Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Immunol ; 211(5): 853-861, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37477694

ABSTRACT

APCs such as dendritic cells and macrophages play a pivotal role in mediating immune tolerance and restoring intestinal immune homeostasis by limiting inflammatory responses against commensal bacteria. However, cell-intrinsic molecular regulators critical for programming intestinal APCs to a regulatory state rather than an inflammatory state are unknown. In this study, we report that the transcription factor retinoid X receptor α (RXRα) signaling in CD11c+ APCs is essential for suppressing intestinal inflammation by imparting an anti-inflammatory phenotype. Using a mouse model of ulcerative colitis, we demonstrated that targeted deletion of RXRα in CD11c+ APCs in mice resulted in the loss of T cell homeostasis with enhanced intestinal inflammation and increased histopathological severity of colonic tissue. This was due to the increased production of proinflammatory cytokines that drive Th1/Th17 responses and decreased expression of immune-regulatory factors that promote regulatory T cell differentiation in the colon. Consistent with these findings, pharmacological activation of the RXRα pathway alleviated colitis severity in mice by suppressing the expression of inflammatory cytokines and limiting Th1/Th17 cell differentiation. These findings identify an essential role for RXRα in APCs in regulating intestinal immune homeostasis and inflammation. Thus, manipulating the RXRα pathway could provide novel opportunities for enhancing regulatory responses and dampening colonic inflammation.


Subject(s)
Colitis , Transcription Factors , Animals , Mice , Colon , Cytokines/metabolism , Homeostasis , Inflammation , Intestinal Mucosa , Intestines/pathology , Mice, Inbred C57BL , Retinoid X Receptor alpha , Transcription Factors/metabolism
2.
J Immunol ; 209(2): 368-378, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35760519

ABSTRACT

Extraintestinal manifestations are common in inflammatory bowel disease and involve several organs, including the kidney. However, the mechanisms responsible for renal manifestation in inflammatory bowel disease are not known. In this study, we show that the Wnt-lipoprotein receptor-related proteins 5 and 6 (LRP5/6) signaling pathway in macrophages plays a critical role in regulating colitis-associated systemic inflammation and renal injury in a murine dextran sodium sulfate-induced colitis model. Conditional deletion of the Wnt coreceptors LRP5/6 in macrophages in mice results in enhanced susceptibility to dextran sodium sulfate colitis-induced systemic inflammation and acute kidney injury (AKI). Furthermore, our studies show that aggravated colitis-associated systemic inflammation and AKI observed in LRP5/6LysM mice are due to increased bacterial translocation to extraintestinal sites and microbiota-dependent increased proinflammatory cytokine levels in the kidney. Conversely, depletion of the gut microbiota mitigated colitis-associated systemic inflammation and AKI in LRP5/6LysM mice. Mechanistically, LRP5/6-deficient macrophages were hyperresponsive to TLR ligands and produced higher levels of proinflammatory cytokines, which are associated with increased activation of MAPKs. These results reveal how the Wnt-LRP5/6 signaling in macrophages controls colitis-induced systemic inflammation and AKI.


Subject(s)
Acute Kidney Injury , Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Microbiota , Acute Kidney Injury/metabolism , Animals , Colitis/chemically induced , Cytokines/metabolism , Dextran Sulfate/toxicity , Inflammation/metabolism , Inflammatory Bowel Diseases/metabolism , Kidney/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Wnt Signaling Pathway/genetics
3.
J Immunol ; 207(5): 1428-1436, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34348977

ABSTRACT

Dendritic cells (DCs) are professional APCs that play a crucial role in initiating robust immune responses against invading pathogens while inducing regulatory responses to the body's tissues and commensal microorganisms. A breakdown of DC-mediated immunological tolerance leads to chronic inflammation and autoimmune disorders. However, cell-intrinsic molecular regulators that are critical for programming DCs to a regulatory state rather than to an inflammatory state are not known. In this study, we show that the activation of the TCF4 transcription factor in DCs is critical for controlling the magnitude of inflammatory responses and limiting neuroinflammation. DC-specific deletion of TCF4 in mice increased Th1/Th17 responses and exacerbated experimental autoimmune encephalomyelitis pathology. Mechanistically, loss of TCF4 in DCs led to heightened activation of p38 MAPK and increased levels of proinflammatory cytokines IL-6, IL-23, IL-1ß, TNF-α, and IL-12p40. Consistent with these findings, pharmacological blocking of p38 MAPK activation delayed experimental autoimmune encephalomyelitis onset and diminished CNS pathology in TCF4ΔDC mice. Thus, manipulation of the TCF4 pathway in DCs could provide novel opportunities for regulating chronic inflammation and represents a potential therapeutic approach to control autoimmune neuroinflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Th1 Cells , Animals , Dendritic Cells , Mice , Mice, Inbred C57BL , Th17 Cells , Transcription Factor 4
4.
Front Immunol ; 12: 691134, 2021.
Article in English | MEDLINE | ID: mdl-34394085

ABSTRACT

For decades, lactate has been considered an innocuous bystander metabolite of cellular metabolism. However, emerging studies show that lactate acts as a complex immunomodulatory molecule that controls innate and adaptive immune cells' effector functions. Thus, recent advances point to lactate as an essential and novel signaling molecule that shapes innate and adaptive immune responses in the intestine and systemic sites. Here, we review these recent advances in the context of the pleiotropic effects of lactate in regulating diverse functions of immune cells in the tissue microenvironment and under pathological conditions.


Subject(s)
Dendritic Cells/immunology , Lactic Acid/immunology , Macrophages/immunology , Animals , Autoimmunity , Cell Cycle Proteins/immunology , Humans , Immunomodulation , Infections/immunology , Inflammatory Bowel Diseases/immunology , Monocarboxylic Acid Transporters/immunology , Neoplasms/immunology , Receptors, G-Protein-Coupled/immunology
5.
J Immunol ; 205(8): 2265-2275, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32917787

ABSTRACT

Loss of immune tolerance to gut microflora is inextricably linked to chronic intestinal inflammation and colitis-associated colorectal cancer (CAC). The LRP5/6 signaling cascade in APCs contributes to immune homeostasis in the gut, but whether this pathway in APCs protects against CAC is not known. In the current study, using a mouse model of CAC, we show that the LRP5/6-ß-catenin-IL-10 signaling axis in intestinal CD11c+ APCs protects mice from CAC by regulating the expression of tumor-promoting inflammatory factors in response to commensal flora. Genetic deletion of LRP5/6 in CD11c+ APCs in mice (LRP5/6ΔCD11c) resulted in enhanced susceptibility to CAC. This is due to a microbiota-dependent increased expression of proinflammatory factors and decreased expression of the immunosuppressive cytokine IL-10. This condition could be improved in LRP5/6ΔCD11c mice by depleting the gut flora, indicating the importance of LRP5/6 in mediating immune tolerance to the gut flora. Moreover, mechanistic studies show that LRP5/6 suppresses the expression of tumor-promoting inflammatory factors in CD11c+ APCs via the ß-catenin-IL-10 axis. Accordingly, conditional activation of ß-catenin specifically in CD11c+ APCs or in vivo administration of IL-10 protected LRP5/6ΔCD11c mice from CAC by suppressing the expression of inflammatory factors. In summary, in this study, we identify a key role for the LRP5/6-ß-catenin-IL-10 signaling pathway in intestinal APCs in resolving chronic intestinal inflammation and protecting against CAC in response to the commensal flora.


Subject(s)
Antigen-Presenting Cells/immunology , Colitis/immunology , Colonic Neoplasms/immunology , Gastrointestinal Microbiome/immunology , Interleukin-10/immunology , Wnt Signaling Pathway/immunology , beta Catenin/immunology , Animals , Antigen-Presenting Cells/pathology , Colitis/complications , Colitis/genetics , Colitis/pathology , Colonic Neoplasms/etiology , Colonic Neoplasms/genetics , Colonic Neoplasms/prevention & control , Gastrointestinal Microbiome/genetics , Interleukin-10/genetics , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics
6.
Oncoimmunology ; 5(4): e1115941, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27141399

ABSTRACT

The tumor microenvironment (TME) contains high levels of the Wnt family of ligands, and aberrant Wnt-signaling occurs in many tumors. Past studies have been directed toward how the Wnt signaling cascade regulates cancer development, progression and metastasis. However, its effects on host antitumor immunity remain unknown. In this report, we show that Wnts in the TME condition dendritic cells (DCs) to a regulatory state and suppress host antitumor immunity. DC-specific deletion of Wnt co-receptors low-density lipoprotein receptor-related protein 5 and 6 (LRP5/6) in mice markedly delayed tumor growth and enhanced host antitumor immunity. Mechanistically, loss of LRP5/6-mediated signaling in DCs resulted in enhanced effector T cell differentiation and decreased regulatory T cell differentiation. This was due to increased production of pro-inflammatory cytokines and decreased production of IL-10, TGF-ß1 and retinoic acid (RA). Likewise, pharmacological inhibition of the Wnts' interaction with its cognate co-receptors LRP5/6 and Frizzled (Fzd) receptors had similar effects on tumor growth and effector T cell responses. Moreover, blocking Wnt-signaling in DCs resulted in enhanced capture of tumor-associated antigens and efficient cross-priming of CD8+ T cells. Hence, blocking the Wnt pathway represents a potential therapeutic to overcome tumor-mediated immune suppression and augment antitumor immunity.

7.
J Immunol ; 196(11): 4739-49, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183583

ABSTRACT

Dietary lipids and their metabolites activate members of the peroxisome proliferative-activated receptor (PPAR) family of transcription factors and are critical for colonic health. The PPARα isoform plays a vital role in regulating inflammation in various disease settings, but its role in intestinal inflammation, commensal homeostasis, and mucosal immunity in the gut are unclear. In this study, we demonstrate that the PPARα pathway in innate immune cells orchestrates gut mucosal immunity and commensal homeostasis by regulating the expression of IL-22 and the antimicrobial peptides RegIIIß, RegIIIγ, and calprotectin. Additionally, the PPARα pathway is critical for imparting regulatory phenotype in intestinal macrophages. PPARα deficiency in mice led to commensal dysbiosis in the gut, resulting in a microbiota-dependent increase in the expression of inflammatory cytokines and enhanced susceptibility to intestinal inflammation. Pharmacological activation of this pathway decreased the expression of inflammatory cytokines and ameliorated colonic inflammation. Taken together, these findings identify a new important innate immune function for the PPARα signaling pathway in regulating intestinal inflammation, mucosal immunity, and commensal homeostasis. Thus, the manipulation of the PPARα pathway could provide novel opportunities for enhancing mucosal immunity and treating intestinal inflammation.


Subject(s)
Gastrointestinal Microbiome/immunology , Homeostasis , Inflammation/prevention & control , PPAR alpha/metabolism , Signal Transduction , Animals , Cells, Cultured , Homeodomain Proteins/immunology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/deficiency
8.
J Immunol ; 194(7): 3295-304, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25710911

ABSTRACT

Breakdown in immunological tolerance to self-Ags or uncontrolled inflammation results in autoimmune disorders. Dendritic cells (DCs) play an important role in regulating the balance between inflammatory and regulatory responses in the periphery. However, factors in the tissue microenvironment and the signaling networks critical for programming DCs to control chronic inflammation and promote tolerance are unknown. In this study, we show that wnt ligand-mediated activation of ß-catenin signaling in DCs is critical for promoting tolerance and limiting neuroinflammation. DC-specific deletion of key upstream (lipoprotein receptor-related protein [LRP]5/6) or downstream (ß-catenin) mediators of canonical wnt signaling in mice exacerbated experimental autoimmune encephalomyelitis pathology. Mechanistically, loss of LRP5/6-ß-catenin-mediated signaling in DCs led to an increased Th1/Th17 cell differentiation but reduced regulatory T cell response. This was due to increased production of proinflammatory cytokines and decreased production of anti-inflammatory cytokines such as IL-10 and IL-27 by DCs lacking LRP5/6-ß-catenin signaling. Consistent with these findings, pharmacological activation of canonical wnt/ß-catenin signaling delayed experimental autoimmune encephalomyelitis onset and diminished CNS pathology. Thus, the activation of canonical wnt signaling in DCs limits effector T cell responses and represents a potential therapeutic approach to control autoimmune neuroinflammation.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway , Animals , Cell Differentiation , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Deletion , Gene Knockout Techniques , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Male , Mice , Mice, Transgenic , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/cytology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Wnt Signaling Pathway/drug effects , Wnt3A Protein/metabolism , beta Catenin/metabolism
9.
Cancer Res ; 75(4): 656-665, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25568183

ABSTRACT

Tumors actively suppress antitumor immunity, creating formidable barriers to successful cancer immunotherapy. The molecular mechanisms underlying tumor-induced immune tolerance are largely unknown. In the present study, we show that dendritic cells (DC) in the tumor microenvironment acquire the ability to metabolize vitamin A to produce retinoic acid (RA), which drives regulatory T-cell responses and immune tolerance. Tolerogenic responses were dependent on induction of vitamin A-metabolizing enzymes via the ß-catenin/T-cell factor (TCF) pathway in DCs. Consistent with this observation, DC-specific deletion of ß-catenin in mice markedly reduced regulatory T-cell responses and delayed melanoma growth. Pharmacologic inhibition of either vitamin A-metabolizing enzymes or the ß-catenin/TCF4 pathway in vivo had similar effects on tumor growth and regulatory T-cell responses. Hence, ß-catenin/TCF4 signaling induces local regulatory DC and regulatory T-cell phenotypes via the RA pathway, identifying this pathway as an important target for anticancer immunotherapy.


Subject(s)
Dendritic Cells/metabolism , Tumor Microenvironment/immunology , Vitamin A/metabolism , beta Catenin/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/pathology , Humans , Mice , Mice, Transgenic , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factor 4 , Tumor Microenvironment/genetics , beta Catenin/metabolism
10.
J Immunol ; 193(8): 4203-13, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25210120

ABSTRACT

Dendritic cells (DCs) sense microbes via multiple innate receptors. Signals from different innate receptors are coordinated and integrated by DCs to generate specific innate and adaptive immune responses against pathogens. Previously, we have shown that two pathogen recognition receptors, TLR2 and dectin-1, which recognize the same microbial stimulus (zymosan) on DCs, induce mutually antagonistic regulatory or inflammatory responses, respectively. How diametric signals from these two receptors are coordinated in DCs to regulate or incite immunity is not known. In this study, we show that TLR2 signaling via AKT activates the ß-catenin/T cell factor 4 pathway in DCs and programs them to drive T regulatory cell differentiation. Activation of ß-catenin/T cell factor 4 was critical to induce regulatory molecules IL-10 (Il-10) and vitamin A metabolizing enzyme retinaldehyde dehydrogenase 2 (Aldh1a2) and to suppress proinflammatory cytokines. Deletion of ß-catenin in DCs programmed them to drive Th17/Th1 cell differentiation in response to zymosan. Consistent with these findings, activation of the ß-catenin pathway in DCs suppressed chronic inflammation and protected mice from Th17/Th1-mediated autoimmune neuroinflammation. Thus, activation of ß-catenin in DCs via the TLR2 receptor is a novel mechanism in DCs that regulates autoimmune inflammation.


Subject(s)
Autoimmunity/immunology , Dendritic Cells/immunology , T-Lymphocytes, Regulatory/cytology , Toll-Like Receptor 2/immunology , beta Catenin/metabolism , Adoptive Transfer , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase 1 Family , Animals , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Inflammation/prevention & control , Interleukin-10/biosynthesis , Lectins, C-Type/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/immunology , Retinal Dehydrogenase , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Transcription Factor 7-Like 2 Protein/immunology , Zymosan/immunology , Zymosan/pharmacology , beta Catenin/genetics
11.
Methods Mol Biol ; 1194: 437-48, 2014.
Article in English | MEDLINE | ID: mdl-25064119

ABSTRACT

Crohn's disease (CD) and ulcerative colitis are two main clinically defined forms of chronic inflammatory bowel disease (IBD). Our understanding of IBD depends largely on rodent models. DSS-induced intestinal inflammation in mice and T cell transfer colitis in SCID mice are most widely used and accepted models that can recapitulate the human diseases. Here, we provide detailed protocols of these two mouse models of experimentally induced intestinal inflammation. We also discuss the protocols for the isolation and analysis of inflammatory T cell from the colon.


Subject(s)
Colitis, Ulcerative , Disease Models, Animal , Acute Disease , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Chronic Disease , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colon/immunology , Colon/pathology , Enzyme Assays , Homeodomain Proteins/metabolism , Humans , Leukocyte Common Antigens/metabolism , Lymph Nodes/immunology , Male , Mice , Peroxidase/metabolism , Staining and Labeling , Tissue Culture Techniques , Tissue and Organ Harvesting
12.
Clin Chim Acta ; 378(1-2): 128-35, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17182021

ABSTRACT

BACKGROUND: Butyrylcholinesterase (BChE; gi:116353) deficiency has adverse effects on the response to succinylcholine and mivacurium. A physiological function of BChE is to inactivate octanoyl ghrelin. We determined the health effect of complete absence of BChE in humans. METHODS: Clinical tests of cardiac, lung, liver, and kidney function, body weight, sperm counts and motility were performed on 5 men, age 20-32 y, in the Vysya community of Coimbatore, India who had silent BChE. Postmortem tissues from 2 cadavers with wild-type BChE were assayed. RESULTS: Test results were normal, except for lung function, which indicated mild obstruction in silent as well as in wild-type BChE subjects. Creatine kinase-MB levels were high in 2 subjects, but there were no other indications of damage to the heart. Body weight was normal. Family histories revealed no trend in disease susceptibility. The human body contains 10 times more BChE than acetylcholinesterase molecules. CONCLUSION: Individuals completely deficient in BChE have only minor abnormalities in clinical test results. However, they respond abnormally to standard doses of succinylcholine and mivacurium. It is expected, but not proven, that they are unusually susceptible to the toxicity of cocaine and organophosphorus pesticides, and resistant to bambuterol and irinotecan. Their normal body weight suggests alternative routes for deactivation of octanoyl ghrelin.


Subject(s)
Butyrylcholinesterase/deficiency , Adult , Body Weight , Butyrylcholinesterase/physiology , Cocaine/toxicity , Electrocardiography , Female , Ghrelin , Humans , Kidney/physiopathology , Liver/physiopathology , Lung/physiopathology , Male , Middle Aged , Peptide Hormones/metabolism , Sperm Motility
13.
Arch Biochem Biophys ; 452(2): 186-8, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16824479

ABSTRACT

Butyrylcholinesterase in human plasma and acetylcholinesterase in human red blood cells have aryl acylamidase activity toward o-nitroacetanilide, hydrolyzing the amide bond to produce o-nitroaniline and acetate. People with a genetic variant of butyrylcholinesterase that had no detectable activity with butyrylthiocholine, nevertheless had aryl acylamidase activity in their plasma. To determine the source of this aryl acylamidase activity we tested fatty acid free human albumin for activity. We found that albumin had aryl acylacylamidase activity and that this activity was inhibited by diisopropylfluorophosphate. Since the esterase activity of albumin is also inhibited by diisopropylfluorophosphate, and since it is known that diisopropylfluorophosphate covalently binds to Tyr 411 of human albumin, we conclude that the active site for aryl acylamidase activity of albumin is Tyr 411. Albumin accounts for about 10% of the aryl acylamidase activity in human plasma.


Subject(s)
Amidohydrolases/chemistry , Fatty Acids/chemistry , Isoflurophate/chemistry , Serum Albumin/chemistry , Enzyme Activation , Humans
14.
Pharmacogenet Genomics ; 16(7): 461-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16788378

ABSTRACT

BACKGROUND: People with genetic variants of butyrylcholinesterase (EC 3.1.1.8, BChE) can have hours of prolonged apnea after a normal dose of succinylcholine or mivacurium. METHODS: Plasma samples from 226 people in the Vysya community in Coimbatore, India were tested for BChE activity. RESULTS: Nine unrelated individuals had no detectable activity. DNA sequencing revealed a novel mutation in exon 2 of the BCHE gene, responsible for the silent phenotype of human serum BChE. All silent BChE samples were homozygous for a point mutation at codon 307 (CTT-->CCT), resulting in substitution of leucine 307 by proline. Western blot analysis with a monoclonal antibody showed no BChE protein in plasma. Silent BChE plasma samples had no organophosphate-reactive BChE, as measured with FP-biotin. Expression of recombinant Leu307Pro BChE in cell culture confirmed that this mutant is expressed at very low levels. The proline substitution most likely destabilizes the BChE structure and causes the protein to be misfolded and rapidly degraded. CONCLUSIONS: This is the first report of a molecularly defined BChE mutation in the Indian population. The frequency of homozygous silent BChE in the Vysya community is 1 in 24, a value 4000-fold higher than the frequency of homozygous silent BChE in European and American populations.


Subject(s)
Butyrylcholinesterase/deficiency , Butyrylcholinesterase/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , DNA Mutational Analysis , Exons , Female , Homozygote , Humans , India , Male , Middle Aged , Phenotype , Point Mutation
15.
Biochem Pharmacol ; 70(11): 1673-84, 2005 Nov 25.
Article in English | MEDLINE | ID: mdl-16213467

ABSTRACT

The goal of this work was to identify the esterases in human plasma and to clarify common misconceptions. The method for identifying esterases was nondenaturing gradient gel electrophoresis stained for esterase activity. We report that human plasma contains four esterases: butyrylcholinesterase (EC 3.1.1.8), paraoxonase (EC 3.1.8.1), acetylcholinesterase (EC 3.1.1.7), and albumin. Butyrylcholinesterase (BChE), paraoxonase (PON1), and albumin are in high enough concentrations to contribute significantly to ester hydrolysis. However, only trace amounts of acetylcholinesterase (AChE) are present. Monomeric AChE is seen in wild-type as well as in silent BChE plasma. Albumin has esterase activity with alpha- and beta-naphthylacetate as well as with p-nitrophenyl acetate. Misconception #1 is that human plasma contains carboxylesterase. We demonstrate that human plasma contains no carboxylesterase (EC 3.1.1.1), in contrast to mouse, rat, rabbit, horse, cat, and tiger that have high amounts of plasma carboxylesterase. Misconception #2 is that lab animals have BChE but no AChE in their plasma. We demonstrate that mice, unlike humans, have substantial amounts of soluble AChE as well as BChE in their plasma. Plasma from AChE and BChE knockout mice allowed identification of AChE and BChE bands without the use of inhibitors. Human BChE is irreversibly inhibited by diisopropylfluorophosphate, echothiophate, and paraoxon, but mouse BChE spontaneously reactivates. Since human plasma contains no carboxylesterase, only BChE, PON1, and albumin esterases need to be considered when evaluating hydrolysis of an ester drug in human plasma.


Subject(s)
Albumins/metabolism , Aryldialkylphosphatase/blood , Butyrylcholinesterase/blood , Acetylcholinesterase/blood , Acetylcholinesterase/genetics , Acetylcholinesterase/metabolism , Animals , Aryldialkylphosphatase/metabolism , Butyrylcholinesterase/genetics , Butyrylcholinesterase/metabolism , Carboxylesterase/blood , Carboxylesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Humans , Mice , Mice, Knockout , Organophosphates/pharmacology , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...