Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 412.607
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 38: 49-77, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340580

ABSTRACT

Mast cells have existed long before the development of adaptive immunity, although they have been given different names. Thus, in the marine urochordate Styela plicata, they have been designated as test cells. However, based on their morphological characteristics (including prominent cytoplasmic granules) and mediator content (including heparin, histamine, and neutral proteases), test cells are thought to represent members of the lineage known in vertebrates as mast cells. So this lineage presumably had important functions that preceded the development of antibodies, including IgE. Yet mast cells are best known, in humans, as key sources of mediators responsible for acute allergic reactions, notably including anaphylaxis, a severe and potentially fatal IgE-dependent immediate hypersensitivity reaction to apparently harmless antigens, including many found in foods and medicines. In this review, we briefly describe the origins of tissue mast cells and outline evidence that these cells can have beneficial as well as detrimental functions, both innately and as participants in adaptive immune responses. We also discuss aspects of mast cell heterogeneity and comment on how the plasticity of this lineage may provide insight into its roles in health and disease. Finally, we consider some currently open questions that are yet unresolved.


Subject(s)
Disease Susceptibility , Inflammation/etiology , Inflammation/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Adaptive Immunity , Animals , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Humans , Immunity, Innate , Inflammation/diagnosis , Inflammation Mediators/metabolism , Signal Transduction
2.
Annu Rev Immunol ; 37: 1-17, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30379594

ABSTRACT

Each of us is a story. Mine is a story of doing science for 60 years, and I am honored to be asked to tell it. Even though this autobiography was written for the Annual Review of Immunology, I have chosen to describe my whole career in science because the segment that was immunology is so intertwined with all else I was doing. This article is an elongation and modification of a talk I gave at my 80th birthday celebration at Caltech on March 23, 2018.


Subject(s)
Allergy and Immunology/history , NF-kappa B/metabolism , RNA Viruses/physiology , Virus Diseases/immunology , Animals , Disease Models, Animal , Gene Rearrangement , History, 20th Century , History, 21st Century , Humans , Mice , Protein-Tyrosine Kinases/metabolism , Reverse Transcription , United States
3.
Annu Rev Immunol ; 36: 755-781, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677472

ABSTRACT

Inflammatory bowel disease (IBD) defines a spectrum of complex disorders. Understanding how environmental risk factors, alterations of the intestinal microbiota, and polygenetic and epigenetic susceptibility impact on immune pathways is key for developing targeted therapies. Mechanistic understanding of polygenic IBD is complemented by Mendelian disorders that present with IBD, pharmacological interventions that cause colitis, autoimmunity, and multiple animal models. Collectively, this multifactorial pathogenesis supports a concept of immune checkpoints that control microbial-host interactions in the gut by modulating innate and adaptive immunity, as well as epithelial and mesenchymal cell responses. In addition to classical immunosuppressive strategies, we discuss how resetting the microbiota and restoring innate immune responses, in particular autophagy and epithelial barrier function, might be key for maintaining remission or preventing IBD. Targeting checkpoints in genetically stratified subgroups of patients with Mendelian disorder-associated IBD increasingly directs treatment strategies as part of personalized medicine.


Subject(s)
Disease Susceptibility/immunology , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/therapy , Animals , Biomarkers , Chronic Disease , Disease Management , Disease Models, Animal , Drug-Related Side Effects and Adverse Reactions , Dysbiosis , Gastrointestinal Microbiome , Genetic Predisposition to Disease , Humans , Inflammatory Bowel Diseases/prevention & control , Molecular Targeted Therapy , Translational Research, Biomedical
4.
Annu Rev Immunol ; 35: 53-84, 2017 04 26.
Article in English | MEDLINE | ID: mdl-27912316

ABSTRACT

Helper T (Th) cell subsets direct immune responses by producing signature cytokines. Th2 cells produce IL-4, IL-5, and IL-13, which are important in humoral immunity and protection from helminth infection and are central to the pathogenesis of many allergic inflammatory diseases. Molecular analysis of Th2 cell differentiation and maintenance of function has led to recent discoveries that have refined our understanding of Th2 cell biology. Epigenetic regulation of Gata3 expression by chromatin remodeling complexes such as Polycomb and Trithorax is crucial for maintaining Th2 cell identity. In the context of allergic diseases, memory-type pathogenic Th2 cells have been identified in both mice and humans. To better understand these disease-driving cell populations, we have developed a model called the pathogenic Th population disease induction model. The concept of defined subsets of pathogenic Th cells may spur new, effective strategies for treating intractable chronic inflammatory disorders.


Subject(s)
Helminthiasis/immunology , Hypersensitivity/immunology , Th2 Cells/immunology , Animals , Cell Differentiation , Disease Models, Animal , Epigenesis, Genetic , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunity, Humoral , Immunologic Memory , Interleukin-13/metabolism , Interleukin-4/metabolism , Interleukin-5/metabolism , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism
5.
Cell ; 187(16): 4129-4143, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39067442

ABSTRACT

Obesity causes significant morbidity and mortality globally. Research in the last three decades has delivered a step-change in our understanding of the fundamental mechanisms that regulate energy homeostasis, building on foundational discoveries in mouse models of metabolic disease. However, not all findings made in rodents have translated to humans, hampering drug discovery in this field. Here, we review how studies in mice and humans have informed our current framework for understanding energy homeostasis, discuss their challenges and limitations, and offer a perspective on how human studies may play an increasingly important role in the discovery of disease mechanisms and identification of therapeutic targets in the future.


Subject(s)
Disease Models, Animal , Metabolic Diseases , Translational Research, Biomedical , Animals , Humans , Mice , Metabolic Diseases/metabolism , Energy Metabolism , Homeostasis , Obesity/metabolism
6.
Cell ; 187(2): 428-445.e20, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38086389

ABSTRACT

A recent case report described an individual who was a homozygous carrier of the APOE3 Christchurch (APOE3ch) mutation and resistant to autosomal dominant Alzheimer's Disease (AD) caused by a PSEN1-E280A mutation. Whether APOE3ch contributed to the protective effect remains unclear. We generated a humanized APOE3ch knock-in mouse and crossed it to an amyloid-ß (Aß) plaque-depositing model. We injected AD-tau brain extract to investigate tau seeding and spreading in the presence or absence of amyloid. Similar to the case report, APOE3ch expression resulted in peripheral dyslipidemia and a marked reduction in plaque-associated tau pathology. Additionally, we observed decreased amyloid response and enhanced microglial response around plaques. We also demonstrate increased myeloid cell phagocytosis and degradation of tau aggregates linked to weaker APOE3ch binding to heparin sulfate proteoglycans. APOE3ch influences the microglial response to Aß plaques, which suppresses Aß-induced tau seeding and spreading. The results reveal new possibilities to target Aß-induced tauopathy.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Apolipoprotein E3 , tau Proteins , Animals , Humans , Mice , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloidogenic Proteins/metabolism , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Brain/metabolism , Disease Models, Animal , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Case Reports as Topic
7.
Cell ; 187(15): 4043-4060.e30, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38878778

ABSTRACT

Inflammation-induced neurodegeneration is a defining feature of multiple sclerosis (MS), yet the underlying mechanisms remain unclear. By dissecting the neuronal inflammatory stress response, we discovered that neurons in MS and its mouse model induce the stimulator of interferon genes (STING). However, activation of neuronal STING requires its detachment from the stromal interaction molecule 1 (STIM1), a process triggered by glutamate excitotoxicity. This detachment initiates non-canonical STING signaling, which leads to autophagic degradation of glutathione peroxidase 4 (GPX4), essential for neuronal redox homeostasis and thereby inducing ferroptosis. Both genetic and pharmacological interventions that target STING in neurons protect against inflammation-induced neurodegeneration. Our findings position STING as a central regulator of the detrimental neuronal inflammatory stress response, integrating inflammation with glutamate signaling to cause neuronal cell death, and present it as a tractable target for treating neurodegeneration in MS.


Subject(s)
Inflammation , Membrane Proteins , Multiple Sclerosis , Neurons , Animals , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Membrane Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Mice , Humans , Inflammation/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Signal Transduction , Autophagy , Mice, Inbred C57BL , Glutamic Acid/metabolism , Ferroptosis , Disease Models, Animal , Female , Male
8.
Cell ; 187(14): 3671-3689.e23, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38866017

ABSTRACT

Ongoing, early-stage clinical trials illustrate the translational potential of human pluripotent stem cell (hPSC)-based cell therapies in Parkinson's disease (PD). However, an unresolved challenge is the extensive cell death following transplantation. Here, we performed a pooled CRISPR-Cas9 screen to enhance postmitotic dopamine neuron survival in vivo. We identified p53-mediated apoptotic cell death as a major contributor to dopamine neuron loss and uncovered a causal link of tumor necrosis factor alpha (TNF-α)-nuclear factor κB (NF-κB) signaling in limiting cell survival. As a translationally relevant strategy to purify postmitotic dopamine neurons, we identified cell surface markers that enable purification without the need for genetic reporters. Combining cell sorting and treatment with adalimumab, a clinically approved TNF-α inhibitor, enabled efficient engraftment of postmitotic dopamine neurons with extensive reinnervation and functional recovery in a preclinical PD mouse model. Thus, transient TNF-α inhibition presents a clinically relevant strategy to enhance survival and enable engraftment of postmitotic hPSC-derived dopamine neurons in PD.


Subject(s)
Cell Survival , Dopaminergic Neurons , NF-kappa B , Tumor Necrosis Factor-alpha , Tumor Suppressor Protein p53 , Dopaminergic Neurons/metabolism , Animals , Humans , NF-kappa B/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Necrosis Factor-alpha/metabolism , Mice , Cell Survival/drug effects , Signal Transduction , Parkinson Disease/metabolism , Pluripotent Stem Cells/metabolism , Apoptosis , Disease Models, Animal , CRISPR-Cas Systems
9.
Cell ; 186(8): 1792-1813, 2023 04 13.
Article in English | MEDLINE | ID: mdl-37059072

ABSTRACT

Despite many advances, metastatic disease remains essentially uncurable. Thus, there is an urgent need to better understand mechanisms that promote metastasis, drive tumor evolution, and underlie innate and acquired drug resistance. Sophisticated preclinical models that recapitulate the complex tumor ecosystem are key to this process. We begin with syngeneic and patient-derived mouse models that are the backbone of most preclinical studies. Second, we present some unique advantages of fish and fly models. Third, we consider the strengths of 3D culture models for resolving remaining knowledge gaps. Finally, we provide vignettes on multiplexed technologies to advance our understanding of metastatic disease.


Subject(s)
Drug Discovery , Neoplasms , Animals , Mice , Disease Models, Animal , Neoplasms/drug therapy
10.
Cell ; 186(23): 5084-5097.e18, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37918394

ABSTRACT

Anti-NMDA receptor (NMDAR) autoantibodies cause NMDAR encephalitis, the most common autoimmune encephalitis, leading to psychosis, seizures, and autonomic dysfunction. Current treatments comprise broad immunosuppression or non-selective antibody removal. We developed NMDAR-specific chimeric autoantibody receptor (NMDAR-CAAR) T cells to selectively eliminate anti-NMDAR B cells and disease-causing autoantibodies. NMDAR-CAARs consist of an extracellular multi-subunit NMDAR autoantigen fused to intracellular 4-1BB/CD3ζ domains. NMDAR-CAAR T cells recognize a large panel of human patient-derived autoantibodies, release effector molecules, proliferate, and selectively kill antigen-specific target cell lines even in the presence of high autoantibody concentrations. In a passive transfer mouse model, NMDAR-CAAR T cells led to depletion of an anti-NMDAR B cell line and sustained reduction of autoantibody levels without notable off-target toxicity. Treatment of patients may reduce side effects, prevent relapses, and improve long-term prognosis. Our preclinical work paves the way for CAAR T cell phase I/II trials in NMDAR encephalitis and further autoantibody-mediated diseases.


Subject(s)
Autoantibodies , Encephalitis , T-Lymphocytes , Animals , Humans , Mice , Autoantibodies/metabolism , Encephalitis/metabolism , Encephalitis/therapy , Receptors, N-Methyl-D-Aspartate , Autoimmune Diseases , Disease Models, Animal
11.
Cell ; 186(23): 5183-5199.e22, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37852258

ABSTRACT

Cellular lineage histories and their molecular states encode fundamental principles of tissue development and homeostasis. Current lineage-recording mouse models have insufficient barcode diversity and single-cell lineage coverage for profiling tissues composed of millions of cells. Here, we developed DARLIN, an inducible Cas9 barcoding mouse line that utilizes terminal deoxynucleotidyl transferase (TdT) and 30 CRISPR target sites. DARLIN is inducible, generates massive lineage barcodes across tissues, and enables the detection of edited barcodes in ∼70% of profiled single cells. Using DARLIN, we examined fate bias within developing hematopoietic stem cells (HSCs) and revealed unique features of HSC migration. Additionally, we established a protocol for joint transcriptomic and epigenomic single-cell measurements with DARLIN and found that cellular clonal memory is associated with genome-wide DNA methylation rather than gene expression or chromatin accessibility. DARLIN will enable the high-resolution study of lineage relationships and their molecular signatures in diverse tissues and physiological contexts.


Subject(s)
Epigenomics , Transcriptome , Animals , Mice , Transcriptome/genetics , Cell Lineage/genetics , Gene Expression Profiling , Disease Models, Animal , DNA
12.
Cell ; 186(4): 786-802.e28, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36754049

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that results from many diverse genetic causes. Although therapeutics specifically targeting known causal mutations may rescue individual types of ALS, these approaches cannot treat most cases since they have unknown genetic etiology. Thus, there is a pressing need for therapeutic strategies that rescue multiple forms of ALS. Here, we show that pharmacological inhibition of PIKFYVE kinase activates an unconventional protein clearance mechanism involving exocytosis of aggregation-prone proteins. Reducing PIKFYVE activity ameliorates ALS pathology and extends survival of animal models and patient-derived motor neurons representing diverse forms of ALS including C9ORF72, TARDBP, FUS, and sporadic. These findings highlight a potential approach for mitigating ALS pathogenesis that does not require stimulating macroautophagy or the ubiquitin-proteosome system.


Subject(s)
Amyotrophic Lateral Sclerosis , Phosphatidylinositol 3-Kinases , Animals , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Motor Neurons , Mutation , RNA-Binding Protein FUS/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal
13.
Cell ; 186(24): 5394-5410.e18, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37922901

ABSTRACT

Parkinson's disease (PD) is a debilitating neurodegenerative disorder. Its symptoms are typically treated with levodopa or dopamine receptor agonists, but its action lacks specificity due to the wide distribution of dopamine receptors in the central nervous system and periphery. Here, we report the development of a gene therapy strategy to selectively manipulate PD-affected circuitry. Targeting striatal D1 medium spiny neurons (MSNs), whose activity is chronically suppressed in PD, we engineered a therapeutic strategy comprised of a highly efficient retrograde adeno-associated virus (AAV), promoter elements with strong D1-MSN activity, and a chemogenetic effector to enable precise D1-MSN activation after systemic ligand administration. Application of this therapeutic approach rescues locomotion, tremor, and motor skill defects in both mouse and primate models of PD, supporting the feasibility of targeted circuit modulation tools for the treatment of PD in humans.


Subject(s)
Genetic Therapy , Parkinson Disease , Animals , Humans , Mice , Corpus Striatum/metabolism , Levodopa/therapeutic use , Levodopa/genetics , Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/therapy , Primates , Receptors, Dopamine D1/metabolism , Disease Models, Animal
14.
Cell ; 186(18): 3882-3902.e24, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37597510

ABSTRACT

Inflammation can trigger lasting phenotypes in immune and non-immune cells. Whether and how human infections and associated inflammation can form innate immune memory in hematopoietic stem and progenitor cells (HSPC) has remained unclear. We found that circulating HSPC, enriched from peripheral blood, captured the diversity of bone marrow HSPC, enabling investigation of their epigenomic reprogramming following coronavirus disease 2019 (COVID-19). Alterations in innate immune phenotypes and epigenetic programs of HSPC persisted for months to 1 year following severe COVID-19 and were associated with distinct transcription factor (TF) activities, altered regulation of inflammatory programs, and durable increases in myelopoiesis. HSPC epigenomic alterations were conveyed, through differentiation, to progeny innate immune cells. Early activity of IL-6 contributed to these persistent phenotypes in human COVID-19 and a mouse coronavirus infection model. Epigenetic reprogramming of HSPC may underlie altered immune function following infection and be broadly relevant, especially for millions of COVID-19 survivors.


Subject(s)
COVID-19 , Epigenetic Memory , Post-Acute COVID-19 Syndrome , Animals , Humans , Mice , Cell Differentiation , COVID-19/immunology , Disease Models, Animal , Hematopoietic Stem Cells , Inflammation/genetics , Trained Immunity , Monocytes/immunology , Post-Acute COVID-19 Syndrome/genetics , Post-Acute COVID-19 Syndrome/immunology , Post-Acute COVID-19 Syndrome/pathology
15.
Cell ; 186(10): 2062-2077.e17, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37075755

ABSTRACT

Entry of enveloped viruses into cells is mediated by viral fusogenic proteins that drive membrane rearrangements needed for fusion between viral and target membranes. Skeletal muscle development also requires membrane fusion events between progenitor cells to form multinucleated myofibers. Myomaker and Myomerger are muscle-specific cell fusogens but do not structurally or functionally resemble classical viral fusogens. We asked whether the muscle fusogens could functionally substitute for viral fusogens, despite their structural distinctiveness, and fuse viruses to cells. We report that engineering of Myomaker and Myomerger on the membrane of enveloped viruses leads to specific transduction of skeletal muscle. We also demonstrate that locally and systemically injected virions pseudotyped with the muscle fusogens can deliver µDystrophin to skeletal muscle of a mouse model of Duchenne muscular dystrophy and alleviate pathology. Through harnessing the intrinsic properties of myogenic membranes, we establish a platform for delivery of therapeutic material to skeletal muscle.


Subject(s)
Bioengineering , Lentivirus , Membrane Proteins , Muscle, Skeletal , Muscular Dystrophy, Duchenne , Animals , Mice , Cell Fusion , Membrane Fusion , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle Development , Muscle, Skeletal/metabolism , Muscle, Skeletal/virology , Bioengineering/methods , Muscular Dystrophy, Duchenne/therapy , Disease Models, Animal , Viral Tropism , Lentivirus/genetics
16.
Annu Rev Immunol ; 33: 823-74, 2015.
Article in English | MEDLINE | ID: mdl-25706096

ABSTRACT

Patients with autoinflammatory diseases present with noninfectious fever flares and systemic and/or disease-specific organ inflammation. Their excessive proinflammatory cytokine and chemokine responses can be life threatening and lead to organ damage over time. Studying such patients has revealed genetic defects that have helped unravel key innate immune pathways, including excessive IL-1 signaling, constitutive NF-κB activation, and, more recently, chronic type I IFN signaling. Discoveries of monogenic defects that lead to activation of proinflammatory cytokines have inspired the use of anticytokine-directed treatment approaches that have been life changing for many patients and have led to the approval of IL-1-blocking agents for a number of autoinflammatory conditions. In this review, we describe the genetically characterized autoinflammatory diseases, we summarize our understanding of the molecular pathways that drive clinical phenotypes and that continue to inspire the search for novel treatment targets, and we provide a conceptual framework for classification.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Genetic Predisposition to Disease , Inflammation/genetics , Inflammation/immunology , Animals , Autoimmune Diseases/metabolism , Autoimmunity , Disease Models, Animal , Humans , Immunity, Innate , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Inflammation/metabolism , Inflammation Mediators/metabolism , Interferons/metabolism , Interleukin-1/metabolism , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , NF-kappa B/metabolism , Signal Transduction
17.
Cell ; 185(22): 4135-4152.e22, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36257314

ABSTRACT

Recent studies have begun to reveal critical roles for the brain's professional phagocytes, microglia, and their receptors in the control of neurotoxic amyloid beta (Aß) and myelin debris accumulation in neurodegenerative disease. However, the critical intracellular molecules that orchestrate neuroprotective functions of microglia remain poorly understood. In our studies, we find that targeted deletion of SYK in microglia leads to exacerbated Aß deposition, aggravated neuropathology, and cognitive defects in the 5xFAD mouse model of Alzheimer's disease (AD). Disruption of SYK signaling in this AD model was further shown to impede the development of disease-associated microglia (DAM), alter AKT/GSK3ß-signaling, and restrict Aß phagocytosis by microglia. Conversely, receptor-mediated activation of SYK limits Aß load. We also found that SYK critically regulates microglial phagocytosis and DAM acquisition in demyelinating disease. Collectively, these results broaden our understanding of the key innate immune signaling molecules that instruct beneficial microglial functions in response to neurotoxic material.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Animals , Mice , Alzheimer Disease/pathology , Amyloid beta-Peptides , Disease Models, Animal , Mice, Transgenic , Microglia/pathology , Phagocytosis
18.
Cell ; 185(8): 1261-1265, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35385685

ABSTRACT

Through studies in mice and in humans, Stuart Orkin showed that GATA-1 is a master transcriptional regulator of hematopoiesis. He has highlighted the role of BCL11A in the fetal-adult hemoglobin switch. The Gairdner Foundation Award recognizes Orkin's contribution to the development of gene therapy of sickle cell disease.


Subject(s)
Anemia, Sickle Cell , Awards and Prizes , Genetic Therapy , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Animals , Disease Models, Animal , Fetal Hemoglobin/genetics , Hematopoiesis/genetics , Humans , Mice , Repressor Proteins/genetics
19.
Cell ; 185(21): 3877-3895.e21, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36152627

ABSTRACT

Williams-Beuren syndrome (WBS) is a rare disorder caused by hemizygous microdeletion of ∼27 contiguous genes. Despite neurodevelopmental and cognitive deficits, individuals with WBS have spared or enhanced musical and auditory abilities, potentially offering an insight into the genetic basis of auditory perception. Here, we report that the mouse models of WBS have innately enhanced frequency-discrimination acuity and improved frequency coding in the auditory cortex (ACx). Chemogenetic rescue showed frequency-discrimination hyperacuity is caused by hyperexcitable interneurons in the ACx. Haploinsufficiency of one WBS gene, Gtf2ird1, replicated WBS phenotypes by downregulating the neuropeptide receptor VIPR1. VIPR1 is reduced in the ACx of individuals with WBS and in the cerebral organoids derived from human induced pluripotent stem cells with the WBS microdeletion. Vipr1 deletion or overexpression in ACx interneurons mimicked or reversed, respectively, the cellular and behavioral phenotypes of WBS mice. Thus, the Gtf2ird1-Vipr1 mechanism in ACx interneurons may underlie the superior auditory acuity in WBS.


Subject(s)
Auditory Cortex/physiology , Williams Syndrome/physiopathology , Animals , Auditory Cortex/cytology , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells , Interneurons/cytology , Interneurons/physiology , Mice , Phenotype , Trans-Activators/genetics , Williams Syndrome/genetics
20.
Cell ; 185(11): 1943-1959.e21, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35545089

ABSTRACT

Parthanatos-associated apoptosis-inducing factor (AIF) nuclease (PAAN), also known as macrophage migration inhibitor factor (MIF), is a member of the PD-D/E(X)K nucleases that acts as a final executioner in parthanatos. PAAN's role in Parkinson's disease (PD) and whether it is amenable to chemical inhibition is not known. Here, we show that neurodegeneration induced by pathologic α-synuclein (α-syn) occurs via PAAN/MIF nuclease activity. Genetic depletion of PAAN/MIF and a mutant lacking nuclease activity prevent the loss of dopaminergic neurons and behavioral deficits in the α-syn preformed fibril (PFF) mouse model of sporadic PD. Compound screening led to the identification of PAANIB-1, a brain-penetrant PAAN/MIF nuclease inhibitor that prevents neurodegeneration induced by α-syn PFF, AAV-α-syn overexpression, or MPTP intoxication in vivo. Our findings could have broad relevance in human pathologies where parthanatos plays a role in the development of cell death inhibitors targeting the druggable PAAN/MIF nuclease.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Parkinson Disease , Animals , Brain/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Endonucleases/metabolism , Mice , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL