; 178(1): 122-134.e12, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31230714

ABSTRACT

Epitranscriptomic regulation controls information flow through the central dogma and provides unique opportunities for manipulating cells at the RNA level. However, both fundamental studies and potential translational applications are impeded by a lack of methods to target specific RNAs with effector proteins. Here, we present CRISPR-Cas-inspired RNA targeting system (CIRTS), a protein engineering strategy for constructing programmable RNA control elements. We show that CIRTS is a simple and generalizable approach to deliver a range of effector proteins, including nucleases, degradation machinery, translational activators, and base editors to target transcripts. We further demonstrate that CIRTS is not only smaller than naturally occurring CRISPR-Cas programmable RNA binding systems but can also be built entirely from human protein parts. CIRTS provides a platform to probe fundamental RNA regulatory processes, and the human-derived nature of CIRTS provides a potential strategy to avoid immune issues when applied to epitranscriptome-modulating therapies.


Subject(s)
Gene Editing/methods , Protein Engineering/methods , RNA, Guide, Kinetoplastida/metabolism , RNA/metabolism , Transcription Activator-Like Effector Nucleases/metabolism , CRISPR-Cas Systems/genetics , Escherichia coli/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Protein Biosynthesis , Proteolysis , RNA, Small Interfering , Transcription Activator-Like Effector Nucleases/genetics , Transfection
5.
Immunity ; 56(9): 2152-2171.e13, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37582369

ABSTRACT

Microglia phenotypes are highly regulated by the brain environment, but the transcriptional networks that specify the maturation of human microglia are poorly understood. Here, we characterized stage-specific transcriptomes and epigenetic landscapes of fetal and postnatal human microglia and acquired corresponding data in induced pluripotent stem cell (iPSC)-derived microglia, in cerebral organoids, and following engraftment into humanized mice. Parallel development of computational approaches that considered transcription factor (TF) co-occurrence and enhancer activity allowed prediction of shared and state-specific gene regulatory networks associated with fetal and postnatal microglia. Additionally, many features of the human fetal-to-postnatal transition were recapitulated in a time-dependent manner following the engraftment of iPSC cells into humanized mice. These data and accompanying computational approaches will facilitate further efforts to elucidate mechanisms by which human microglia acquire stage- and disease-specific phenotypes.


Subject(s)
Induced Pluripotent Stem Cells , Microglia , Humans , Mice , Animals , Gene Regulatory Networks , Brain , Gene Expression Regulation
6.
Cell ; 171(7): 1508-1519.e13, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29198523

ABSTRACT

Humans and yeast are separated by a billion years of evolution, yet their conserved histones retain central roles in gene regulation. Here, we "reset" yeast to use core human nucleosomes in lieu of their own (a rare event taking 20 days), which initially only worked with variant H3.1. The cells adapt by acquiring suppressor mutations in cell-division genes or by acquiring certain aneuploid states. Converting five histone residues to their yeast counterparts restored robust growth. We reveal that humanized nucleosomes are positioned according to endogenous yeast DNA sequence and chromatin-remodeling network, as judged by a yeast-like nucleosome repeat length. However, human nucleosomes have higher DNA occupancy, globally reduce RNA content, and slow adaptation to new conditions by delaying chromatin remodeling. These humanized yeasts (including H3.3) pose fundamental new questions about how chromatin is linked to many cell processes and provide a platform to study histone variants via yeast epigenome reprogramming.


Subject(s)
Histones/chemistry , Nucleosomes/chemistry , Saccharomyces cerevisiae/chemistry , Chromatin Assembly and Disassembly , DNA-Directed RNA Polymerases/metabolism , Gene Expression Regulation , HeLa Cells , Histones/metabolism , Humans , Mutation , Saccharomyces cerevisiae/metabolism , Species Specificity , Transcription, Genetic
7.
Cell ; 168(6): 1086-1100.e10, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28283063

ABSTRACT

Innate lymphoid cells (ILCs) represent innate versions of T helper and cytotoxic T cells that differentiate from committed ILC precursors (ILCPs). How ILCPs give rise to mature tissue-resident ILCs remains unclear. Here, we identify circulating and tissue ILCPs in humans that fail to express the transcription factors and cytokine outputs of mature ILCs but have these signature loci in an epigenetically poised configuration. Human ILCPs robustly generate all ILC subsets in vitro and in vivo. While human ILCPs express low levels of retinoic acid receptor (RAR)-related orphan receptor C (RORC) transcripts, these cells are found in RORC-deficient patients and retain potential for EOMES+ natural killer (NK) cells, interferon gamma-positive (IFN-γ+) ILC1s, interleukin (IL)-13+ ILC2s, and for IL-22+, but not for IL-17A+ ILC3s. Our results support a model of tissue ILC differentiation ("ILC-poiesis"), whereby diverse ILC subsets are generated in situ from systemically distributed ILCPs in response to local environmental signals.


Subject(s)
Lymphocytes/cytology , Stem Cells/cytology , Animals , Antigens, CD34/analysis , Cell Differentiation , Cell Lineage , Fetal Blood/cytology , Fetus/cytology , Humans , Immunity, Innate , Interleukin-17 , Liver/cytology , Lung/cytology , Lymphocytes/immunology , Lymphoid Tissue/cytology , Mice , Proto-Oncogene Proteins c-kit/analysis , Transcription, Genetic
8.
Immunity ; 55(8): 1343-1353, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35947979

ABSTRACT

While inbred mice have informed most of what we know about the immune system in the modern era, they have clear limitations with respect to their ability to be informative regarding genetic heterogeneity or microbial influences. They have also not been very predictive as models of human disease or vaccination results. Although there are concerted attempts to compensate for these flaws, the rapid rise of human studies, driven by both technical and conceptual advances, promises to fill in these gaps, as well as provide direct information about human diseases and vaccination responses. Work on human immunity has already provided important additional perspectives on basic immunology such as the importance of clonal deletion to self-tolerance, and while many challenges remain, it seems inevitable that "the human model" will continue to inform us about the immune system and even allow for the discovery of new mechanisms.


Subject(s)
Clonal Deletion , Immune System , Animals , Humans , Immune System/physiology , Mice , Self Tolerance , Vaccination
9.
Immunity ; 54(2): 259-275.e7, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33382972

ABSTRACT

The study of human macrophages and their ontogeny is an important unresolved issue. Here, we use a humanized mouse model expressing human cytokines to dissect the development of lung macrophages from human hematopoiesis in vivo. Human CD34+ hematopoietic stem and progenitor cells (HSPCs) generated three macrophage populations, occupying separate anatomical niches in the lung. Intravascular cell labeling, cell transplantation, and fate-mapping studies established that classical CD14+ blood monocytes derived from HSPCs migrated into lung tissue and gave rise to human interstitial and alveolar macrophages. In contrast, non-classical CD16+ blood monocytes preferentially generated macrophages resident in the lung vasculature (pulmonary intravascular macrophages). Finally, single-cell RNA sequencing defined intermediate differentiation stages in human lung macrophage development from blood monocytes. This study identifies distinct developmental pathways from circulating monocytes to lung macrophages and reveals how cellular origin contributes to human macrophage identity, diversity, and localization in vivo.


Subject(s)
Hematopoietic Stem Cells/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Monocytes/immunology , Antigens, CD34/metabolism , Biodiversity , Cell Differentiation , Cell Movement , Cells, Cultured , Fetal Blood/cytology , Humans , Lipopolysaccharide Receptors/metabolism , Lung/blood supply , Receptors, IgG/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Stem Cell Niche
10.
Immunity ; 50(6): 1513-1529.e9, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31126879

ABSTRACT

Broadly neutralizing antibodies (bNAbs) against HIV-1 envelope (Env) inform vaccine design and are potential therapeutic agents. We identified SF12 and related bNAbs with up to 62% neutralization breadth from an HIV-infected donor. SF12 recognized a glycan-dominated epitope on Env's silent face and was potent against clade AE viruses, which are poorly covered by V3-glycan bNAbs. A 3.3Å cryo-EM structure of a SF12-Env trimer complex showed additional contacts to Env protein residues by SF12 compared with VRC-PG05, the only other known donor-derived silentface antibody, explaining SF12's increased neutralization breadth, potency, and resistance to Env mutation routes. Asymmetric binding of SF12 was associated with distinct N-glycan conformations across Env protomers, demonstrating intra-Env glycan heterogeneity. Administrating SF12 to HIV-1-infected humanized mice suppressed viremia and selected for viruses lacking the N448gp120 glycan. Effective bNAbs can therefore be raised against HIV-1 Env's silent face, suggesting their potential for HIV-1 prevention, therapy, and vaccine development.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Amino Acid Sequence , Antibodies, Neutralizing/isolation & purification , Antibody Affinity/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Epitopes/chemistry , Epitopes/immunology , Glycosylation , HIV Antibodies/isolation & purification , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/immunology , HIV Infections/virology , HIV-1/classification , HIV-1/genetics , Humans , Models, Molecular , Phylogeny , Polysaccharides/chemistry , Polysaccharides/metabolism , Protein Binding/immunology , Protein Conformation , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/metabolism
11.
Proc Natl Acad Sci U S A ; 121(28): e2403442121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968107

ABSTRACT

Plasmodium falciparum causes severe malaria and assembles a protein translocon (PTEX) complex at the parasitophorous vacuole membrane (PVM) of infected erythrocytes, through which several hundred proteins are exported to facilitate growth. The preceding liver stage of infection involves growth in a hepatocyte-derived PVM; however, the importance of protein export during P. falciparum liver infection remains unexplored. Here, we use the FlpL/FRT system to conditionally excise genes in P. falciparum sporozoites for functional liver-stage studies. Disruption of PTEX members ptex150 and exp2 did not affect sporozoite development in mosquitoes or infectivity for hepatocytes but attenuated liver-stage growth in humanized mice. While PTEX150 deficiency reduced fitness on day 6 postinfection by 40%, EXP2 deficiency caused 100% loss of liver parasites, demonstrating that PTEX components are required for growth in hepatocytes to differing degrees. To characterize PTEX loss-of-function mutations, we localized four liver-stage Plasmodium export element (PEXEL) proteins. P. falciparum liver specific protein 2 (LISP2), liver-stage antigen 3 (LSA3), circumsporozoite protein (CSP), and a Plasmodium berghei LISP2 reporter all localized to the periphery of P. falciparum liver stages but were not exported beyond the PVM. Expression of LISP2 and CSP but not LSA3 was reduced in ptex150-FRT and exp2-FRT liver stages, suggesting that expression of some PEXEL proteins is affected directly or indirectly by PTEX disruption. These results show that PTEX150 and EXP2 are important for P. falciparum development in hepatocytes and emphasize the emerging complexity of PEXEL protein trafficking.


Subject(s)
Hepatocytes , Liver , Malaria, Falciparum , Plasmodium falciparum , Protozoan Proteins , Sporozoites , Plasmodium falciparum/growth & development , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Animals , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Sporozoites/metabolism , Sporozoites/growth & development , Mice , Liver/parasitology , Liver/metabolism , Humans , Hepatocytes/parasitology , Hepatocytes/metabolism , Malaria, Falciparum/parasitology
12.
Hum Mol Genet ; 33(12): 1074-1089, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38520741

ABSTRACT

We have generated using CRISPR/Cas9 technology a partially humanized mouse model of the neurometabolic disease phenylketonuria (PKU), carrying the highly prevalent PAH variant c.1066-11G>A. This variant creates an alternative 3' splice site, leading to the inclusion of 9 nucleotides coding for 3 extra amino acids between Q355 and Y356 of the protein. Homozygous Pah c.1066-11A mice, with a partially humanized intron 10 sequence with the variant, accurately recapitulate the splicing defect and present almost undetectable hepatic PAH activity. They exhibit fur hypopigmentation, lower brain and body weight and reduced survival. Blood and brain phenylalanine levels are elevated, along with decreased tyrosine, tryptophan and monoamine neurotransmitter levels. They present behavioral deficits, mainly hypoactivity and diminished social interaction, locomotor deficiencies and an abnormal hind-limb clasping reflex. Changes in the morphology of glial cells, increased GFAP and Iba1 staining signals and decreased myelinization are observed. Hepatic tissue exhibits nearly absent PAH protein, reduced levels of chaperones DNAJC12 and HSP70 and increased autophagy markers LAMP1 and LC3BII, suggesting possible coaggregation of mutant PAH with chaperones and subsequent autophagy processing. This PKU mouse model with a prevalent human variant represents a useful tool for pathophysiology research and for novel therapies development.


Subject(s)
Disease Models, Animal , Phenylalanine Hydroxylase , Phenylketonurias , Animals , Mice , Phenylketonurias/genetics , Phenylketonurias/pathology , Phenylketonurias/metabolism , Humans , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Brain/metabolism , Brain/pathology , CRISPR-Cas Systems , Autophagy/genetics , Mutation , Liver/metabolism , Liver/pathology
13.
RNA ; 30(2): 149-170, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38071476

ABSTRACT

Intron branchpoint (BP) recognition by the U2 snRNP is a critical step of splicing, vulnerable to recurrent cancer mutations and bacterial natural product inhibitors. The BP binds a conserved pocket in the SF3B1 (human) or Hsh155 (yeast) U2 snRNP protein. Amino acids that line this pocket affect the binding of splicing inhibitors like Pladienolide-B (Plad-B), such that organisms differ in their sensitivity. To study the mechanism of splicing inhibitor action in a simplified system, we modified the naturally Plad-B resistant yeast Saccharomyces cerevisiae by changing 14 amino acids in the Hsh155 BP pocket to those from human. This humanized yeast grows normally, and splicing is largely unaffected by the mutation. Splicing is inhibited within minutes after the addition of Plad-B, and different introns appear inhibited to different extents. Intron-specific inhibition differences are also observed during cotranscriptional splicing in Plad-B using single-molecule intron tracking to minimize gene-specific transcription and decay rates that cloud estimates of inhibition by standard RNA-seq. Comparison of Plad-B intron sensitivities to those of the structurally distinct inhibitor Thailanstatin-A reveals intron-specific differences in sensitivity to different compounds. This work exposes a complex relationship between the binding of different members of this class of inhibitors to the spliceosome and intron-specific rates of BP recognition and catalysis. Introns with variant BP sequences seem particularly sensitive, echoing observations from mammalian cells, where monitoring individual introns is complicated by multi-intron gene architecture and alternative splicing. The compact yeast system may hasten the characterization of splicing inhibitors, accelerating improvements in selectivity and therapeutic efficacy.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Humans , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Introns/genetics , Ribonucleoprotein, U2 Small Nuclear/chemistry , Saccharomyces cerevisiae Proteins/metabolism , RNA Splicing , Spliceosomes/genetics , Amino Acids/genetics , RNA Precursors/genetics
14.
Semin Immunol ; 60: 101652, 2022 03.
Article in English | MEDLINE | ID: mdl-36162228

ABSTRACT

The two γ-herpesviruses Epstein Barr virus (EBV) and Kaposi sarcoma associated herpesvirus (KSHV) are each associated with more than 1% of all tumors in humans. While EBV establishes persistent infection in nearly all adult individuals, KSHV benefits from this widespread EBV prevalence for its own persistence. Interestingly, EBV infection expands early differentiated NKG2A+KIR- NK cells that protect against lytic EBV infection, while KSHV co-infection drives accumulation of poorly functional CD56-CD16+ NK cells. Thus persistent γ-herpesvirus infections are sculptors of human NK cell repertoires and the respectively stimulated NK cell subsets should be considered for immunotherapies of EBV and KSHV associated malignancies.


Subject(s)
Epstein-Barr Virus Infections , Herpesviridae Infections , Herpesvirus 8, Human , Neoplasms , Adult , Humans , Herpesvirus 4, Human/physiology , Herpesvirus 8, Human/physiology , Killer Cells, Natural
15.
Proc Natl Acad Sci U S A ; 120(37): e2306965120, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37669366

ABSTRACT

Fibrosis is regulated by interactions between immune and mesenchymal cells. However, the capacity of cell types to modulate human fibrosis pathology is poorly understood due to lack of a fully humanized model system. MISTRG6 mice were engineered by homologous mouse/human gene replacement to develop an immune system like humans when engrafted with human hematopoietic stem cells (HSCs). We utilized MISTRG6 mice to model scleroderma by transplantation of healthy or scleroderma skin from a patient with pansclerotic morphea to humanized mice engrafted with unmatched allogeneic HSC. We identified that scleroderma skin grafts contained both skin and bone marrow-derived human CD4 and CD8 T cells along with human endothelial cells and pericytes. Unlike healthy skin, fibroblasts in scleroderma skin were depleted and replaced by mouse fibroblasts. Furthermore, HSC engraftment alleviated multiple signatures of fibrosis, including expression of collagen and interferon genes, and proliferation and activation of human T cells. Fibrosis improvement correlated with reduced markers of T cell activation and expression of human IL-6 by mesenchymal cells. Mechanistic studies supported a model whereby IL-6 trans-signaling driven by CD4 T cell-derived soluble IL-6 receptor complexed with fibroblast-derived IL-6 promoted excess extracellular matrix gene expression. Thus, MISTRG6 mice transplanted with scleroderma skin demonstrated multiple fibrotic responses centered around human IL-6 signaling, which was improved by the presence of healthy bone marrow-derived immune cells. Our results highlight the importance of IL-6 trans-signaling in pathogenesis of scleroderma and the ability of healthy bone marrow-derived immune cells to mitigate disease.


Subject(s)
Basidiomycota , Scleroderma, Localized , Humans , Animals , Mice , Interleukin-6 , Endothelial Cells , Skin , Disease Models, Animal
16.
Proc Natl Acad Sci U S A ; 120(10): e2217199120, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36848564

ABSTRACT

COVID-19 remains a global pandemic of an unprecedented magnitude with millions of people now developing "COVID lung fibrosis." Single-cell transcriptomics of lungs of patients with long COVID revealed a unique immune signature demonstrating the upregulation of key proinflammatory and innate immune effector genes CD47, IL-6, and JUN. We modeled the transition to lung fibrosis after COVID and profiled the immune response with single-cell mass cytometry in JUN mice. These studies revealed that COVID mediated chronic immune activation reminiscent to long COVID in humans. It was characterized by increased CD47, IL-6, and phospho-JUN (pJUN) expression which correlated with disease severity and pathogenic fibroblast populations. When we subsequently treated a humanized COVID lung fibrosis model by combined blockade of inflammation and fibrosis, we not only ameliorated fibrosis but also restored innate immune equilibrium indicating possible implications for clinical management of COVID lung fibrosis in patients.


Subject(s)
COVID-19 , Pulmonary Fibrosis , Humans , Animals , Mice , Pulmonary Fibrosis/etiology , Post-Acute COVID-19 Syndrome , CD47 Antigen , Interleukin-6/genetics , Immunity, Innate
17.
Proc Natl Acad Sci U S A ; 120(19): e2217887120, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37126704

ABSTRACT

Treatment of HIV-1ADA-infected CD34+ NSG-humanized mice with long-acting ester prodrugs of cabotegravir, lamivudine, and abacavir in combination with native rilpivirine was followed by dual CRISPR-Cas9 C-C chemokine receptor type five (CCR5) and HIV-1 proviral DNA gene editing. This led to sequential viral suppression, restoration of absolute human CD4+ T cell numbers, then elimination of replication-competent virus in 58% of infected mice. Dual CRISPR therapies enabled the excision of integrated proviral DNA in infected human cells contained within live infected animals. Highly sensitive nucleic acid nested and droplet digital PCR, RNAscope, and viral outgrowth assays affirmed viral elimination. HIV-1 was not detected in the blood, spleen, lung, kidney, liver, gut, bone marrow, and brain of virus-free animals. Progeny virus from adoptively transferred and CRISPR-treated virus-free mice was neither detected nor recovered. Residual HIV-1 DNA fragments were easily seen in untreated and viral-rebounded animals. No evidence of off-target toxicities was recorded in any of the treated animals. Importantly, the dual CRISPR therapy demonstrated statistically significant improvements in HIV-1 cure percentages compared to single treatments. Taken together, these observations underscore a pivotal role of combinatorial CRISPR gene editing in achieving the elimination of HIV-1 infection.


Subject(s)
HIV Infections , HIV Seropositivity , Mice , Animals , Humans , Anti-Retroviral Agents/therapeutic use , Gene Editing , Proviruses/genetics , Receptors, CCR5
18.
Proc Natl Acad Sci U S A ; 120(1): e2217883120, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36574685

ABSTRACT

Antibody heavy chain (HC) and light chain (LC) variable region exons are assembled by V(D)J recombination. V(D)J junctional regions encode complementarity-determining-region 3 (CDR3), an antigen-contact region immensely diversified through nontemplated nucleotide additions ("N-regions") by terminal deoxynucleotidyl transferase (TdT). HIV-1 vaccine strategies seek to elicit human HIV-1 broadly neutralizing antibodies (bnAbs), such as the potent CD4-binding site VRC01-class bnAbs. Mice with primary B cells that express receptors (BCRs) representing bnAb precursors are used as vaccination models. VRC01-class bnAbs uniformly use human HC VH1-2 and commonly use human LCs Vκ3-20 or Vκ1-33 associated with an exceptionally short 5-amino-acid (5-aa) CDR3. Prior VRC01-class models had nonphysiological precursor levels and/or limited precursor diversity. Here, we describe VRC01-class rearranging mice that generate more physiological primary VRC01-class BCR repertoires via rearrangement of VH1-2, as well as Vκ1-33 and/or Vκ3-20 in association with diverse CDR3s. Human-like TdT expression in mouse precursor B cells increased LC CDR3 length and diversity and also promoted the generation of shorter LC CDR3s via N-region suppression of dominant microhomology-mediated Vκ-to-Jκ joins. Priming immunization with eOD-GT8 60mer, which strongly engages VRC01 precursors, induced robust VRC01-class germinal center B cell responses. Vκ3-20-based responses were enhanced by N-region addition, which generates Vκ3-20-to-Jκ junctional sequence combinations that encode VRC01-class 5-aa CDR3s with a critical E residue. VRC01-class-rearranging models should facilitate further evaluation of VRC01-class prime and boost immunogens. These new VRC01-class mouse models establish a prototype for the generation of vaccine-testing mouse models for other HIV-1 bnAb lineages that employ different HC or LC Vs.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Vaccines , Mice , Humans , Animals , Broadly Neutralizing Antibodies , Antibodies, Neutralizing , HIV-1/genetics , HIV Antibodies , DNA Nucleotidylexotransferase , Complementarity Determining Regions/genetics , HIV Infections/prevention & control
19.
J Biol Chem ; 300(6): 107340, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705390

ABSTRACT

Triclosan (TCS) is an antimicrobial toxicant found in a myriad of consumer products and has been detected in human tissues, including breastmilk. We have evaluated the impact of lactational TCS on UDP-glucuronosyltransferase 1A1 (UGT1A1) expression and bilirubin metabolism in humanized UGT1 (hUGT1) neonatal mice. In hUGT1 mice, expression of the hepatic UGT1A1 gene is developmentally delayed resulting in elevated total serum bilirubin (TSB) levels. We found that newborn hUGT1 mice breastfed or orally treated with TCS presented lower TSB levels along with induction of hepatic UGT1A1. Lactational and oral treatment by gavage with TCS leads to the activation of hepatic nuclear receptors constitutive androstane receptor (CAR), peroxisome proliferator-activated receptor alpha (PPARα), and stress sensor, activating transcription factor 4 (ATF4). When CAR-deficient hUGT1 mice (hUGT1/Car-/-) were treated with TCS, TSB levels were reduced with a robust induction of hepatic UGT1A1, leaving us to conclude that CAR is not tied to UGT1A1 induction. Alternatively, when PPARα-deficient hUGT1 mice (hUGT1/Pparα-/-) were treated with TCS, hepatic UGT1A1 was not induced. Additionally, we had previously demonstrated that TCS is a potent inducer of ATF4, a transcriptional factor linked to the integrated stress response. When ATF4 was deleted in liver of hUGT1 mice (hUGT1/Atf4ΔHep) and these mice treated with TCS, we observed superinduction of hepatic UGT1A1. Oxidative stress genes in livers of hUGT1/Atf4ΔHep treated with TCS were increased, suggesting that ATF4 protects liver from excessive oxidative stress. The increase oxidative stress may be associated with superinduction of UGT1A1. The expression of ATF4 in neonatal hUGT1 hepatic tissue may play a role in the developmental repression of UGT1A1.


Subject(s)
Activating Transcription Factor 4 , Animals, Newborn , Bilirubin , Glucuronosyltransferase , Liver , PPAR alpha , Triclosan , Animals , Glucuronosyltransferase/metabolism , Glucuronosyltransferase/genetics , PPAR alpha/metabolism , PPAR alpha/genetics , Mice , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Triclosan/pharmacology , Humans , Bilirubin/pharmacology , Bilirubin/metabolism , Liver/metabolism , Liver/drug effects , Mice, Knockout , Female , Constitutive Androstane Receptor , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics
20.
EMBO J ; 40(12): e107346, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33934394

ABSTRACT

Mutations in the shelterin protein POT1 are associated with chronic lymphocytic leukemia (CLL), Hodgkin lymphoma, angiosarcoma, melanoma, and other cancers. These cancer-associated POT1 (caPOT1) mutations are generally heterozygous, missense, or nonsense mutations occurring throughout the POT1 reading frame. Cancers with caPOT1 mutations have elongated telomeres and show increased genomic instability, but which of the two phenotypes promotes tumorigenesis is unclear. We tested the effects of CAS9-engineered caPOT1 mutations in human embryonic and hematopoietic stem cells (hESCs and HSCs, respectively). HSCs with caPOT1 mutations did not show overt telomere damage. In vitro and in vivo competition experiments showed the caPOT1 mutations did not confer a selective disadvantage. Since DNA damage signaling is known to affect the fitness of HSCs, the data argue that caPOT1 mutations do not cause significant telomere damage. Furthermore, hESC lines with caPOT1 mutations showed no detectable telomere damage response while showing consistent telomere elongation. Thus, caPOT1 mutations are likely selected for during cancer progression because of their ability to elongate telomeres and extend the proliferative capacity of the incipient cancer cells.


Subject(s)
Neoplasms/genetics , Telomere-Binding Proteins/genetics , Telomere , Animals , DNA Damage , Female , Humans , K562 Cells , Male , Mice , Mutation , Shelterin Complex , Stem Cells
SELECTION OF CITATIONS
SEARCH DETAIL