Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 211(1): 34-42, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37212607

RESUMO

We recently discovered a (to our knowledge) new neuroimmune interaction named the gateway reflex, in which the activation of specific neural circuits establishes immune cell gateways at specific vessel sites in organs, leading to the development of tissue-specific autoimmune diseases, including a multiple sclerosis (MS) mouse model, experimental autoimmune encephalomyelitis (EAE). We have reported that peripheral-derived myeloid cells, which are CD11b+MHC class II+ and accumulate in the fifth lumbar (L5) cord during the onset of a transfer model of EAE (tEAE), play a role in the pain-mediated relapse via the pain-gateway reflex. In this study, we investigated how these cells survive during the remission phase to cause the relapse. We show that peripheral-derived myeloid cells accumulated in the L5 cord after tEAE induction and survive more than other immune cells. These myeloid cells, which highly expressed GM-CSFRα with common ß chain molecules, grew in number and expressed more Bcl-xL after GM-CSF treatment but decreased in number by blockade of the GM-CSF pathway, which suppressed pain-mediated relapse of neuroinflammation. Therefore, GM-CSF is a survival factor for these cells. Moreover, these cells were colocalized with blood endothelial cells (BECs) around the L5 cord, and BECs expressed a high level of GM-CSF. Thus, GM-CSF from BECs may have an important role in the pain-mediated tEAE relapse caused by peripheral-derived myeloid cells in the CNS. Finally, we found that blockade of the GM-CSF pathway after pain induction suppressed EAE development. Therefore, GM-CSF suppression is a possible therapeutic approach in inflammatory CNS diseases with relapse, such as MS.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Camundongos , Animais , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Doenças Neuroinflamatórias , Células Endoteliais/metabolismo , Sistema Nervoso Central , Dor/metabolismo , Células Mieloides , Recidiva
2.
Cancer Immunol Immunother ; 72(4): 851-864, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36104597

RESUMO

Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype characterized by a lack of therapeutic targets. The paucity of effective treatment options motivated a number of studies to tackle this problem. Immunosuppressive cells infiltrated into the tumor microenvironment (TME) of TNBC are currently considered as candidates for new therapeutic targets. Myeloid-derived suppressor cells (MDSCs) have been reported to populate in the TME of TNBC, but their roles in the clinical and biological features of TNBC have not been clarified. This study identified that interleukin-34 (IL-34) released by TNBC cells is a crucial immunomodulator to regulate MDSCs accumulation in the TME. We provide evidence that IL-34 induces a differentiation of myeloid stem cells into monocytic MDSCs (M-MDSCs) that recruits regulatory T (Treg) cells, while suppressing a differentiation into polymorphonuclear MDSCs (PMN-MDSCs). As a result, the increase in M-MDSCs contributes to the creation of an immunosuppressive TME, and the decrease in PMN-MDSCs suppresses angiogenesis, leading to an acquisition of resistance to chemotherapy. Accordingly, blockade of M-MDSC differentiation with an estrogen receptor inhibitor or anti-IL-34 monoclonal antibody suppressed M-MDSCs accumulation causing retardation of tumor growth and restores chemosensitivity of the tumor by promoting PMN-MDSCs accumulation. This study demonstrates previously poorly understood mechanisms of MDSCs-mediated chemoresistance in the TME of TNBC, which is originated from the existence of IL-34, suggesting a new rationale for TNBC treatment.


Assuntos
Células Supressoras Mieloides , Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/patologia , Microambiente Tumoral , Linfócitos T Reguladores/patologia , Interleucinas
3.
Acta Neuropathol ; 145(5): 637-650, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36879070

RESUMO

A missense variant from methionine to arginine at codon 232 (M232R) of the prion protein gene accounts for ~ 15% of Japanese patients with genetic prion diseases. However, pathogenic roles of the M232R substitution for the induction of prion disease have remained elusive because family history is usually absent in patients with M232R. In addition, the clinicopathologic phenotypes of patients with M232R are indistinguishable from those of sporadic Creutzfeldt-Jakob disease patients. Furthermore, the M232R substitution is located in the glycosylphosphatidylinositol (GPI)-attachment signal peptide that is cleaved off during the maturation of prion proteins. Therefore, there has been an argument that the M232R substitution might be an uncommon polymorphism rather than a pathogenic mutation. To unveil the role of the M232R substitution in the GPI-attachment signal peptide of prion protein in the pathogenesis of prion disease, here we generated a mouse model expressing human prion proteins with M232R and investigated the susceptibility to prion disease. The M232R substitution accelerates the development of prion disease in a prion strain-dependent manner, without affecting prion strain-specific histopathologic and biochemical features. The M232R substitution did not alter the attachment of GPI nor GPI-attachment site. Instead, the substitution altered endoplasmic reticulum translocation pathway of prion proteins by reducing the hydrophobicity of the GPI-attachment signal peptide, resulting in the reduction of N-linked glycosylation and GPI glycosylation of prion proteins. To the best of our knowledge, this is the first time to show a direct relationship between a point mutation in the GPI-attachment signal peptide and the development of disease.


Assuntos
Síndrome de Creutzfeldt-Jakob , Doenças Priônicas , Príons , Animais , Camundongos , Humanos , Proteínas Priônicas/genética , Mutação Puntual , Glicosilfosfatidilinositóis/genética , Glicosilfosfatidilinositóis/metabolismo , Sinais Direcionadores de Proteínas/genética , Doenças Priônicas/genética , Doenças Priônicas/patologia , Síndrome de Creutzfeldt-Jakob/genética , Síndrome de Creutzfeldt-Jakob/patologia , Príons/genética , Príons/metabolismo , Mutação/genética
4.
Cell Tissue Res ; 381(2): 273-284, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32418131

RESUMO

The anterior pituitary gland is composed of five types of hormone-producing cells and folliculo-stellate cells. Folliculo-stellate cells do not produce anterior pituitary hormones but they are thought to play important roles as stem cells, phagocytes, or supporting cells of hormone-producing cells in the anterior pituitary. S100ß protein has been used as a folliculo-stellate cell marker in some animals, including rats. However, since no reliable molecular marker for folliculo-stellate cells has been reported in mice, genetic approaches for the investigation of folliculo-stellate cells in mice are not yet available. Aldolase C/Zebrin II is a brain-type isozyme and is a fructose-1,6-bisphosphate aldolase. In the present study, we first used immunohistochemistry to verify that aldolase C was produced in the anterior pituitary of rats. Moreover, using transgenic rats expressing green fluorescent protein under the control of the S100ß gene promoter, we identified aldolase C-immunoreactive signals in folliculo-stellate cells and marginal cells located in the parenchyma of the anterior pituitary and around Rathke's cleft, respectively. We also identified aldolase C-expressing cells in the mouse pituitary using immunohistochemistry and in situ hybridization. Aldolase C was not produced in any pituitary hormone-producing cells, while aldolase C-immunopositive signal co-localized with E-cadherin- and SOX2-positive cells. Using post-embedding immunoelectron microscopy, aldolase C-immunoreactive products were observed in the cytoplasm of marginal cells and folliculo-stellate cells of the mouse pituitary. Taken together, aldolase C is a common folliculo-stellate cell marker in the anterior pituitary gland of rodents.


Assuntos
Frutose-Bifosfato Aldolase/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Adeno-Hipófise , Animais , Biomarcadores/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Adeno-Hipófise/citologia , Adeno-Hipófise/metabolismo , Ratos , Ratos Transgênicos
5.
Biochem Biophys Res Commun ; 513(3): 708-713, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-30987827

RESUMO

Neutrophils rapidly migrate to infection sites after the recognition of invaders. During chemotaxis, neutrophils require energy supplied by mitochondria oxidative phosphorylation (OXPHOS), whereas neutrophils rely heavily on glycolysis under normal conditions. Mitochondrial OXPHOS correlates with mitochondrial morphology. Here, we examined the mitochondrial morphology of neutrophil-like differentiated HL-60 cells after chemoattractant N-formyl-Met-Leu-Phe (fMLP) stimulation. We found that mitochondrial morphology changes to a tubular form after fMLP stimulation. Mitochondrial OXPHOS activity and mitochondrial complex II significantly increased after fMLP stimulation. On the other hand, the silencing of mitochondrial fusion protein mitofusin 2 (MFN2) suppresses mitochondrial morphological changes. Furthermore, MFN2 silencing suppressed OXPHOS activation and chemotaxis after fMLP stimulation. These results suggest that MFN2 is involved in chemotaxis of differentiated HL-60 cells depending on mitochondria.


Assuntos
Quimiotaxia de Leucócito , GTP Fosfo-Hidrolases/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Neutrófilos/citologia , Células HL-60 , Humanos , Mitocôndrias/ultraestrutura , Neutrófilos/metabolismo , Fosforilação Oxidativa
6.
Biochem Biophys Res Commun ; 514(4): 1115-1121, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31101337

RESUMO

Cancer cells can invade as a population in various cancer tissues. This phenomenon is called collective invasion, which is associated with the metastatic potential and prognosis of cancer patients. The collectiveness of cancer cells is necessary for collective invasion. However, the mechanism underlying the generation of collectiveness by cancer cells is not well known. In this study, the phenomenon of contact following, where neighboring cells move in the same direction via intercellular adhesion, was investigated. An experimental system was created to observe the two-dimensional invasion using a collagen gel overlay to study contact following in collective invasion. The role of integrin-ß1, one of the major extracellular matrix (ECM) receptors, in contact following was examined through the experimental system. Integrin-ß1 was localized to the intercellular site in squamous carcinoma cells. Moreover, the intercellular adhesion and contact following were suppressed by treatment of an integrin-ß1 inhibitory antibody. ECM proteins such as laminin-332 and type-XVII collagen were also localized to the intercellular site and critical for contact following. Collectively, it was demonstrated that the activity of integrin-ß1 and expression of ECM proteins in the intercellular site promote contact following in the collective invasion of a cancer cell population.


Assuntos
Autoantígenos/metabolismo , Carcinoma de Células Escamosas/metabolismo , Moléculas de Adesão Celular/metabolismo , Integrina beta1/metabolismo , Colágenos não Fibrilares/metabolismo , Neoplasias Cutâneas/metabolismo , Autoantígenos/biossíntese , Carcinoma de Células Escamosas/patologia , Adesão Celular , Moléculas de Adesão Celular/biossíntese , Humanos , Integrina beta1/biossíntese , Colágenos não Fibrilares/biossíntese , Neoplasias Cutâneas/patologia , Células Tumorais Cultivadas , Calinina , Colágeno Tipo XVII
7.
Biochem Biophys Res Commun ; 517(1): 146-154, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31351587

RESUMO

The phosphinositide PtdIns(3)P plays an important role in autophagy; however, the detailed mechanism of its activity remains unclear. Here, we used a Systematic Evolution of Ligands by EXponential enrichment (SELEX) screening approach to identify an RNA aptamer of 40 nucleotides that specifically recognizes and binds to intracellular lysosomal PtdIns(3)P. Binding occurs in a magnesium concentration- and pH-dependent manner, and consequently inhibits autophagy as determined by LC3II/I conversion, p62 degradation, formation of LC3 puncta, and lysosomal accumulation of Phafin2. These effects in turn inhibited lysosomal acidification, and the subsequent hydrolytic activity of cathepsin D following induction of autophagy. Given the essential role of PtdIns(3)P as a key targeting molecule for autophagy induction, identification of this novel PtdIns(3)P RNA aptamer provides new opportunities for investigating the biological functions and mechanisms of phosphoinositides.


Assuntos
Aptâmeros de Nucleotídeos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/farmacologia , Autofagia/efeitos dos fármacos , Sequência de Bases , Linhagem Celular , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Técnica de Seleção de Aptâmeros , Proteínas de Transporte Vesicular/metabolismo
8.
Exp Eye Res ; 172: 1-9, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29596849

RESUMO

The hyaloid vasculature constitutes a transitory system nourishing the internal structures of the developing eye, but the mechanism of vascular regression and its cell biological characteristics are not fully understood. The present study aimed to reveal the specificity of the hyaloid vessels by a systematic immunohistochemical approach for marker substances of myeloid cells and the extracellular matrix (ECM) in neonatal mice. Macrophages immunoreactive for F4/80, cathepsin D, and LYVE-1 gathered around the vasa hyaloidea propria (VHP), while small round cells in vascular lumen of VHP were selectively immunoreactive for galectin-3; their segmented nuclei and immunoreactivities for Ly-6G, CD11b, and myeloperoxidase indicated their neutrophilic origin. VHP possessed thick ECM and a dense pericyte envelope as demonstrated by immunostaining for laminin, type IV collagen, integrin ß1, and NG2. The galectin-3+ cells loosely aggregated with numerous erythrocytes in the lumen of hyaloid vessels in a manner reminiscent of vascular congestion. Galectin-3 is known to polymerize and form a complex with ECM and NG2 as well as recruit leukocytes on the endothelium. Observation of galectin-3 KO mice implicated the involvement of galectin-3 in the regression of hyaloid vasculature. Since macrophages may play central roles including blocking of the blood flow and the induction of apoptosis in the regression, galectin-3+ neutrophils may play a supportive role in the macrophage-mediated involution of the hyaloid vascular system.


Assuntos
Vasos Sanguíneos/patologia , Corpo Vítreo/irrigação sanguínea , Animais , Animais Recém-Nascidos , Antígenos de Diferenciação/metabolismo , Antígenos Ly/metabolismo , Atrofia , Biomarcadores/metabolismo , Vasos Sanguíneos/metabolismo , Antígeno CD11b/metabolismo , Catepsina D/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Macrófagos/metabolismo , Camundongos , Microscopia de Fluorescência , Neutrófilos/metabolismo , Pericitos/metabolismo , Peroxidase/metabolismo , Gravidez , Proteínas de Transporte Vesicular/metabolismo
9.
Mol Hum Reprod ; 23(5): 271-281, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28333263

RESUMO

STUDY QUESTION: Do intraluteal prostaglandins (PG) contribute to luteal regulation in women? SUMMARY ANSWER: Prostaglandin E (PGE), which is produced in human granulosa-lutein cells stimulated with luteotropic hCG, exerts similar luteotropic effects to hCG, and the expression of PG synthetic and metabolic enzymes in the human CL is driven toward less PGE but more prostaglandin F (PGF) during luteolysis. WHAT IS KNOWN ALREADY: Uterine PGF is a major luteolysin in many non-primate species but not in women. Increases in the PGF synthase, aldo-ketoreductase family one member C3 (AKR1C3), have been observed in the CL of marmoset monkeys during luteolysis. PGE prevents spontaneous or induced luteolysis in domestic animals. STUDY DESIGN, SIZE, DURATION: Human CL tissues staged as the early-luteal (n = 6), mid-luteal (n = 6), late-luteal (n = 5) and menstrual (n = 3) phases were obtained at the time of hysterectomy for benign gynecological conditions. Luteinized granulosa cells (LGCs) were purified from follicular fluids obtained from patients undergoing assisted conception. PARTICIPANTS/MATERIALS, SETTING, METHODS: Upon collection, one half of the CL was snap-frozen and the other was fixed with formalin and processed for immunohistochemical analysis of a PGE synthase (PTGES). Quantitative RT-PCR was employed to examine changes in the mRNA abundance of PG synthetic and metabolic enzymes, steroidogenic enzymes, and luteolytic molecules in the staged human CL and in human LGCs in vitro treated with hCG, PGE and PGF. A PGE withdrawal experiment was also conducted in order to reveal the effects of the loss of PGE in LGCs. Progesterone concentrations in the culture medium were measured. MAIN RESULTS AND THE ROLE OF CHANCE: The key enzyme for PGE synthesis, PTGES mRNA was abundant in the functional CL during the mid-luteal phase (P < 0.01), while mRNA abundance for genes involved in PGF synthesis (AKR1B1 and AKR1C1-3) increased in the CL during the late-luteal phase and menstruation (P < 0.05-0.001). PTGES mRNA expression positively correlated with that of 3ß-hydroxysteroid dehydrogenase (HSD3B1; r = 0.7836, P < 0.001), while AKR1C3 expression inversely correlated with that of HSD3B1 (r = -0.7514, P = 0.0012) and PTGES (r = -0.6923, P = 0.0042). PGE exerted similar effects to hCG-promoting genes, such as steroidogenic acute regulatory protein (STAR) and HSD3B1, to produce progesterone and luteotropic PGE, suppress PGF synthetic enzymes and down-regulate luteolytic molecules such as ßA- and ßB-inhibin subunits (INHBA and INHBB) and bone morphogenetic proteins (BMP2, BMP4 and BMP6). PGE withdrawal resulted in reductions in the enzymes that produce progesterone (STAR; P < 0.001) and PGE (PTGES; P < 0.001), and the capacity to produce PGE decreased, while the capacity to produce PGF increased during the culture. The addition of PGF did not recapitulate the luteolytic effects of PGE withdrawal. LARGE SCALE DATA: None. LIMITATIONS, REASONS FOR CAUTION: Changes in mRNA expression of PG synthetic and metabolic enzymes may not represent actual increases in PGF during luteolysis in the CL. The effects of PGF on luteal cells currently remain unclear and the mechanisms responsible for decreases in the synthesis of PGE in vitro and at luteolysis have not been elucidated in detail. WIDER IMPLICATIONS OF THE FINDINGS: The results obtained strongly support a luteotropic function of PGE in regulation of the human CL. They suggest that the main PG produced in human luteal tissue changes from PGE to PGF during the maturation and regression of the CL, and the loss of PGE is more important than the effects of PGF during luteolysis in women. This may be accompanied by reduced effects of LH/hCG in luteal cells, particularly decreased activation of cAMP/protein kinase A; however, the underlying mechanisms remain unknown. STUDY FUNDING AND COMPETING INTEREST(S): This study was supported by the Cunningham Trust to WCD, a Postdoctoral Fellowship for Research Abroad from the Japan Society for the Promotion of Science and the Suntory Foundation for Life Sciences to J.N.-K.; W.C.D. is supported by an MRC Centre Grant G1002033 and a Scottish Senior Clinical Fellowship. The authors have nothing to disclose.


Assuntos
Corpo Lúteo/metabolismo , Células da Granulosa/metabolismo , Luteinização/fisiologia , Luteólise/genética , Prostaglandinas E/genética , 20-Hidroxiesteroide Desidrogenases/genética , 20-Hidroxiesteroide Desidrogenases/metabolismo , Aldeído Redutase/genética , Aldeído Redutase/metabolismo , Animais , Gonadotropina Coriônica/farmacologia , Corpo Lúteo/citologia , Corpo Lúteo/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Humanos , Subunidades beta de Inibinas/genética , Subunidades beta de Inibinas/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Fase Luteal/fisiologia , Menstruação/fisiologia , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fator de Crescimento Placentário/farmacologia , Cultura Primária de Células , Progesterona/biossíntese , Progesterona/metabolismo , Progesterona Redutase/genética , Progesterona Redutase/metabolismo , Prostaglandina-E Sintases/genética , Prostaglandina-E Sintases/metabolismo , Prostaglandinas E/deficiência , Prostaglandinas E/farmacologia , Transdução de Sinais , Esteroide Isomerases/genética , Esteroide Isomerases/metabolismo
10.
Biol Reprod ; 95(1): 17, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27281709

RESUMO

The corpus luteum (CL) is essential for establishing pregnancy. If pregnancy does not occur during the estrous cycle, luteolysis is induced by prostaglandin (PG) F2alpha secreted from the uterus. Galectin-1, a beta-galactose-binding protein, is expressed in the functional CL of cows and increases the viability of bovine luteal steroidogenic cells (LSCs) by modifying the functions of membrane glycoproteins. The binding of galectin-1 to glycoproteins is blocked by alpha2,6-sialylation of the terminal galactose residues of glycoconjugates, which is catalyzed by a sialyltransferase (ST6Gal-I). However, the physiological role of alpha2,6-sialic acid in bovine CL is unclear. The level of alpha2,6-sialylation of the bovine CL was higher during the regressed-luteal stage than in other luteal stages. Lectin histochemistry revealed that alpha2,6-sialylated glycoconjugates were localized to luteal endothelial cells throughout the estrous cycle. In addition, alpha2,6-sialylated glycoconjugates concentrated to the membrane of LSCs during the regressed-luteal stage. PGF2alpha treatment for 72 h enhanced the expression of ST6Gal-I mRNA and the level of alpha2,6-sialylated glycoproteins in mid-LSCs. The level of alpha2,6-sialylated glycoproteins of late-stage LSCs (Days 15-17 after ovulation) was higher than that of mid-stage LSCs (Days 8-12 after ovulation), and galectin-1 increased the viability of mid-LSCs but not that of late-stage LSCs. Furthermore, galectin-1 increased the viability of late-LSCs when alpha2,6-sialic acid residues were removed by neuraminidase. The overall findings suggest that alpha2,6-sialylation stimulated by PGF2alpha contributes to luteolysis by inhibiting the luteotropic effects of galectin-1 in bovine CL.


Assuntos
Corpo Lúteo/metabolismo , Dinoprosta/farmacologia , Galectina 1/metabolismo , Células Lúteas/metabolismo , Luteólise/fisiologia , Animais , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Corpo Lúteo/efeitos dos fármacos , Feminino , Galectina 1/farmacologia , Células Lúteas/efeitos dos fármacos , Luteólise/efeitos dos fármacos
11.
Mol Reprod Dev ; 83(12): 1083-1091, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27704662

RESUMO

Sialylation creates a negative charge on the cell surface that can interfere with blastocyst implantation. For example, α2,6-sialylation on terminal galactose, catalyzed by the sialyltransferase ST6GAL1, inhibits the binding of galectin-1, a ß-galactoside-binding lectin. We recently reported the potential involvement of galectin-1 and -3 in the pathogenesis of tubal ectopic pregnancy; however, the precise role of galectins and their ligand glycoconjugates remain unclear. Here, we investigated the expression of the genes encoding α2,3- and α2,6-galactoside sialyltransferases (ST3GAL1-6 and ST6GAL1-2) and the localization of sialic acids in the Fallopian tube of women with or without ectopic implantation. ST6GAL1 expression was higher in the mid-secretory phase than the proliferative phase of non-pregnant women (P < 0.0001), whereas ST6GAL1 (P < 0.0001), ST3GAL3 (P = 0.0029), ST3GAL5 (P = 0.0089), and ST3GAL6 (P = 0.0018) were all lower in Fallopian tubes with ectopic implantations. α2,3- and α2,6-sialic acids, however, both remained enriched on the surface of Fallopian tube epithelium. Cigarette smoking, a major risk factor for tubal ectopic pregnancy, was associated with reduced mid-secretory-phase expression of ST6GAL1 (P = 0.0298), but elevated expression of ST3GAL5 (P = 0.0006), an enzyme known to be involved in ciliogenesis. Indeed, sialic acid-containing ciliated inclusion cysts, which are associated with abnormal ciliogenesis, were observed within the epithelium at a higher frequency in women who smoked (P = 0.0177), suggesting that abnormal ciliogenesis is associated with smoking. Thus, cigarette smoking alters sialylation in the Fallopian tube epithelium, and is potentially a source of decreased tubal transport and increased receptivity for blastocyst in the human Fallopian tube. Mol. Reprod. Dev. 83: 1083-1091, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Tubas Uterinas/metabolismo , Regulação Enzimológica da Expressão Gênica , Ácido N-Acetilneuramínico/metabolismo , Gravidez Ectópica/metabolismo , Sialiltransferases/biossíntese , Fumar/efeitos adversos , Adolescente , Adulto , Tubas Uterinas/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Gravidez , Gravidez Ectópica/etiologia , Gravidez Ectópica/patologia , Fumar/metabolismo , Fumar/patologia
12.
Mol Hum Reprod ; 21(8): 645-54, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26003810

RESUMO

Intense macrophage infiltration is observed during luteolysis in various animals including women; however, we still do not know how macrophage infiltration into the human corpus luteum (CL) during luteolysis is regulated. In this study, we examined the expression, localization and regulation of an important chemokine for the recruitment of monocyte/macrophage lineages, C-C motif ligand 2 (CCL2), in the human CL across the luteal phase and in cultured human luteinized granulosa cells (LGCs), with special reference to the number of infiltrating macrophages and luteal cell function. CCL2 mRNA increased in the non-functional regressing CL during menstruation (P < 0.01), corresponding to an elevated mRNA expression of a macrophage-derived cytokine, tumor necrosis factor (TNF), and an increased number of infiltrating macrophages positively stained with a macrophage marker, CD68. CCL2 protein was immunohistochemically localized to the cytoplasm of granulosa-lutein and theca-lutein cells, and CCL2 mRNA was significantly reduced by hCG both in vivo (P < 0.05) and in vitro (P < 0.01). CCL2 was also down-regulated by luteotrophic prostaglandin (PG) E (P < 0.0001), but up-regulated by luteolytic PGF (P < 0.05) in vitro. Administration of TNF significantly enhanced the CCL2 mRNA expression in cultured LGCs (P < 0.01). A greater abundance of infiltrating macrophages were found around granulosa-lutein cells lacking 3ß-HSD or PGE synthase (PGES) immunostaining. CCL2 mRNA expression was negatively correlated with both HSD3B1 and PGES, suggesting that locally produced progesterone and PGE suppress macrophage infiltration into the CL. Taken together, the infiltration of macrophages in the human CL is regulated by endocrine and paracrine molecules via regulation of the CCL2 expression in luteal cells.


Assuntos
Quimiocina CCL2/metabolismo , Corpo Lúteo/metabolismo , Células Lúteas/citologia , Células Lúteas/metabolismo , Luteólise/genética , Macrófagos/citologia , Macrófagos/metabolismo , Apoptose/genética , Apoptose/fisiologia , Células Cultivadas , Corpo Lúteo/citologia , Feminino , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Luteólise/fisiologia
13.
Cells Tissues Organs ; 200(6): 424-34, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26359845

RESUMO

Galectin-1 and galectin-3 are abundantly expressed at implantation sites in the uterus, suggesting their involvement in the establishment of pregnancy. In this study, we examined the expression and localization of galectin-1 and galectin-3 in fallopian tubes from nonpregnant women, and in those presenting with tubal ectopic pregnancy. There was no significant difference in the expression of either galectin-1 (LGALS1) or galectin-3 (LGALS3) transcripts in the fallopian tube across the menstrual cycle. Their expressions in the fallopian tube were inversely correlated to each other (r = -0.5134, p < 0.0001) and differentially localized. Galectin-1 protein was abundant in the stroma of nonpregnant fallopian tubes, whereas galectin-3 was mainly localized to the epithelium, notably to the cilia of ciliated cells and the apical cytoplasm of secretory cells. In ectopic pregnancies, LGALS3 expression was significantly reduced (p < 0.0001), but LGALS1 expression did not change when compared to nonpregnant fallopian tubes collected during the mid-secretory phase. The percentage of fallopian tube epithelial cells expressing galectin-3 in cilia tended to be reduced (p = 0.0685), with an accompanying loss of a normal ciliary structure, while nuclear galectin-3 increased (p < 0.05) in ectopic pregnancies. Epithelial immunostaining for galectin-1 tended to be elevated in fallopian tubes from women with ectopic pregnancy. Coculture of human trophoblast origin SW71 cells significantly increased LGALS1 expression in human fallopian tube epithelial OE-E6/E7 cells, suggesting that trophoblast-derived products regulate LGALS1 expression in the oviductal epithelium. These findings imply a differential contribution of galectin-1 and galectin-3 in the homeostasis of human fallopian tubes and in the pathophysiology of ectopic pregnancy.


Assuntos
Tubas Uterinas/patologia , Galectina 1/análise , Galectina 3/análise , Regulação da Expressão Gênica , Gravidez Tubária/genética , Gravidez Tubária/patologia , Adolescente , Adulto , Linhagem Celular , Tubas Uterinas/metabolismo , Feminino , Galectina 1/genética , Galectina 3/genética , Humanos , Pessoa de Meia-Idade , Gravidez , Gravidez Tubária/sangue , Adulto Jovem
14.
J Reprod Dev ; 61(5): 439-48, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26155753

RESUMO

The corpus luteum (CL) is a temporary endocrine gland producing a large amount of progesterone, which is essential for the establishment and maintenance of pregnancy. Galectin-1 is a ß-galactose-binding protein that can modify functions of membrane glycoproteins and is expressed in the CL of mice and women. However, the physiological role of galectin-1 in the CL is unclear. In the present study, we investigated the expression and localization of galectin-1 in the bovine CL and the effect of galectin-1 on cultured luteal steroidogenic cells (LSCs) with special reference to its binding to the glycans on vascular endothelial growth factor receptor-2 (VEGFR-2). Galectin-1 protein was highly expressed at the mid and late luteal stages in the membrane fraction of bovine CL tissue and was localized to the surface of LSCs in a carbohydrate-dependent manner. Galectin-1 increased the viability in cultured LSCs. However, the viability of LSCs was decreased by addition of ß-lactose, a competitive carbohydrate inhibitor of galectin-1 binding activity. VEGFR-2 protein, like galectin-1, is also highly expressed in the mid CL, and it was modified by multi-antennary glycans, which can be recognized by galectin-1. An overlay assay using biotinylated galectin-1 revealed that galectin-1 directly binds to asparagine-linked glycans (N-glycans) on VEGFR-2. Enhancement of LSC viability by galectin-1 was suppressed by a selective inhibitor of VEGFR-2. The overall findings suggest that galectin-1 plays a role as a survival factor in the bovine CL, possibly by binding to N-glycans on VEGFR-2.


Assuntos
Corpo Lúteo/metabolismo , Galectina 1/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células Lúteas/metabolismo , Luteinização , Polissacarídeos/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/agonistas , Animais , Animais Endogâmicos , Ligação Competitiva , Bovinos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Corpo Lúteo/citologia , Corpo Lúteo/efeitos dos fármacos , Feminino , Galectina 1/antagonistas & inibidores , Galectina 1/genética , Humanos , Lactose/análogos & derivados , Lactose/metabolismo , Células Lúteas/citologia , Células Lúteas/efeitos dos fármacos , Luteinização/efeitos dos fármacos , Polissacarídeos/química , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Proteínas Recombinantes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Propriedades de Superfície , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/química , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
Biol Reprod ; 91(1): 2, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24855102

RESUMO

Luteolysis is characterized by a reduction in progesterone (P4) production and tissue degeneration in the corpus luteum (CL). One of major events during luteolysis is luteal cell death. Galectin-3, a ubiquitously expressed protein involved in many cellular processes, serves as an antiapoptotic and/or proapoptotic factor in various cell types. Although galectin-3 is detected in the bovine CL, its role remains unclear. The expression of galectin-3 in the bovine CL was higher at the regressed stage than at the other luteal stages. Galectin-3 was localized on luteal steroidogenic cells (LSCs). When cultured LSCs were exposed to prostaglandin F2alpha (PGF) for 48 h, the expression and secretion of galectin-3 increased. When the cultured LSCs were treated with galectin-3 for 24 h, cleaved caspase-3 expression was increased, and the cell viability was decreased, whereas P4 production did not change. Beta 1 integrin, a target protein of galectin-3, was expressed in bovine CL and possessed glycans, which galectin-3 binds. Furthermore, galectin-3 bound to glycans of luteal beta 1 integrin. The decreased cell viability of cultured LSCs by galectin-3 was suppressed by beta 1 integrin antibody. The overall findings suggest that the secreted galectin-3 stimulated by PGF plays a role in structural luteolysis by binding to beta 1 integrin.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Corpo Lúteo/metabolismo , Galectina 3/metabolismo , Integrina beta1/metabolismo , Células Lúteas/metabolismo , Luteólise/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Caspase 3/metabolismo , Bovinos , Sobrevivência Celular/fisiologia , Células Cultivadas , Corpo Lúteo/citologia , Corpo Lúteo/efeitos dos fármacos , Dinoprosta/farmacologia , Feminino , Galectina 3/farmacologia , Células Lúteas/citologia , Células Lúteas/efeitos dos fármacos , Luteólise/efeitos dos fármacos
16.
Cells ; 13(7)2024 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-38607051

RESUMO

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) featuring numerous neuropathologies, including optic neuritis (ON) in some patients. However, the molecular mechanisms of ON remain unknown. Galectins, ß-galactoside-binding lectins, are involved in various pathophysiological processes. We previously showed that galectin-3 (gal-3) is associated with the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of MS. In the current study, we investigated the expression of gal-3 in the visual pathway in EAE mice to clarify its role in the pathogenesis of ON. Immunohistochemical analysis revealed upregulation of gal-3 in the visual pathway of the EAE mice during the peak stage of the disease, compared with naïve and EAE mice during the chronic stage. Gal-3 was detected mainly in microglia/macrophages and astrocytes in the visual pathway in EAE mice. In addition, gal-3+/Iba-1+ cells, identified as phagocytic by immunostaining for cathepsin D, accumulated in demyelinating lesions in the visual pathway during the peak disease stage of EAE. Moreover, NLRP3 expression was detected in most gal-3+/Iba-1+ cells. These results strongly suggest that gal-3 regulates NLRP3 signaling in microglia/macrophages and neuroinflammatory demyelination in ON. In astrocytes, gal-3 was expressed from the peak to the chronic disease stages. Taken together, our findings suggest a critical role of gal-3 in the pathogenesis of ON. Thus, gal-3 in glial cells may serve as a potential therapeutic target for ON.


Assuntos
Galectina 3 , Neurite Óptica , Animais , Humanos , Camundongos , Encefalomielite Autoimune Experimental/patologia , Galectina 3/metabolismo , Galectinas/metabolismo , Esclerose Múltipla/patologia , Doenças Neuroinflamatórias , Proteína 3 que Contém Domínio de Pirina da Família NLR , Neurite Óptica/patologia , Vias Visuais/patologia
17.
Mol Hum Reprod ; 19(2): 82-92, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23160862

RESUMO

Members of the transforming growth factor-ß (TGF-ß) superfamily are likely to have major roles in the regulation of tissue and vascular remodelling in the corpus luteum (CL). There are four inhibitor-of-differentiation (ID1-4) genes that are regulated by members of the TGF-ß superfamily and are involved in the transcriptional regulation of cell growth and differentiation. We studied their expression, localization and regulation in dated human corpora lutea from across the luteal phase (n = 22) and after human chorionic gonadotrophin (hCG) administration in vivo (n = 5), and in luteinized granulosa cells (LGCs), using immunohistochemistry and quantitative RT-PCR. ID1-4 can be localized to multiple cell types in the CL across the luteal phase. Endothelial cell ID3 (P < 0.05) and ID4 (P < 0.05) immunostaining intensities peak at the time of angiogenesis but overall ID1 (P < 0.05) and ID3 (P < 0.05) expression peaks at the time of luteolysis, and luteal ID3 expression is inhibited by hCG in vivo (P < 0.01). In LGC cultures in vitro, hCG had no effect on ID1, down-regulated ID3 (P < 0.001), and up-regulated ID2 (P < 0.001) and ID4 (P < 0.01). Bone morphogenic proteins (BMPs) had no effect on ID4 expression but up-regulated ID1 (P < 0.01 to P < 0.005). BMP up-regulation of ID2 (P < 0.05) was additive to the hCG up-regulation of ID2 expression (P < 0.001), while BMP cancelled out the down regulative effect of hCG on ID3 regulation. As well as documenting regulation patterns specific for ID1, ID2, ID3 and ID4, we have shown that IDs are located and differentially regulated in the human CL, suggesting a role in the transcriptional regulation of luteal cells during tissue and vascular remodelling.


Assuntos
Corpo Lúteo/metabolismo , Proteínas Inibidoras de Diferenciação/metabolismo , Proteínas Morfogenéticas Ósseas/farmacologia , Células Cultivadas , Gonadotropina Coriônica/farmacologia , Corpo Lúteo/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Humanos , Proteína 1 Inibidora de Diferenciação/metabolismo , Proteína 2 Inibidora de Diferenciação/metabolismo , Proteínas Inibidoras de Diferenciação/genética , Proteínas de Neoplasias/metabolismo
18.
Reprod Fertil Dev ; 25(2): 362-71, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22951108

RESUMO

The ovary is a key tissue in the study of physiological neo-vascularisation in the adult and its study has highlighted important molecules involved in the regulation of angiogenesis in vivo. These include vascular endothelial growth factor, delta-like ligand 4, thrombospondin-1, prokineticin-1 and prostaglandin E2. Targeting these molecular pathways has therapeutic potential and their manipulation has an increasing preclinical and clinical role in the management of the pathological ovary. Targeting angiogenic pathways has utility in the promotion of ovarian angiogenesis to improve tissue and follicle survival and function as well as the prevention and management of ovarian hyperstimulation syndrome. There is a theoretical possibility that targeting angiogenesis may improve the function of the polycystic ovary and a real role for targeting angiogenesis in ovarian cancer.


Assuntos
Corpo Lúteo/irrigação sanguínea , Regulação da Expressão Gênica/fisiologia , Modelos Biológicos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Doenças Ovarianas/fisiopatologia , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Aminoquinolinas/farmacologia , Proteínas de Ligação ao Cálcio , Gonadotropina Coriônica/farmacologia , Corpo Lúteo/efeitos dos fármacos , Dinoprostona/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Hormônio Luteinizante/metabolismo , Doenças Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Síndrome do Ovário Policístico/tratamento farmacológico , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Reproduction ; 144(5): 617-24, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22956518

RESUMO

Galectin-1 and galectin-3, ß-galactoside-binding lectins, are specifically expressed in the regressing corpus luteum (CL) of mice; however, their function remains unclear. In this study, we examined the effects of prolactin (PRL) and prostaglandin F(2) (α) (PGF(2) (α)), two main regulatory molecules of mouse CL function, on galectin expression. In situ hybridization analysis clearly demonstrated an initial increase in galectin-1 in the newly formed CL (CLN) after postpartum ovulation 48  h after compulsory weaning. This was accompanied by a decline in 3ß-hydroxysteroid dehydrogenase (3ß-HSD) and LH receptor (LH-R) expression, suggesting a withdrawal of PRL stimulation. At 72  h after the weaning, the expression of both galectins in CLN was remarkably increased, being associated with an intense expression of progesterone degradation enzyme (20α-HSD). Compulsory weaning did not significantly alter both galectin expression in the remaining CL of pregnancy (CLP), while PGF(2) (α) strongly upregulated both galectin expression only in the remaining CLP, which lacked LH-R in postpartum mice. Administration of bromocriptine, an antagonist for PRL secretion, to nonpregnant cyclic mice induced an accumulation of galectin-1 - but not galectin-3 - in all CL of various generations, and additional PRL treatment reduced its accumulation, suggesting a direct suppressive effect of PRL on galectin-1 expression. Although the function and regulatory mechanism of galectin in the CL is not fully understood, PGF(2) (α) is an excellent candidate that regulates galectin expression, but its effect may be abolished by LH-R-mediated signal. PRL withdrawal seems to be necessary for an initiation of luteolysis and the following PGF(2) (α)-induced galectin expression.


Assuntos
Corpo Lúteo/metabolismo , Dinoprosta/farmacologia , Galectina 1/genética , Galectina 3/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Prolactina/farmacologia , Animais , Bromocriptina/farmacologia , Corpo Lúteo/química , Feminino , Galectina 1/análise , Galectina 3/análise , Hibridização In Situ , Camundongos , Ovário/metabolismo , Período Pós-Parto , Gravidez , RNA Mensageiro/análise , Desmame
20.
Biomed Res ; 43(5): 161-171, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36244794

RESUMO

Mammals express a set of chitinase family proteins, comprising chitinases, which can hydrolyze chitin, and chitinase-like proteins without the chitinase activity but possessing chitin-binding properties. They act as endogenous lectins, regulating various physiological/pathological events. Ym1, originally identified as an eosinophil chemotactic factor or a macrophage-derived protein in parasite-infected mice, is a rodent-specific chitinase-like protein. Ym1 is also purified from eosinophilic crystals formed in the lung and urinary system in various disease models. We previously reported that major cellular sources of murine Ym1 are alveolar macrophages in the lung and neutrophils/monocytes lineage cells of the spleen and bone marrow under normal conditions. We here analyzed the detailed cellular expression of Ym1 in Mesocestoides corti (M. corti)-infected mice. Ym1 was significantly increased in the liver containing the larvae, lung, and peritoneal exudate cells in M. corti-infected mice, where activated macrophages expressed Ym1. Characteristic needle-shaped eosinophilic crystals appeared in the larvae-free lung, and Ym1 was localized to endoplasmic reticulum of activated alveolar macrophages. Moreover, swollen mesothelial cells covering the liver, spleen, and heart expressed Ym1 abundantly. Although the role of Ym1 in parasitic infection remains unclear, our findings focusing on an endogenous lectin may help in better understanding defense mechanism against parasites.


Assuntos
Quitinases , Mesocestoides , Animais , Camundongos , beta-N-Acetil-Hexosaminidases/metabolismo , Fatores Quimiotáticos , Quitina , Quitinases/química , Quitinases/genética , Lectinas/química , Lectinas/metabolismo , Mamíferos/metabolismo , Mesocestoides/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA