Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 16(753): eadl3758, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38924428

ABSTRACT

Vitamin B12 is critical for hematopoiesis and myelination. Deficiency can cause neurologic deficits including loss of coordination and cognitive decline. However, diagnosis relies on measurement of vitamin B12 in the blood, which may not accurately reflect the concentration in the brain. Using programmable phage display, we identified an autoantibody targeting the transcobalamin receptor (CD320) in a patient with progressive tremor, ataxia, and scanning speech. Anti-CD320 impaired cellular uptake of cobalamin (B12) in vitro by depleting its target from the cell surface. Despite a normal serum concentration, B12 was nearly undetectable in her cerebrospinal fluid (CSF). Immunosuppressive treatment and high-dose systemic B12 supplementation were associated with increased B12 in the CSF and clinical improvement. Optofluidic screening enabled isolation of a patient-derived monoclonal antibody that impaired B12 transport across an in vitro model of the blood-brain barrier (BBB). Autoantibodies targeting the same epitope of CD320 were identified in seven other patients with neurologic deficits of unknown etiology, 6% of healthy controls, and 21.4% of a cohort of patients with neuropsychiatric lupus. In 132 paired serum and CSF samples, detection of anti-CD320 in the blood predicted B12 deficiency in the brain. However, these individuals did not display any hematologic signs of B12 deficiency despite systemic CD320 impairment. Using a genome-wide CRISPR screen, we found that the low-density lipoprotein receptor serves as an alternative B12 uptake pathway in hematopoietic cells. These findings dissect the tissue specificity of B12 transport and elucidate an autoimmune neurologic condition that may be amenable to immunomodulatory treatment and nutritional supplementation.


Subject(s)
Autoantibodies , Vitamin B 12 Deficiency , Vitamin B 12 , Humans , Vitamin B 12 Deficiency/immunology , Vitamin B 12/blood , Autoantibodies/blood , Autoantibodies/immunology , Female , Receptors, Cell Surface/metabolism , Antigens, CD/metabolism , Middle Aged , Autoimmune Diseases/immunology , Autoimmune Diseases/blood , Blood-Brain Barrier/metabolism , Male
2.
Proc Natl Acad Sci U S A ; 120(11): e2215376120, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36897988

ABSTRACT

The Siglecs (sialic acid-binding immunoglobulin-like lectins) are glycoimmune checkpoint receptors that suppress immune cell activation upon engagement of cognate sialoglycan ligands. The cellular drivers underlying Siglec ligand production on cancer cells are poorly understood. We find the MYC oncogene causally regulates Siglec ligand production to enable tumor immune evasion. A combination of glycomics and RNA-sequencing of mouse tumors revealed the MYC oncogene controls expression of the sialyltransferase St6galnac4 and induces a glycan known as disialyl-T. Using in vivo models and primary human leukemias, we find that disialyl-T functions as a "don't eat me" signal by engaging macrophage Siglec-E in mice or the human ortholog Siglec-7, thereby preventing cancer cell clearance. Combined high expression of MYC and ST6GALNAC4 identifies patients with high-risk cancers and reduced tumor myeloid infiltration. MYC therefore regulates glycosylation to enable tumor immune evasion. We conclude that disialyl-T is a glycoimmune checkpoint ligand. Thus, disialyl-T is a candidate for antibody-based checkpoint blockade, and the disialyl-T synthase ST6GALNAC4 is a potential enzyme target for small molecule-mediated immune therapy.


Subject(s)
Neoplasms , Proto-Oncogene Proteins c-myc , Sialic Acid Binding Immunoglobulin-like Lectins , Animals , Humans , Mice , Antigens, CD/metabolism , Ligands , Macrophages/metabolism , Neoplasms/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Proto-Oncogene Proteins c-myc/metabolism
3.
Sci Transl Med ; 13(622): eabg2919, 2021 12.
Article in English | MEDLINE | ID: mdl-34851695

ABSTRACT

Lysosome dysfunction is a shared feature of rare lysosomal storage diseases and common age-related neurodegenerative diseases. Microglia, the brain-resident macrophages, are particularly vulnerable to lysosome dysfunction because of the phagocytic stress of clearing dying neurons, myelin, and debris. CD22 is a negative regulator of microglial homeostasis in the aging mouse brain, and soluble CD22 (sCD22) is increased in the cerebrospinal fluid of patients with Niemann-Pick type C disease (NPC). However, the role of CD22 in the human brain remains unknown. In contrast to previous findings in mice, here, we show that CD22 is expressed by oligodendrocytes in the human brain and binds to sialic acid­dependent ligands on microglia. Using unbiased genetic and proteomic screens, we identify insulin-like growth factor 2 receptor (IGF2R) as the binding partner of sCD22 on human myeloid cells. Targeted truncation of IGF2R revealed that sCD22 docks near critical mannose 6-phosphate­binding domains, where it disrupts lysosomal protein trafficking. Interfering with the sCD22-IGF2R interaction using CD22 blocking antibodies ameliorated lysosome dysfunction in human NPC1 mutant induced pluripotent stem cell­derived microglia-like cells without harming oligodendrocytes in vitro. These findings reinforce the differences between mouse and human microglia and provide a candidate microglia-directed immunotherapeutic to treat NPC.


Subject(s)
Microglia , Niemann-Pick Disease, Type C , Animals , Humans , Lysosomes/metabolism , Macrophages/metabolism , Mice , Microglia/metabolism , Niemann-Pick Disease, Type C/drug therapy , Proteomics , Sialic Acid Binding Ig-like Lectin 2/metabolism , Sialic Acid Binding Ig-like Lectin 2/therapeutic use
5.
Nat Rev Neurosci ; 21(5): 298, 2020 05.
Article in English | MEDLINE | ID: mdl-32203300

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

7.
Nat Rev Neurosci ; 21(2): 93-102, 2020 02.
Article in English | MEDLINE | ID: mdl-31913356

ABSTRACT

A rapidly ageing population and a limited therapeutic toolbox urgently necessitate new approaches to treat neurodegenerative diseases. Brain ageing, the key risk factor for neurodegeneration, involves complex cellular and molecular processes that eventually result in cognitive decline. Although cell-intrinsic defects in neurons and glia may partially explain this decline, cell-extrinsic changes in the systemic environment, mediated by blood, have recently been shown to contribute to brain dysfunction with age. Here, we review the current understanding of how systemic factors mediate brain ageing, how these factors are regulated and how we can translate these findings into therapies for neurodegenerative diseases.


Subject(s)
Aging/physiology , Brain/physiology , Homeostasis , Neurodegenerative Diseases/physiopathology , Aging/immunology , Animals , Brain/immunology , Endothelial Cells/immunology , Endothelial Cells/physiology , Exercise/physiology , Humans , Microbiota/immunology , Microbiota/physiology , Neurodegenerative Diseases/immunology , Neuroglia/immunology , Neuroglia/physiology , Neurons/immunology , Neurons/physiology
8.
Nature ; 577(7790): 399-404, 2020 01.
Article in English | MEDLINE | ID: mdl-31915375

ABSTRACT

Alzheimer's disease is an incurable neurodegenerative disorder in which neuroinflammation has a critical function1. However, little is known about the contribution of the adaptive immune response in Alzheimer's disease2. Here, using integrated analyses of multiple cohorts, we identify peripheral and central adaptive immune changes in Alzheimer's disease. First, we performed mass cytometry of peripheral blood mononuclear cells and discovered an immune signature of Alzheimer's disease that consists of increased numbers of CD8+ T effector memory CD45RA+ (TEMRA) cells. In a second cohort, we found that CD8+ TEMRA cells were negatively associated with cognition. Furthermore, single-cell RNA sequencing revealed that T cell receptor (TCR) signalling was enhanced in these cells. Notably, by using several strategies of single-cell TCR sequencing in a third cohort, we discovered clonally expanded CD8+ TEMRA cells in the cerebrospinal fluid of patients with Alzheimer's disease. Finally, we used machine learning, cloning and peptide screens to demonstrate the specificity of clonally expanded TCRs in the cerebrospinal fluid of patients with Alzheimer's disease to two separate Epstein-Barr virus antigens. These results reveal an adaptive immune response in the blood and cerebrospinal fluid in Alzheimer's disease and provide evidence of clonal, antigen-experienced T cells patrolling the intrathecal space of brains affected by age-related neurodegeneration.


Subject(s)
Alzheimer Disease/immunology , CD8-Positive T-Lymphocytes/immunology , Cerebrospinal Fluid/immunology , Aged , Amino Acid Sequence , Cohort Studies , Humans , Immunologic Memory , Middle Aged , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Sequence Analysis, Protein
9.
Nat Neurosci ; 23(2): 194-208, 2020 02.
Article in English | MEDLINE | ID: mdl-31959936

ABSTRACT

Microglia become progressively activated and seemingly dysfunctional with age, and genetic studies have linked these cells to the pathogenesis of a growing number of neurodegenerative diseases. Here we report a striking buildup of lipid droplets in microglia with aging in mouse and human brains. These cells, which we call 'lipid-droplet-accumulating microglia' (LDAM), are defective in phagocytosis, produce high levels of reactive oxygen species and secrete proinflammatory cytokines. RNA-sequencing analysis of LDAM revealed a transcriptional profile driven by innate inflammation that is distinct from previously reported microglial states. An unbiased CRISPR-Cas9 screen identified genetic modifiers of lipid droplet formation; surprisingly, variants of several of these genes, including progranulin (GRN), are causes of autosomal-dominant forms of human neurodegenerative diseases. We therefore propose that LDAM contribute to age-related and genetic forms of neurodegeneration.


Subject(s)
Aging/pathology , Brain/pathology , Lipids , Microglia/pathology , Animals , Humans , Inflammation/pathology , Mice
10.
Haematologica ; 105(4): 905-913, 2020 04.
Article in English | MEDLINE | ID: mdl-31171641

ABSTRACT

Healthy bone marrow progenitors yield a co-ordinated balance of hematopoietic lineages. This balance shifts with aging toward enhanced granulopoiesis with diminished erythropoiesis and lymphopoiesis, changes which likely contribute to the development of bone marrow disorders in the elderly. In this study, RUNX3 was identified as a hematopoietic stem and progenitor cell factor whose levels decline with aging in humans and mice. This decline is exaggerated in hematopoietic stem and progenitor cells from subjects diagnosed with unexplained anemia of the elderly. Hematopoietic stem cells from elderly unexplained anemia patients had diminished erythroid but unaffected granulocytic colony forming potential. Knockdown studies revealed human hematopoietic stem and progenitor cells to be strongly influenced by RUNX3 levels, with modest deficiencies abrogating erythroid differentiation at multiple steps while retaining capacity for granulopoiesis. Transcriptome profiling indicated control by RUNX3 of key erythroid transcription factors, including KLF1 and GATA1 These findings thus implicate RUNX3 as a participant in hematopoietic stem and progenitor cell aging, and a key determinant of erythroid-myeloid lineage balance.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Aged , Aging , Animals , Cell Differentiation , Core Binding Factor Alpha 3 Subunit/genetics , Erythropoiesis , Humans , Mice
11.
Nature ; 571(7764): 205-210, 2019 07.
Article in English | MEDLINE | ID: mdl-31270459

ABSTRACT

The mammalian brain contains neurogenic niches that comprise neural stem cells and other cell types. Neurogenic niches become less functional with age, but how they change during ageing remains unclear. Here we perform single-cell RNA sequencing of young and old neurogenic niches in mice. The analysis of 14,685 single-cell transcriptomes reveals a decrease in activated neural stem cells, changes in endothelial cells and microglia, and an infiltration of T cells in old neurogenic niches. T cells in old brains are clonally expanded and are generally distinct from those in old blood, which suggests that they may experience specific antigens. T cells in old brains also express interferon-γ, and the subset of neural stem cells that has a high interferon response shows decreased proliferation in vivo. We find that T cells can inhibit the proliferation of neural stem cells in co-cultures and in vivo, in part by secreting interferon-γ. Our study reveals an interaction between T cells and neural stem cells in old brains, opening potential avenues through which to counteract age-related decline in brain function.


Subject(s)
Aging/physiology , Brain/cytology , Cell Movement , Neural Stem Cells/cytology , Neurogenesis , Single-Cell Analysis , Stem Cell Niche/physiology , T-Lymphocytes/cytology , Animals , Blood , Cell Proliferation , Clone Cells/cytology , Coculture Techniques , Endothelial Cells/cytology , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Sequence Analysis, RNA , Signal Transduction , T-Lymphocytes/metabolism , Transcriptome/genetics
12.
Nature ; 568(7751): 187-192, 2019 04.
Article in English | MEDLINE | ID: mdl-30944478

ABSTRACT

Microglia maintain homeostasis in the central nervous system through phagocytic clearance of protein aggregates and cellular debris. This function deteriorates during ageing and neurodegenerative disease, concomitant with cognitive decline. However, the mechanisms of impaired microglial homeostatic function and the cognitive effects of restoring this function remain unknown. We combined CRISPR-Cas9 knockout screens with RNA sequencing analysis to discover age-related genetic modifiers of microglial phagocytosis. These screens identified CD22, a canonical B cell receptor, as a negative regulator of phagocytosis that is upregulated on aged microglia. CD22 mediates the anti-phagocytic effect of α2,6-linked sialic acid, and inhibition of CD22 promotes the clearance of myelin debris, amyloid-ß oligomers and α-synuclein fibrils in vivo. Long-term central nervous system delivery of an antibody that blocks CD22 function reprograms microglia towards a homeostatic transcriptional state and improves cognitive function in aged mice. These findings elucidate a mechanism of age-related microglial impairment and a strategy to restore homeostasis in the ageing brain.


Subject(s)
Aging/physiology , Brain/cytology , Homeostasis/drug effects , Microglia/drug effects , N-Acetylneuraminic Acid/pharmacology , Phagocytosis/drug effects , Sialic Acid Binding Ig-like Lectin 2/antagonists & inhibitors , Aging/drug effects , Aging/genetics , Animals , Brain/drug effects , Brain/physiology , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Cognition/drug effects , Cognition/physiology , Female , Homeostasis/genetics , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , N-Acetylneuraminic Acid/chemistry , Phagocytosis/genetics , Sequence Analysis, RNA , Sialic Acid Binding Ig-like Lectin 2/genetics , Sialic Acid Binding Ig-like Lectin 2/metabolism
13.
Neuron ; 93(3): 468-470, 2017 Feb 08.
Article in English | MEDLINE | ID: mdl-28182900

ABSTRACT

In this issue of Neuron, Tufail et al. present an underlying mechanism for microglia-mediated elimination of virally transduced cells in the central nervous system. These findings could contribute to the development of improved gene therapies for various neurological disorders by exploring why microglia destroy viable cells following viral infection.


Subject(s)
Central Nervous System , Microglia , Gene Transfer Techniques , Genetic Therapy , Humans , Neurons
14.
Proc Natl Acad Sci U S A ; 112(31): 9638-43, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195758

ABSTRACT

Determining the developmental pathway leading to erythrocytes and being able to isolate their progenitors are crucial to understanding and treating disorders of red cell imbalance such as anemia, myelodysplastic syndrome, and polycythemia vera. Here we show that the human erythrocyte progenitor (hEP) can be prospectively isolated from adult bone marrow. We found three subfractions that possessed different expression patterns of CD105 and CD71 within the previously defined human megakaryocyte/erythrocyte progenitor (hMEP; Lineage(-) CD34(+) CD38(+) IL-3Rα(-) CD45RA(-)) population. Both CD71(-) CD105(-) and CD71(+) CD105(-) MEPs, at least in vitro, still retained bipotency for the megakaryocyte (MegK) and erythrocyte (E) lineages, although the latter subpopulation is skewed in differentiation toward the erythroid lineage. Notably, the proliferative and differentiation output of the CD71(intermediate(int)/+) CD105(+) subset of cells within the MEP population was completely restricted to the erythroid lineage with the loss of MegK potential. CD71(+) CD105(-) MEPs are erythrocyte-biased MEPs (E-MEPs) and CD71(int/+) CD105(+) cells are EPs. These previously unclassified populations may facilitate further understanding of the molecular mechanisms governing human erythroid development and serve as potential therapeutic targets in disorders of the erythroid lineage.


Subject(s)
Cell Lineage , Cell Separation/methods , Erythroid Precursor Cells/cytology , Antigens, CD/metabolism , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Fractionation , Cell Lineage/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Erythrocytes/cytology , Erythrocytes/drug effects , Erythroid Precursor Cells/drug effects , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Immunophenotyping , Megakaryocytes/cytology , Megakaryocytes/drug effects , Models, Biological , Transforming Growth Factor beta/pharmacology , Up-Regulation/drug effects
15.
J Vis Exp ; (93): e52056, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25407120

ABSTRACT

Osteoclasts are highly specialized cells that are derived from the monocyte/macrophage lineage of the bone marrow. Their unique ability to resorb both the organic and inorganic matrices of bone means that they play a key role in regulating skeletal remodeling. Together, osteoblasts and osteoclasts are responsible for the dynamic coupling process that involves both bone resorption and bone formation acting together to maintain the normal skeleton during health and disease. As the principal bone-resorbing cell in the body, changes in osteoclast differentiation or function can result in profound effects in the body. Diseases associated with altered osteoclast function can range in severity from lethal neonatal disease due to failure to form a marrow space for hematopoiesis, to more commonly observed pathologies such as osteoporosis, in which excessive osteoclastic bone resorption predisposes to fracture formation. An ability to isolate osteoclasts in high numbers in vitro has allowed for significant advances in the understanding of the bone remodeling cycle and has paved the way for the discovery of novel therapeutic strategies that combat these diseases. Here, we describe a protocol to isolate and cultivate osteoclasts from mouse bone marrow that will yield large numbers of osteoclasts.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Cell Separation/methods , Osteoclasts/cytology , Animals , Macrophages/cytology , Mice , Mice, Inbred C57BL
16.
Proc Natl Acad Sci U S A ; 110(8): 3011-6, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23388639

ABSTRACT

Myelodysplastic syndromes (MDS) are a group of disorders characterized by variable cytopenias and ineffective hematopoiesis. Hematopoietic stem cells (HSCs) and myeloid progenitors in MDS have not been extensively characterized. We transplanted purified human HSCs from MDS samples into immunodeficient mice and show that HSCs are the disease-initiating cells in MDS. We identify a recurrent loss of granulocyte-macrophage progenitors (GMPs) in the bone marrow of low risk MDS patients that can distinguish low risk MDS from clinical mimics, thus providing a simple diagnostic tool. The loss of GMPs is likely due to increased apoptosis and increased phagocytosis, the latter due to the up-regulation of cell surface calreticulin, a prophagocytic marker. Blocking calreticulin on low risk MDS myeloid progenitors rescues them from phagocytosis in vitro. However, in the high-risk refractory anemia with excess blasts (RAEB) stages of MDS, the GMP population is increased in frequency compared with normal, and myeloid progenitors evade phagocytosis due to up-regulation of CD47, an antiphagocytic marker. Blocking CD47 leads to the selective phagocytosis of this population. We propose that MDS HSCs compete with normal HSCs in the patients by increasing their frequency at the expense of normal hematopoiesis, that the loss of MDS myeloid progenitors by programmed cell death and programmed cell removal are, in part, responsible for the cytopenias, and that up-regulation of the "don't eat me" signal CD47 on MDS myeloid progenitors is an important transition step leading from low risk MDS to high risk MDS and, possibly, to acute myeloid leukemia.


Subject(s)
Hematopoietic Stem Cells/pathology , Myelodysplastic Syndromes/pathology , Animals , Antigens, CD/immunology , Hematopoietic Stem Cells/immunology , Humans , In Situ Hybridization, Fluorescence , Mice , Myelodysplastic Syndromes/immunology , Phagocytosis
17.
Blood ; 118(13): 3622-33, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21788341

ABSTRACT

Reduced gene dosage of ribosomal protein subunits has been implicated in 5q- myelodysplastic syndrome and Diamond Blackfan anemia, but the cellular and pathophysiologic defects associated with these conditions are enigmatic. Using conditional inactivation of the ribosomal protein S6 gene in laboratory mice, we found that reduced ribosomal protein gene dosage recapitulates cardinal features of the 5q- syndrome, including macrocytic anemia, erythroid hypoplasia, and megakaryocytic dysplasia with thrombocytosis, and that p53 plays a critical role in manifestation of these phenotypes. The blood cell abnormalities are accompanied by a reduction in the number of HSCs, a specific defect in late erythrocyte development, and suggest a disease-specific ontogenetic pathway for megakaryocyte development. Further studies of highly purified HSCs from healthy patients and from those with myelodysplastic syndrome link reduced expression of ribosomal protein genes to decreased RBC maturation and suggest an underlying and common pathophysiologic pathway for additional subtypes of myelodysplastic syndrome.


Subject(s)
Gene Dosage , Myelodysplastic Syndromes/genetics , Ribosomal Proteins/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Down-Regulation/genetics , Female , Gene Dosage/genetics , Gene Dosage/physiology , Genetic Predisposition to Disease , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/metabolism , Ribosomal Proteins/metabolism , Risk Factors , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...