Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(43): e2205277119, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36252012

RESUMO

Mucins are the main macrocomponents of the mucus layer that protects the digestive tract from pathogens. Fucosylation of mucins increases mucus viscoelasticity and its resistance to shear stress. These properties are altered in patients with ulcerative colitis (UC), which is marked by a chronic inflammation of the distal part of the colon. Here, we show that levels of Fucosyltransferase 8 (FUT8) and specific mucins are increased in the distal inflamed colon of UC patients. Recapitulating this FUT8 overexpression in mucin-producing HT29-18N2 colonic cell line increases delivery of MUC1 to the plasma membrane and extracellular release of MUC2 and MUC5AC. Mucins secreted by FUT8 overexpressing cells are more resistant to removal from the cell surface than mucins secreted by FUT8-depleted cells (FUT8 KD). FUT8 KD causes intracellular accumulation of MUC1 and alters the ratio of secreted MUC2 to MUC5AC. These data fit well with the Fut8-/- mice phenotype, which are protected from UC. Fut8-/- mice exhibit a thinner proximal colon mucus layer with an altered ratio of neutral to acidic mucins. Together, our data reveal that FUT8 modifies the biophysical properties of mucus by controlling levels of cell surface MUC1 and quantity and quality of secreted MUC2 and MUC5AC. We suggest that these changes in mucus viscoelasticity likely facilitate bacterial-epithelial interactions leading to inflammation and UC progression.


Assuntos
Colite Ulcerativa , Fucosiltransferases , Animais , Camundongos , Colite Ulcerativa/genética , Colite Ulcerativa/metabolismo , Fucosiltransferases/genética , Inflamação , Mucina-2/genética , Mucina-2/metabolismo , Células HT29
2.
Am J Physiol Lung Cell Mol Physiol ; 322(5): L683-L698, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35348023

RESUMO

Excessive production, secretion, and retention of abnormal mucus is a pathological feature of many obstructive airways diseases including asthma. Azithromycin is an antibiotic that also possesses immunomodulatory and mucoregulatory activities, which may contribute to the clinical effectiveness of azithromycin in asthma. The current study investigated these nonantibiotic activities of azithromycin in mice exposed daily to intranasal house dust mite (HDM) extract for 10 days. HDM-exposed mice exhibited airways hyperresponsiveness to aerosolized methacholine, a pronounced mixed eosinophilic and neutrophilic inflammatory response, increased airway smooth muscle (ASM) thickness, and elevated levels of epithelial mucin staining. Azithromycin (50 mg/kg sc, 2 h before each HDM exposure) attenuated HDM-induced airways hyperresponsiveness to methacholine, airways inflammation (bronchoalveolar lavage eosinophil and neutrophils numbers, and IL-4, IL-5, IL-6, IL-10, IL-12, IL-13, and RANTES levels), and epithelial mucin staining (mucous metaplasia) by at least 50% (compared with HDM-exposed mice, P < 0.05). Isolated tracheal segments of HDM-exposed mice secreted Muc5ac and Muc5b (above baseline levels) in response to exogenous ATP. Moreover, ATP-induced secretion of mucins was attenuated in segments obtained from azithromycin-treated, HDM-exposed mice (P < 0.05). In additional ex vivo studies, ATP-induced secretion of Muc5ac (but not muc5b) from HDM-exposed tracheal segments was inhibited by in vitro exposure to azithromycin. In vitro azithromycin also inhibited ATP-induced secretion of Muc5ac and Muc5b in tracheal segments from IL-13-exposed mice. In summary, azithromycin inhibited ATP-induced mucin secretion and airways inflammation in HDM-exposed mice, both of which are likely to contribute to suppression of airways hyperresponsiveness.


Assuntos
Asma , Pyroglyphidae , Trifosfato de Adenosina , Alérgenos , Animais , Asma/patologia , Azitromicina/farmacologia , Modelos Animais de Doenças , Inflamação/tratamento farmacológico , Interleucina-13 , Metaplasia , Cloreto de Metacolina , Camundongos , Mucinas , Muco
3.
Int J Mol Sci ; 23(11)2022 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-35682912

RESUMO

Specialized pro-resolving mediators (SPMs), including Maresins (MaR)-1 and 2, contribute to tear film homeostasis and resolve conjunctival inflammation. We investigated MaR2's signaling pathways in goblet cells (GC) from rat conjunctiva. Agonist-induced [Ca2+]i and high-molecular weight glycoconjugate secretion were measured. MaR2 increased [Ca2+]i and stimulated secretion. MaR2 and MaR1 stimulate conjunctival goblet cell function, especially secretion, by activating different but overlapping GPCR and signaling pathways, and furthermore counter-regulate histamine stimulated increase in [Ca2+]i. Thus, MaR2 and MaR1 play a role in maintaining the ocular surface and tear film homeostasis in health and disease. As MaR2 and MaR1 modulate conjunctival goblet cell function, they each may have potential as novel, but differing, options for the treatment of ocular surface inflammatory diseases including allergic conjunctivitis and dry eye disease. We conclude that in conjunctival GC MaR2 and MaR1, both increase the [Ca2+]i and stimulate secretion to maintain homeostasis by using one set of different, but overlapping, signaling pathways to increase [Ca2+]i and another set to stimulate secretion. MaR2 also resolves ocular allergy.


Assuntos
Células Caliciformes , Mucinas , Animais , Células Cultivadas , Túnica Conjuntiva/metabolismo , Ácidos Docosa-Hexaenoicos , Células Caliciformes/metabolismo , Mucinas/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
4.
Ecotoxicol Environ Saf ; 221: 112448, 2021 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-34174739

RESUMO

Atmospheric PM2.5 can induce airway inflammation and mucin secretion. MUC5B is required for airway defense. However, the research on the role of MUC5B in airway inflammation induced by atmospheric PM2.5 remains limited. This study was designed to explore the role of MUC5B in airway inflammation induced by atmospheric PM2.5. In vivo, Wistar rats were exposed to 0, 1.5, 7.5, 37.5 mg/ kg PM2.5 saline suspension via intratracheal instillation. HE staining and AB-PAS staining were used to observe the airway inflammation and goblet cell hyperplasia. In vitro, normal A549 cells and MUC5B-knockdown A549 cells were exposed to 0, 100, 200 and 400 µg/mL PM2.5 for 6 h, 12 h, 24 h and 48 h. ELISA was used to measure the levels of TNF-α and IL-1ß in serum and bronchoalveolar lavage fluid of rats and in cell culture. Real time-PCR and ELISA were used to quantify the mRNA and protein levels of MUC5B in trachea and lung of rats and in A549 cells. PM2.5 could cause the infiltration of inflammatory cells and increase the mucus secretions and goblet cell metaplasia. MUC5B is related to rats' airway inflammation induced by PM2.5. A549 cells exposed to PM2.5 in higher concentration and longer time, the protein level of MUC5B was significantly increased, while the levels of IL-1ß, TNF-α and MUC5B mRNA were significantly decreased. Compared with normal A549 cells, the levels of IL-1ß and TNF-α were significantly higher in Muc5b-knockdown cells. Atmospheric PM2.5 can induce airway inflammation and mucin secretion. MUC5B played a critical role in controlling the inflammatory response induced by PM2.5.


Assuntos
Inflamação/metabolismo , Mucina-5B/metabolismo , Material Particulado/toxicidade , Células A549 , Animais , Líquido da Lavagem Broncoalveolar/química , Feminino , Humanos , Interleucina-1beta/sangue , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Pulmão/metabolismo , Masculino , Mucina-5B/genética , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 306(10): L925-36, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24705720

RESUMO

MARCKS (myristoylated alanine-rich C kinase substrate) is postulated to regulate the passage of secretory granules through cortical actin in the early phase of exocytosis. There are, however, three proposed mechanisms of action, all of which were derived from studies using synthetic peptides representing either the central phosphorylation site domain or the upstream, NH2-terminal domain: it tethers actin to the plasma membrane and/or to secretory granules, and/or it sequesters PIP2. Using MARCKS-null mice, we probed for a loss of function secretory phenotype in mast cells harvested from embryonic livers and maturated in vivo [embryonic hepatic-derived mast cells (eHMCs)]. Both wild-type (WT) and MARCKS-null eHMCs exhibited full exocytic responses upon FcϵRI receptor activation with DNP-BSA (2,4-dinitrophenyl-BSA), whether they were in suspension or adherent. The secretory responses of MARCKS-null eHMCs were consistently higher than those of WT cells, but the differences had sporadic statistical significance. The MARCKS-null cells exhibited faster secretory kinetics, however, achieving the plateau phase of the response with a t½ ∼2.5-fold faster. Hence, MARCKS appears to be a nonessential regulatory protein in mast cell exocytosis but exerts a negative modulation. Surprisingly, the MARCKS NH2-terminal peptide, MANS, which has been reported to inhibit mucin secretion from airway goblet cells (Li Y, Martin LD, Spizz G, Adler KB. J Biol Chem 276: 40982-40990, 2001), inhibited hexosaminidase secretion from WT and MARCKS-null eHMCs, leading us to reexamine its effects on mucin secretion. Results from studies using peptide inhibitors with human bronchial epithelial cells and with binding assays using purified mucins suggested that MANS inhibited the mucin binding assay, rather than the secretory response.


Assuntos
Células Caliciformes/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Mastócitos/metabolismo , Proteínas de Membrana/fisiologia , Mucinas/metabolismo , Animais , Brônquios/citologia , Células Cultivadas , Exocitose , Hexosaminidases/metabolismo , Humanos , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Substrato Quinase C Rico em Alanina Miristoilada , Fragmentos de Peptídeos/fisiologia
6.
Am J Physiol Gastrointest Liver Physiol ; 307(12): G1169-79, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25342048

RESUMO

In antral mucous cells, acetylcholine (ACh, 1 µM) activates Ca(2+)-regulated exocytosis, consisting of a peak in exocytotic events that declines rapidly (initial phase) followed by a second slower decline (late phase) lasting during ACh stimulation. GW7647 [a peroxisome proliferation activation receptor α (PPARα) agonist] enhanced the ACh-stimulated initial phase, and GW6471 (a PPARα antagonist) abolished the GW7647-induced enhancement. However, GW6471 produced the delayed, but transient, increase in the ACh-stimulated late phase, and it also decreased the initial phase and produced the delayed increase in the late phase during stimulation with ACh alone. A similar delayed increase in the ACh-stimulated late phase is induced by an inhibitor of the PKG, Rp8BrPETcGMPS, suggesting that GW6471 inhibits cGMP accumulation. An inhibitor of nitric oxide synthase 1 (NOS1), N(5)-[imino(propylamino)methyl]-L-ornithine hydrochloride (N-PLA), also abolished the GW7647-induced-enhancement of ACh-stimulated initial phase but produced the delayed increase in the late phase. However, in the presence of N-PLA, an NO donor or 8BrcGMP enhanced the ACh-stimulated initial phase and abolished the delayed increase in the late phase. Moreover, GW7647 and ACh stimulated NO production and cGMP accumulation in antral mucosae, which was inhibited by GW6471 or N-PLA. Western blotting and immunohistochemistry revealed that NOS1 and PPARα colocalize in antral mucous cells. In conclusion, during ACh stimulation, a PPARα autocrine mechanism, which accumulates NO via NOS1 leading to cGMP accumulation, modulates the Ca(2+)-regulated exocytosis in antral mucous cells.


Assuntos
Comunicação Autócrina/fisiologia , Exocitose/fisiologia , Células Caliciformes/metabolismo , PPAR alfa/metabolismo , Antro Pilórico/metabolismo , Animais , Comunicação Autócrina/efeitos dos fármacos , Butiratos/farmacologia , Cálcio/metabolismo , GMP Cíclico/metabolismo , Exocitose/efeitos dos fármacos , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/metabolismo , Células Caliciformes/efeitos dos fármacos , Cobaias , Masculino , Óxido Nítrico/metabolismo , Oxazóis/farmacologia , PPAR alfa/agonistas , PPAR alfa/antagonistas & inibidores , Compostos de Fenilureia/farmacologia , Antro Pilórico/efeitos dos fármacos , Tirosina/análogos & derivados , Tirosina/farmacologia
7.
J Control Release ; 373: 306-318, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39004103

RESUMO

Dry eye disease (DED) is a prevalent ocular disorder characterized by unstable tear film condition with loss of aqueous or mucin, excessive oxidative stress, and inflammation, leading to discomfort and potential damage to the ocular surface. Current DED therapies have shown restricted therapeutic effects such as frequent dosing and temporary relief with potential unwanted side effects, urgently necessitating the development of innovative efficient therapeutic approaches. Herein, we developed rosmarinic acid (RosA) conjugated gelatin nanogels loading diquafosol sodium (DQS), DRGNG, for simultaneous ROS-scavenging and mucin-secreting DED treatment. Mechanically, DRGNG suppressed the ROS production, reduced inflammatory factors, and prompted mucin secretion in vitro and in vivo. The whole transcriptome RNA sequencing in vitro further provided a detailed analysis of the upregulation of anti-oxidant, anti-inflammatory, and mucin-promotion pathways. Therapeutically, both in evaporative DED and aqueous deficient DED models, the dual-functional DRGNG could prolong the retention time at the ocular surface, efficiently suppress the oxidative stress response, reverse ocular surface morphology, and recover tear film homeostasis, thus alleviating the DED when the dosage is halved compared to the commercial Diquas®. Our findings contribute to developing innovative therapies for DED and offer insights into the broader applications of nanogels in ocular drug delivery and oxidative stress-related conditions.

8.
Am J Physiol Cell Physiol ; 304(10): C976-84, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23467297

RESUMO

Nucleotides within the airway surface liquid promote fluid secretion via activation of airway epithelial purinergic receptors. ATP is stored within and released from mucin granules as co-cargo with mucins, but the mechanism by which ATP, and potentially other nucleotides, enter the lumen of mucin granules is not known. We assessed the contribution of the recently identified SLC17A9 vesicle nucleotide transporter (VNUT) to the nucleotide availability within isolated mucin granules and further examined the involvement of VNUT in mucin granule secretion-associated nucleotide release. RT-PCR and Western blot analyses indicated that VNUT is abundantly expressed in airway epithelial goblet-like Calu-3 cells, migrating as a duplex with apparent mobility of 55 and 60 kDa. Subcellular fractionation studies indicated that VNUT55 was associated with high-density mucin granules, whereas VNUT60 was associated with low-density organelles. Immunofluorescence studies showed that recombinant VNUT localized to mucin granules and other organelles. Mucin granules isolated from VNUT short hairpin RNA-expressing cells exhibited a marked reduction of ATP, ADP, AMP, and UTP levels within granules. Ca(2+)-regulated vesicular ATP release was markedly reduced in these cells, but mucin secretion was not affected. These results suggest that VNUT is the relevant nucleotide transporter responsible for the uptake of cytosolic nucleotides into mucin granules. By controlling the entry of nucleotides into mucin granules, VNUT contributes to the release of purinergic signaling molecules necessary for the proper hydration of co-released mucins.


Assuntos
Células Caliciformes/metabolismo , Proteínas de Transporte de Nucleotídeos/metabolismo , Nucleotídeos/metabolismo , Sistema Respiratório/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Difosfato de Adenosina/biossíntese , Monofosfato de Adenosina/biossíntese , Trifosfato de Adenosina/biossíntese , Trifosfato de Adenosina/metabolismo , Transporte Biológico , Linhagem Celular , Grânulos Citoplasmáticos/metabolismo , Humanos , Mucinas/genética , Proteínas de Transporte de Nucleotídeos/biossíntese , RNA Interferente Pequeno , Vesículas Secretórias/metabolismo , Uridina Trifosfato/biossíntese
9.
Am J Physiol Gastrointest Liver Physiol ; 304(9): G773-80, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23449671

RESUMO

In antral mucous cells, acetylcholine (ACh, 1 µM) activates Ca(2+)-regulated exocytosis, consisting of an initial peak that declines rapidly (initial transient phase) followed by a second slower decline (late phase) lasting during ACh stimulation. The addition of 8-bromo-cGMP (8-BrcGMP) enhanced the initial phase, which was inhibited by the protein kinase G (PKG) inhibitor guanosine 3',5'-cyclic monophosphorothoiate, ß-phenyl-1,N(2)-etheno-8-bromo, Rp-isomer, sodium salt (Rp-8-BrPETcGMPS, 100 nM). However, Rp-8-BrPETcGMPS produced a delayed, but transient, increase in the exocytotic frequency during the late phase that was abolished by a protein kinase A (PKA) inhibitor (PKI-amide), suggesting that Rp-8-BrPETcGMPS accumulates cAMP. The cGMP-dependent phosphodiesterase 2 (PDE2), which degrades cAMP, may exist in antral mucous cells. The PDE2 inhibitor BAY-60-7550 (250 nM) mimicked the effect of Rp-8-BrPETcGMPS on ACh-stimulated exocytosis. Measurement of the cGMP and cAMP contents in antral mucosae revealed that ACh stimulates the accumulation of cGMP and that BAY-60-7550 accumulates cAMP similarly to Rp-8-BrPETcGMPS during ACh stimulation. Analyses of Western blot and immunohistochemistry demonstrated that PDE2A exists in antral mucous cells. In conclusion, Rp-8-BrPETcGMPS accumulates cAMP by inhibiting PDE2 in ACh-stimulated antral mucous cells, leading to the delayed, but transient, increase in the frequency of Ca(2+)-regulated exocytosis. PDE2 may prevent antral mucous cells from excessive mucin secretion caused by the cAMP accumulation.


Assuntos
Cálcio/fisiologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , GMP Cíclico/análogos & derivados , Exocitose/efeitos dos fármacos , Antro Pilórico/fisiologia , Acetilcolina/farmacologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , GMP Cíclico/metabolismo , GMP Cíclico/farmacologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/antagonistas & inibidores , Dinoprostona/metabolismo , Mucosa Gástrica/efeitos dos fármacos , Cobaias , Masculino , Inibidores de Proteínas Quinases/farmacologia , Antro Pilórico/efeitos dos fármacos
10.
J Mol Biol ; 435(1): 167853, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36243149

RESUMO

Synaptic neurotransmitter release is mediated by an orchestra of presynaptic proteins that precisely control and trigger fusion between synaptic vesicles and the neuron terminal at the active zone upon the arrival of an action potential. Critical to this process are the neuronal SNAREs (Soluble N-ethylmaleimide sensitive factor Attachment protein REceptor), the Ca2+-sensor synaptotagmin, the activator/regulator complexin, and other factors. Here, we review the interactions between the SNARE complex and synaptotagmin, with focus on the so-called primary interface between synaptotagmin and the SNARE complex that has been validated in terms of its physiological relevance. We discuss several other but less validated interfaces as well, including the so-called tripartite interface, and we discuss the pros and cons for these possible alternative interfaces. We also present new molecular dynamics simulations of the tripartite interface and new data of an inhibitor of the primary interface in a reconstituted system of synaptic vesicle fusion.


Assuntos
Fusão de Membrana , Transmissão Sináptica , Vesículas Sinápticas , Cálcio/metabolismo , Neurônios/metabolismo , Proteínas SNARE/metabolismo , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/metabolismo , Sinaptotagminas/metabolismo
11.
Front Pharmacol ; 13: 891041, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35814209

RESUMO

The so-called primary interface between the SNARE complex and synaptotagmin-1 (Syt1) is essential for Ca2+-triggered neurotransmitter release in neuronal synapses. The interacting residues of the primary interface are conserved across different species for synaptotagmins (Syt1, Syt2, Syt9), SNAP-25, and syntaxin-1A homologs involved in fast synchronous release. This Ca2+-independent interface forms prior to Ca2+-triggering and plays a role in synaptic vesicle priming. This primary interface is also conserved in the fusion machinery that is responsible for mucin granule membrane fusion. Ca2+-stimulated mucin secretion is mediated by the SNAREs syntaxin-3, SNAP-23, VAMP8, Syt2, and other proteins. Here, we designed and screened a series of hydrocarbon-stapled peptides consisting of SNAP-25 fragments that included some of the key residues involved in the primary interface as observed in high-resolution crystal structures. We selected a subset of four stapled peptides that were highly α-helical as assessed by circular dichroism and that inhibited both Ca2+-independent and Ca2+-triggered ensemble lipid-mixing with neuronal SNAREs and Syt1. In a single-vesicle content-mixing assay with reconstituted neuronal SNAREs and Syt1 or with reconstituted airway SNAREs and Syt2, the selected peptides also suppressed Ca2+-triggered fusion. Taken together, hydrocarbon-stapled peptides that interfere with the primary interface consequently inhibit Ca2+-triggered exocytosis. Our inhibitor screen suggests that these compounds may be useful to combat mucus hypersecretion, which is a major cause of airway obstruction in the pathophysiology of COPD, asthma, and cystic fibrosis.

12.
Biomed Pharmacother ; 139: 111595, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33862492

RESUMO

BACKGROUND: Akkermansia spp. plays important roles in maintenance of host health. Increasing evidence reveals that berberine (BBR) may exert its pharmacological effects via, at least partially, promotion of Akkermansia spp. However, how BBR stimulates Akkermansia remains largely unknown. PURPOSE: In this study, we investigated the mechanism underlying the Akkermansia-promoting effect of BBR. MATERIALS AND METHODS: The effect of BBR on Akkermansia was assessed in BBR-gavaged mice and direct incubation. The influence of BBR on intestinal mucin production was determined by alcian-blue staining and real-time PCR. The feces were analysis by gas chromatography-time-of-flight mass spectrometry (GC-TOF/MS) metabolomics. The role of polyamines in BBR-elicited mucin secretion and Akkermansia growth was evaluated by administration of difluoromethylornithine (DFMO) in mice. RESULTS: Gavage of BBR dose-dependently and time-dependently increased the abundance of Akkermansia in mice. However, it did not stimulate Akkermansia growth in direct incubation, suggesting that BBR may promote Akkermansia in a host-dependent way. Oral administration of BBR significantly increased the transcription of mucin-producing genes and mucin secretion in colon. Untargeted metabolomics analysis showed that BBR increased polyamines production in feces which are known to stimulate goblet cell proliferation and differentiation, but treatment with eukaryotic polyamine synthase inhibitor DFMO did not abolish the stimulating effect of BBR on mucin secretion and Akkermansia growth, indicating that the gut bacteria-derived but not the host-derived polyamines may involve in the BBR-promoted Akkermansia growth. CONCLUSIONS: Our results reveal that BBR is a promising prebiotic for Akkermansia, and it promotes Akkermansia growth via stimulating mucin secretion in colon.


Assuntos
Akkermansia/efeitos dos fármacos , Berberina/farmacologia , Colo/efeitos dos fármacos , Mucinas/metabolismo , Prebióticos , Akkermansia/crescimento & desenvolvimento , Animais , Colo/metabolismo , Dieta Hiperlipídica , Fezes/química , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Proteína Jagged-1/genética , Masculino , Metabolômica , Camundongos Endogâmicos ICR , Poliaminas/metabolismo , RNA Ribossômico 16S , Receptor Notch1/genética , Fatores de Transcrição HES-1/genética
13.
Inflamm Bowel Dis ; 27(5): 732-741, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33772551

RESUMO

Ulcerative colitis (UC) is an idiopathic, long-term inflammatory disorder of the colon, characterized by a continuous remitting and relapsing course. The intestinal mucus barrier is the first line at the interface between the host and microbiota and acts to protect intestinal epithelial cells from invasion. Data from patients and animal studies have shown that an impaired mucus barrier is closely related to the severity of UC. Depletion of the mucus barrier is not just the strongest but is also the only independent risk factor predicting relapse in patients with UC. Peroxisome proliferator-activated receptor gamma (PPARγ), a nuclear transcription regulator, is involved in the regulation of inflammatory cytokine expression. It is also known to promote mucus secretion under pathological conditions to expel pathogenic bacteria or toxins. More important, PPARγ has been shown to affect host-microbiota interactions by modulating the energy metabolism of colonocytes and the oxygen availability of the intestinal microbiome. It is well known that gut microbiota homeostasis is essential for butyrate generation by the commensal bacteria to supply energy resources for colonocytes. Therefore, it can be speculated that PPARγ, as a central coordinator of the mucus barrier, may be a promising target for the development of effective agents to combat UC.


Assuntos
Colite Ulcerativa , PPAR gama , Animais , Bactérias , Humanos , Mucosa Intestinal , Muco
14.
Biomed Pharmacother ; 139: 111583, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33901875

RESUMO

TMEM16A is a Ca2+-activated Cl- channel involved in mucus secretion in inflamed airways and proposed as a drug target for diseases associated with mucus hypersecretion including asthma. This study aimed to identify novel inhibitors of TMEM16A-mediated Cl- secretion in airway epithelial cells from a collection of compounds isolated from fungi indigenous in Thailand and examine its potential utility in mitigating airway mucus secretion using Calu-3 cells as a study model. Screening of > 400 fungal metabolites revealed purpactin A isolated from a soil-derived fungus Penicillium aculeatum PSU-RSPG105 as an inhibitor of TMEM16A-mediated Cl- transport with an IC50 value of ~2 µM. A consistent inhibitory effect of purpactin A on TMEM16A were observed regardless of TMEM16A activators or in the presence of an inhibitor of Ca2+/calmodulin-dependent protein kinase II (CaMKII), a negative regulator of TMEM16A. In addition, purpactin A did not affect cell viability, epithelial barrier integrity and activities of membrane transport proteins essential for maintaining airway hydration including CFTR Cl- channels and apical BK K+ channels. Intriguingly, purpactin A prevented a Ca2+-induced mucin release in cytokine-treated airway cells. Taken together, purpactin A represents the first class of TMEM16A inhibitor derived from fungus, which may be beneficial for the treatment of diseases associated with mucus hypersecretion.


Assuntos
Anoctamina-1/antagonistas & inibidores , Células Epiteliais/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis/farmacologia , Mucinas/metabolismo , Animais , Anoctamina-1/metabolismo , Anoctamina-1/fisiologia , Cálcio/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Humanos , Ratos Endogâmicos F344 , Sistema Respiratório/citologia , Talaromyces
15.
Microsc Res Tech ; 82(8): 1339-1344, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31070847

RESUMO

Inflammatory bowel disease (IBD) is a global, chronic intractable disease. The functions of drugs and food components have been evaluated in models of IBD induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS). Here, we used transmission (TEM) and osmium-maceration scanning (SEM) electron microscopy to evaluate the ultrastructure of colonic epithelial cells in rat models of IBD induced by TNBS. Histological evaluation revealed that the intestinal crypts in the most regions of the IBD-model colons were deformed and we classified them as having high cell migration rates (HMIG). The remaining regions in the intestinal crypts retained a relatively normal structure and we classified them as having low cell migration rates (LMIG). Osmium-maceration SEM revealed the mucosal fluid flowing in spaces without secretory granules in crypt goblet cells of both HMIG and LMIG regions, indicating the depletion of goblet cell mucin that is found in patients with IBD. The Golgi apparatus in absorptive cells was stacked and curled in both regions. Osmium-maceration SEM showed membrane network structures resembling endoplasmic reticulum that were large and expanded in absorptive cells with HMIG rather than with LMIG regions in IBD-model colons. These findings indicated that endoplasmic reticulum stress is associated with susceptibility to IBD and that the effects of various agents can be evaluated according to endoplasmic reticulum stress revealed by using electron microscopy in models of IBD induced by TNBS.


Assuntos
Colo/citologia , Células Epiteliais/ultraestrutura , Doenças Inflamatórias Intestinais/patologia , Animais , Colo/patologia , Modelos Animais de Doenças , Retículo Endoplasmático/patologia , Retículo Endoplasmático/ultraestrutura , Células Epiteliais/patologia , Células Caliciformes/ultraestrutura , Complexo de Golgi/patologia , Complexo de Golgi/ultraestrutura , Doenças Inflamatórias Intestinais/induzido quimicamente , Masculino , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Mucinas , Ratos , Ácido Trinitrobenzenossulfônico/administração & dosagem
16.
Front Physiol ; 9: 357, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29681865

RESUMO

The main dietary flavonoid quercetin, is known to preserve the integrity of gastrointestinal barrier and to have anti-inflammatory, anti-cancer, anti-fibrotic, and other beneficial properties. Many of the biological effects of quercetin appear to be associated to the modulation of cell signaling pathways, rather than to its antioxidant activity. In spite of the large number of data available on the molecular and cellular mechanisms by which quercetin exerts its biological effects, including protection of intestinal barrier function, there is a lack of data about the role of this substance on the expression and/or the secretion of mucins released by intestinal goblet cells. Here we investigated the effects of quercetin on the secretion and the gene expression of the main intestinal gel-forming mucins, MUC2 and MUC5AC, and the signaling mechanisms underlined, in human intestinal goblet cell-like LS174T. We found that quercetin increases intracellular Ca2+ levels and induces MUC2 and MUC5AC secretion in a Ca2+-dependent manner. Quercetin also induces mRNA levels of both secretory mucins. Quercetin stimulation of LS174T cells increases phosphorylation levels of extracellular signal regulated kinase (ERK)1-2 and protein kinase C (PKC) α and the induction of MUC2 and MUC5AC secretion and mRNA relies on phospholipase C (PLC), PKC, and ERK1-2 signaling pathways since the PLC inhibitor U73122, the PKC inhibitor bisindolylmaleimide (BIM) and the ERK1-2 pathway inhibitor PD98059, all revert the stimulatory effects of quercetin. We also demonstrated that the induction of mucin gene expression by quercetin is not limited to goblet cells. Indeed, quercetin induces mRNA levels of MUC2 and MUC5AC via PKCα/ERK1-2 pathway also in the human intestinal epithelial Caco-2 cells. These data highlight a novel mechanism thereby quercetin, regulating the secretory function of intestinal goblet cells and mucin levels in enterocytes may exert its protective effects on intestinal mucosal barrier.

17.
Artigo em Inglês | MEDLINE | ID: mdl-30337943

RESUMO

BACKGROUND: To assess the effects of Rhinosectan® spray, a medical device containing xyloglucan, on nasal ciliary function (in MucilAir™Nasal cells). METHODS: MucilAir™Nasal, a three-dimensional organotypic airway tissue model (with different cell types), was treated with Rhinosectan® (30 µl) or with a control (saline solution). The effects of Rhinosectan® were evaluated at 15 and 60 min post-exposure by: measurement of the cilia beating frequency (Hz), mucin detection (Enzyme-Linked Lectin Assay-ELLA), mucociliary clearance (µm/s) and phagocytosis assay (fluorescence). RESULTS: Exposure of MucilAir™ to Rhinosectan® did not alter the cilia beating frequency at 15 and 60 min post-exposure (diluted and undiluted). Exposure to Rhinosectan® (undiluted) during 60 min increased mucociliary clearance (93.3 ± 2.1 µm/s vs. 80.9 ± 1.8 µm/s; p < 0.01) and phagocytic activity (1.89-fold increase) in comparison with saline solution. Moreover, a significant decrease in mucin concentration was observed after 15 min of exposure (171.4 ng/ml vs. 306.5 ng/ml; p < 0.01) and at 60 min post-treatment (242.7 ng/ml vs. 339 ng/ml; p < 0.05). CONCLUSIONS: The application of Rhinosectan® to nasal epithelial cells does not impair ciliary movement, enhances mucociliary clearance and facilitates phagocytosis while reducing mucin secretion, which are optimal properties for the management of rhinitis and associated conditions.

18.
Elife ; 72018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30272559

RESUMO

Regulated mucin secretion from specialized goblet cells by exogenous agonist-dependent (stimulated) and -independent (baseline) manner is essential for the function of the epithelial lining. Over extended periods, baseline release of mucin can exceed quantities released by stimulated secretion, yet its regulation remains poorly characterized. We have discovered that ryanodine receptor-dependent intracellular Ca2+ oscillations effect the dissociation of the Ca2+-binding protein, KChIP3, encoded by KCNIP3 gene, from mature mucin-filled secretory granules, allowing for their exocytosis. Increased Ca2+ oscillations, or depleting KChIP3, lead to mucin hypersecretion in a human differentiated colonic cell line, an effect reproduced in the colon of Kcnip3-/- mice. Conversely, overexpressing KChIP3 or abrogating its Ca2+-sensing ability, increases KChIP3 association with granules, and inhibits baseline secretion. KChIP3 therefore emerges as the high-affinity Ca2+ sensor that negatively regulates baseline mucin secretion. We suggest KChIP3 marks mature, primed mucin granules, and functions as a Ca2+ oscillation-dependent brake to control baseline secretion. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Colo/metabolismo , Proteínas Interatuantes com Canais de Kv/metabolismo , Mucina-5AC/metabolismo , Animais , Células Caliciformes/metabolismo , Células HEK293 , Células HT29 , Humanos , Proteínas Interatuantes com Canais de Kv/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Mucina-5AC/genética , Vesículas Secretórias/metabolismo
19.
Virchows Arch ; 471(1): 65-76, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28550497

RESUMO

Intraductal papillary epithelial neoplasms of the pancreatobiliary system (intraductal papillary neoplasm of the bile duct (IPNB) and intraductal papillary mucinous neoplasm (IPMN)) seem to share many clinicopathological features; however, IPNB has not been fully characterized. In order to understand the clinicopathological/immunohistochemical features of IPNB better, we compared 52 cases of IPNB with 42 cases of IPMNs with mural nodules. The IPNB cases were divided into two groups according to their histological similarity and according to five key histological findings. All IPNB and IPMN cases mainly affected middle-aged to elderly people, predominantly men. Mucin hypersecretion was less frequent in IPNB compared to IPMN. Group 2 IPNB more frequently had a higher histopathological grade and more extensive stromal invasion than IPMN. Group 1 IPNB and IPMN were further classified into four subtypes (gastric, intestinal, pancreatobiliary, and oncocytic). Although each subtype of IPNB and IPMN showed similar histology, the immunohistochemical results were different. The gastric type of IPNB was less frequently positive for CDX2, and intestinal IPNB was more frequently positive for MUC1 and less frequently positive for MUC2, MUC5AC, and CDX2 compared to each subtype of IPMN, respectively. In conclusion, IPNB and IPMN have some clinicopathological features in common, but mucin hypersecretion was less frequent both in IPNBs than in IPMN. Group 2 IPNB differed from IPMN in several parameters of tumor aggressiveness. Additional clinicopathological and molecular studies should be performed with respect to the subtypes of IPNB and IPMN.


Assuntos
Adenocarcinoma Papilar/patologia , Neoplasias dos Ductos Biliares/patologia , Biomarcadores Tumorais/análise , Adenocarcinoma Mucinoso/patologia , Idoso , Carcinoma Ductal Pancreático/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/patologia
20.
Artigo em Inglês | MEDLINE | ID: mdl-27865847

RESUMO

INTRODUCTION: The aim of this study was to develop two dynamic ex vivo airway explant systems, a perfusion-superfusion system and a ventilation-superfusion system, for the study of toxic airborne substances, such as the prevalent smoke constituent acrolein. METHODS: Mouse isolated tracheal segments were perfused with physiological media or ventilated with humidified air at 37°C to mimic dynamic flow conditions, and superfused with media over the exterior surface. At selected time points, the histological and functional integrity of segments was evaluated. The perfusion-superfusion system was subsequently used to examine mucin secretory responses elicited by acrolein in airways in which mucous metaplasia had been induced with lipopolysaccharide (LPS; 1µgml-1) prior to 24h of media perfusion, followed by stimulation with acrolein or ATP for 15min. Epithelial mucin levels were determined by quantitative analysis of periodic acid-Schiff's reagent (PAS)-stained sections. RESULTS: Epithelial morphology was successfully preserved in the perfusion-superfusion and ventilation-superfusion systems for at least 24h and up to 18h, respectively. At these time points, the contractile and relaxation responses of perfused and ventilated tracheal segments to carbachol, the neuropeptide substance P, and the prostanoid PGE2 were also preserved. Using the perfusion-superfusion system, acute exposure to acrolein caused a dose-dependent reduction in the levels of PAS-positive mucin stores induced by LPS, consistent with mucin secretion. DISCUSSION: Both the perfusion-superfusion and ventilation-superfusion systems successfully preserved the viability of mouse isolated tracheal segments on a histological and functional level, and the perfusion-superfusion system was used to characterise the mucin secretory responses elicited by acrolein. Thus, this system may be a useful model through which to conduct further toxicological studies in mammalian airways.


Assuntos
Músculo Liso/fisiologia , Perfusão/métodos , Traqueia/fisiologia , Acroleína/farmacologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Liso/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Perfusão/instrumentação , Traqueia/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA