Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Mol Neurodegener ; 19(1): 42, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802940

ABSTRACT

Microglia play diverse pathophysiological roles in Alzheimer's disease (AD), with genetic susceptibility factors skewing microglial cell function to influence AD risk. CD33 is an immunomodulatory receptor associated with AD susceptibility through a single nucleotide polymorphism that modulates mRNA splicing, skewing protein expression from a long protein isoform (CD33M) to a short isoform (CD33m). Understanding how human CD33 isoforms differentially impact microglial cell function in vivo has been challenging due to functional divergence of CD33 between mice and humans. We address this challenge by studying transgenic mice expressing either of the human CD33 isoforms crossed with the 5XFAD mouse model of amyloidosis and find that human CD33 isoforms have opposing effects on the response of microglia to amyloid-ß (Aß) deposition. Mice expressing CD33M have increased Aß levels, more diffuse plaques, fewer disease-associated microglia, and more dystrophic neurites compared to 5XFAD control mice. Conversely, CD33m promotes plaque compaction and microglia-plaque contacts, and minimizes neuritic plaque pathology, highlighting an AD protective role for this isoform. Protective phenotypes driven by CD33m are detected at an earlier timepoint compared to the more aggressive pathology in CD33M mice that appears at a later timepoint, suggesting that CD33m has a more prominent impact on microglia cell function at earlier stages of disease progression. In addition to divergent roles in modulating phagocytosis, scRNAseq and proteomics analyses demonstrate that CD33m+ microglia upregulate nestin, an intermediate filament involved in cell migration, at plaque contact sites. Overall, our work provides new functional insights into how CD33, as a top genetic susceptibility factor for AD, modulates microglial cell function.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Mice, Transgenic , Microglia , Protein Isoforms , Sialic Acid Binding Ig-like Lectin 3 , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Microglia/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , Humans , Mice , Protein Isoforms/metabolism , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
2.
ACS Cent Sci ; 10(2): 447-459, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38435526

ABSTRACT

Streptococcus gordonii is a Gram-positive bacterial species that typically colonizes the human oral cavity, but can also cause local or systemic diseases. Serine-rich repeat (SRR) glycoproteins exposed on the S. gordonii bacterial surface bind to sialylated glycans on human salivary, plasma, and platelet glycoproteins, which may contribute to oral colonization as well as endocardial infections. Despite a conserved overall domain organization of SRR adhesins, the Siglec-like binding regions (SLBRs) are highly variable, affecting the recognition of a wide range of sialoglycans. SLBR-N from the SRR glycoprotein of S. gordonii UB10712 possesses the remarkable ability to recognize complex core 2 O-glycans. We here employed a multidisciplinary approach, including flow cytometry, native mass spectrometry, isothermal titration calorimetry, NMR spectroscopy from both protein and ligand perspectives, and computational methods, to investigate the ligand specificity and binding preferences of SLBR-N when interacting with mono- and disialylated core 2 O-glycans. We determined the means by which SLBR-N preferentially binds branched α2,3-disialylated core 2 O-glycans: a selected conformation of the 3'SLn branch is accommodated into the main binding site, driving the sTa branch to further interact with the protein. At the same time, SLBR-N assumes an open conformation of the CD loop of the glycan-binding pocket, allowing one to accommodate the entire complex core 2 O-glycan. These findings establish the basis for the generation of novel tools for the detection of specific complex O-glycan structures and pave the way for the design and development of potential therapeutics against streptococcal infections.

3.
ACS Chem Biol ; 18(10): 2156-2162, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37556411

ABSTRACT

CRISPR-Cas9 is currently the most versatile technique to perform gene editing in living organisms. In this approach, the Cas9 endonuclease is guided toward its DNA target sequence by the guide RNA (gRNA). Chemical synthesis of a functional single gRNA (sgRNA) is nontrivial because of the length of the RNA strand. Recently we demonstrated that a sgRNA can be stitched together from three smaller fragments through a copper-catalyzed azide-alkyne cycloaddition, making the process highly modular. Here we further advance this approach by leveraging this modulator platform by incorporating chemically modified nucleotides at both ends of the modular sgRNA to increase resistance against ribonucleases. Modified nucleotides consisted of a 2'-O-Me group and a phosphorothioate backbone in varying number at both the 5'- and 3'-ends of the sgRNA. It was observed that three modified nucleotides at both ends of the sgRNA significantly increased the success of Cas9 in knocking out a gene of interest. Using these chemically stabilized sgRNAs facilitates multigene editing at the protein level, as demonstrated by successful knockout of both Siglec-3 and Siglec-7 using two fluorophores in conjunction with fluorescence-activated cell sorting. These results demonstrate the versatility of this modular platform for assembling sgRNAs from small, chemically modified strands to simultaneously disrupt the gene expression of two proteins.


Subject(s)
Gene Editing , RNA, Guide, CRISPR-Cas Systems , Gene Editing/methods , CRISPR-Cas Systems/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Nucleotides
4.
Bioconjug Chem ; 33(5): 858-868, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35436106

ABSTRACT

Gene-editing systems such as CRISPR-Cas9 readily enable individual gene phenotypes to be studied through loss of function. However, in certain instances, gene compensation can obfuscate the results of these studies, necessitating the editing of multiple genes to properly identify biological pathways and protein function. Performing multiple genetic modifications in cells remains difficult due to the requirement for multiple rounds of gene editing. While fluorescently labeled guide RNAs (gRNAs) are routinely used in laboratories for targeting CRISPR-Cas9 to disrupt individual loci, technical limitations in single gRNA (sgRNA) synthesis hinder the expansion of this approach to multicolor cell sorting. Here, we describe a modular strategy for synthesizing sgRNAs where each target sequence is conjugated to a unique fluorescent label, which enables fluorescence-activated cell sorting (FACS) to isolate cells that incorporate the desired combination of gene-editing constructs. We demonstrate that three short strands of RNA functionalized with strategically placed 5'-azide and 3'-alkyne terminal deoxyribonucleotides can be assembled in a one-step, template-assisted, copper-catalyzed alkyne-azide cycloaddition to generate fully functional, fluorophore-modified sgRNAs. Using these synthetic sgRNAs in combination with FACS, we achieved selective cleavage of two targeted genes, either separately as a single-color experiment or in combination as a dual-color experiment. These data indicate that our strategy for generating double-clicked sgRNA allows for Cas9 activity in cells. By minimizing the size of each RNA fragment to 41 nucleotides or less, this strategy is well suited for custom, scalable synthesis of sgRNAs.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Alkynes , Azides/metabolism , CRISPR-Cas Systems/genetics , Gene Editing/methods , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
5.
ACS Chem Biol ; 16(11): 2673-2689, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34661385

ABSTRACT

The immunomodulatory family of Siglecs recognizes sialic acid-containing glycans as "self", which is exploited in cancer for immune evasion. The biochemical nature of Siglec ligands remains incompletely understood, with emerging evidence suggesting the importance of carbohydrate sulfation. Here, we investigate how specific sulfate modifications affect Siglec ligands by overexpressing eight carbohydrate sulfotransferases (CHSTs) in five cell lines. Overexpression of three CHSTs─CHST1, CHST2, or CHST4─significantly enhance the binding of numerous Siglecs. Unexpectedly, two other CHSTs (Gal3ST2 and Gal3ST3) diminish Siglec binding, suggesting a new mode to modulate Siglec ligands via sulfation. Results are cell type dependent, indicating that the context in which sulfated glycans are presented is important. Moreover, a pharmacological blockade of N- and O-glycan maturation reveals a cell-type-specific pattern of importance for either class of glycan. Production of a highly homogeneous Siglec-3 (CD33) fragment enabled a mass-spectrometry-based binding assay to determine ≥8-fold and ≥2-fold enhanced affinity for Neu5Acα2-3(6-O-sulfo)Galß1-4GlcNAc and Neu5Acα2-3Galß1-4(6-O-sulfo)GlcNAc, respectively, over Neu5Acα2-3Galß1-4GlcNAc. CD33 shows significant additivity in affinity (≥28-fold) for the disulfated ligand, Neu5Acα2-3(6-O-sulfo)Galß1-4(6-O-sulfo)GlcNAc. Moreover, joint overexpression of CHST1 with CHST2 in cells greatly enhanced the binding of CD33 and several other Siglecs. Finally, we reveal that CHST1 is upregulated in numerous cancers, correlating with poorer survival rates and sodium chlorate sensitivity for the binding of Siglecs to cancer cell lines. These results provide new insights into carbohydrate sulfation as a general mechanism for tuning Siglec ligands on cells, including in cancer.


Subject(s)
Carbohydrate Metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Sulfates/metabolism , Cell Line , Down-Regulation , Humans , Ligands , Mass Spectrometry , N-Acetylneuraminic Acid/metabolism , Neoplasms/metabolism , Protein Binding , Protein Processing, Post-Translational , Up-Regulation
6.
J Control Release ; 338: 680-693, 2021 10 10.
Article in English | MEDLINE | ID: mdl-34517042

ABSTRACT

CD33 is an immunomodulatory receptor expressed by microglia and genetically linked to Alzheimer's disease (AD) susceptibility. While antibodies targeting CD33 have entered clinical trials to treat neurodegeneration, it is unknown whether the glycan-binding properties of CD33 can be exploited to modulate microglia. Here, we use liposomes that multivalently display glycan ligands of CD33 (CD33L liposomes) to engage CD33. We find that CD33L liposomes increase phagocytosis of cultured monocytic cells and microglia in a CD33-dependent manner. Enhanced phagocytosis strongly correlates with loss of CD33 from the cell surface and internalization of liposomes. Increased phagocytosis by treatment with CD33L liposomes is dependent on a key intracellular signaling motif on CD33 as well as the glycan-binding ability of CD33. These effects are specific to trans engagement of CD33 by CD33L liposomes, as cis engagement through insertion of lipid-linked CD33L into cells produces the opposite effect on phagocytosis. Moreover, intracerebroventricular injection of CD33L liposomes into transgenic mice expressing human CD33 in the microglial cell lineage enhances phagocytosis of microglia in a CD33-dependent manner. These results demonstrate that multivalent engagement of CD33 with glycan ligands can modulate microglial cell function.


Subject(s)
Alzheimer Disease , Microglia , Alzheimer Disease/drug therapy , Animals , Ligands , Liposomes , Mice , Phagocytosis , Polysaccharides
7.
Mol Neurodegener ; 16(1): 19, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33766097

ABSTRACT

BACKGROUND: CD33 is genetically linked to Alzheimer's disease (AD) susceptibility through differential expression of isoforms in microglia. The role of the human CD33 short isoform (hCD33m), preferentially encoded by an AD-protective CD33 allele (rs12459419T), is unknown. Here, we test whether hCD33m represents a loss-of-function or gain-of-function variant. METHODS: We have developed two models to test the role of hCD33m. The first is a new strain of transgenic mice expressing hCD33m in the microglial cell lineage. The second is U937 cells where the CD33 gene was disrupted by CRISPR/Cas9 and complemented with different variants of hCD33. Primary microglia and U937 cells were tested in phagocytosis assays and single cell RNA sequencing (scRNAseq) was carried out on the primary microglia. Furthermore, a new monoclonal antibody was developed to detect hCD33m more efficiently. RESULTS: In both primary microglia and U937 cells, we find that hCD33m enhances phagocytosis. This contrasts with the human CD33 long isoform (hCD33M) that represses phagocytosis, as previously demonstrated. As revealed by scRNAseq, hCD33m+ microglia are enriched in a cluster of cells defined by an upregulated expression and gene regulatory network of immediate early genes, which was further validated within microglia in situ. Using a new hCD33m-specific antibody enabled hCD33m expression to be examined, demonstrating a preference for an intracellular location. Moreover, this newly discovered gain-of-function role for hCD33m is dependent on its cytoplasmic signaling motifs, dominant over hCD33M, and not due to loss of glycan ligand binding. CONCLUSIONS: These results provide strong support that hCD33m represents a gain-of-function isoform and offers insight into what it may take to therapeutically capture the AD-protective CD33 allele.


Subject(s)
Amyloid beta-Peptides/metabolism , Microglia/physiology , Peptide Fragments/metabolism , Phagocytosis/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Alleles , Animals , CRISPR-Cas Systems , Crosses, Genetic , Female , Gain of Function Mutation , Gene Editing , Gene Regulatory Networks , Genes, Immediate-Early , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Polysaccharides/metabolism , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA-Seq , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 3/physiology , Single-Cell Analysis , U937 Cells
8.
Ophthalmol Sci ; 1(2): 100028, 2021 Jun.
Article in English | MEDLINE | ID: mdl-36249295

ABSTRACT

Purpose: Peroxisomal biogenesis disorders (PBDs) represent a spectrum of conditions that result in vision loss, sensorineural hearing loss, neurologic dysfunction, and other abnormalities resulting from aberrant peroxisomal function caused by mutations in PEX genes. With no treatments currently available, we sought to investigate the disease mechanism in a patient with a PBD caused by defects in PEX6 and to probe whether overexpression of PEX6 could restore peroxisome function and potentially offer therapeutic benefit. Design: Laboratory-based study. Participants: A 12-year-old boy sought treatment with hearing loss and retinopathy. After negative results in an Usher syndrome panel, targeted genetic testing revealed compound heterozygous mutations in PEX6. These included a 14-nucleotide deletion (c.802_815del: p.(Asp268Cysfs∗8)) and a milder missense variant (c.35T→C:(p.Phe12Ser)). Methods: Patient-derived skin fibroblasts were cultured, and a PEX6 knockout cell line was developed using clustered regularly interspaced short palindromic repeats and Cas9 technology in HEK293T cells to emulate a more severe disease phenotype. Immunoblot analysis of whole cell lysates was performed to assess peroxisome number. Immunofluorescence studies used antibodies against components of the peroxisomal protein import pathway to interrogate the effects of mutations in PEX6 on protein trafficking. Main Outcome Measures: Primary outcome measures were peroxisome abundance and matrix protein import. Results: Peroxisome number was not significantly different between control fibroblasts and patient fibroblasts; however, fewer peroxisomes were observed in PEX6 knockout cells compared with wild-type cells (P = 0.04). Analysis by immunofluorescent microscopy showed significantly impaired peroxisomal targeting signal 1- and peroxisomal targeting signal 2-mediated matrix protein import in both patient fibroblasts and PEX6 knockout cells. Overexpressing PEX6 resulted in improved matrix protein import in PEX6 knockout cells. Conclusions: Mutations in PEX6 were responsible for combined hearing loss and retinopathy in our patient. The primary peroxisomal defect in our patient's skin fibroblasts was impaired peroxisomal protein import as opposed to reduction in the number of peroxisomes. Genetic strategies that introduce wild-type PEX6 into cells deficient in PEX6 protein show promise in restoring peroxisome function. Future studies of patient-specific induced pluripotent stem cell-derived retinal pigment epithelium cells may clarify the role of PEX6 in the retina and the potential for gene therapy in these patients.

9.
Nat Commun ; 11(1): 5091, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037195

ABSTRACT

Sialic acid-binding immunoglobulin-type lectins (Siglecs) are immunomodulatory receptors that are regulated by their glycan ligands. The connections between Siglecs and human disease motivate improved methods to detect Siglec ligands. Here, we describe a new versatile set of Siglec-Fc proteins for glycan ligand detection. Enhanced sensitivity and selectivity are enabled through multimerization and avoiding Fc receptors, respectively. Using these Siglec-Fc proteins, Siglec ligands are systematically profiled on healthy and cancerous cells and tissues, revealing many unique patterns. Additional features enable the production of small, homogenous Siglec fragments and development of a quantitative ligand-binding mass spectrometry assay. Using this assay, the ligand specificities of several Siglecs are clarified. For CD33 (Siglec-3), we demonstrate that it recognizes both α2-3 and α2-6 sialosides in solution and on cells, which has implications for its link to Alzheimer's disease susceptibility. These soluble Siglecs reveal the abundance of their glycan ligands on host cells as self-associated molecular patterns.


Subject(s)
Polysaccharides/analysis , Sialic Acid Binding Immunoglobulin-like Lectins/chemistry , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Breast Neoplasms/metabolism , CHO Cells , Cricetulus , Female , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , K562 Cells , Mass Spectrometry , Polysaccharides/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Sialic Acid Binding Immunoglobulin-like Lectins/isolation & purification , Sialic Acids/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism , Spleen/cytology , Spleen/metabolism , Streptavidin/metabolism
11.
Commun Biol ; 2: 450, 2019.
Article in English | MEDLINE | ID: mdl-31815204

ABSTRACT

CD33 is an immunomodulatory receptor linked to Alzheimer's disease (AD) susceptibility via regulation of phagocytosis in microglia. Divergent features between human CD33 (hCD33) and murine CD33 (mCD33) include a unique transmembrane lysine in mCD33 and cytoplasmic tyrosine in hCD33. The functional consequences of these differences in restraining phagocytosis remains poorly understood. Using a new αmCD33 monoclonal antibody, we show that mCD33 is expressed at high levels on neutrophils and low levels on microglia. Notably, cell surface expression of mCD33 is entirely dependent on Dap12 due to an interaction with the transmembrane lysine in mCD33. In RAW264.7 cultured macrophages, BV-2 cultured microglia, primary neonatal and adult microglia, uptake of cargo - including aggregated Aß1-42 - is not altered upon genetic ablation of mCD33. Alternatively, deletion of hCD33 in monocytic cell lines increased cargo uptake. Moreover, transgenic mice expressing hCD33 in the microglial cell lineage showed repressed cargo uptake in primary microglia. Therefore, mCD33 and hCD33 have divergent roles in regulating phagocytosis, highlighting the importance of studying hCD33 in AD susceptibility.

12.
Antiviral Res ; 171: 104566, 2019 11.
Article in English | MEDLINE | ID: mdl-31348951

ABSTRACT

As a part of innate immune defense, the role of mast cells during viral replication has been incompletely understood. In this study, we characterized and compared the responses of the human mast cell line, LAD2, and human lung epithelial cell line, Calu-3, against three influenza A virus strains; A/PR/8/34 (H1N1), A/WS/33 (H1N1) and A/HK/8/68 (H3N2). We found that there were strain-dependent mast cell responses, and different profiles of cytokine, chemokine and antiviral gene expression between the two cell types. All three strains did not induce histamine or ß-hexosaminidase release in LAD2. A/HK/8/68 induced release of prostaglandin D2 in LAD2, whereas A/PR/8/34 and A/WS/33 did not. We found that, among those examined, only CCL4 (by A/PR/8/34) was statistically significantly released from LAD2 cells. Furthermore, there was increased mRNA expression of viral recognition receptors (RIG-I and MDA5) and antiviral protein, viperin, but levels and kinetics of the expression were different among the cell types, as well as by the strains examined. Our findings highlight the variability in innate response to different strains of influenza A virus in two human cell types, indicating that further investigation is needed to understand better the role of mast cells and epithelial cells in innate immunity against influenza A viruses.


Subject(s)
Epithelial Cells/immunology , Epithelial Cells/virology , Host-Pathogen Interactions/immunology , Influenza A virus/immunology , Influenza, Human/immunology , Mast Cells/immunology , Mast Cells/virology , Biomarkers , Cell Line , Cytokines/metabolism , Disease Resistance/genetics , Epithelial Cells/metabolism , Humans , Influenza A virus/classification , Influenza, Human/genetics , Influenza, Human/metabolism , Influenza, Human/virology , Mast Cells/metabolism , Virus Release , Virus Replication
13.
Curr Opin Ophthalmol ; 28(5): 410-415, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28520608

ABSTRACT

PURPOSE OF REVIEW: Although much has been written to define the phenotype and genotype of choroideremia (CHM), research continues to provide new insights that serve to better understand its pathogenesis and the directions for potential experimental therapies. RECENT FINDINGS: We would like to highlight new findings, expanding the type of disease-causing mutations to include mutations in the CHM promoter that will dramatically influence gene expression. Information derived from careful phenotyping of patients points increasingly to the central role of the retinal pigment epithelium as the key cell layer affected in the degenerative process. Finally, we will review the current initiatives that are testing vector-mediated gene replacement approaches in humans, including our current understanding of the likelihood of success by this approach. SUMMARY: Clinical and basic vision science have benefited greatly by the active engagement of patients with CHM in clinical research studies. The impetus for their involvement in these studies has been generated by the initial results of safety from subretinal injection of and AAV2.REP1 vector in humans. Follow-up studies in the next few years are expected to show if this approach will modify disease progression.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Choroideremia , Genetic Therapy/methods , Mutation , Retinal Pigment Epithelium/pathology , Choroideremia/diagnosis , Choroideremia/genetics , Choroideremia/therapy , Humans , Phenotype
14.
Am J Physiol Regul Integr Comp Physiol ; 312(6): R1004-R1016, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28381457

ABSTRACT

Calcium-binding protein spermatid-specific 1 (CABS1) is expressed in the human submandibular gland and has an anti-inflammatory motif similar to that in submandibular rat 1 in rats. Here, we investigate CABS1 in human saliva and its association with psychological and physiological distress and inflammation in humans. Volunteers participated across three studies: 1) weekly baseline measures; 2) a psychosocial speech and mental arithmetic stressor under evaluative threat; and 3) during academic exam stress. Salivary samples were analyzed for CABS1 and cortisol. Additional measures included questionnaires of perceived stress and negative affect; exhaled nitric oxide; respiration and cardiac activity; lung function; and salivary and nasal inflammatory markers. We identified a CABS1 immunoreactive band at 27 kDa in all participants and additional molecular mass forms in some participants. One week temporal stability of the 27-kDa band was satisfactory (test-retest reliability estimate = 0.62-0.86). Acute stress increased intensity of 18, 27, and 55 kDa bands; 27-kDa increases were associated with more negative affect and lower heart rate, sympathetic activity, respiration rate, and minute ventilation. In both acute and academic stress, changes in 27 kDa were positively associated with salivary cortisol. The 27-kDa band was also positively associated with VEGF and salivary leukotriene B4 levels. Participants with low molecular weight CABS1 bands showed reduced habitual stress and negative affect in response to acute stress. CABS1 is readily detected in human saliva and is associated with psychological and physiological indicators of stress. The role of CABS1 in inflammatory processes, stress, and stress resilience requires careful study.


Subject(s)
Calcium-Binding Proteins/metabolism , Saliva/metabolism , Stress, Psychological/metabolism , Adolescent , Adult , Affect , Asthma/metabolism , Asthma/physiopathology , Asthma/psychology , Biomarkers/metabolism , Female , Forced Expiratory Volume , Heart Rate , Humans , Hydrocortisone/metabolism , Leukotriene B4/metabolism , Male , Mathematical Concepts , Middle Aged , Molecular Weight , Respiratory Rate , Speech , Stress, Psychological/etiology , Stress, Psychological/physiopathology , Stress, Psychological/psychology , Time Factors , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , Young Adult
15.
Am J Physiol Regul Integr Comp Physiol ; 308(7): R569-75, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25632019

ABSTRACT

Salivary glands are involved in the production and exocrine and endocrine secretion of biologically active proteins, polypeptides, and hormones involved in growth and differentiation, homeostasis, and digestion. We have previously studied the prohormone submandibular rat 1 (SMR1), product of the Vcsa1 gene, which is highly expressed in the testes and salivary glands of rats, and can be cleaved to produce polypeptides with analgesic, erectile function, and anti-inflammatory activities. Humans lack the Vcsa1 gene, but homologous sequences and functions for analgesia and erectile function exist in the human genes Prol1, SMR3a, and SMR3b located on the human chromosomal region close to where Vcsa1 lies in the rat. Here we show the human protein calcium-binding protein spermatid-specific 1 (CABS1) contains a similar sequence to the anti-inflammatory sequence in rat SMR1, thus CABS1 may be another human gene with homologous function to Vcsa1. Using Western blot and PCR, we discovered that the human protein CABS1, previously thought to only be expressed in the testes, is also expressed in the salivary glands and lung, in a tissue-specific manner. Peptides derived from CABS1 were tested in an in vivo mouse model of lipopolysaccharide (LPS)-induced neutrophilia and an ex vivo rat model of antigen-induced intestinal anaphylaxis and significantly reduced both neutrophil accumulation in bronchoalveolar lavage fluid and antigen-induced ileal contractions, respectively. Thus human CABS1 has a peptide motif homologous to the anti-inflammatory peptide sequence of rat SMR1. Whether this similarity of CABS1 extends to the neuroendocrine regulation of the anti-inflammatory activity seen for SMR1 remains to be determined.


Subject(s)
Anaphylaxis/prevention & control , Anti-Inflammatory Agents/pharmacology , Calcium-Binding Proteins/pharmacology , Peptide Fragments/pharmacology , Pneumonia/prevention & control , Salivary Glands/metabolism , Salivary Proteins and Peptides/pharmacology , Amino Acid Motifs , Anaphylaxis/chemically induced , Anaphylaxis/immunology , Anaphylaxis/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Lipopolysaccharides , Male , Mice, Inbred BALB C , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Ovalbumin , Peptide Fragments/metabolism , Pneumonia/chemically induced , Pneumonia/immunology , Pneumonia/metabolism , Protein Precursors/metabolism , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Salivary Proteins and Peptides/genetics , Salivary Proteins and Peptides/metabolism
16.
Methods Mol Biol ; 1220: 339-45, 2015.
Article in English | MEDLINE | ID: mdl-25388261

ABSTRACT

The production of nitric oxide in mast cells has been difficult to measure due to the low amounts made by mast cells, as well as limitations in the specificity and sensitivity of the assays available. We present here a sensitive and specific 96-well plate-based method to directly measure NO using the cell-permeable fluorescent compound DAF-FM diacetate.


Subject(s)
Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Mast Cells/metabolism , Nitric Oxide/metabolism , Spectrometry, Fluorescence/methods , Animals , Cell Line , Humans , Mice
17.
Immunol Res ; 56(1): 32-43, 2013 May.
Article in English | MEDLINE | ID: mdl-23055084

ABSTRACT

Mast cells are important in innate immunity and protective against certain bacterial infections. However, there is limited evidence that mast cells respond to viruses. As mast cells are abundant in mucosal tissues of the lung, they are in a prime location to detect and respond to influenza virus. In this study, we characterized for the first time the replication cycle of influenza A virus in human mast cells by measuring influenza A virus transcription, RNA replication, protein synthesis, and formation of infectious virus as compared to the replication cycle in epithelial cells. We detected the presence of influenza A viral genomic RNA transcription, replication, and protein synthesis in human mast cells and epithelial cells. However, there was no significant release of infectious influenza A virus from mast cells, whereas epithelial cells produce ~100-fold virus compared with the inoculating dose. We confirmed that influenza A virus infects human mast cells, begins to replicate, but the production of new virus is aborted. Thus, mast cells may lack critical factors essential for productive infection or there are intrinsic or inducible anti-influenza A mechanisms in mast cells.


Subject(s)
Influenza A virus/physiology , Influenza, Human/immunology , Influenza, Human/virology , Mast Cells/virology , RNA, Viral/analysis , Virus Replication , Cells, Cultured , Epithelial Cells/immunology , Epithelial Cells/virology , Humans , Immunity, Innate , Influenza A virus/pathogenicity , Lung/pathology , Mast Cells/immunology , Transcription, Genetic , Virion/growth & development , Virion/immunology
18.
Chem Immunol Allergy ; 98: 176-95, 2012.
Article in English | MEDLINE | ID: mdl-22767064

ABSTRACT

The cervical sympathetic nerves which innervate the medial basal hypothalamus-hypophyseal complex, primary and secondary lymph organs, and numerous glands, such as the pineal, thyroid, parathyroid and salivary glands form a relevant neuroimmunoendocrine structure that is involved in the regulation of systemic homeostasis. The superior cervical ganglia and the submandibular glands form a 'neuroendocrine axis' called the cervical sympathetic trunk submandibular gland (CST-SMG) axis. The identification of this axis usurps the traditional view of salivary glands as accessory digestive structures and reinforces the view that they are important sources of systemically active immunoregulatory and anti-inflammatory factors whose release is intimately controlled by the autonomic nervous system, and in particular the sympathetic branch. An end component of the CST-SMG axis is the synthesis, processing and release of submandibular rat-1 protein (SMR1), a prohormone, that generates several different peptides, one from near its N-terminus called sialorphin and another from its C-terminus called - submandibular gland peptide-T (SGP-T). SGP-T formed the template for tripeptide fragment (FEG) and its metabolically stable D-isomeric peptide feG, which are potent inhibitors of allergy and asthma (IgE-mediated allergic reactions) and several non-IgE-mediated inflammations. The translation from rat genetics and proteomics to humans has yielded structural and functional correlates that hopefully will lead to the development of new medications and therapeutic approaches for difficult to treat disorders. Although the CST-SMG axis has barely been explored in humans recognition of the importance of this axis could facilitate an understanding and improved management of periodontal disease, and other diseases with a more systemic and nervous system basis such as asthma, autoimmunity, graft-versus-host disease and even Parkinson's disease.


Subject(s)
Salivary Glands/physiopathology , Sympathetic Nervous System/physiopathology , Animals , Genomics , Humans , Immunoglobulin E/metabolism , Inflammation/physiopathology , Isomerism , Neuroimmunomodulation , Neurosecretory Systems/physiopathology , Oligopeptides/metabolism , Protein Precursors/metabolism , Proteomics , Rats , Salivary Glands/innervation , Salivary Proteins and Peptides/metabolism , Submandibular Gland/innervation , Submandibular Gland/physiopathology
19.
Ann Allergy Asthma Immunol ; 108(3): 172-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22374200

ABSTRACT

BACKGROUND: Nitric oxide (NO) has various roles in airway physiology and pathophysiology. Monitoring exhaled NO levels is increasingly common to measure airways inflammation and inhaled NO studied for its therapeutic value in premature infants and adult respiratory distress syndrome. NO is produced by 3 isoforms of NO synthase (NOS1, 2, 3), and each can play distinct and perhaps overlapping roles in the airways. However, the distribution, regulation, and functions of NOS in various cells in the upper airways, particularly in leukocytes, are incompletely understood. OBJECTIVE: To characterize the expression of NOS isoforms in leukocytes in normal middle turbinate tissues (MT) and in inflammatory nasal tissue (nasal polyps, NP). METHODS: Normal MT tissue was collected from surgical specimens that were to be discarded. The NP samples were from surgical tissue archives of 15 patients with chronic rhinosinusitis. Isoforms of NOS in cells were identified by double immunostaining using NOS isoform-specific and leukocyte-specific (mast cell, eosinophil, macrophage, neutrophil, or T cell) antibodies. RESULTS: The proportion of total cells below the epithelium that were positive for each isoform of NOS was higher in NP than in MT. Each isoform of NOS was found in all leukocyte populations studied, and there were significant differences in the percentage of leukocytes expressing NOS isoforms between MT and NP. CONCLUSION: All isoforms of NOS are expressed in leukocytes in MT and NP, and their expression varies among leukocyte types. Our data provide a basis to investigate the regulation, cell distribution, and distinct functions of NOS isoforms in normal and inflamed nasal tissues.


Subject(s)
Leukocytes/enzymology , Nasal Polyps/enzymology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type I/metabolism , Adult , Eosinophils/enzymology , Eosinophils/immunology , Female , Humans , Inflammation/immunology , Leukocytes/immunology , Macrophages/enzymology , Macrophages/immunology , Male , Mast Cells/enzymology , Mast Cells/immunology , Middle Aged , Nasal Mucosa/enzymology , Nasal Mucosa/immunology , Nasal Mucosa/pathology , Nasal Polyps/immunology , Nasal Polyps/pathology , Nitric Oxide/biosynthesis , Turbinates/enzymology
20.
Am J Physiol Lung Cell Mol Physiol ; 297(6): L1091-102, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19801448

ABSTRACT

Acute lung injury (ALI) is an inflammatory disorder associated with recruitment and activation of neutrophils in lungs. Rac2, a member of the Rho GTPase subfamily, is an essential regulator of neutrophil degranulation, superoxide release, and chemotaxis. Here, we hypothesized that Rac2 is important in mediating lung injury. Using a model of IgG immune complex-mediated ALI, we showed that injury was attenuated in rac2(-/-) mice compared with wild-type (WT) mice undergoing ALI, with significant decreases in alveolar leukocyte numbers, vascular leakage, and the inflammatory mediators, myeloperoxidase (MPO) and matrix metalloproteinases (MMPs). Reduced injury in rac2(-/-) mice was not associated with diminished cytokine and chemokine production, since bronchoalveolar lavage (BAL) levels of IL-17, TNF, CCL3, CXCL1, and CXCL2 were similarly increased in WT and rac2(-/-) mice with ALI compared with sham-treated mice (no ALI). BAL levels of MMP-2 and MMP-9 were significantly decreased in the airways of rac2(-/-) mice with ALI. Immunohistochemical analysis revealed that MMP-2 and MMP-9 expression was evident in alveolar macrophages and interstitial neutrophils in WT ALI. In contrast, MMP-positive cells were less prominent in rac2(-/-) mice with ALI. Chimeric mice showed that Rac2-mediated lung injury was dependent on hematopoietic cells derived from bone marrow. We propose that lung injury in response to immune complex deposition is dependent on Rac2 in alveolar macrophages and neutrophils.


Subject(s)
Acute Lung Injury/complications , Acute Lung Injury/immunology , Antigen-Antibody Complex/immunology , Inflammation/complications , Inflammation/immunology , rac GTP-Binding Proteins/metabolism , Acute Lung Injury/enzymology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Movement , Chemokines/biosynthesis , Epithelial Cells/pathology , Hematopoietic Stem Cells/pathology , Inflammation Mediators/metabolism , Lung/enzymology , Lung/immunology , Lung/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Neutrophils/pathology , Peroxidase/metabolism , Superoxides/metabolism , rac GTP-Binding Proteins/deficiency , RAC2 GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...