Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
J Am Chem Soc ; 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38981108

ABSTRACT

The antibiotic cerulenin is a fungal natural product identified as a covalent inhibitor of ketosynthases within fatty acid and polyketide biosynthesis. Due to its selective and potent inhibitory activity, cerulenin has found significant utility in multidisciplinary biochemical, biomedical, and clinical studies. Although its covalent inhibition profile has been confirmed, cerulenin's mechanism has not been fully determined at a molecular level, frustrating the drug development of related analogues. Herein, we describe the use of stable isotopic tracking with NMR and MS methods to unravel the covalent mechanism of cerulenin against type II fatty acid ketosynthases. We detail the discovery of a unique C2-C3 retro-aldol bond cleavage and a structural rearrangement upon covalent inhibition of cerulenin at the active cysteine residue in E. coli type II fatty acid ketosynthases FabB and FabF.

2.
Immunol Cell Biol ; 102(5): 321-325, 2024.
Article in English | MEDLINE | ID: mdl-38403985

ABSTRACT

Monash Sensory Science is a scientific outreach initiative specifically tailored to members of the community who are blind, have low vision and have diverse needs. The purpose of this initiative is to showcase Australian science and encourage greater participation in science from these often-overlooked communities. This article presents our experience in establishing Monash Sensory Science at Monash University and inspiring other institutions to launch similar outreach events.


Subject(s)
Vision, Low , Humans , Australia , Blindness
3.
Cytokine ; 155: 155887, 2022 07.
Article in English | MEDLINE | ID: mdl-35512531

ABSTRACT

Interleukin-36 gamma (IL-36G) is a member of the IL-36 subfamily of cytokines and acts as a potent driver of inflammation. IL-36G has been extensively characterized in the pathogenesis of psoriasis and has been recently described to play roles in wound healing particularly in the gastrointestinal tract. However, the effects of IL-36G during cancer development including gastric cancer remain unexplored. Here, we show that IL-36G induced ERK1/2 activation in AGS, MKN1 and MKN45 human gastric cancer cell lines. Moreover, IL-36G induced colony formation, migration and invasion of these gastric cancer cell lines that was inhibited by the natural antagonist, IL-36 receptor antagonist (RA). Interrogation of TCGA stomach adenocarcinoma patient datasets revealed highly elevated IL-36G gene expression in human gastric cancer compared to normal tissue independent of tumor stage, and high IL-36G expression corresponded with poorer patient survival. Collectively, our results indicate for the first time that IL-36G supports a neoplastic phenotype in human gastric cancer cells.


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , Signal Transduction , Stomach Neoplasms/pathology
4.
Catheter Cardiovasc Interv ; 98(5): E668-E676, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34329539

ABSTRACT

OBJECTIVES: This study evaluated myocardial viability as well as global and regional functional recovery after successful chronic coronary total occlusion (CTO) percutaneous coronary intervention (PCI) using sequential quantitative cardiac magnetic resonance (CMR) imaging. BACKGROUND: The patient benefits of CTO PCI are being questioned. METHODS: In a single high-volume CTO PCI center patients were prospectively scheduled for CMR at baseline and 3 months after successful CTO PCI between 2013 and 2018. Segmental wall thickening (SWT) and percentage late gadolinium enhancement (LGE) were quantitatively measured per segment. Viability was defined as dysfunctional myocardium (<2.84 mm SWT) with no or limited scar (≤50% LGE). RESULTS: A total of 132 patients were included. Improvement of left ventricular ejection fraction was modest after CTO PCI (from 48.1 ± 11.8 to 49.5 ± 12.1%, p < 0.01). CTO segments with viability (N = 216, [31%]) demonstrated a significantly higher increase in SWT (0.80 ± 1.39 mm) compared to CTO segments with pre-procedural preserved function (N = 456 [65%], 0.07 ± 1.43 mm, p < 0.01) or extensive scar (LGE >50%, N = 26 [4%], -0.08 ± 1.09 mm, p < 0.01). Patients with ≥2 CTO segments viability showed more SWT increase in the CTO territory compared to patients with 0-1 segment viability (0.49 ± 0.93 vs. 0.12 ± 0.98 mm, p = 0.03). CONCLUSIONS: Detection of dysfunctional myocardial segments without extensive scar (≤50% LGE) as a marker for viability on CMR aids in identifying patients with significant regional functional recovery after CTO PCI.


Subject(s)
Coronary Occlusion , Percutaneous Coronary Intervention , Chronic Disease , Contrast Media , Coronary Occlusion/diagnostic imaging , Coronary Occlusion/therapy , Gadolinium , Humans , Stroke Volume , Treatment Outcome , Ventricular Function, Left
5.
Eur J Vasc Endovasc Surg ; 61(6): 1008-1016, 2021 06.
Article in English | MEDLINE | ID: mdl-33858751

ABSTRACT

OBJECTIVE: Perivascular adipose tissue (PVAT) contributes to vascular homeostasis and is increasingly linked to vascular pathology. PVAT density and volume were associated with abdominal aortic aneurysm (AAA) presence and dimensions on imaging. However, mechanisms underlying the role of PVAT in AAA have not been clarified. This study aimed to explore differences in PVAT from AAA using gene expression and functional tests. METHODS: Human aortic PVAT and control subcutaneous adipose tissue were collected during open AAA surgery. Gene analyses and functional tests were performed. The control group consisted of healthy aorta from non-living renal transplant donors. Gene expression tests were performed to study genes potentially involved in various inflammatory processes and AAA related genes. Live PVAT and subcutaneous adipose tissue (SAT) from AAA were used for ex vivo co-culture with smooth muscle cells (SMCs) retrieved from non-pathological aortas. RESULTS: Adipose tissue was harvested from 27 AAA patients (n [gene expression] = 22, n [functional tests] = 5) and five control patients. An increased inflammatory gene expression of PTPRC (p = .008), CXCL8 (p = .033), LCK (p = .003), CCL5 (p = .004) and an increase in extracellular matrix breakdown marker MMP9 (p = .016) were found in AAA compared with controls. Also, there was a decreased anti-inflammatory gene expression of PPARG in AAA compared with controls (p = .040). SMC co-cultures from non-pathological aortas with PVAT from AAA showed increased MMP9 (p = .033) and SMTN (p = .008) expression and SAT increased SMTN expression in these SMC. CONCLUSION: The data revealed that PVAT from AAA shows an increased pro-inflammatory and matrix metallopeptidase gene expression and decreased anti-inflammatory gene expression. Furthermore, increased expression of genes involved in aneurysm formation was found in healthy SMC co-culture with PVAT of AAA patients. Therefore, PVAT from AAA might contribute to inflammation of the adjacent aortic wall and thereby plays a possible role in AAA pathophysiology. These proposed pathways of inflammatory induction could reveal new therapeutic targets in AAA treatment.


Subject(s)
Aortic Aneurysm, Abdominal/genetics , Chemokine CCL5/genetics , Interleukin-8/genetics , Leukocyte Common Antigens/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Matrix Metalloproteinase 9/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Aged , Aged, 80 and over , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Case-Control Studies , Chemokine CCL5/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Female , Humans , Interleukin-8/metabolism , Leukocyte Common Antigens/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Middle Aged , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Messenger/metabolism
6.
Int J Mol Sci ; 22(20)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34681666

ABSTRACT

Multiplex immunohistochemistry (mIHC) enables simultaneous staining of multiple immune markers on a single tissue section. Mounting studies have demonstrated the versatility of mIHC in evaluating immune infiltrates in different diseases and the tumour microenvironment (TME). However, the majority of published studies are limited to the analysis of human patient samples. Performing mIHC on formalin-fixed paraffin-embedded (FFPE) mouse tissues, particularly with sensitive antigens, remain challenging. The aim of our study was to develop a robust and reproducible protocol to uncover the immune landscape in mouse FFPE tissues. Effective antibody stripping while maintaining sensitivity to antigens and tissue adhesion to the glass slide is critical in developing an mIHC panel to allow successive rounds of staining. Thus, we identified a highly efficient stripping method that preserves signal intensity and antigenicity to allow multiple rounds of staining. We subsequently optimised an mIHC workflow with antibodies specific against CD4, CD8α, FOXP3 and B220 to identify distinct T and B cell populations on mouse FFPE tissues. Lastly, the application of this mIHC panel was validated in a mouse model of inflammatory bowel cancer, two allograft mouse models of spontaneous colon adenocarcinoma and a sporadic mouse model of colon cancer. Together, these demonstrate the utility of the aforementioned protocol in establishing the quantity and spatial localisation of immune cells in different pathological tissues.


Subject(s)
Colitis/pathology , Colonic Neoplasms/pathology , Immunohistochemistry/methods , Animals , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Colitis/chemically induced , Colitis/metabolism , Colonic Neoplasms/metabolism , Disease Models, Animal , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/pathology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
7.
Cell Microbiol ; 20(7): e12837, 2018 07.
Article in English | MEDLINE | ID: mdl-29504253

ABSTRACT

Porphyromonas gingivalis is a keystone pathogen in chronic periodontitis. Its expression of gingipain proteases (Kgp and RgpA/B) is central to the stimulation of chronic inflammation. In this study, we investigated the inflammatory response of oral epithelial cells to P. gingivalis. The cells responded by upregulating the expression of the orphan chemokine CXCL14. The stimulation of CXCL14 expression was largely triggered by the gingipain proteases and was dependent on the host protease-activated receptor PAR-3. Significantly, CXCL14 expression was transcriptionally repressed in response to epidermal growth factor (EGF)-induced activation of the MEK-ERK1/2 pathway. P. gingivalis overcomes the repression of CXCL14 via the gingipain protease-mediated degradation of EGF. Therefore, P. gingivalis not only directly stimulates CXCL14 expression via PAR-3 but also promotes its expression by antagonising EGF signalling. In addition to chemotactic activity, some chemokines also have antimicrobial activities. CXCL14 was demonstrated to have bactericidal activity, against commensal oral streptococci associated with health. Notably though, P. gingivalis was not susceptible to killing by CXCL14, potentially because the gingipain proteases can degrade CXCL14. This suggests that the stimulation of dysregulated CXCL14 expression by P. gingivalis may help promote dysbiosis and the development of chronic periodontitis.


Subject(s)
Cell Cycle Proteins/metabolism , Chemokines, CXC/metabolism , Epidermal Growth Factor/metabolism , Epithelial Cells/microbiology , Host-Pathogen Interactions , Membrane Proteins/metabolism , Porphyromonas gingivalis/growth & development , Signal Transduction , Adaptor Proteins, Signal Transducing , Cell Line , Epithelial Cells/immunology , Gene Expression Regulation , Humans
8.
Semin Cancer Biol ; 45: 13-22, 2017 08.
Article in English | MEDLINE | ID: mdl-28647610

ABSTRACT

Intercellular communication between tumor cells, immune cells and the stroma characterises the tumor microenvironment, which is instrumental for establishing the ecological niche that fosters tumor growth and metastasis. While tumor cell intrinsic STAT3 signaling provides a crucial axis to support cell proliferation and survival, it also regulates many activities of the non-transformed cells that collectively make up the tumor microenvironment. Accordingly, excessive activation of STAT3 is a hallmark of many malignancies, and often occurs in response to cytokines of the IL-6 and IL-10 families. However, tumor extrinsic STAT3 signaling also regulates the effector function of tumor-associated immune and stromal cells, which support the growth of tumors by suppressing the host's anti-tumor immune response. Given that STAT3 mediates tumorigenic effects in many cell types, the molecular players of STAT3 signaling and its upstream JAK kinases provide viable therapeutic targets for the treatment of cancer. Here we provide an update on novel insights into the role of STAT3 in immune suppression and describe current therapeutic strategies that target the JAK/STAT3 signaling axis for the treatment of malignancies.


Subject(s)
Janus Kinases/metabolism , Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers , Cell Communication , Clinical Trials as Topic , Humans , Immunomodulation , Immunotherapy , Molecular Targeted Therapy , Mutation , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/pathology , Signal Transduction/drug effects , Tumor Microenvironment
9.
J Immunol ; 196(5): 2230-8, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26819203

ABSTRACT

IFN regulatory factors (IRFs) help to shape the immune response to pathogens by imparting signaling specificity to individual TLRs. We recently demonstrated that IRF6 provides specificity to TLR2 signaling in oral epithelial cells. TLR2 plays an important role in eliciting inflammation to Porphyromonas gingivalis, a keystone pathogen in periodontitis. Therefore, we investigated a role for IRF6 in mediating the inflammatory cytokine response of oral epithelial cells to P. gingivalis. IRF6 expression was strongly upregulated when human oral epithelial cells were challenged with P. gingivalis. Moreover, gene silencing and gene promoter experiments indicated that IRF6 acts downstream of IL-1R-associated kinase 1 to stimulate the expression of the IL-1 family cytokine IL-36γ in response to P. gingivalis. IRF6 and IL-1R-associated kinase 1 also regulated the stimulation of IL-36γ expression by a TLR2 agonist. IL-36γ was shown to elicit inflammatory responses by human monocyte-derived dendritic cells and macrophages, including the expression of the neutrophil chemokines IL-8 and CXCL1, as well as the Th17 chemokine CCL20. IL-36γ similarly stimulated their expression by human oral epithelial cells. Significantly, the Th17 cytokine IL-17 not only stimulated the expression of important regulators of neutrophil recruitment and survival by oral epithelial cells, but IL-17 also stimulated them to express IL-36γ. Thus, our findings suggest that IRF6 is likely to promote inflammation to P. gingivalis through its regulation of IL-36γ.


Subject(s)
Gene Expression Regulation , Interferon Regulatory Factors/metabolism , Interleukin-1/genetics , Mouth Mucosa/metabolism , Mouth Mucosa/virology , Porphyromonas gingivalis/immunology , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epithelial Cells , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-17/metabolism , Macrophages/immunology , Macrophages/metabolism , Models, Biological , Mouth Mucosa/immunology , Toll-Like Receptor 2/metabolism , Up-Regulation
10.
Infect Immun ; 85(5)2017 05.
Article in English | MEDLINE | ID: mdl-28289145

ABSTRACT

We recently demonstrated that the expression of the interferon regulatory factor 6 (IRF6) transcription factor in oral keratinocytes was stimulated by the periodontal pathogen Porphyromonas gingivalis Here, we have established that IRF6 promotes the differentiation of oral keratinocytes in response to P. gingivalis This was evidenced by the IRF6-dependent upregulation of specific markers of keratinocyte terminal differentiation (e.g., involucrin [IVL] and keratin 13 [KRT13]), together with additional transcriptional regulators of keratinocyte differentiation, including Grainyhead-like 3 (GRHL3) and Ovo-like zinc finger 1 (OVOL1). We have previously established that the transactivator function of IRF6 is activated by receptor-interacting protein kinase 4 (RIPK4). Consistently, the silencing of RIPK4 inhibited the stimulation of IVL, KRT13, GRHL3, and OVOL1 gene expression. IRF6 was shown to also regulate the stimulation of transglutaminase-1 (TGM1) gene expression by P. gingivalis, as well as that of small proline-rich proteins (e.g., SPRR1), which are covalently cross-linked by TGM1 to other proteins, including IVL, during cornification. The expression of the tight junction protein occludin (OCLN) was found to also be upregulated in an IRF6-dependent manner. IRF6 was demonstrated to be important for the barrier function of oral keratinocytes; specifically, silencing of IRF6 increased P. gingivalis-induced intercellular permeability and cell invasion. Taken together, our findings potentially position IRF6 as an important mediator of barrier defense against P. gingivalis.


Subject(s)
Cell Differentiation , Host-Pathogen Interactions , Interferon Regulatory Factors/metabolism , Keratinocytes/physiology , Porphyromonas gingivalis/growth & development , Biomarkers/analysis , Cell Line , Gene Expression Profiling , Humans
11.
Diabetologia ; 58(6): 1329-32, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25748329

ABSTRACT

AIMS/HYPOTHESIS: Metabolomic profiling in populations with impaired glucose tolerance has revealed that branched chain and aromatic amino acids (BCAAs) are predictive of type 2 diabetes. Because gestational diabetes mellitus (GDM) shares pathophysiological similarities with type 2 diabetes, the metabolite profile predictive of type 2 diabetes could potentially identify women who will develop GDM. METHODS: We conducted a nested case-control study of 18- to 40-year-old women who participated in the Massachusetts General Hospital Obstetrical Maternal Study between 1998 and 2007. Participants were enrolled during their first trimester of a singleton pregnancy and fasting serum samples were collected. The women were followed throughout pregnancy and identified as having GDM or normal glucose tolerance (NGT) in the third trimester. Women with GDM (n = 96) were matched to women with NGT (n = 96) by age, BMI, gravidity and parity. Liquid chromatography-mass spectrometry was used to measure the levels of 91 metabolites. RESULTS: Data analyses revealed the following characteristics (mean ± SD): age 32.8 ± 4.4 years, BMI 28.3 ± 5.6 kg/m(2), gravidity 2 ± 1 and parity 1 ± 1. Six metabolites (anthranilic acid, alanine, glutamate, creatinine, allantoin and serine) were identified as having significantly different levels between the two groups in conditional logistic regression analyses (p < 0.05). The levels of the BCAAs did not differ significantly between GDM and NGT. CONCLUSIONS/INTERPRETATION: Metabolic markers identified as being predictive of type 2 diabetes may not have the same predictive power for GDM. However, further study in a racially/ethnically diverse population-based cohort is necessary.


Subject(s)
Blood Glucose/analysis , Diabetes, Gestational/blood , Metabolomics/methods , Adolescent , Adult , Amino Acids, Branched-Chain/blood , Body Mass Index , Case-Control Studies , Chromatography, Liquid , Diabetes Mellitus, Type 2/blood , Female , Glucose Tolerance Test , Humans , Logistic Models , Mass Spectrometry , Massachusetts , Pregnancy , Pregnancy Trimester, First , Young Adult
12.
J Biol Chem ; 289(45): 31077-87, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25246526

ABSTRACT

Receptor-interacting protein kinase 4 (RIPK4) and interferon regulatory factor 6 (IRF6) are critical regulators of keratinocyte differentiation, and their mutation causes the related developmental epidermal disorders Bartsocas-Papas syndrome and popliteal pterygium syndrome, respectively. However, the signaling pathways in which RIPK4 and IRF6 operate to regulate keratinocyte differentiation are poorly defined. Here we identify and mechanistically define a direct functional relationship between RIPK4 and IRF6. Gene promoter reporter and in vitro kinase assays, coimmunoprecipitation experiments, and confocal microscopy demonstrated that RIPK4 directly regulates IRF6 trans-activator activity and nuclear translocation. Gene knockdown and overexpression studies indicated that the RIPK4-IRF6 signaling axis controls the expression of key transcriptional regulators of keratinocyte differentiation, including Grainyhead-like 3 and OVO-like 1. Additionally, we demonstrate that the p.Ile121Asn missense mutation in RIPK4, which has been identified recently in Bartsocas-Papas syndrome, inhibits its kinase activity, thereby preventing RIPK4-mediated IRF6 activation and nuclear translocation. We show, through mutagenesis-based experiments, that Ser-413 and Ser-424 in IRF6 are important for its activation by RIPK4. RIPK4 is also important for the regulation of IRF6 expression by the protein kinase C pathway. Therefore, our findings not only provide important mechanistic insights into the regulation of keratinocyte differentiation by RIPK4 and IRF6, but they also suggest one mechanism by which mutations in RIPK4 may cause epidermal disorders (e.g. Bartsocas-Papas syndrome), namely by the impaired activation of IRF6 by RIPK4.


Subject(s)
Cell Differentiation , Interferon Regulatory Factors/metabolism , Keratinocytes/cytology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Cells, Cultured , DNA-Binding Proteins/metabolism , Epidermis/metabolism , Gene Silencing , Genetic Vectors , Glutathione Transferase/metabolism , HEK293 Cells , Humans , Mutation, Missense , Phosphorylation , Promoter Regions, Genetic , Protein Kinase C/metabolism , Serine/chemistry , Transcription Factors/metabolism
13.
J Biol Chem ; 289(28): 19758-68, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24872416

ABSTRACT

Epidermal and mucosal epithelial cells are integral to host defense. They not only act as a physical barrier but also utilize pattern recognition receptors, such as the Toll-like receptors (TLRs), to detect and respond to pathogens. Members of the interferon regulatory factor (IRF) family of transcription factors are key components of TLR signaling as they impart specificity to downstream responses. Although IRF6 is a critical regulator of epithelial cell proliferation and differentiation, its role in TLR signaling has not previously been addressed. We show here that IRF6 is activated by IRAK1 as well as by MyD88 but not by TRIF or TBK1. Co-immunoprecipitation experiments further demonstrated that IRF6 can interact with IRAK1. Gene silencing in epithelial cells along with gene promoter reporter assays showed that IRAK1 mediates TLR2-inducible CCL5 gene expression at least in part by promoting IRF6 activation. Conversely, IRAK1 regulated CXCL8 gene expression independently of IRF6, thus identifying a molecular mechanism by which TLR2 signaling differentially regulates the expression of specific chemokines in epithelial cells. Bioinformatics analysis and mutagenesis-based experiments identified Ser-413 and Ser-424 as key regulatory sites in IRF6. Phosphomimetic mutation of these residues resulted in greatly enhanced IRF6 dimerization and trans-activator function. Collectively, our findings suggest that, in addition to its importance for epithelial barrier function, IRF6 also contributes to host defense by providing specificity to the regulation of inflammatory chemokine expression by TLR2 in epithelial cells.


Subject(s)
Chemokine CCL5/biosynthesis , Epithelial Cells/metabolism , Gene Expression Regulation/physiology , Interferon Regulatory Factors/metabolism , Interleukin-8/biosynthesis , Signal Transduction/physiology , Toll-Like Receptor 2/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Amino Acid Substitution , Cell Line , Chemokine CCL5/genetics , Epithelial Cells/cytology , Humans , Interferon Regulatory Factors/genetics , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-8/genetics , Mutation, Missense , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Toll-Like Receptor 2/genetics
14.
Depress Anxiety ; 32(10): 774-82, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26130074

ABSTRACT

INTRODUCTION: The relationship between gestational diabetes mellitus (GDM) and postpregnancy mental health disorders has been inconsistently reported. Additionally, race/ethnicity data are limited. We sought to elucidate the intersection of these relationships. METHODS: We analyzed 18,109 women aged 18-40 with self-reported race/ethnicity. Women with (n = 659) and without (n = 14,461) GDM were followed for a median of 4.4 (interquartile range 1.4-6.8) and 4.0 (1.5-6.4) years, respectively, for incident mental health disorders. Multivariable repeated measures analyses were conducted to examine associations between GDM and postpregnancy mental health disorders, race/ethnicity, and the interaction of these factors. RESULTS: Women with compared to women without GDM were older (mean ± standard deviation, 32 ± 5 vs. 30 ± 5 years; P < .001) and had higher body mass index (29.0 ± 7.2 vs. 25.3 ± 5.2 kg/m(2) ; P < .001). GDM was associated with increased risk for depression and anxiety after adjusting for age and pregnancy complications; however, loss of significance in the fully adjusted model for depression (odds ratio [95% CI]: 1.29 [0.98, 1.70]; P = .064) and anxiety (1.14 [0.83, 1.57], P = .421) suggested that clinical and socioeconomic factors influence this relationship. Hispanic compared to white women had a greater risk for depression (1.40 [1.15, 1.72]; P = .001), even after multivariable adjustment. The interaction between GDM and race was evident in complication-adjusted but not fully adjusted models. CONCLUSIONS: The incidence of mental health disorders subsequent to GDM was attenuated after adjustment for clinical and socioeconomic factors. Moreover, race/ethnicity influenced this relationship. Further investigation is warranted to clarify potential underlying mechanisms.


Subject(s)
Diabetes, Gestational/psychology , Hispanic or Latino/psychology , Hispanic or Latino/statistics & numerical data , Mental Disorders/ethnology , Pregnancy Complications/psychology , White People/psychology , White People/statistics & numerical data , Adult , Body Mass Index , Diabetes, Gestational/epidemiology , Ethnicity/statistics & numerical data , Female , Humans , Incidence , Odds Ratio , Pregnancy , Pregnancy Complications/epidemiology , Risk
15.
Biochem J ; 461(3): 469-76, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24854174

ABSTRACT

A key factor in oncogenesis is the transport into the nucleus of oncogenic signalling molecules, such as Gli1 (glioma-associated oncogene homologue 1), the central transcriptional activator in the Hedgehog signalling pathway. Little is known, however, how factors such as Gli are transported into the nucleus and how this may be regulated by interaction with other cellular factors, such as the negative regulator suppressor of fused (SuFu). In the present study we show for the first time that nuclear entry of Gli1 is regulated by a unique mechanism through mutually exclusive binding by its nuclear import factor Impß1 (importin ß1) and SuFu. Using quantitative live mammalian cell imaging, we show that nuclear accumulation of GFP-Gli1 fusion proteins, but not of a control protein, is specifically inhibited by co-expression of SuFu. Using a direct binding assay, we show that Impß1 exhibits a high nanomolar affinity to Gli1, with specific knockdown of Impß1 expression being able to inhibit Gli1 nuclear accumulation, thus implicating Impß1 as the nuclear transporter for Gli1 for the first time. SuFu also binds to Gli1 with a high nanomolar affinity, intriguingly being able to compete with Impß1 for binding to Gli1, through the fact that the sites for SuFu and Impß1 binding overlap at the Gli1 N-terminus. The results indicate for the first time that the relative intracellular concentrations of SuFu and Impß1 are likely to determine the localization of Gli1, with implications for its action in cancer, as well as in developmental systems.


Subject(s)
Adenocarcinoma/metabolism , Cell Nucleus/metabolism , Neoplasm Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , beta Karyopherins/metabolism , Adenocarcinoma/pathology , Animals , Binding Sites , Binding, Competitive , COS Cells , Cell Nucleus/ultrastructure , Chlorocebus aethiops , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Transport , RNA Interference , RNA, Small Interfering , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Zinc Finger Protein GLI1 , beta Karyopherins/antagonists & inhibitors , beta Karyopherins/chemistry , beta Karyopherins/genetics
16.
Diabetologia ; 57(12): 2453-64, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25193282

ABSTRACT

AIMS/HYPOTHESIS: Gestational diabetes mellitus is associated with adverse maternal and fetal outcomes during, as well as subsequent to, pregnancy, including increased risk of type 2 diabetes and cardiovascular disease. Because of the importance of early risk stratification in preventing these complications, improved first-trimester biomarker determination for diagnosing gestational diabetes would enhance our ability to optimise both maternal and fetal health. Metabolomic profiling, the systematic study of small molecule products of biochemical pathways, has shown promise in the identification of key metabolites associated with the pathogenesis of several metabolic diseases, including gestational diabetes. This article provides a systematic review of the current state of research on biomarkers and gestational diabetes and discusses the clinical relevance of metabolomics in the prediction, diagnosis and management of gestational diabetes. METHODS: We conducted a systematic search of MEDLINE (PubMed) up to the end of February 2014 using the key term combinations of 'metabolomics,' 'metabonomics,' 'nuclear magnetic spectroscopy,' 'mass spectrometry,' 'metabolic profiling' and 'amino acid profile' combined (AND) with 'gestational diabetes'. Additional articles were identified through searching the reference lists from included studies. Quality assessment of included articles was conducted through the use of QUADOMICS. RESULTS: This systematic review included 17 articles. The biomarkers most consistently associated with gestational diabetes were asymmetric dimethylarginine and NEFAs. After QUADOMICS analysis, 13 of the 17 included studies were classified as 'high quality'. CONCLUSIONS/INTERPRETATION: Existing metabolomic studies of gestational diabetes present inconsistent findings regarding metabolite profile characteristics. Further studies are needed in larger, more racially/ethnically diverse populations.


Subject(s)
Diabetes, Gestational/metabolism , Metabolomics , Biomarkers , Female , Humans , Pregnancy
17.
Biochim Biophys Acta ; 1833(10): 2348-56, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23773962

ABSTRACT

The importin (IMP) superfamily of nuclear transport proteins is essential to key developmental pathways, including in the murine testis where expression of the 6 distinct IMPα proteins is highly dynamic. Present predominantly from the spermatocyte stage onwards, IMPα4 is unique in showing a striking nuclear localization, a property we previously found to be linked to maintenance of pluripotency in embryonic stem cells and to the cellular stress response in cultured cells. Here we examine the role of IMPα4 in vivo for the first time using a novel transgenic mouse model in which we overexpress an IMPα4-EGFP fusion protein from the protamine 1 promoter to recapitulate endogenous testicular germ cell IMPα4 expression in spermatids. IMPα4 overexpression did not affect overall fertility, testis morphology/weight or spermatogenic progression under normal conditions, but conferred significantly (>30%) increased resistance to oxidative stress specifically in the spermatid subpopulation expressing the transgene. Consistent with a cell-specific role for IMPα4 in protecting against oxidative stress, haploid germ cells from IMPα4 null mice were significantly (c. 30%) less resistant to oxidative stress than wild type controls. These results from two unique and complementary mouse models demonstrate a novel protective role for IMPα4 in stress responses specifically within haploid male germline cells, with implications for male fertility and genetic integrity.


Subject(s)
Active Transport, Cell Nucleus/genetics , Cell Nucleus/genetics , Germ Cells/metabolism , Oxidative Stress , Spermatids/metabolism , Spermatogenesis , Testis/metabolism , alpha Karyopherins/physiology , Animals , Blotting, Southern , Blotting, Western , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , DNA/genetics , Fertility , Flow Cytometry , Germ Cells/cytology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoenzyme Techniques , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Promoter Regions, Genetic/genetics , Protamines/genetics , Real-Time Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spermatids/cytology , Testis/cytology
18.
Biochem Biophys Res Commun ; 450(1): 812-7, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24953690

ABSTRACT

We previously showed that increased intracellular calcium can modulate Importin (Imp)ß1-dependent nuclear import of SRY-related chromatin remodeling proteins. Here we extend this work to show for the first time that high intracellular calcium inhibits Impα/ß1- or Impß1-dependent nuclear protein import generally. The basis of this relates to the mislocalisation of the transport factors Impß1 and Ran, which show significantly higher nuclear localization in contrast to various other factors, and RCC1, which shows altered subnuclear localisation. The results here establish for the first time that intracellular calcium modulates conventional nuclear import through direct effects on the nuclear transport machinery.


Subject(s)
Active Transport, Cell Nucleus/physiology , Calcium/metabolism , Cell Cycle Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Intracellular Fluid/metabolism , Nuclear Proteins/metabolism , beta Karyopherins/metabolism , ran GTP-Binding Protein/metabolism , Animals , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Humans
19.
Article in English | MEDLINE | ID: mdl-38860700

ABSTRACT

INTRODUCTION: This systematic review aimed to discuss the current knowledge of possibly useful circulatory biomarkers (other than D-dimers) in the diagnosis of patients with an acute aortic dissection (AAD), to distinguish these patients from patients with Acute Myocardial Infarction (AMI). EVIDENCE ACQUISITION: This study followed the PRISMA guidelines. The databases PubMed, EMBASE and Scopus were systematically searched from inception to May 2023. Studies were included if they presented measurements of biomarker(s) in the blood/plasma/serum samples from adult patients with AAD versus AMI. Articles were excluded if aortic dissection was subacute or chronic (>14 days), if they lack a control group (AMI), or if they were animal studies, revisions, or editorials. The main outcome was the identification of biomarkers that exhibited diagnostic potential to differentiate patients with AAD versus AMI. EVIDENCE SYNTHESIS: The research query resulted in 1342 articles after the removal of duplicates, from which seven were included in the systematic review. The biomarkers identified included general blood assessment, metabolomics, products of the degradation of fibrin, extracellular matrix markers and an ischemia-associated molecule. Most studies lack diagnostic validity such as sensitivity and specificity. In six studies, the concentration of a total of six biomarkers showed significative differences between AAD and AMI group. CONCLUSIONS: A great heterogeneity of molecules has been studied as putative diagnostic markers of AAD versus AMI. Studies of better quality are needed, presenting the diagnostic validity of the molecules under analysis and the putative synergic diagnostic value of the molecules identified so far.

20.
J Neurosci Res ; 91(6): 854-60, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23553842

ABSTRACT

Morphine differs from most opiates its poor ability to internalize µ opioid receptors (µORs). However, chronic treatment with morphine produces adaptational changes at the dynamin level, which enhance the efficiency of acute morphine stimulation to promote µOR internalization in enteric neurons. This study tested the effect of chronic treatment with fentanyl, a µOR-internalizing agonist, on ligand-induced endocytosis and the expression of the intracellular trafficking proteins, dynamin and ß-arrestin, in enteric neurons using organotypic cultures of the guinea pig ileum. In enteric neurons from guinea pigs chronically treated with fentanyl, µOR immunoreactivity was predominantly at the cell surface after acute exposure to morphine with a low level of µOR translocation, slightly higher than in neurons from naïve animals. This internalization was not due to morphine's direct effect, because it was also observed in neurons exposed to medium alone. By contrast, D-Ala2-N-Me-Phe4-Gly-ol5-enkephalin (DAMGO), a potent µOR-internalizing agonist, induced pronounced and rapid µOR endocytosis in enteric neurons from animals chronically treated with fentanyl or from naïve animals. Chronic fentanyl treatment did not alter dynamin or ß-arrestin expression. These findings indicate that prolonged activation of µORs with an internalizing agonist such as fentanyl does not enhance the ability of acute morphine to trigger µOR endocytosis or induce changes in intracellular trafficking proteins, as observed with prolonged activation of µORs with a poorly internalizing agonist such as morphine. Cellular adaptations induced by chronic opiate treatment might be ligand dependent and vary with the agonist efficiency to induce receptor internalization.


Subject(s)
Analgesics, Opioid/pharmacology , Enteric Nervous System/drug effects , Fentanyl/pharmacology , Neurons/drug effects , Receptors, Opioid, mu/metabolism , Animals , Endocytosis/drug effects , Enteric Nervous System/metabolism , Guinea Pigs , Ileum/drug effects , Ileum/metabolism , Immunohistochemistry , Ligands , Male , Morphine/pharmacology , Neurons/metabolism , Organ Culture Techniques , Protein Transport/drug effects , Protein Transport/physiology
SELECTION OF CITATIONS
SEARCH DETAIL