Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
Add more filters

Publication year range
1.
Cell ; 173(6): 1439-1453.e19, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29856956

ABSTRACT

The absence of cancer-restricted surface markers is a major impediment to antigen-specific immunotherapy using chimeric antigen receptor (CAR) T cells. For example, targeting the canonical myeloid marker CD33 in acute myeloid leukemia (AML) results in toxicity from destruction of normal myeloid cells. We hypothesized that a leukemia-specific antigen could be created by deleting CD33 from normal hematopoietic stem and progenitor cells (HSPCs), thereby generating a hematopoietic system resistant to CD33-targeted therapy and enabling specific targeting of AML with CAR T cells. We generated CD33-deficient human HSPCs and demonstrated normal engraftment and differentiation in immunodeficient mice. Autologous CD33 KO HSPC transplantation in rhesus macaques demonstrated long-term multilineage engraftment of gene-edited cells with normal myeloid function. CD33-deficient cells were impervious to CD33-targeting CAR T cells, allowing for efficient elimination of leukemia without myelotoxicity. These studies illuminate a novel approach to antigen-specific immunotherapy by genetically engineering the host to avoid on-target, off-tumor toxicity.


Subject(s)
Hematopoietic Stem Cells/cytology , Immunotherapy/methods , Leukemia, Myeloid, Acute/therapy , RNA, Guide, Kinetoplastida/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , T-Lymphocytes/immunology , Animals , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Electroporation , Female , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/immunology , Macaca mulatta , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Transplantation , Reactive Oxygen Species , T-Lymphocytes/cytology
2.
PLoS Comput Biol ; 19(2): e1009894, 2023 02.
Article in English | MEDLINE | ID: mdl-36780558

ABSTRACT

Modeling biological mechanisms is a key for disease understanding and drug-target identification. However, formulating quantitative models in the field of Alzheimer's Disease is challenged by a lack of detailed knowledge of relevant biochemical processes. Additionally, fitting differential equation systems usually requires time resolved data and the possibility to perform intervention experiments, which is difficult in neurological disorders. This work addresses these challenges by employing the recently published Variational Autoencoder Modular Bayesian Networks (VAMBN) method, which we here trained on combined clinical and patient level gene expression data while incorporating a disease focused knowledge graph. Our approach, called iVAMBN, resulted in a quantitative model that allowed us to simulate a down-expression of the putative drug target CD33, including potential impact on cognitive impairment and brain pathophysiology. Experimental validation demonstrated a high overlap of molecular mechanism predicted to be altered by CD33 perturbation with cell line data. Altogether, our modeling approach may help to select promising drug targets.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Bayes Theorem , Artificial Intelligence , Sialic Acid Binding Ig-like Lectin 3/chemistry , Sialic Acid Binding Ig-like Lectin 3/genetics , Sialic Acid Binding Ig-like Lectin 3/metabolism
3.
J Biol Chem ; 298(6): 101960, 2022 06.
Article in English | MEDLINE | ID: mdl-35452678

ABSTRACT

Alzheimer's disease (AD) is characterized by accumulation of misfolded proteins. Genetic studies implicate microglia, brain-resident phagocytic immune cells, in AD pathogenesis. As positive effectors, microglia clear toxic proteins, whereas as negative effectors, they release proinflammatory mediators. An imbalance of these functions contributes to AD progression. Polymorphisms of human CD33, an inhibitory microglial receptor, are linked to AD susceptibility; higher CD33 expression correlates with increased AD risk. CD33, also called Siglec-3, is a member of the sialic acid-binding immunoglobulin-type lectin (Siglec) family of immune regulatory receptors. Siglec-mediated inhibition is initiated by binding to complementary sialoglycan ligands in the tissue environment. Here, we identify a single sialoglycoprotein in human cerebral cortex that binds CD33 as well as Siglec-8, the most abundant Siglec on human microglia. The ligand, which we term receptor protein tyrosine phosphatase zeta (RPTPζ)S3L, is composed of sialylated keratan sulfate chains carried on a minor isoform/glycoform of RPTPζ (phosphacan) and is found in the extracellular milieu of the human brain parenchyma. Brains from human AD donors had twofold higher levels of RPTPζS3L than age-matched control donors, raising the possibility that RPTPζS3L overexpression limits misfolded protein clearance contributing to AD pathology. Mice express the same structure, a sialylated keratan sulfate RPTPζ isoform, that binds mouse Siglec-F and crossreacts with human CD33 and Siglec-8. Brains from mice engineered to lack RPTPζ, the sialyltransferase St3gal4, or the keratan sulfate sulfotransferase Chst1 lacked Siglec binding, establishing the ligand structure. The unique CD33 and Siglec-8 ligand, RPTPζS3L, may contribute to AD progression.


Subject(s)
Alzheimer Disease , Sialic Acid Binding Immunoglobulin-like Lectins , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Humans , Keratan Sulfate/metabolism , Ligands , Mice , Microglia/metabolism , Protein Isoforms/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Sialic Acid Binding Ig-like Lectin 3/genetics , Sialic Acid Binding Ig-like Lectin 3/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism
4.
Blood ; 137(8): 1037-1049, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33094319

ABSTRACT

Emerging immunotherapies such as chimeric antigen receptor T cells have advanced the treatment of acute lymphoblastic leukemia. In contrast, long-term control of acute myeloid leukemia (AML) cannot be achieved by single lineage-specific targeting while sparing benign hematopoiesis. In addition, heterogeneity of AML warrants combinatorial targeting, and several suitable immunotargets (HAVCR2/CD33 and HAVCR2/CLEC12A) have been identified in adult AML. However, clinical and biologic characteristics of AML differ between children and the elderly. Here, we analyzed 36 bone marrow (BM) samples of pediatric AML patients and 13 age-matched healthy donors using whole RNA sequencing of sorted CD45dim and CD34+CD38-CD45dim BM populations and flow cytometry for surface expression of putative target antigens. Pediatric AML clusters apart from healthy myeloid BM precursors in principal-component analysis. Known immunotargets of adult AML, such as IL3RA, were not overexpressed in pediatric AML compared with healthy precursors by RNA sequencing. CD33 and CLEC12A were the most upregulated immunotargets on the RNA level and showed the highest surface expression on AML detected by flow cytometry. KMT2A-mutated infant AML clusters separately by RNA sequencing and overexpresses FLT3, and hence, CD33/FLT3 cotargeting is an additional specific option for this subgroup. CLEC12A and CD33/CLEC12Adouble-positive expression was absent in CD34+CD38-CD45RA-CD90+ hematopoietic stem cells (HSCs) and nonhematopoietic tissue, while CD33 and FLT3 are expressed on HSCs. In summary, we show that expression of immunotargets in pediatric AML differs from known expression profiles in adult AML. We identify CLEC12A and CD33 as preferential generic combinatorial immunotargets in pediatric AML and CD33 and FLT3 as immunotargets specific for KMT2A-mutated infant AML.


Subject(s)
Gene Expression Regulation, Leukemic , Lectins, C-Type/genetics , Leukemia, Myeloid, Acute/genetics , Receptors, Mitogen/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Adolescent , Child , Child, Preschool , Female , Humans , Immunotherapy , Infant , Lectins, C-Type/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Male , Receptors, Mitogen/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Transcriptome , Up-Regulation
5.
Int J Cancer ; 150(7): 1141-1155, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34766343

ABSTRACT

Immunotherapies, such as chimeric antigen receptor (CAR) modified T cells and antibody-drug conjugates (ADCs), have revolutionized the treatment of cancer, especially of lymphoid malignancies. The application of targeted immunotherapy to patients with acute myeloid leukemia (AML) has been limited in particular by the lack of a tumor-specific target antigen. Gemtuzumab ozogamicin (GO), an ADC targeting CD33, is the only approved immunotherapeutic agent in AML. In our study, we introduce a CD33-directed third-generation CAR T-cell product (3G.CAR33-T) for the treatment of patients with AML. 3G.CAR33-T cells could be expanded up to the end-of-culture, that is, 17 days after transduction, and displayed significant cytokine secretion and robust cytotoxic activity when incubated with CD33-positive cells including cell lines, drug-resistant cells, primary blasts as well as normal hematopoietic stem and progenitor cells (HSPCs). When compared to second-generation CAR33-T cells, 3G.CAR33-T cells exhibited higher viability, increased proliferation and stronger cytotoxicity. Also, GO exerted strong antileukemia activity against CD33-positive AML cells. Upon genomic deletion of CD33 in HSPCs, 3G.CAR33-T cells and GO preferentially killed wildtype leukemia cells, while sparing CD33-deficient HSPCs. Our data provide evidence for the applicability of CD33-targeted immunotherapies in AML and its potential implementation in CD33 genome-edited stem cell transplantation approaches.


Subject(s)
Gemtuzumab/therapeutic use , Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Leukemia, Myeloid, Acute/therapy , Receptors, Chimeric Antigen/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Gene Editing , Hematopoietic Stem Cells/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Sialic Acid Binding Ig-like Lectin 3/analysis , Sialic Acid Binding Ig-like Lectin 3/genetics
6.
Hum Mol Genet ; 29(17): 2920-2935, 2020 10 10.
Article in English | MEDLINE | ID: mdl-32803224

ABSTRACT

Neuroinflammation is a key contributor to the pathology of Alzheimer's disease (AD). CD33 (Siglec-3) is a transmembrane sialic acid-binding receptor on the surface of microglial cells. CD33 is upregulated on microglial cells from post-mortem AD patient brains, and high levels of CD33 inhibit uptake and clearance of amyloid beta (Aß) in microglial cell cultures. Furthermore, knockout of CD33 reduces amyloid plaque burden in mouse models of AD. Here, we tested whether a gene therapy strategy to reduce CD33 on microglia in AD could decrease Aß plaque load. Intracerebroventricular injection of an adeno-associated virus (AAV) vector-based system encoding an artificial microRNA targeting CD33 (miRCD33) into APP/PS1 mice reduced CD33 mRNA and TBS-soluble Aß40 and Aß42 levels in brain extracts. Treatment of APP/PS1 mice with miRCD33 vector at an early age (2 months) was more effective at reducing Aß plaque burden than intervening at later times (8 months). Furthermore, early intervention downregulated several microglial receptor transcripts (e.g. CD11c, CD47 and CD36) and pro-inflammatory activation genes (e.g. Tlr4 and Il1b). Marked reductions in the chemokine Ccl2 and the pro-inflammatory cytokine Tnfα were observed at the protein level in the brain of APP/PS1 mice treated with miRCD33 vector. Overall, our data indicate that CD33 is a viable target for AAV-based knockdown strategies to reduce AD pathology. One Sentence Summary: A gene therapy approach for Alzheimer's disease using adeno-associated virus vector-based knockdown of CD33 reduced amyloid beta accumulation and neuroinflammation.


Subject(s)
Alzheimer Disease/therapy , Genetic Therapy , Presenilin-1/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Brain/pathology , Dependovirus/genetics , Disease Models, Animal , Humans , Mice , Mice, Transgenic/genetics , Microglia/metabolism , Microglia/pathology , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors
7.
Blood ; 135(8): 542-546, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31880804

ABSTRACT

Acute myeloid leukemia (AML) is a highly heterogeneous disease both in terms of genetic background and response to chemotherapy. Although molecular aberrations are routinely used to stratify AML patients into prognostic subgroups when receiving standard chemotherapy, the predictive value of the genetic background and co-occurring mutations remains to be assessed when using newly approved antileukemic drugs. In the present study, we retrospectively addressed the question of the predictive value of molecular events on the benefit of the addition of gemtuzumab ozogamicin (GO) to standard front-line chemotherapy. Using the more recent European LeukemiaNet (ELN) 2017 risk classification, we confirmed that the benefit of GO was restricted to the favorable (hazard ratio [HR], 0.54, 95% confidence interval [CI], 0.30-0.98) and intermediate (HR, 0.57; 95% CI, 0.33-1.00) risk categories, whereas it did not influence the outcome of patients within the adverse risk subgroup (HR, 0.93; 95% CI, 0.61-1.43). Interestingly, the benefit of GO was significant for patients with activating signaling mutations (HR, 0.43; 95% CI, 0.28-0.65), which correlated with higher CD33 expression levels. These results suggest that molecular aberrations could be critical for future differentially tailored treatments based on integrated genetic profiles that are able to predict the benefit of GO on outcome.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Gemtuzumab/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Mutation , Aged , Antineoplastic Agents, Immunological/adverse effects , Gemtuzumab/adverse effects , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid, Acute/genetics , Middle Aged , Proportional Hazards Models , Sialic Acid Binding Ig-like Lectin 3/genetics
8.
Blood ; 135(2): 85-96, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31765470

ABSTRACT

The acute myeloid leukemia (AML) treatment landscape has changed substantially since 2017. New targeted drugs have emerged, including venetoclax to target B-cell lymphoma 2, midostaurin and gilteritinib to target FLT3, and ivosidenib and enasidenib to target mutant isocitrate dehydrogenase 1 and 2, respectively. Other additions include reapproval of gemtuzumab ozogomycin to target CD33, glasdegib to target the hedgehog pathway, and a liposomal formulation of daunorubicin and cytarabine (CPX-351). Genomically heterogeneous AML has a tendency to evolve, particularly under selective treatment pressure. For decades, treatment decisions have largely centered around chemotherapy drug intensity. Physicians now have access to an increasing number of drugs with novel mechanisms of action and distinctive side-effect profiles. Key issues faced by hematologists in this era of new drugs include (1) the timely identification of actionable mutations at diagnosis and at relapse; (2) deciding which drug to use among several therapeutic options; and (3) increasing awareness of how to anticipate, mitigate, and manage common complications associated with these new agents. This article will use 3 case presentations to discuss some of the new treatment challenges encountered in AML management, with the goal of providing practical guidance to aid the practicing physician.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Leukemia, Myeloid, Acute/drug therapy , Molecular Targeted Therapy , Mutation , Adult , Aged , Aminopyridines/therapeutic use , Aniline Compounds/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Female , Glycine/analogs & derivatives , Glycine/therapeutic use , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Prognosis , Pyrazines/therapeutic use , Pyridines/therapeutic use , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/genetics , Staurosporine/analogs & derivatives , Staurosporine/therapeutic use , Sulfonamides/therapeutic use , Triazines/therapeutic use , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics
9.
Gerontology ; 68(4): 421-430, 2022.
Article in English | MEDLINE | ID: mdl-34569532

ABSTRACT

BACKGROUND/AIMS: In despite of conflicting results among different ethnic groups, the rs3865444 of CD33 gene has previously been identified as a risk factor for late-onset Alzheimer's disease (LOAD).This study was aimed to evaluate the association between rs3865444 SNP with LOAD occurrence, and to investigate whether CD33 mRNA expression will change in the leukocytes of peripheral blood in LOAD patients. METHODS: The rs3865444 polymorphism was genotyped in 233 LOAD and 238 control subjects using the Tetra-ARMS-PCR method. CD33 mRNAs expression in leukocytes were assessed and analyzed using the real-time qPCR method. We used in silico approach to analyze potential effects imparted by rs3865444 polymorphism in LOAD pathogenesis. RESULTS: Our results show a significant increase in CD33 mRNA expression levels in white blood cells of LOAD patients, however, the association between CD33 rs3865444 polymorphism and LOAD was found to be not significant. We also noticed that LOAD patients with the C/A genotype had higher CD33 mRNA levels in their peripheral blood than those of the control group. CONCLUSIONS: rs3865444, located upstream of the 5'CD33 coding region, might positively influence CD33 mRNAs expression in leukocytes of LOAD versus healthy people. This is likely to happen through interfering rs3865444 (C) with the functional activity of several other transcription factors given that rs3865444 is in linkage disequilibrium with other functional polymorphisms in this coding region according to an in silico study. We propose that CD33 mRNAs elevation in peripheral immune cells - as a potential biomarker in LOAD - is related to peripheral immune system impairment.


Subject(s)
Alzheimer Disease , Genetic Predisposition to Disease , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Humans , Leukocytes/metabolism , Polymorphism, Single Nucleotide , RNA, Messenger/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics
10.
Int J Mol Sci ; 23(7)2022 Mar 26.
Article in English | MEDLINE | ID: mdl-35408990

ABSTRACT

The CD33 gene encodes for a member of the sialic-acid-binding immunoglobulin-type lectin (Siglec) family, and is one of the top-ranked Alzheimer's disease (AD) risk genes identified by genome-wide association studies (GWAS). Many CD33 polymorphisms are associated with an increased risk of AD, but the function and potential mechanism of many CD33 single-nucleotide polymorphisms (SNPs) in promoting AD have yet to be elucidated. We recently identified the CD33 SNP rs2455069-A>G (R69G) in a familial form of dementia. Here, we demonstrate an association between the G allele of the rs2455069 gene variant and the presence of AD in a cohort of 195 patients from southern Italy. We carried out in silico analysis of the 3D structures of CD33 carrying the identified SNP to provide insights into its functional effect. Structural models of the CD33 variant carrying the R69G amino acid change were compared to the CD33 wild type, and used for the docking analysis using sialic acid as the ligand. Our analysis demonstrated that the CD33-R69G variant may bind sialic acid at additional binding sites compared to the wild type, thus potentially increasing its affinity/specificity for this molecule. Our results led to a new hypothesis of rs2455069-A>G SNP as a risk factor for AD, suggesting that a long-term cumulative effect of the CD33-R69G variant results from the binding of sialic acid, acting as an enhancer of the CD33 inhibitory effects on amyloid plaque degradation.


Subject(s)
Alzheimer Disease , Polymorphism, Single Nucleotide , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Genome-Wide Association Study , Humans , Microglia/metabolism , N-Acetylneuraminic Acid/metabolism , Sialic Acid Binding Ig-like Lectin 3/genetics
11.
Glia ; 69(6): 1393-1412, 2021 06.
Article in English | MEDLINE | ID: mdl-33539598

ABSTRACT

Genome-wide association studies demonstrated that polymorphisms in the CD33/sialic acid-binding immunoglobulin-like lectin 3 gene are associated with late-onset Alzheimer's disease (AD). CD33 is expressed on myeloid immune cells and mediates inhibitory signaling through protein tyrosine phosphatases, but the exact function of CD33 in microglia is still unknown. Here, we analyzed CD33 knockout human THP1 macrophages and human induced pluripotent stem cell-derived microglia for immunoreceptor tyrosine-based activation motif pathway activation, cytokine transcription, phagocytosis, and phagocytosis-associated oxidative burst. Transcriptome analysis of the macrophage lines showed that knockout of CD33 as well as knockdown of the CD33 signaling-associated protein tyrosine phosphatase, nonreceptor type 6 (PTPN6) led to constitutive activation of inflammation-related pathways. Moreover, deletion of CD33 or expression of Exon 2-deleted CD33 (CD33ΔE2 /CD33m) led to increased phosphorylation of the kinases spleen tyrosine kinase (SYK) and extracellular signal-regulated kinase 1 and 2 (ERK1 and 2). Transcript analysis by quantitative real-time polymerase chain reaction confirmed increased levels of interleukin (IL) 1B, IL8, and IL10 after knockout of CD33 in macrophages and microglia. In addition, upregulation of the gene transcripts of the AD-associated phosphatase INPP5D was observed after knockout of CD33. Functional analysis of macrophages and microglia showed that phagocytosis of aggregated amyloid-ß1-42 and bacterial particles were increased after knockout of CD33 or CD33ΔE2 expression and knockdown of PTPN6. Furthermore, the phagocytic oxidative burst during uptake of amyloid-ß1-42 or bacterial particles was increased after CD33 knockout but not in CD33ΔE2 -expressing microglia. In summary, deletion of CD33 or expression of CD33ΔE2 in human macrophages and microglia resulted in putative beneficial phagocytosis of amyloid ß1-42 , but potentially detrimental oxidative burst and inflammation, which was absent in CD33ΔE2 -expressing microglia.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Alzheimer Disease/genetics , Amyloid beta-Peptides , Genome-Wide Association Study , Humans , Inflammation , Microglia , Phenotype , Sialic Acid Binding Ig-like Lectin 3/genetics
12.
J Neurochem ; 158(2): 297-310, 2021 07.
Article in English | MEDLINE | ID: mdl-33720433

ABSTRACT

CD33 is a Siglec (sialic acid-binding immunoglobulin-type lectin) receptor on microglia. Human CD33 can be alternatively spliced into two isoforms: the long isoform (CD33M) and a shorter isoform (CD33m) that lacks the sialic acid-binding site. CD33m appears to protect against Alzheimer's disease; however, it remains unclear how. To investigate potential mechanisms by which CD33m may confer protection, we expressed the CD33m and CD33M isoforms of human CD33 in mouse BV-2 and human CHME3 microglial cells and assessed microglia functions. In the BV-2 cells, CD33M inhibited microglial phagocytosis of beads, synapses, debris and dead cells, while CD33m increased phagocytosis of beads, debris and cells. RNAi knockdown of the endogenous mouse CD33 increased phagocytosis and prevented CD33m's (but not CD33M's) effect on phagocytosis. CD33M increased cell attachment but inhibited cell proliferation, while CD33m did the opposite. We also found that CD33M inhibited cell migration. In human CHME3 cells, CD33M increased cell attachment, but inhibited phagocytosis, proliferation and migration, whereas CD33m did the opposite. We conclude that CD33M inhibits microglial phagocytosis, inhibits migration and increases adhesion, while CD33m increases phagocytosis, proliferation and inhibits adhesion. Thus, CD33m might protect against Alzheimer's disease by increasing microglial proliferation, movement and phagocytosis of debris and dead cells.


Subject(s)
Alzheimer Disease/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Microglia/drug effects , Phagocytosis/drug effects , Sialic Acid Binding Ig-like Lectin 3/genetics , Alzheimer Disease/genetics , Animals , Cell Line , Encephalitis/genetics , Gene Knockdown Techniques , Genetic Variation , Humans , Mice , Neuraminidase/chemistry , RNA Interference , Sialic Acid Binding Ig-like Lectin 3/metabolism
13.
Cancer Immunol Immunother ; 70(3): 803-815, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33000418

ABSTRACT

Colorectal cancer (CRC) has high mortality rates, especially in patients with advanced disease stages, who often do not respond to therapy. The cellular components of the tumor microenvironment are essentially responsible for dictating disease progression and response to therapy. Expansion of different myeloid cell subsets in CRC tumors has been reported previously. However, tumor-infiltrating myeloid cells have both pro- and anti-tumor roles in disease progression. In this study, we performed transcriptomic profiling of cells of myeloid lineage (CD33+) from bulk CRC tumors at varying disease stages. We identified differentially expressed genes and pathways between CRC patients with advanced stage and early stages. We found that pro-angiogenic and hypoxia-related genes were upregulated, while genes related to immune and inflammatory responses were downregulated in CD33+ myeloid cells from patients with advanced stages, implying that immune cell recruitment and activation could be compromised in advanced disease stages. Moreover, we identified a unique "poor prognosis CD33+ gene signature" by aligning top upregulated and downregulated genes in tumor-infiltrating myeloid cells from our analyses with data from The Cancer Genome Atlas. Our results showed that this gene signature is an independent prognostic indicator for disease-specific survival in CRC patients, potentially reflecting its clinical importance.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Myeloid Cells/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , Biomarkers , Colorectal Neoplasms/pathology , Female , Gene Expression Profiling , Genomics/methods , Humans , Male , Myeloid Cells/pathology , Neoplasm Grading , Neoplasm Staging , Sialic Acid Binding Ig-like Lectin 3/genetics
14.
FASEB J ; 34(2): 1928-1938, 2020 02.
Article in English | MEDLINE | ID: mdl-31907986

ABSTRACT

Humans and orcas are among the very rare species that have a prolonged post-reproductive lifespan (PRLS), during which the aging process continues. Reactive oxygen species (ROS) derived from mitochondria and from the NADPH oxidase (NOX) enzymes of innate immune cells are known to contribute to aging, with the former thought to be dominant. CD33-related-Siglecs are immune receptors that recognize self-associated-molecular-patterns and modulate NOX-derived-ROS. We herewith demonstrate a strong correlation of lifespan with CD33rSIGLEC gene number in 26 species, independent of body weight or phylogeny. The correlation is stronger when considering total CD33rSIGLEC gene number rather than those encoding inhibitory and activating subsets, suggesting that lifetime balancing of ROS is important. Combining independent lines of evidence including the short half-life and spontaneous activation of neutrophils, we calculate that even without inter-current inflammation, a major source of lifetime ROS exposure may actually be neutrophil NOX-derived. However, genomes of human supercentenarians (>110 years) do not harbor a significantly higher number of functional CD33rSIGLEC genes. Instead, lifespan correlation with CD33rSIGLEC gene number was markedly strengthened by excluding the post-reproductive lifespan of humans and orcas (R2  = 0.83; P < .0001). Thus, CD33rSIGLEC modulation of ROS likely contributes to maximum reproductive lifespan, but other unknown mechanisms could be important to PRLS.


Subject(s)
Gene Dosage , Longevity , NADPH Oxidases , Neutrophils/immunology , Reactive Oxygen Species/immunology , Sialic Acid Binding Ig-like Lectin 3 , Animals , Humans , Longevity/genetics , Longevity/immunology , NADPH Oxidases/genetics , NADPH Oxidases/immunology , Sialic Acid Binding Ig-like Lectin 3/genetics , Sialic Acid Binding Ig-like Lectin 3/immunology , Whale, Killer
15.
Transfusion ; 61(3): 960-967, 2021 03.
Article in English | MEDLINE | ID: mdl-33638187

ABSTRACT

BACKGROUND: A 30-year-old man underwent double umbilical cord blood transplantation (UCBT) for acute myeloid leukemia (AML) with reduced intensity conditioning. The cords had identical HLA types and were each a 5/6 match to the patient. Following transplantation, cord 2 initially dominated all tested cell populations. At day +306, we observed an unusual reversal of dominance chimerism pattern in which cord 1 instead dominated all tested populations. STUDY DESIGN & METHODS: Polymerase chain reaction (PCR)-based short tandem repeat (STR) assays were performed on the peripheral blood and bone marrow samples. The white blood cell (WBC) populations from the peripheral blood were manipulated for testing to create subpopulations enriched for CD3, CD33, and CD56. RESULTS: Chimerism studies on day +77 showed the following: cord 1: 44%-CD3; 0%-CD33; 16%-CD56; cord 2: 56%-CD3; 100%-CD33; 84%-CD56. Cord 2 initially dominated in all tested cell populations. Chimerism studies performed on post-transplantation day +306 uncovered a reversal of dominance chimerism pattern in which cord 1 now dominated in all cell populations (cord 1: 82%-CD3; >95%-CD33; 67%-CD56; cord 2: 18%-CD3; <5%-CD33; 33%-CD56). Between days +127 and +244, the patient's blood type shifted from B Rh-positive to A Rh-negative. CONCLUSION: The change in the patient's blood type identified a late reversal of dominance chimerism pattern. This is a rare occurrence, previously cited only once, which is inconsistent with published data that early high CD3 counts and unseparated bone marrow chimerism predominance at day +100 predict long-term cord dominance in double UCBT in the vast majority of cases.


Subject(s)
Chimerism , Cord Blood Stem Cell Transplantation , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/therapy , Leukocytes/metabolism , Adult , Blood Grouping and Crossmatching , Bone Marrow/metabolism , CD3 Complex/blood , CD3 Complex/genetics , CD56 Antigen/blood , CD56 Antigen/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Male , Polymerase Chain Reaction , Sialic Acid Binding Ig-like Lectin 3/blood , Sialic Acid Binding Ig-like Lectin 3/genetics
16.
Cell Biol Int ; 45(10): 2086-2095, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34184811

ABSTRACT

Prostate cancer (PCa) is one of the most epidemic types of cancer in men. The tumor microenvironment (TME) of PCa is involved in the emergence of immunosuppressive factors such as myeloid-derived suppressor cells (MDSC), which regulate the immune system by several mechanisms, including interleukin (IL)-10 production. On the other hand, IL-17+ helper T cells (Th17) induce MDSCs and chronic inflammation in TME by producing IL-17. This study demonstrated that the frequency of CD33+ pSTAT3+ MDSC and IL-17+ lymphocyte as well as IL-10 messenger RNA (mRNA) expression were significantly higher in the PCa patients than in the benign prostatic hyperplasia (BPH) group. Moreover, there was no significant relationship between the frequency of CD33+ pSTAT3+ MDSC, and IL-17+ lymphocyte with Gleason scores in the PCa group. We suggested that the higher frequency of CD33+ pSTAT3+ MDSC and IL-17+ lymphocyte and the more frequent expression of IL-10 mRNA in PCa patients may play roles in tumor progression from BPH to PCa.


Subject(s)
Interleukin-17/metabolism , Lymphocytes/immunology , Myeloid-Derived Suppressor Cells/immunology , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Th17 Cells/immunology , Apoptosis , Case-Control Studies , Cell Proliferation , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-17/genetics , Male , Myeloid-Derived Suppressor Cells/metabolism , Prognosis , Prostatic Hyperplasia/immunology , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , STAT3 Transcription Factor/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Sialic Acid Binding Ig-like Lectin 3/metabolism , Tumor Cells, Cultured , Tumor Microenvironment
17.
Future Oncol ; 17(3): 263-277, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33356566

ABSTRACT

The aim of this study was to establish the therapeutic relevance of the CD33D2 isoform by developing novel antibodies targeting the IgC domain of CD33. Two novel IgC-targeting antibodies, HL2541 and 5C11-2, were developed, and CD33 isoforms were assessed using multiple assays in cells overexpressing either CD33FL or CD33D2 isoforms, unmodified acute myeloid leukemia (AML) cell lines and primary AML specimens representing different genotypes for the CD33 splicing single nucleotide polymorphism. CD33D2 was recognized on cells overexpressing CD33D2 and unmodified AML cell lines; however, minimal/no cell surface detection of CD33D2 was observed in primary AML specimens. Both isoforms were detected intracellularly using novel antibodies. Minimal cell surface expression of CD33D2 on primary AML/progenitor cells warrants further studies on anti-CD33D2 immunotherapeutics.


Subject(s)
Antibodies, Monoclonal/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Sialic Acid Binding Ig-like Lectin 3/metabolism , Adolescent , Animals , Antibodies, Monoclonal/therapeutic use , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Line, Tumor , Child , Child, Preschool , Female , Genotype , Humans , Immunoglobulin Domains/immunology , Infant , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Protein Isoforms , Sialic Acid Binding Ig-like Lectin 3/chemistry , Sialic Acid Binding Ig-like Lectin 3/genetics
18.
Mol Ther ; 28(9): 1974-1986, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32526203

ABSTRACT

The successful implementation of chimeric antigen receptor (CAR)-T cell therapy in the clinical context of B cell malignancies has paved the way for further development in the more critical setting of acute myeloid leukemia (AML). Among the potentially targetable AML antigens, CD33 is insofar one of the main validated molecules. Here, we describe the feasibility of engineering cytokine-induced killer (CIK) cells with a CD33.CAR by using the latest optimized version of the non-viral Sleeping Beauty (SB) transposon system "SB100X-pT4." This offers the advantage of improving CAR expression on CIK cells, while reducing the amount of DNA transposase as compared to the previously employed "SB11-pT" version. SB-modified CD33.CAR-CIK cells exhibited significant antileukemic activity in vitro and in vivo in patient-derived AML xenograft models, reducing AML development when administered as an "early treatment" and delaying AML progression in mice with established disease. Notably, by exploiting an already optimized xenograft chemotherapy model that mimics human induction therapy in mice, we demonstrated for the first time that CD33.CAR-CIK cells are also effective toward chemotherapy resistant/residual AML cells, further supporting its future clinical development and implementation within the current standard regimens.


Subject(s)
Cell Engineering/methods , Cell Transplantation/methods , Cytokine-Induced Killer Cells/immunology , Drug Resistance, Neoplasm , Genetic Therapy/methods , Heterografts , Immunotherapy, Adoptive/methods , Leukemia, Experimental/therapy , Leukemia, Myeloid, Acute/therapy , Receptors, Chimeric Antigen/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Animals , Feasibility Studies , Gene Transfer Techniques , Humans , Mice , Mice, Inbred NOD , Mice, SCID , THP-1 Cells , Transposases/genetics , Transposases/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
19.
Int J Immunogenet ; 47(5): 397-405, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32333488

ABSTRACT

CD33 rs3865444:C>A single nucleotide polymorphism (SNP) has been previously associated with the risk of late-onset Alzheimer's disease (LOAD); however, the results have been inconsistent across different populations. CD33 is a transmembrane receptor that plays an important role in AD pathogenesis by inhibiting amyloid ß42 uptake by microglial cells. In this study, we aimed to validate the association between rs3865444 and LOAD risk in the Slovak population and to evaluate whether it was affected by the carrier status of the major LOAD risk allele apolipoprotein (APOE) ε4. CD33 rs3865444 and APOE variants were genotyped in 206 LOAD patients and 487 control subjects using the polymerase chain reaction-restriction fragment length polymorphism method and direct sequencing, respectively. Logistic regression analysis revealed a significant association of rs3865444 A allele with a reduced LOAD risk that was only present in APOE ε4 allele carriers (AA + CA versus CC: p = .0085; OR = 0.45; 95% CI = 0.25-0.82). On the other hand, no such association was found in subjects without the APOE ε4 (p = .75; OR = 0.93; 95% CI = 0.61-1.42). Moreover, regression analysis detected a significant interaction between CD33 rs3865444 A and APOE ε4 alleles (p = .021 for APOE ε4 allele dosage and p = .051 for APOE ε4 carriage status), with synergy factor (SF) value of 0.49 indicating an antagonistic effect between the two alleles in LOAD risk. In conclusion, our results suggest that CD33 rs3865444:C˃A substitution may reduce the risk of LOAD in Slovaks by antagonizing the effect conferred by the major susceptibility allele APOE ε4.


Subject(s)
Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Apolipoproteins E/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Aged , Alleles , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Apolipoprotein E4/immunology , Apolipoproteins E/immunology , Female , Gene Frequency , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Risk Factors , Sialic Acid Binding Ig-like Lectin 3/immunology , Slovakia
20.
Int J Mol Sci ; 21(1)2020 Jan 02.
Article in English | MEDLINE | ID: mdl-31906437

ABSTRACT

Nanobodies (Nbs) are the smallest antigen-binding, single domain fragments derived from heavy-chain-only antibodies from Camelidae. Among the several advantages over conventional monoclonal antibodies, their small size (12-15 kDa) allows them to extravasate rapidly, to show improved tissue penetration, and to clear rapidly from blood, which are important characteristics for cancer imaging and targeted radiotherapy. Herein, we identified Nbs against CD33, a marker for acute myeloid leukemia (AML). A total of 12 Nbs were generated against recombinant CD33 protein, out of which six bound natively CD33 protein, expressed on the surface of acute myeloid leukemia THP-1 cells. The equilibrium dissociation constants (KD) of these six Nbs and CD33 range from 4 to 270 nM, and their melting temperature (Tm) varies between 52.67 and 67.80 °C. None of these Nbs showed leukemogenicity activity in vitro. The selected six candidates were radiolabeled with 99mTc, and their biodistribution was evaluated in THP-1-tumor-bearing mice. The imaging results demonstrated the fast tumor-targeting capacity of the Nbs in vivo. Among the anti-CD33 Nbs, Nb_7 showed the highest tumor uptake (2.53 ± 0.69 % injected activity per gram (IA/g), with low background signal, except in the kidneys and bladder. Overall, Nb_7 exhibits the best characteristics to be used as an anti-CD33 targeting vehicle for future diagnostic or therapeutic applications.


Subject(s)
Leukemia, Myeloid, Acute/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Single-Domain Antibodies/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Epitopes/immunology , Female , Humans , Kinetics , Mice , Mice, SCID , Sialic Acid Binding Ig-like Lectin 3/genetics , Single-Domain Antibodies/genetics , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Transition Temperature
SELECTION OF CITATIONS
SEARCH DETAIL