Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Clin Transl Med ; 11(10): e540, 2021 10.
Article in English | MEDLINE | ID: mdl-34709764

ABSTRACT

Natural killer (NK) cells preferentially accumulate at maternal-foetal interface and are believed to play vital immune-modulatory roles during early pregnancy and related immunological dysfunction may result in pregnant failure such as recurrent miscarriage (RM). However, the mechanisms underlying the establishment of maternal-foetal immunotolerance are complex but clarifying the roles of decidual NK (dNK) cells offers the potential to design immunotherapeutic strategies to assist RM patients. In this report, we analysed RNA sequencing on peripheral NK (pNK) and decidual NK cells during early pregnancy; we identified an immunomodulatory dNK subset CXCR4+ CD56bright dNK and investigated its origin and phenotypic and functional characteristics. CXCR4+ CD56bright dNK displayed a less activated and cytotoxic phenotype but an enhanced immunomodulatory potential relative to the CXCR4 negative subset. CXCR4+ CD56bright dNK promote Th2 shift in an IL-4-dependent manner and can be recruited from peripheral blood and reprogramed by trophoblasts, as an active participant in the establishment of immune-tolerance during early pregnancy. Diminished CXCR4+ dNK cells and their impaired ability to induce Th2 differentiation were found in RM patients and mouse models of spontaneous abortion. Moreover, adoptive transfer of CXCR4+ dNK cells to NK-deficient (Nfil3-/-) mice showed great therapeutic potential of CXCR4+ dNK via recovering the Th2/Th1 bias and reducing embryo resorption rates. The identification of this new dNK cell subset may lay the foundation for understanding NK cell mechanisms in early pregnancy and provide potential prognostic factors for the diagnosis and therapy of RM.


Subject(s)
Abortion, Habitual/prevention & control , Immune Tolerance/immunology , Killer Cells, Natural/immunology , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Abortion, Habitual/blood , Abortion, Habitual/immunology , Animals , Decidua/immunology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred BALB C , Neural Cell Adhesion Molecules/blood , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/immunology , Pregnancy , Pregnancy Trimester, First , Receptors, CXCR4/blood
2.
BMJ Open ; 7(7): e015666, 2017 Jul 20.
Article in English | MEDLINE | ID: mdl-28729316

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the relationship between receipt of the substitutable-for-fee vaccines (SFV) and completion of the expanded programme on immunisation (EPI). DESIGN AND SETTINGS: A cross-sectional study was conducted in Fujian province, China. PARTICIPANTS: Children who were born from 1 September 2009 to 31 August 2011, and who had been residing in the township for at least 3 months, were randomly recruited from 34 townships. MAIN OUTCOMES MEASURES: Outcomes were completion rate of the EPI and coverage rate of the SFV. RESULTS: The study included 1428 children, of whom 1350 (94.5%) finished the EPI and 282 (19.7%) received at least one dose of the SFV. Administration of the SFV was associated with an increased likelihood of completing the EPI (OR=3.2, 95% CI 1.3 to 7.6 in the total sample and OR=4.0, 95% CI 1.7 to 9.6 in the subsample of children in regions with the SFV accessibility). The impact of the SFV administration on completion of the EPI was larger among children whose parents have junior school education or less (97.8% and 97.9% vs 92.5% and 91.9%, both p<0.001) and among those with a timely hepatitis B vaccine first dose (98.5% vs 94.0%, p<0.001). CONCLUSIONS: Receipt of SFV is associated with increased likelihood of completion of the EPI in Fujian, China.


Subject(s)
Immunization Programs/statistics & numerical data , Vaccination/statistics & numerical data , Vaccines/economics , Child, Preschool , China , Cross-Sectional Studies , Female , Humans , Immunization Schedule , Logistic Models , Male , Multivariate Analysis , Proportional Hazards Models
5.
Exp Mol Pathol ; 100(3): 506-13, 2016 06.
Article in English | MEDLINE | ID: mdl-27163202

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is frequently altered in human malignancies and Akt over-expression and/or activation induces malignant transformation and chemoresistance. However, the role of Akt in the mechanisms of chemoresistance remains elusive. Here we reported that cisplatin treatment of chemosensitive, but not resistant, ovarian cancer cells (OVCAs) markedly increased the cell proportion in sub-G1 phase. Cisplatin however caused a significant accumulation of the resistant cells in S and G2/M phases, which was associated with a rapid and sustained checkpoint kinase 1 (Chk1) activation. In contrast, while cisplatin also elicited a rapid activation of Chk1 in sensitive cells, it markedly decreased total ChK1 and phospho-Chk1 contents over 12 h. Over-expression of dominant negative (DN)-AKT alone increased phospho-Chk1 content, and induced G2/M arrest and apoptosis. However, it inhibited Chk1 activation and G2/M arrest with combination of cisplatin treatment, resulting in p53-independent apoptosis. Furthermore, the responses of the chemoresistant cells to cisplatin were attenuated with forced expression of constitutive active AKT2. Chk1 knock-down also facilitated cisplatin-induced apoptosis in chemoresistant cells. Our studies implicate that, in addition to its cell survival and anti-apoptotic actions, Akt might also play an important role in the regulation of G2-M transition, possibly via up-regulation of Chk1 activity and stability. These data provide strong support for the concept that Akt is important in cell cycle regulation in the control of chemosensitivity in OVCAs and offers an alternate regulatory pathway for the development of rationale therapy for cisplatin-resistant ovarian cancer.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , S Phase Cell Cycle Checkpoints/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Female , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference
6.
Cell Mol Immunol ; 13(1): 73-81, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25578313

ABSTRACT

Decidual natural killer (dNK) cells actively participate in the establishment and maintenance of maternal-fetal immune tolerance and act as local guardians against infection. However, how dNK cells maintain the immune balance between tolerance and anti-infection immune responses during pregnancy remains unknown. Here, we demonstrated that the inhibitory molecule T-cell immunoglobulin domain and mucin domain-containing molecule-3 (Tim-3) are expressed on over 60% of dNK cells. Tim-3(+) dNK cells display higher interleukin (IL)-4 and lower tumor necrosis factor (TNF)-α and perforin production. Human trophoblast cells can induce the transformation of peripheral NK cells into a dNK-like phenotype via the secretion of galectin-9 (Gal-9) and the interaction between Gal-9 and Tim-3. In addition, trophoblasts inhibit lipopolysaccharide (LPS)-induced pro-inflammatory cytokine and perforin production by dNK cells, which can be attenuated by Tim-3 neutralizing antibodies. Interestingly, a decreased percentage of Tim-3-expressing dNK cells were observed in human miscarriages and murine abortion-prone models. Moreover, T helper (Th)2-type cytokines were decreased and Th1-type cytokines were increased in Tim-3(+) but not Tim-3(-) dNK cells from human and mouse miscarriages. Therefore, our results suggest that the Gal-9/Tim-3 signal is important for the regulation of dNK cell function, which is beneficial for the maintenance of a normal pregnancy.


Subject(s)
Abortion, Spontaneous/genetics , Galectins/immunology , Killer Cells, Natural/immunology , Membrane Proteins/immunology , Trophoblasts/immunology , Abortion, Spontaneous/immunology , Abortion, Spontaneous/pathology , Adult , Animals , Antibodies, Neutralizing/pharmacology , Coculture Techniques , Decidua/immunology , Decidua/pathology , Female , Galectins/genetics , Gene Expression Regulation , Hepatitis A Virus Cellular Receptor 2 , Humans , Immune Tolerance , Interleukin-4/genetics , Interleukin-4/immunology , Killer Cells, Natural/pathology , Lipopolysaccharides/pharmacology , Maternal-Fetal Exchange/immunology , Membrane Proteins/genetics , Mice , Mice, Inbred CBA , Mice, Inbred DBA , Perforin/genetics , Perforin/immunology , Pregnancy , Primary Cell Culture , Signal Transduction , Trophoblasts/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
Cell Mol Immunol ; 12(1): 77-86, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24793405

ABSTRACT

Decidual natural killer (dNK) cells are believed to be critical for maintaining maternal/fetal tolerance and regulating placental vascular remodeling based upon their abundance and unique phenotype during early pregnancy. However, the mechanism for how the dNK cells play such important roles in successful pregnancy remains undefined. Here, we identified a subtype of dNK cells characterized as having a CD3(-)CD56(bright)CD25(+) phenotype. We found that CD56(bright)CD25(+) NK cells preferentially localize to the maternal/fetal interface during early human pregnancy. CD25(+) dNK cells account for approximately 75% of CD25-expressing decidual immune cells (DICs). However, less than 5% of CD25-positive peripheral blood mononuclear cells are CD25(+) NK cells. Furthermore, CD25(+) and CD25(-) dNK cells exhibit distinct phenotypes: CD25(+) dNK cells display a more activated phenotype and greater cytokine-secreting capacity. Interestingly, coculture of peripheral NK (pNK) cells with primary trophoblasts upregulates the percentage of CD25-expressing pNK cells, resulting in increased expression of activation markers and cytokine production by pNK cells. In addition, we demonstrated that the CXCL12/CXCR4 axis is crucial for the recruitment of CD25(+) dNK cells and contributes to the accumulation of CD3(-)CD56(bright)CD25(+) dNK cells at the maternal/fetal interface. Thus, our data reveal that the crosstalk between trophoblasts and pNK cells leads to the accumulation of CD3(-)CD56(bright)CD25(+) dNK cells, which exert a regulating effect at the maternal/fetal interface.


Subject(s)
Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Trophoblasts/immunology , CD56 Antigen/metabolism , Cell Communication , Cell Movement , Cells, Cultured , Chemokine CXCL12/metabolism , Coculture Techniques , Cytokines/metabolism , Female , Humans , Immunophenotyping , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Maternal-Fetal Exchange/immunology , Pregnancy/immunology , Receptors, CXCR4
8.
J Immunol ; 192(4): 1502-11, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24453244

ABSTRACT

Physiological pregnancy requires the maternal immune system to recognize and tolerate embryonic Ags. Although multiple mechanisms have been proposed, it is not yet clear how the fetus evades the maternal immune system. In this article, we demonstrate that trophoblast-derived thymic stromal lymphopoietin (TSLP) instructs decidual CD11c(+) dendritic cells (dDCs)with increased costimulatory molecules; MHC class II; and Th2/3-type, but not Th1-type, cytokines. TSLP-activated dDCs induce proliferation and differentiation of decidual CD4(+)CD25(-) T cells into CD4(+)CD25(+)FOXP3(+) regulatory T cells (Tregs) through TGF-ß1. TSLP-activated dDC-induced Tregs display immunosuppressive features and express Th2-type cytokines. In addition, decidual CD4(+)CD25(+)FOXP3(+) Tregs promote invasiveness and HLA-G expression of trophoblasts, resulting in preferential production of Th2 cytokines and reduced cytotoxicity in decidual CD56(bright)CD16(-) NK cells. Of interest, decreased TSLP expression and reduced numbers of Tregs were observed at the maternal-fetal interface during miscarriage. Our study identifies a novel feedback loop between embryo-derived trophoblasts and maternal decidual leukocytes, which induces a tolerogenic immune response to ensure a successful pregnancy.


Subject(s)
Cytokines/metabolism , Dendritic Cells/metabolism , Histocompatibility, Maternal-Fetal/immunology , T-Lymphocytes, Regulatory/metabolism , Abortion, Spontaneous/metabolism , Adult , CD11c Antigen/immunology , CD4 Antigens/metabolism , CD56 Antigen/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/immunology , Decidua/cytology , Decidua/metabolism , Dendritic Cells/immunology , Female , Forkhead Transcription Factors/metabolism , Histocompatibility Antigens Class II , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Killer Cells, Natural/immunology , Pregnancy , Receptors, IgG/metabolism , Th2 Cells/metabolism , Transforming Growth Factor beta1/metabolism , Trophoblasts/immunology , Trophoblasts/metabolism , Young Adult , Thymic Stromal Lymphopoietin
9.
Int J Clin Exp Pathol ; 6(10): 1999-2010, 2013.
Article in English | MEDLINE | ID: mdl-24133577

ABSTRACT

Our previous studies have demonstrated that cyclosporin A (CsA) promotes the proliferation and migration of human trophoblasts via the mitgen-activated protein kinase-3/1 (MAPK3/1) pathway. In the present study, we further investigated the role of nuclear factor (NF)-κB in the CsA-induced trophoblast proliferating cell nuclear antigen (PCNA) expression and migration, and its relationship to MAPK3/1 signal. Flow cytometry was used to analyze the expression of PCNA in trophoblasts. The migration of human primary trophoblasts was determined by wound-healing assay and transwell migration assay. Western blot analysis was performed to evaluate the activation of NF-κB p65 and NF-κB inhibitory protein I-κB in human trophoblasts. We found that treatment with CsA promotes PCNA expression and migration of human trophoblast in a dose-associated manner. Blocking of the MAPK3/1 signal abrogated the enhanced PCNA expression and migration in trophoblasts by CsA. In addition, CsA increased the phosphorylation of NF-κB p65 and the inhibitor I-κB in human trophoblasts in a time-related manner. Pretreatment with MAPK3/1 inhibitor U0126 abrogated the phosphorylation of NF-κB p65 and I-κB. Accordingly, the CsA-induced enhancement of PCNA expression and migration in trophoblasts was also decreased. This CsA-induced enhancement in the expression and migration of trophoblasts was abolished by pretreatment with pyrrolidine dithiocarbamate, a specific NF-κB inhibitor. Thus, our results suggest that CsA promotes PCNA expression and migration of human trophoblasts via MAPK-mediated NF-κB activation.


Subject(s)
Cell Movement/drug effects , Cyclosporine/pharmacology , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Trophoblasts/cytology , Butadienes/pharmacology , Cell Movement/physiology , Enzyme Inhibitors/pharmacology , Female , Humans , Nitriles/pharmacology , Phosphorylation/drug effects , Phosphorylation/physiology , Placenta/cytology , Placenta/drug effects , Placenta/metabolism , Pregnancy , Signal Transduction/drug effects , Signal Transduction/physiology , Trophoblasts/drug effects , Trophoblasts/metabolism
10.
PLoS One ; 8(9): e74812, 2013.
Article in English | MEDLINE | ID: mdl-24069351

ABSTRACT

Hyaluronan (HA) and its receptor CD44 are expressed at the maternal-fetal interface, but its role in early pregnancy remains unclear. Here, we found that primary decidual stromal cells (DSCs) continuously secreted HA and expressed its receptor CD44. Pregnancy-associated hormones up-regulated HA synthetase (HAS) 2 transcription and HA release from DSCs. High molecular weight-HA (HMW-HA), but not medium molecular weight (MMW-HA) or low molecular weight (LMW-HA), promoted proliferation and inhibited apoptosis of DSCs in a CD44-dependent manner. The in-cell Western analysis revealed HMW-HA activated PI3K/AKT and mitogen-activated protein kinase (MAPK)/ERK1/2 signaling pathways time-dependently. Blocking these pathways by specific inhibitor LY294002 or U0126 abrogated HMW-HA-regulated DSc proliferation and apoptosis. Finally, we have found that HA content, HA molecular weight, HAS2 mRNA level, and CD44 expression were significantly decreased in DSCs from unexplained miscarriage compared with the normal pregnancy. Collectively, our results indicate that higher level and greater molecular mass of HA at maternal-fetal interface contributes to DSc growth and maintenance of DSCs in human early pregnancy.


Subject(s)
Decidua/cytology , Decidua/metabolism , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Stromal Cells/metabolism , Abortion, Spontaneous/genetics , Abortion, Spontaneous/metabolism , Adult , Apoptosis , Cell Proliferation/drug effects , Female , Gene Expression Regulation/drug effects , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Gonadal Steroid Hormones/pharmacology , Humans , Hyaluronan Synthases , Hyaluronic Acid/chemistry , Immunohistochemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Weight , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Pregnancy Trimester, First , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stromal Cells/drug effects , Transcription, Genetic , Young Adult
11.
Mol Hum Reprod ; 19(10): 676-86, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23737337

ABSTRACT

Spontaneous abortion is the most common complication of pregnancy. Immune activation and the subsequent inflammation-induced tissue injury are often observed at the maternal-fetal interface as the final pathological assault in recurrent spontaneous abortion. However, the precise mechanisms responsible for spontaneous abortion involving inflammation are not fully understood. Chemokine CCL28 and its receptors CCR3 and CCR10 are important regulators in inflammatory process. Here, we examined the expression of CCL28 and its receptors in decidual stromal cells (DSCs) by immunochemistry and flow cytometry (FCM), and compared their expression level in DSCs from normal pregnancy versus spontaneous abortion, and their relationship to inflammatory cytokines production by DSCs. We further analyzed regulation of the pro-inflammatory cytokines on CCL28 expression in DSCs by real-time polymerase chain reaction, In-cell Western and FCM. The effects of CCL28-CCR3/CCR10 interaction on DSC apoptosis was investigated by Annexin V staining and FCM analysis or DAPI staining and nuclear morphology. Higher levels of the inflammatory cytokines interleukin (IL)-1ß, IL-17A and tumor necrosis factor-α, and increased CCR3/CCR10 expression were observed in DSCs from spontaneous abortion compared with normal pregnancy. Treatment with inflammatory cytokines differently affected CCL28 and CCR3/CCR10 expression in DSCs. Human recombinant CCL28 promoted DSC apoptosis, which was eliminated by pretreatment with neutralizing antibodies against CCR3/CCR10 and CCL28. However, CCL28 did not affect DSC growth. These results suggest that the inflammation-promoted up-regulation of CCL28 and its receptors interaction in DSCs is involved in human spontaneous abortion via inducing DSC apoptosis.


Subject(s)
Abortion, Spontaneous/metabolism , Chemokines, CC/metabolism , Decidua/cytology , Receptors, CCR10/metabolism , Receptors, CCR3/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Abortion, Spontaneous/genetics , Adult , Apoptosis/drug effects , Apoptosis/genetics , Cells, Cultured , Chemokines, CC/genetics , Enzyme-Linked Immunosorbent Assay , Female , Humans , In Vitro Techniques , Interleukin-17/pharmacology , Interleukin-1beta/pharmacology , Pregnancy , Receptors, CCR10/genetics , Receptors, CCR3/genetics , Stromal Cells/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Young Adult
12.
PLoS One ; 7(9): e45275, 2012.
Article in English | MEDLINE | ID: mdl-23028901

ABSTRACT

Our previous study has demonstrated that cyclosporine A (CsA) administration in vivo induces Th2 bias at the maternal-fetal interface, leading to improved murine pregnancy outcomes. Here, we investigated how CsA treatment in vitro induced Th2 bias at the human maternal-fetal interface in early pregnancy. The cell co-culture in vitro in different combination of component cells at the maternal-fetal interface was established to investigate the regulation of CsA on cytokine production from the interaction of these cells. It was found that interferon (IFN)-γ was produced only by decidual immune cells (DICs), and not by trophoblasts or decidual stromal cells (DSCs); all these cells secreted interleukin (IL)-4, IL-10, and tumor necrosis factor (TNF)-α. Treatment with CsA completely blocked IFN-γ production in DICs and inhibited TNF-α production in all examined cells. CsA increased IL-10 and IL-4 production in trophoblasts co-cultured with DSCs and DICs although CsA treatment did not affect IL-10 or IL-4 production in any of the cells when cultured alone. These results suggest that CsA promotes Th2 bias at the maternal-fetal interface by increasing Th2-type cytokine production in trophoblasts with the aid of DSCs and DICs, while inhibiting Th1-type cytokine production in DICs and TNF-α production in all investigated cells. Our study might be useful in clinical therapeutics for spontaneous pregnancy wastage and other pregnancy complications.


Subject(s)
Cyclosporine/pharmacology , Decidua/metabolism , Immunosuppressive Agents/pharmacology , Th1 Cells/metabolism , Th2 Cells/metabolism , Trophoblasts/metabolism , Adult , Cell Communication , Coculture Techniques , Decidua/cytology , Decidua/drug effects , Female , Humans , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-4/biosynthesis , Pregnancy , Pregnancy Trimester, First , Th1 Cells/cytology , Th1 Cells/drug effects , Th1-Th2 Balance , Th2 Cells/cytology , Th2 Cells/drug effects , Trophoblasts/cytology , Trophoblasts/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
13.
Cell Mol Immunol ; 9(5): 423-30, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22885527

ABSTRACT

The regulatory mechanism of Th2 bias at the maternal/fetal interface remains unclear. In this study, we characterized cytokine production in decidual stromal cells (DSCs), decidual immune cells (DICs) and embryo-derived trophoblast cells, and investigated the regulation of CXCL12/CXCR4 interaction on Th2 bias at the maternal/fetal interface in early human pregnancy. We found differential production of Th1-type and Th2-type cytokines by trophoblasts, DSCs and DICs. The secretion of these cytokines varied in different cell cocultures, conduced to Th2 bias. Flow cytometry showed that coculture of trophoblasts with DSCs and DICs significantly increased IL-4 and IL-10 production in trophoblasts, and IL-10 production in DSCs. However, the coculture of trophoblasts with DSCs and DICs significantly increased interferon (IFN)-γ expression in DSCs, and tumor-necrosis factor (TNF)-α expression in DICs. No change was seen in Th1-type cytokine production in trophoblasts, and in Th2-type cytokine production in DICs in all cocultures. Furthermore, pre-treatment with anti-CXCR4 neutralizing antibody upregulated the production of the Th1-type cytokines IFN-γ and TNF-α, and downregulated the production of the Th2-type cytokines IL-4 and IL-10, in trophoblasts, DSCs, DICs or their cocultures. Interestingly, rhCXCL12 inhibited production of the Th1-type cytokine TNF-α and enhanced the expression of the Th2-type cytokines such as IL-4 and IL-10 in DICs; this effect was abrogated by anti-CXCR4 antibody. Our present study has elucidated the individual contributions of component cells to the shaping of Th2 bias, and uncovered a complicated cross-talk via the CXCL12/CXCR4 signal at the maternal/fetal interface in early human pregnancy.


Subject(s)
Chemokine CXCL12/metabolism , Decidua/metabolism , Pregnancy/metabolism , Receptors, CXCR4/metabolism , Trophoblasts/metabolism , Adult , Cells, Cultured , Coculture Techniques , Cytokines/biosynthesis , Decidua/cytology , Decidua/immunology , Female , Flow Cytometry , Humans , Pregnancy/immunology , Pregnancy Trimester, First , Stromal Cells/cytology , Stromal Cells/immunology , Stromal Cells/metabolism , Th1-Th2 Balance , Trophoblasts/cytology , Trophoblasts/immunology
14.
Eukaryot Cell ; 10(1): 63-71, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21097665

ABSTRACT

Hph1 and Hph2 are homologous integral endoplasmic reticulum (ER) membrane proteins required for Saccharomyces cerevisiae survival under environmental stress conditions. To investigate the molecular functions of Hph1 and Hph2, we carried out a split-ubiquitin-membrane-based yeast two-hybrid screen and identified their interactions with Sec71, a subunit of the Sec63/Sec62 complex, which mediates posttranslational translocation of proteins into the ER. Hph1 and Hph2 likely function in posttranslational translocation, as they interact with other Sec63/Sec62 complex subunits, i.e., Sec72, Sec62, and Sec63. hph1Δ hph2Δ cells display reduced vacuole acidification; increased instability of Vph1, a subunit of vacuolar proton ATPase (V-ATPase); and growth defects similar to those of mutants lacking V-ATPase activity. sec71Δ cells exhibit similar phenotypes, indicating that Hph1/Hph2 and the Sec63/Sec62 complex function during V-ATPase biogenesis. Hph1/Hph2 and the Sec63/Sec62 complex may act together in this process, as vacuolar acidification and Vph1 stability are compromised to the same extent in hph1Δ hph2Δ and hph1Δ hph2Δ sec71Δ cells. In contrast, loss of Pkr1, an ER protein that promotes posttranslocation assembly of Vph1 with V-ATPase subunits, further exacerbates hph1Δ hph2Δ phenotypes, suggesting that Hph1 and Hph2 function independently of Pkr1-mediated V-ATPase assembly. We propose that Hph1 and Hph2 aid Sec63/Sec62-mediated translocation of specific proteins, including factors that promote efficient biogenesis of V-ATPase, to support yeast cell survival during environmental stress.


Subject(s)
Heat-Shock Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Transport Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vacuolar Proton-Translocating ATPases/biosynthesis , Gene Expression Regulation, Fungal , Hydrogen-Ion Concentration , Intracellular Signaling Peptides and Proteins/genetics , Multiprotein Complexes , Protein Binding , Saccharomyces cerevisiae Proteins/genetics , Stress, Physiological , Vacuoles/metabolism
15.
EMBO J ; 26(7): 1963-71, 2007 Apr 04.
Article in English | MEDLINE | ID: mdl-17363896

ABSTRACT

Adaptor proteins play important endocytic roles including recognition of internalization signals in transmembrane cargo. Sla1p serves as the adaptor for uptake of transmembrane proteins containing the NPFxD internalization signal, and is essential for normal functioning of the actin cytoskeleton during endocytosis. The Sla1p homology domain 1 (SHD1) within Sla1p is responsible for recognition of the NPFxD signal. This study presents the NMR structure of the NPFxD-bound state of SHD1 and a model for the protein-ligand complex. The alpha+beta structure of the protein reveals an SH3-like topology with a solvent-exposed hydrophobic ligand binding site. NMR chemical shift perturbations and effects of structure-based mutations on ligand binding in vitro define residues that are key for NPFxD binding. Mutations that abolish ligand recognition in vitro also abolish NPFxD-mediated receptor internalization in vivo. Thus, SHD1 is a novel functional domain based on SH3-like topology, which employs a unique binding site to recognize the NPFxD endocytic internalization signal. Its distant relationship with the SH3 fold endows this superfamily with a new role in endocytosis.


Subject(s)
Amino Acid Motifs , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Endocytosis , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Binding Sites , Cell Wall/metabolism , Cytoskeletal Proteins , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutagenesis , Protein Binding , Protein Folding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Solutions , Substrate Specificity
16.
Mol Biol Cell ; 18(1): 57-65, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17065552

ABSTRACT

The actin-associated protein Sla1p, through its SHD1 domain, acts as an adaptor for the NPFX(1,2)D endocytic targeting signal in yeast. Here we report that Wsc1p, a cell wall stress sensor, depends on this signal-adaptor pair for endocytosis. Mutation of NPFDD in Wsc1p or expression of Sla1p lacking SHD1 blocked Wsc1p internalization. By live cell imaging, endocytically defective Wsc1p was not concentrated at sites of endocytosis. Polarized distribution of Wsc1p to regions of cell growth was lost in the absence of endocytosis. Mutations in genes necessary for endosome to Golgi traffic caused redistribution of Wsc1p from the cell surface to internal compartments, indicative of recycling. Inhibition of Wsc1p endocytosis caused defects in polarized deposition of the cell wall and increased sensitivity to perturbation of cell wall synthesis. Our results reveal that the NPFX(1,2)D-Sla1p system is responsible for directing Wsc1p into an endocytosis and recycling pathway necessary to maintain yeast cell wall polarity. The dynamic localization of Wsc1p, a sensor of the extracellular wall in yeast, resembles polarized distribution of certain extracellular matrix-sensing integrins through endocytic recycling.


Subject(s)
Cell Polarity , Cell Wall/metabolism , Endocytosis , Membrane Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Signal Transduction , Amino Acid Motifs , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cell Wall/ultrastructure , Cytoskeletal Proteins , Membrane Proteins/chemistry , Protein Binding , Protein Structure, Tertiary , Protein Transport , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry
17.
Cell ; 126(6): 1109-20, 2006 Sep 22.
Article in English | MEDLINE | ID: mdl-16990135

ABSTRACT

Abscisic acid (ABA) is a phytohormone critical for plant growth, development, and adaptation to various stress conditions. Plants have to adjust ABA levels constantly to respond to changing physiological and environmental conditions. To date, the mechanisms for fine-tuning ABA levels remain elusive. Here we report that AtBG1, a beta-glucosidase, hydrolyzes glucose-conjugated, biologically inactive ABA to produce active ABA. Loss of AtBG1 causes defective stomatal movement, early germination, abiotic stress-sensitive phenotypes, and lower ABA levels, whereas plants with ectopic AtBG1 accumulate higher ABA levels and display enhanced tolerance to abiotic stress. Dehydration rapidly induces polymerization of AtBG1, resulting in a 4-fold increase in enzymatic activity. Furthermore, diurnal increases in ABA levels are attributable to polymerization-mediated AtBG1 activation. We propose that the activation of inactive ABA pools by polymerized AtBG1 is a mechanism by which plants rapidly adjust ABA levels and respond to changing environmental cues.


Subject(s)
Abscisic Acid/biosynthesis , Arabidopsis/physiology , Enzyme Activation/physiology , beta-Glucosidase/metabolism , Adaptation, Physiological/physiology , Arabidopsis/enzymology , Arabidopsis/genetics , Circadian Rhythm/physiology , Dehydration/enzymology , Environment , Plants, Genetically Modified/enzymology , Plants, Genetically Modified/genetics , Plants, Genetically Modified/physiology , Polymers/metabolism , Time Factors , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...