Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Discov Ment Health ; 3(1): 27, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38036718

ABSTRACT

Schizophrenia is a debilitating condition necessitating more efficacious therapies. Previous studies suggested that schizophrenia development is associated with aberrant synaptic pruning by glial cells. We pursued an interdisciplinary approach to understand whether therapeutic reduction in glial cell-specifically astrocytic-phagocytosis might benefit neuropsychiatric patients. We discovered that beta-2 adrenergic receptor (ADRB2) agonists reduced phagocytosis using a high-throughput, phenotypic screen of over 3200 compounds in primary human fetal astrocytes. We used protein interaction pathways analysis to associate ADRB2, to schizophrenia and endocytosis. We demonstrated that patients with a pediatric exposure to salmeterol, an ADRB2 agonist, had reduced in-patient psychiatry visits using a novel observational study in the electronic health record. We used a mouse model of inflammatory neurodegenerative disease and measured changes in proteins associated with endocytosis and vesicle-mediated transport after ADRB2 agonism. These results provide substantial rationale for clinical consideration of ADRB2 agonists as possible therapies for patients with schizophrenia.

2.
Ecol Evol ; 13(3): e9853, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36911312

ABSTRACT

Wild pigs (Sus scrofa) are invading many areas globally and impacting biodiversity and economies in their non-native range. Thus, wild pigs are often targeted for eradication efforts. Age- and sex-specific body measurements are important for informing these eradication efforts because they reflect body condition, resource availability, and fecundity, which are common indicators of population trajectory. However, body mass is often difficult to collect, especially on large individuals that require specialized equipment or multiple people to weigh. Measurements that can be rapidly taken by a single land or wildlife manager on any size wild pig without aid from specialized equipment would be beneficial if they accurately infer wild pig body mass. Our goals were to assess whether morphometric measurements could accurately predict wild pig body mass, and to provide tools to directly input these measures and estimate wild pig body mass. Using linear models, we quantified the relationship between body mass and morphometric measurements (i.e., body length, chest girth, ear length, eye to snout length, hindfoot length, shoulder length, and tail length) from a subset (n = 102) of wild pigs culled at the Mississippi Alluvial Valley, Mississippi, USA. We evaluated separate models for each individual morphometric measurement. We then used the model coefficients to develop equations to predict wild pig body mass. We validated these equations predicting body mass of 1592 individuals collected across eight areas in Australia, Guam, and the USA for cross-validation. Each developed equation remained accurate when cross-validated across regions. Body length, chest girth, and shoulder length were the morphometrics that best predicted wild pig body mass. Our analyses indicated it is possible to use the presented equations to infer wild pig body mass from simple metrics.

3.
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
4.
Nat Cancer ; 3(8): 976-993, 2022 08.
Article in English | MEDLINE | ID: mdl-35817829

ABSTRACT

Immunotherapy with anti-GD2 antibodies has advanced the treatment of children with high-risk neuroblastoma, but nearly half of patients relapse, and little is known about mechanisms of resistance to anti-GD2 therapy. Here, we show that reduced GD2 expression was significantly correlated with the mesenchymal cell state in neuroblastoma and that a forced adrenergic-to-mesenchymal transition (AMT) conferred downregulation of GD2 and resistance to anti-GD2 antibody. Mechanistically, low-GD2-expressing cell lines demonstrated significantly reduced expression of the ganglioside synthesis enzyme ST8SIA1 (GD3 synthase), resulting in a bottlenecking of GD2 synthesis. Pharmacologic inhibition of EZH2 resulted in epigenetic rewiring of mesenchymal neuroblastoma cells and re-expression of ST8SIA1, restoring surface expression of GD2 and sensitivity to anti-GD2 antibody. These data identify developmental lineage as a key determinant of sensitivity to anti-GD2 based immunotherapies and credential EZH2 inhibitors for clinical testing in combination with anti-GD2 antibody to enhance outcomes for children with neuroblastoma.


Subject(s)
Gangliosides , Neuroblastoma , Antibodies, Monoclonal , Child , Humans , Immunotherapy , Neoplasm Recurrence, Local/chemically induced , Neuroblastoma/drug therapy
5.
Nat Med ; 28(2): 333-344, 2022 02.
Article in English | MEDLINE | ID: mdl-35027753

ABSTRACT

The disialoganglioside GD2 is overexpressed on several solid tumors, and monoclonal antibodies targeting GD2 have substantially improved outcomes for children with high-risk neuroblastoma. However, approximately 40% of patients with neuroblastoma still relapse, and anti-GD2 has not mediated significant clinical activity in any other GD2+ malignancy. Macrophages are important mediators of anti-tumor immunity, but tumors resist macrophage phagocytosis through expression of the checkpoint molecule CD47, a so-called 'Don't eat me' signal. In this study, we establish potent synergy for the combination of anti-GD2 and anti-CD47 in syngeneic and xenograft mouse models of neuroblastoma, where the combination eradicates tumors, as well as osteosarcoma and small-cell lung cancer, where the combination significantly reduces tumor burden and extends survival. This synergy is driven by two GD2-specific factors that reorient the balance of macrophage activity. Ligation of GD2 on tumor cells (a) causes upregulation of surface calreticulin, a pro-phagocytic 'Eat me' signal that primes cells for removal and (b) interrupts the interaction of GD2 with its newly identified ligand, the inhibitory immunoreceptor Siglec-7. This work credentials the combination of anti-GD2 and anti-CD47 for clinical translation and suggests that CD47 blockade will be most efficacious in combination with monoclonal antibodies that alter additional pro- and anti-phagocytic signals within the tumor microenvironment.


Subject(s)
Bone Neoplasms , CD47 Antigen , Animals , Cell Line, Tumor , Humans , Immunotherapy , Mice , Neoplasm Recurrence, Local , Phagocytosis , Tumor Microenvironment
6.
Brain ; 144(8): 2361-2374, 2021 09 04.
Article in English | MEDLINE | ID: mdl-34145876

ABSTRACT

Autoantibodies are a hallmark of numerous neurological disorders, including multiple sclerosis, autoimmune encephalitides and neuromyelitis optica. Whilst well understood in peripheral myeloid cells, the pathophysiological significance of autoantibody-induced Fc receptor signalling in microglia remains unknown, in part due to the lack of a robust in vivo model. Moreover, the application of therapeutic antibodies for neurodegenerative disease also highlights the importance of understanding Fc receptor signalling in microglia. Here, we describe a novel in vivo experimental paradigm that allows for selective engagement of Fc receptors within the CNS by peripherally injecting anti-myelin oligodendrocyte glycoprotein (MOG) monoclonal antibodies into normal wild-type mice. MOG antigen-bound immunoglobulins were detected throughout the CNS and triggered a rapid and tightly regulated proliferative response in both brain and spinal cord microglia. This microglial response was abrogated when anti-MOG antibodies were deprived of Fc receptor effector function or injected into Fcγ receptor knockout mice and was associated with the downregulation of Fc receptors in microglia, but not peripheral myeloid cells, establishing that this response was dependent on central Fc receptor engagement. Downstream of the Fc receptors, BTK was a required signalling node for this response, as microglia proliferation was amplified in BtkE41K knock-in mice expressing a constitutively active form of the enzyme and blunted in mice treated with a CNS-penetrant small molecule inhibitor of BTK. Finally, this response was associated with transient and stringently regulated changes in gene expression predominantly related to cellular proliferation, which markedly differed from transcriptional programs typically associated with Fc receptor engagement in peripheral myeloid cells. Together, these results establish a physiologically-meaningful functional response to Fc receptor and BTK signalling in microglia, while providing a novel in vivo tool to further dissect the roles of microglia-specific Fc receptor and BTK-driven responses to both pathogenic and therapeutic antibodies in CNS homeostasis and disease.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Autoantibodies/immunology , Brain/pathology , Microglia/pathology , Myelin-Oligodendrocyte Glycoprotein/immunology , Receptors, Fc/metabolism , Spinal Cord/pathology , Animals , Brain/immunology , Brain/metabolism , Cell Proliferation/physiology , Mice , Microglia/immunology , Microglia/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism
7.
Cell ; 184(12): 3109-3124.e22, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34004145

ABSTRACT

Glycans modify lipids and proteins to mediate inter- and intramolecular interactions across all domains of life. RNA is not thought to be a major target of glycosylation. Here, we challenge this view with evidence that mammals use RNA as a third scaffold for glycosylation. Using a battery of chemical and biochemical approaches, we found that conserved small noncoding RNAs bear sialylated glycans. These "glycoRNAs" were present in multiple cell types and mammalian species, in cultured cells, and in vivo. GlycoRNA assembly depends on canonical N-glycan biosynthetic machinery and results in structures enriched in sialic acid and fucose. Analysis of living cells revealed that the majority of glycoRNAs were present on the cell surface and can interact with anti-dsRNA antibodies and members of the Siglec receptor family. Collectively, these findings suggest the existence of a direct interface between RNA biology and glycobiology, and an expanded role for RNA in extracellular biology.


Subject(s)
Cell Membrane/metabolism , Polysaccharides/metabolism , RNA/metabolism , Animals , Antibodies/metabolism , Base Sequence , Biosynthetic Pathways , Cell Line , Cell Survival , Humans , Mass Spectrometry , N-Acetylneuraminic Acid/metabolism , Polyadenylation , Polysaccharides/chemistry , RNA/chemistry , RNA/genetics , RNA, Untranslated/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Staining and Labeling
9.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Article in English | MEDLINE | ID: mdl-33495350

ABSTRACT

Glyco-immune checkpoint receptors, molecules that inhibit immune cell activity following binding to glycosylated cell-surface antigens, are emerging as attractive targets for cancer immunotherapy. Defining biologically relevant ligands that bind and activate such receptors, however, has historically been a significant challenge. Here, we present a CRISPRi genomic screening strategy that allowed unbiased identification of the key genes required for cell-surface presentation of glycan ligands on leukemia cells that bind the glyco-immune checkpoint receptors Siglec-7 and Siglec-9. This approach revealed a selective interaction between Siglec-7 and the mucin-type glycoprotein CD43. Further work identified a specific N-terminal glycopeptide region of CD43 containing clusters of disialylated O-glycan tetrasaccharides that form specific Siglec-7 binding motifs. Knockout or blockade of CD43 in leukemia cells relieves Siglec-7-mediated inhibition of immune killing activity. This work identifies a potential target for immune checkpoint blockade therapy and represents a generalizable approach to dissection of glycan-receptor interactions in living cells.


Subject(s)
Antigens, Differentiation, Myelomonocytic/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Genome, Human , Lectins/metabolism , Polysaccharides/metabolism , Amino Acid Motifs , Antigens, Differentiation, Myelomonocytic/chemistry , Cell Line, Tumor , Cell Membrane/metabolism , Glycopeptides/metabolism , Humans , Immunological Synapses/metabolism , Killer Cells, Natural/metabolism , Lectins/chemistry , Leukosialin/chemistry , Leukosialin/metabolism , Ligands , Protein Binding
10.
Nat Rev Drug Discov ; 20(3): 217-243, 2021 03.
Article in English | MEDLINE | ID: mdl-33462432

ABSTRACT

Carbohydrates - namely glycans - decorate every cell in the human body and most secreted proteins. Advances in genomics, glycoproteomics and tools from chemical biology have made glycobiology more tractable and understandable. Dysregulated glycosylation plays a major role in disease processes from immune evasion to cognition, sparking research that aims to target glycans for therapeutic benefit. The field is now poised for a boom in drug development. As a harbinger of this activity, glycobiology has already produced several drugs that have improved human health or are currently being translated to the clinic. Focusing on three areas - selectins, Siglecs and glycan-targeted antibodies - this Review aims to tell the stories behind therapies inspired by glycans and to outline how the lessons learned from these approaches are paving the way for future glycobiology-focused therapeutics.


Subject(s)
Mammals/metabolism , Polysaccharides/metabolism , Selectins/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Animals , Drug Development/methods , Humans
11.
Nat Chem Biol ; 15(10): 949-958, 2019 10.
Article in English | MEDLINE | ID: mdl-31451760

ABSTRACT

Antibody-drug conjugates (ADCs) selectively deliver chemotherapeutic agents to target cells and are important cancer therapeutics. However, the mechanisms by which ADCs are internalized and activated remain unclear. Using CRISPR-Cas9 screens, we uncover many known and novel endolysosomal regulators as modulators of ADC toxicity. We identify and characterize C18ORF8/RMC1 as a regulator of ADC toxicity through its role in endosomal maturation. Through comparative analysis of screens with ADCs bearing different linkers, we show that a subset of late endolysosomal regulators selectively influence toxicity of noncleavable linker ADCs. Surprisingly, we find cleavable valine-citrulline linkers can be processed rapidly after internalization without lysosomal delivery. Lastly, we show that sialic acid depletion enhances ADC lysosomal delivery and killing in diverse cancer cell types, including with FDA (US Food and Drug Administration)-approved trastuzumab emtansine (T-DM1) in Her2-positive breast cancer cells. Together, these results reveal new regulators of endolysosomal trafficking, provide important insights for ADC design and identify candidate combination therapy targets.


Subject(s)
CRISPR-Cas Systems , Genome-Wide Association Study , Immunoconjugates/toxicity , Maytansine/analogs & derivatives , N-Acetylneuraminic Acid/pharmacology , Trastuzumab/pharmacology , Ado-Trastuzumab Emtansine , Antineoplastic Agents, Immunological/pharmacology , Carrier Proteins , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Lysosomes , Maytansine/pharmacology
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.
Neurobiol Dis ; 124: 276-288, 2019 04.
Article in English | MEDLINE | ID: mdl-30381260

ABSTRACT

Aggregation of α-synuclein (α-syn) is neuropathologically and genetically linked to Parkinson's disease (PD). Since stereotypic cell-to-cell spreading of α-syn pathology is believed to contribute to disease progression, immunotherapy with antibodies directed against α-syn is considered a promising therapeutic approach for slowing disease progression. Here we report the identification, binding characteristics, and efficacy in PD mouse models of the human-derived α-syn antibody BIIB054, which is currently under investigation in a Phase 2 clinical trial for PD. BIIB054 was generated by screening human memory B-cell libraries from healthy elderly individuals. Epitope mapping studies conducted using peptide scanning, X-ray crystallography, and mutagenesis show that BIIB054 binds to α-syn residues 1-10. BIIB054 is highly selective for aggregated forms of α-syn with at least an 800-fold higher apparent affinity for fibrillar versus monomeric recombinant α-syn and a strong preference for human PD brain tissue. BIIB054 discriminates between monomers and oligomeric/fibrillar forms of α-syn based on high avidity for aggregates, driven by weak monovalent affinity and fast binding kinetics. In efficacy studies in three different mouse models with intracerebrally inoculated preformed α-syn fibrils, BIIB054 treatment attenuated the spreading of α-syn pathology, rescued motor impairments, and reduced the loss of dopamine transporter density in dopaminergic terminals in striatum. The preclinical data reported here provide a compelling rationale for clinical development of BIIB054 for the treatment and prevention of PD.


Subject(s)
Antibodies, Monoclonal/pharmacology , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology , alpha-Synuclein/antagonists & inhibitors , Animals , Humans , Mice , Phenotype , Protein Aggregates
14.
Br J Hosp Med (Lond) ; 79(7): 372-377, 2018 Jul 02.
Article in English | MEDLINE | ID: mdl-29995540

ABSTRACT

Immunotherapy is a novel type of anti-cancer treatment that works by upregulating the host's immune system to fight against cancer cells. Landmark immunotherapy trials have demonstrated improvements in response rates and survival compared to cytotoxic chemotherapy. Specific immunotherapies known as checkpoint inhibitors are now routinely used in a range of cancers including melanoma, lung, renal and urological cancers. Immunotherapies are associated with immune-related adverse events which are very different to those seen with traditional cytotoxic chemotherapies. This can present a new challenge to oncologists, acute physicians and the wider team of health-care professionals who look after patients receiving immunotherapy. Generally, these side effects are easily managed but some, if untreated, can be subtle and potentially life-threatening. Patients on immunotherapy may present to a wide variety of medical professionals including the emergency department, primary care and general medical admissions units. It is therefore vital that there is increased awareness and education to identify and manage side effects of immunotherapy effectively.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Colitis , Hepatitis , Hypophysitis , Pneumonia , Colitis/diagnosis , Colitis/etiology , Colitis/therapy , Hepatitis/diagnosis , Hepatitis/etiology , Hepatitis/therapy , Humans , Hypophysitis/diagnosis , Hypophysitis/etiology , Hypophysitis/therapy , Immunotherapy , Neoplasms/drug therapy , Pneumonia/diagnosis , Pneumonia/etiology , Pneumonia/therapy , Referral and Consultation
15.
Sci Rep ; 8(1): 6412, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29686315

ABSTRACT

Aducanumab, a human-derived antibody targeting amyloid-ß (Aß), is in Phase 3 clinical trials for the treatment of Alzheimer's disease. Biochemical and structural analyses show that aducanumab binds a linear epitope formed by amino acids 3-7 of the Aß peptide. Aducanumab discriminates between monomers and oligomeric or fibrillar aggregates based on weak monovalent affinity, fast binding kinetics and strong avidity for epitope-rich aggregates. Direct comparative studies with analogs of gantenerumab, bapineuzumab and solanezumab demonstrate clear differentiation in the binding properties of these antibodies. The crystal structure of the Fab fragment of aducanumab bound to its epitope peptide reveals that aducanumab binds to the N terminus of Aß in an extended conformation, distinct from those seen in structures with other antibodies that target this immunodominant epitope. Aducanumab recognizes a compact epitope that sits in a shallow pocket on the antibody surface. In silico analyses suggest that aducanumab interacts weakly with the Aß monomer and may accommodate a variety of peptide conformations, further supporting its selectivity for Aß aggregates. Our studies provide a structural rationale for the low affinity of aducanumab for non-pathogenic monomers and its greater selectivity for aggregated forms than is seen for other Aß-targeting antibodies.


Subject(s)
Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Binding Sites, Antibody , Enzyme-Linked Immunosorbent Assay , Humans , Immunotherapy , Kinetics , Molecular Docking Simulation , Protein Conformation , Surface Plasmon Resonance
16.
Blood ; 126(1): 103-12, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-25972159

ABSTRACT

The successful treatment of acute leukemias with allogeneic hematopoietic cell transplantation (allo-HCT) is limited by acute graft-versus-host disease (GVHD). Because microRNA-155 (miR-155) regulates activation of the innate immune system, we aimed to determine its function in dendritic cells (DCs) during GVHD in an experimental model. We observed that miR-155 deficiency of the recipient led to improved survival, reduced serum levels of proinflammatory cytokines, and lower GVHD histopathology scores. In addition, miR-155(-/-) bone marrow chimeric mice receiving allo-HCT and miR-155(-/-) DCs showed that miR-155 deficiency in the DC compartment was responsible for protection from GVHD. Activated miR-155(-/-) DCs displayed lower expression of various purinergic receptors and impaired migration toward adenosine triphosphate (ATP). Microarray analysis of lipopolysaccharide/ATP-stimulated miR-155(-/-) DCs revealed mitogen-activated protein kinase pathway dysregulation and reduced inflammasome-associated gene expression. Consistent with this gene expression data, we observed reduced ERK activation, caspase-1 cleavage, and IL-1ß production in miR-155(-/-) DCs. The connection between miR-155 and inflammasome activation was supported by the fact that Nlrp3/miR-155 double-knockout allo-HCT recipient mice had no increased protection from GVHD compared with Nlrp3(-/-) recipients. This study indicates that during GVHD, miR-155 promotes DC migration toward sites of ATP release accompanied by inflammasome activation. Inhibiting proinflammatory miR-155 by antagomir treatment could help reduce this complication of allo-HCT.


Subject(s)
Cell Movement , Dendritic Cells/immunology , Graft vs Host Disease/genetics , Inflammasomes/metabolism , MicroRNAs/genetics , Animals , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Dendritic Cells/physiology , Female , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Inflammasomes/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/metabolism , Severity of Illness Index , Transplantation, Homologous/adverse effects
17.
Blood ; 125(3): 570-80, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25352130

ABSTRACT

The common γ chain (CD132) is a subunit of the interleukin (IL) receptors for IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Because levels of several of these cytokines were shown to be increased in the serum of patients developing acute and chronic graft-versus-host disease (GVHD), we reasoned that inhibition of CD132 could have a profound effect on GVHD. We observed that anti-CD132 monoclonal antibody (mAb) reduced acute GVHD potently with respect to survival, production of tumor necrosis factor, interferon-γ, and IL-6, and GVHD histopathology. Anti-CD132 mAb afforded protection from GVHD partly via inhibition of granzyme B production in CD8 T cells, whereas exposure of CD8 T cells to IL-2, IL-7, IL-15, and IL-21 increased granzyme B production. Also, T cells exposed to anti-CD132 mAb displayed a more naive phenotype in microarray-based analyses and showed reduced Janus kinase 3 (JAK3) phosphorylation upon activation. Consistent with a role of JAK3 in GVHD, Jak3(-/-) T cells caused less severe GVHD. Additionally, anti-CD132 mAb treatment of established chronic GVHD reversed liver and lung fibrosis, and pulmonary dysfunction characteristic of bronchiolitis obliterans. We conclude that acute GVHD and chronic GVHD, caused by T cells activated by common γ-chain cytokines, each represent therapeutic targets for anti-CD132 mAb immunomodulation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Marrow Transplantation/adverse effects , Cytokines/metabolism , Graft vs Host Disease/prevention & control , Interleukin Receptor Common gamma Subunit/antagonists & inhibitors , Acute Disease , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Chronic Disease , Female , Flow Cytometry , Fluorescent Antibody Technique , Graft vs Host Disease/etiology , Graft vs Host Disease/mortality , Humans , Janus Kinase 3/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
18.
Methods Enzymol ; 540: 95-117, 2014.
Article in English | MEDLINE | ID: mdl-24630103

ABSTRACT

The actin cytoskeleton is very dynamic and highly regulated by multiple associated proteins in vivo. Understanding how this system of proteins functions in the processes of actin network assembly and disassembly requires methods to dissect the mechanisms of activity of individual factors and of multiple factors acting in concert. The advent of single-filament and single-molecule fluorescence imaging methods has provided a powerful new approach to discovering actin-regulatory activities and obtaining direct, quantitative insights into the pathways of molecular interactions that regulate actin network architecture and dynamics. Here we describe techniques for acquisition and analysis of single-molecule data, applied to the novel challenges of studying the filament assembly and disassembly activities of actin-associated proteins in vitro. We discuss the advantages of single-molecule analysis in directly visualizing the order of molecular events, measuring the kinetic rates of filament binding and dissociation, and studying the coordination among multiple factors. The methods described here complement traditional biochemical approaches in elucidating actin-regulatory mechanisms in reconstituted filamentous networks.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/analysis , Actins/metabolism , Microscopy, Fluorescence/methods , Optical Imaging/methods , Actin Cytoskeleton/chemistry , Equipment Design , Fluorescent Dyes/analysis , Microscopy, Fluorescence/instrumentation , Optical Imaging/instrumentation , Staining and Labeling/methods
19.
Elife ; 2: e01008, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-24015360

ABSTRACT

During cell locomotion and endocytosis, membrane-tethered WASP proteins stimulate actin filament nucleation by the Arp2/3 complex. This process generates highly branched arrays of filaments that grow toward the membrane to which they are tethered, a conflict that seemingly would restrict filament growth. Using three-color single-molecule imaging in vitro we revealed how the dynamic associations of Arp2/3 complex with mother filament and WASP are temporally coordinated with initiation of daughter filament growth. We found that WASP proteins dissociated from filament-bound Arp2/3 complex prior to new filament growth. Further, mutations that accelerated release of WASP from filament-bound Arp2/3 complex proportionally accelerated branch formation. These data suggest that while WASP promotes formation of pre-nucleation complexes, filament growth cannot occur until it is triggered by WASP release. This provides a mechanism by which membrane-bound WASP proteins can stimulate network growth without restraining it. DOI:http://dx.doi.org/10.7554/eLife.01008.001.


Subject(s)
Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Color , Kinetics
20.
Proc Natl Acad Sci U S A ; 110(4): 1285-90, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23292935

ABSTRACT

Actin filament nucleation by actin-related protein (Arp) 2/3 complex is a critical process in cell motility and endocytosis, yet key aspects of its mechanism are unknown due to a lack of real-time observations of Arp2/3 complex through the nucleation process. Triggered by the verprolin homology, central, and acidic (VCA) region of proteins in the Wiskott-Aldrich syndrome protein (WASp) family, Arp2/3 complex produces new (daughter) filaments as branches from the sides of preexisting (mother) filaments. We visualized individual fluorescently labeled Arp2/3 complexes dynamically interacting with and producing branches on growing actin filaments in vitro. Branch formation was strikingly inefficient, even in the presence of VCA: only ~1% of filament-bound Arp2/3 complexes yielded a daughter filament. VCA acted at multiple steps, increasing both the association rate of Arp2/3 complexes with mother filament and the fraction of filament-bound complexes that nucleated a daughter. The results lead to a quantitative kinetic mechanism for branched actin assembly, revealing the steps that can be stimulated by additional cellular factors.


Subject(s)
Actin-Related Protein 2-3 Complex/chemistry , Actin-Related Protein 2-3 Complex/metabolism , Actins/chemistry , Actins/metabolism , Actin-Related Protein 2-3 Complex/ultrastructure , Actins/ultrastructure , Animals , Kinetics , Microscopy, Fluorescence, Multiphoton , Models, Molecular , Protein Interaction Domains and Motifs , Protein Multimerization , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Wiskott-Aldrich Syndrome Protein Family/chemistry , Wiskott-Aldrich Syndrome Protein Family/metabolism , Wiskott-Aldrich Syndrome Protein Family/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...