Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Immunol ; 211(5): 853-861, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37477694

RESUMEN

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.


Asunto(s)
Colitis , Factores de Transcripción , Animales , Ratones , Colon , Citocinas/metabolismo , Homeostasis , Inflamación , Mucosa Intestinal , Intestinos/patología , Ratones Endogámicos C57BL , Receptor alfa X Retinoide , Factores de Transcripción/metabolismo
2.
J Immunol ; 209(2): 368-378, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35760519

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda , Colitis , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Microbiota , Lesión Renal Aguda/metabolismo , Animales , Colitis/inducido químicamente , Citocinas/metabolismo , Sulfato de Dextran/toxicidad , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Riñón/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Vía de Señalización Wnt/genética
3.
J Immunol ; 207(5): 1428-1436, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34348977

RESUMEN

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.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Células TH1 , Animales , Células Dendríticas , Ratones , Ratones Endogámicos C57BL , Células Th17 , Factor de Transcripción 4
4.
Front Immunol ; 12: 691134, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34394085

RESUMEN

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.


Asunto(s)
Células Dendríticas/inmunología , Ácido Láctico/inmunología , Macrófagos/inmunología , Animales , Autoinmunidad , Proteínas de Ciclo Celular/inmunología , Humanos , Inmunomodulación , Infecciones/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Transportadores de Ácidos Monocarboxílicos/inmunología , Neoplasias/inmunología , Receptores Acoplados a Proteínas G/inmunología
5.
J Immunol ; 205(8): 2265-2275, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32917787

RESUMEN

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.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Colitis/inmunología , Neoplasias del Colon/inmunología , Microbioma Gastrointestinal/inmunología , Interleucina-10/inmunología , Vía de Señalización Wnt/inmunología , beta Catenina/inmunología , Animales , Células Presentadoras de Antígenos/patología , Colitis/complicaciones , Colitis/genética , Colitis/patología , Neoplasias del Colon/etiología , Neoplasias del Colon/genética , Neoplasias del Colon/prevención & control , Microbioma Gastrointestinal/genética , Interleucina-10/genética , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Vía de Señalización Wnt/genética , beta Catenina/genética
6.
Oncoimmunology ; 5(4): e1115941, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27141399

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-27183583

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Homeostasis , Inflamación/prevención & control , PPAR alfa/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Proteínas de Homeodominio/inmunología , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR alfa/deficiencia
8.
J Immunol ; 194(7): 3295-304, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25710911

RESUMEN

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.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Vía de Señalización Wnt , Animales , Diferenciación Celular , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Eliminación de Gen , Técnicas de Inactivación de Genes , Mediadores de Inflamación/metabolismo , Interleucina-10/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Masculino , Ratones , Ratones Transgénicos , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células TH1/citología , Células TH1/metabolismo , Células Th17/citología , Células Th17/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo
9.
Cancer Res ; 75(4): 656-665, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25568183

RESUMEN

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.


Asunto(s)
Células Dendríticas/metabolismo , Microambiente Tumoral/inmunología , Vitamina A/metabolismo , beta Catenina/genética , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/inmunología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/patología , Humanos , Ratones , Ratones Transgénicos , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Transcripción 4 , Microambiente Tumoral/genética , beta Catenina/metabolismo
10.
J Immunol ; 193(8): 4203-13, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25210120

RESUMEN

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.


Asunto(s)
Autoinmunidad/inmunología , Células Dendríticas/inmunología , Linfocitos T Reguladores/citología , Receptor Toll-Like 2/inmunología , beta Catenina/metabolismo , Traslado Adoptivo , Aldehído Deshidrogenasa/biosíntesis , Familia de Aldehído Deshidrogenasa 1 , Animales , Diferenciación Celular/inmunología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/inmunología , Inflamación/prevención & control , Interleucina-10/biosíntesis , Lectinas Tipo C/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/inmunología , Retinal-Deshidrogenasa , Células TH1/citología , Células TH1/inmunología , Células Th17/citología , Células Th17/inmunología , Proteína 2 Similar al Factor de Transcripción 7/inmunología , Zimosan/inmunología , Zimosan/farmacología , beta Catenina/genética
11.
Methods Mol Biol ; 1194: 437-48, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25064119

RESUMEN

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.


Asunto(s)
Colitis Ulcerosa , Modelos Animales de Enfermedad , Enfermedad Aguda , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Enfermedad Crónica , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Colon/inmunología , Colon/patología , Pruebas de Enzimas , Proteínas de Homeodominio/metabolismo , Humanos , Antígenos Comunes de Leucocito/metabolismo , Ganglios Linfáticos/inmunología , Masculino , Ratones , Peroxidasa/metabolismo , Coloración y Etiquetado , Técnicas de Cultivo de Tejidos , Recolección de Tejidos y Órganos
12.
Clin Chim Acta ; 378(1-2): 128-35, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17182021

RESUMEN

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.


Asunto(s)
Butirilcolinesterasa/deficiencia , Adulto , Peso Corporal , Butirilcolinesterasa/fisiología , Cocaína/toxicidad , Electrocardiografía , Femenino , Ghrelina , Humanos , Riñón/fisiopatología , Hígado/fisiopatología , Pulmón/fisiopatología , Masculino , Persona de Mediana Edad , Hormonas Peptídicas/metabolismo , Motilidad Espermática
13.
Arch Biochem Biophys ; 452(2): 186-8, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16824479

RESUMEN

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.


Asunto(s)
Amidohidrolasas/química , Ácidos Grasos/química , Isoflurofato/química , Albúmina Sérica/química , Activación Enzimática , Humanos
14.
Pharmacogenet Genomics ; 16(7): 461-8, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16788378

RESUMEN

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.


Asunto(s)
Butirilcolinesterasa/deficiencia , Butirilcolinesterasa/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Análisis Mutacional de ADN , Exones , Femenino , Homocigoto , Humanos , India , Masculino , Persona de Mediana Edad , Fenotipo , Mutación Puntual
15.
Biochem Pharmacol ; 70(11): 1673-84, 2005 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-16213467

RESUMEN

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.


Asunto(s)
Albúminas/metabolismo , Arildialquilfosfatasa/sangre , Butirilcolinesterasa/sangre , Acetilcolinesterasa/sangre , Acetilcolinesterasa/genética , Acetilcolinesterasa/metabolismo , Animales , Arildialquilfosfatasa/metabolismo , Butirilcolinesterasa/genética , Butirilcolinesterasa/metabolismo , Carboxilesterasa/sangre , Carboxilesterasa/metabolismo , Inhibidores de la Colinesterasa/farmacología , Humanos , Ratones , Ratones Noqueados , Organofosfatos/farmacología , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA