Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Nutr ; 154(4): 1130-1140, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38237669

ABSTRACT

BACKGROUND: Fish oil with the ω-3 fatty acids EPA and DHA is an FDA-approved treatment of patients with severe hypertriglyceridemia. Furthermore, EPA is an FDA-approved treatment of patients with high risk of cardiovascular disease (CVD); however, the cardioprotective mechanisms are unclear. OBJECTIVES: We aimed to determine if fish oil supplementation is cardioprotective due to beneficial modifications in HDL particles. METHODS: Seven fish oil naïve subjects without a history of CVD were recruited to take a regimen of fish oil (1125 mg EPA and 875 mg DHA daily) for 30 d, followed by a 30-d washout period wherein no fish oil supplements were taken. HDL isolated from fasting whole blood at each time point via 2-step ultracentrifugation (ucHDL) was assessed for proteome, lipidome, cholesterol efflux capacity (CEC), and anti-inflammatory capacity. RESULTS: Following fish oil supplementation, the HDL-associated proteins immunoglobulin heavy constant γ1, immunoglobulin heavy constant α1, apolipoprotein D, and phospholipid transfer protein decreased compared to baseline (P < 0.05). The HDL-associated phospholipid families sphingomyelins, phosphatidylcholines, and phosphatidylserines increased after fish oil supplementation relative to baseline (P < 0.05). Compared to baseline, fish oil supplementation increased serum HDL's CEC (P = 0.002). Fish oil-induced changes (Post compared with Baseline) in serum HDL's CEC positively correlated with plasma EPA levels (R2 = 0.7256; P = 0.015). Similarly, fish oil-induced changes in ucHDL's CEC positively correlated with ucHDL's ability to reduce interleukin 10 (R2 = 0.7353; P = 0.014) and interleukin 6 mRNA expression (R2 = 0.6322; P =0.033) in a human macrophage cell line. CONCLUSIONS: Overall, fish oil supplementation improved HDL's sterol efflux capacity through comprehensive modifications to its proteome and lipidome.


Subject(s)
Cardiovascular Diseases , Fatty Acids, Omega-3 , Adult , Humans , Fish Oils/pharmacology , Proteome , Lipidomics , Lipoproteins, HDL , Dietary Supplements , Immunoglobulins , Docosahexaenoic Acids , Eicosapentaenoic Acid , Triglycerides
2.
PLoS Genet ; 12(4): e1006002, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27119146

ABSTRACT

Collagen is a major component of the extracellular matrix and its integrity is essential for connective tissue and organ function. The importance of proteins involved in intracellular collagen post-translational modification, folding and transport was recently highlighted from studies on recessive forms of osteogenesis imperfecta (OI). Here we describe the critical role of SC65 (Synaptonemal Complex 65, P3H4), a leprecan-family member, as part of an endoplasmic reticulum (ER) complex with prolyl 3-hydroxylase 3. This complex affects the activity of lysyl-hydroxylase 1 potentially through interactions with the enzyme and/or cyclophilin B. Loss of Sc65 in the mouse results in instability of this complex, altered collagen lysine hydroxylation and cross-linking leading to connective tissue defects that include low bone mass and skin fragility. This is the first indication of a prolyl-hydroxylase complex in the ER controlling lysyl-hydroxylase activity during collagen synthesis.


Subject(s)
Autoantigens/metabolism , Collagen/biosynthesis , Endoplasmic Reticulum/metabolism , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism , Procollagen-Proline Dioxygenase/metabolism , Protein Processing, Post-Translational/physiology , Animals , Autoantigens/genetics , Bone and Bones/physiology , Cell Line , Collagen/metabolism , Cyclophilins/metabolism , Extracellular Matrix/metabolism , Hydroxylation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics
3.
Biol Reprod ; 92(6): 140, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25904015

ABSTRACT

Krüppel-like Factor (KLF) 13 and the closely related KLF9 are members of the Sp/KLF family of transcription factors that have collectively emerged as essential regulators of tissue development, differentiation, proliferation, and programmed cell death. Steroid hormone-responsive tissues express multiple KLFs that are linked to progesterone receptor (PGR) and estrogen receptor (ESR) actions either as integrators or as coregulators. Endometriosis is a chronic disease characterized by progesterone resistance and dysregulated estradiol signaling; nevertheless, distinct KLF members' contributions to endometriosis remain largely undefined. We previously demonstrated promotion of ectopic lesion establishment by Klf9 null endometrium in a mouse model of endometriosis. Here we evaluated whether KLF13 loss of expression in endometrial cells may equally contribute to lesion formation. KLF13 transcript levels were lower in the eutopic endometria of women with versus women without endometriosis at menstrual midsecretory phase. In wild-type (WT) mouse recipients intraperitoneally administered WT or Klf13 null endometrial fragments, lesion incidence did not differ with donor genotype. No differences were noted for lesion volume, number, proliferation status, and apoptotic index as well. Klf13 null lesions displayed reduced total PGR and ESR1 (RNA and immunoreactive protein) and altered expression of several PGR and ESR1 target genes, relative to WT lesions. Unlike for Klf9 null lesions, changes in transcript levels for PGR-A, ESR1, and Notch/Hedgehog-associated pathway components were not observed for Klf13 null lesions. Results demonstrate lack of a causative relationship between endometrial KLF13 deficiency and lesion establishment in mice, and they support the broader participation of multiple signaling pathways, besides those mediated by steroid receptors, in the pathology of endometriosis.


Subject(s)
Cell Cycle Proteins/metabolism , Endometriosis/metabolism , Endometrium/metabolism , Kruppel-Like Transcription Factors/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Repressor Proteins/metabolism , Signal Transduction/genetics , Animals , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Differentiation/genetics , Disease Models, Animal , Endometriosis/genetics , Endometriosis/pathology , Endometrium/pathology , Estradiol/blood , Female , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Progesterone/blood , Repressor Proteins/genetics
4.
Biol Reprod ; 87(5): 115, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22993382

ABSTRACT

The ovarian hormones estrogen and progesterone promote uterine receptivity and successful pregnancy through their cognate receptors functioning in concert with context-dependent nuclear coregulators. Previously, we showed that the transcription factor Krüppel-like factor (KLF) 9 is a progesterone receptor (PGR) coactivator in the uterus and that mice null for Klf9 exhibit subfertility and reduced progesterone sensitivity. The highly related family member KLF13 displays increased expression in uteri of pregnant and nonpregnant Klf9 null mice and similarly regulates PGR-mediated transactivation in endometrial stromal cells. However, a uterine phenotype with loss of Klf13 has not been reported. In the present study, we demonstrate that Klf13 deficiency in mice did not compromise female fertility and pregnancy outcome. Klf13 null females had litter sizes, numbers of implanting embryos, uterine morphology, and ovarian steroid hormone production comparable to those of wild-type (WT) counterparts. Further, pregnant WT and Klf13 null females at Day Postcoitum (DPC) 3.5 had similar uterine Pgr, estrogen receptor, and Wnt-signaling component transcript levels. Nuclear levels of KLF9 were higher in Klf13 null than in WT uteri at DPC 3.5, albeit whole-tissue KLF9 protein and transcript levels did not differ between genotypes. The lack of a similar induction of nuclear KLF9 levels in uteri of virgin Klf13((-/-)) mice relative to WT uteri was associated with lower stromal PGR expression. In differentiating human endometrial stromal cells, coincident KLF9/KLF13 knockdown by small interfering RNA targeting reduced decidualization-associated PRL expression, whereas KLF9 and KLF13 knockdowns alone reduced transcript levels of WNT4 and BMP2, respectively. Results suggest that KLF9 and KLF13 functionally compensate in peri-implantation uterus for pregnancy success.


Subject(s)
Cell Cycle Proteins/physiology , Embryo Implantation/physiology , Kruppel-Like Transcription Factors/physiology , Repressor Proteins/physiology , Reproduction/genetics , Uterus/physiology , Alkaline Phosphatase/metabolism , Animals , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Differentiation , Female , Gene Expression , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Phenotype , Pregnancy , RNA, Small Interfering/pharmacology , Repressor Proteins/deficiency , Repressor Proteins/genetics , Stromal Cells
5.
Biol Reprod ; 85(2): 378-85, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21543766

ABSTRACT

Endometrial cancer is the most commonly diagnosed female genital tract malignancy. Krüppel-like factor 9 (KLF9), a member of the evolutionarily conserved Sp family of transcription factors, is expressed in uterine stroma and glandular epithelium, where it affects cellular proliferation, differentiation, and apoptosis. Deregulated expression of a number of Sp proteins has been associated with multiple types of human tumors, but a role for KLF9 in endometrial cancer development and/or progression is unknown. Here, we evaluated KLF9 expression in endometrial tumors and adjacent uninvolved endometrium of women with endometrial carcinoma. KLF9 mRNA and protein levels were lower in endometrial tumors coincident with decreased expression of family member KLF4 and growth-regulators FBJ murine osteosarcoma viral oncogene homolog (FOS) and myelocytomatosis viral oncogene homolog (MYC) and with increased expression of telomerase reverse transcriptase (TERT) and the chromatin-modifying enzymes DNA methyltransferase 1 (DNMT1) and histone deacetylase 3 (HDAC3). Expression of estrogen receptor alpha (ESR1) and the tumor-suppressor phosphatase and tensin homolog deleted in chromosome 10 (PTEN) did not differ between tumor and normal tissue. The functional relevance of attenuated KLF9 expression in endometrial carcinogenesis was further evaluated in the human endometrial carcinoma cell line Ishikawa by siRNA targeting. KLF9 depletion resulted in loss of normal cellular response to the proliferative effects of estrogen concomitant with reductions in KLF4 and MYC and with enhancement of TERT and ESR1 gene expression. Silencing of KLF4 did not mimic the effects of silencing KLF9 in Ishikawa cells. We suggest that KLF9 loss-of-expression accompanying endometrial carcinogenesis may predispose endometrial epithelial cells to mechanisms of escape from estrogen-mediated growth regulation, leading to progression of established neoplasms.


Subject(s)
Carcinoma/metabolism , Endometrial Neoplasms/metabolism , Estrogens/pharmacology , Gene Expression Regulation, Neoplastic/physiology , Kruppel-Like Transcription Factors/metabolism , Adult , Aged , Carcinoma/genetics , Cell Proliferation/drug effects , Cells, Cultured , Endometrium/cytology , Female , Gene Silencing , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Middle Aged
6.
Genesis ; 48(8): 485-91, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20506374

ABSTRACT

NR4A nuclear receptors are a diverse group of orphan nuclear receptors with critical roles in regulating cell proliferation and cell differentiation. The ortholog of the NR4A nuclear receptor in Caenorhabditis elegans, NHR-6, also has a role in cell proliferation and cell differentiation during organogenesis of the spermatheca. Here we show that NHR-6 is able to bind the canonical NR4A monomer response element and can transactivate from this site in mammalian HEK293 cells. Using a functional GFP-tagged NHR-6 fusion, we also demonstrate that NHR-6 is nuclear localized during development of the spermatheca. Mutation of the DNA-binding domain of NHR-6 abolishes its activity in genetic rescue assays, demonstrating a requirement for the DNA-binding domain. This study represents the first genetic demonstration of an in vivo requirement for an NR4A nuclear receptor DNA-binding domain in a whole organism.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Mutation , Organogenesis/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Animals, Genetically Modified , Binding Sites/genetics , Binding Sites/physiology , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/metabolism , Cell Nucleus/metabolism , DNA/genetics , DNA/metabolism , Genetic Complementation Test , Gonads/cytology , Gonads/growth & development , Gonads/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Microscopy, Fluorescence , Protein Binding , Receptors, Cytoplasmic and Nuclear/metabolism , Response Elements/genetics , Transfection
7.
Endocrinology ; 157(7): 2870-82, 2016 07.
Article in English | MEDLINE | ID: mdl-27175969

ABSTRACT

Endometriosis is a benign gynecological condition that causes considerable morbidity due to associated infertility, debilitating pelvic pain and inflammatory dysfunctions. Diet is a highly modifiable risk factor for many chronic diseases, but its contribution to endometriosis has not been extensively investigated, due partly to the paradoxical inverse association between obesity and disease incidence. Nevertheless, chronic exposure to dietary high-fat intake has been linked to greater systemic inflammation and oxidative stress, both features of women with endometriosis. Here, we evaluated the effects of a high-fat diet (HFD) (45% fat kcal) on endometriosis progression using an immunocompetent mouse model where ectopic lesion incidence was induced in wild-type recipients by ip administration of endometrial fragments from transcription factor Krüppel-like factor 9-null donor mice. We show that HFD significantly increased ectopic lesion numbers in recipient mice with no significant weight gain and modifications in systemic ovarian steroid hormone and insulin levels, relative to control diet-fed (17% fat kcal) mice. HFD promotion of lesion establishment was associated with reductions in stromal estrogen receptor 1 isoform and progesterone receptor expression, increased F4/80-positive macrophage infiltration, higher stromal but not glandular epithelial proliferation, and enhanced expression of proinflammatory and prooxidative stress pathway genes. Lesion-bearing HFD-fed mice also displayed higher peritoneal fluid TNFα and elevated local and systemic redox status than control diet-fed counterparts. Our results suggest that HFD intake exacerbates endometriosis outcome in the absence of ovarian dysfunction and insulin resistance in mice and warrants further consideration with respect to clinical management of endometriosis progression and recurrence in nonobese patients.


Subject(s)
Diet, High-Fat , Endometriosis/metabolism , Inflammation/metabolism , Insulin Resistance/physiology , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Disease Models, Animal , Disease Progression , Endometriosis/pathology , Female , Inflammation/pathology , Mice , Oxidation-Reduction
8.
J Mol Endocrinol ; 54(2): R89-R101, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25654975

ABSTRACT

Female reproductive tract pathologies arise largely from dysregulation of estrogen and progesterone receptor signaling, leading to aberrant cell proliferation, survival, and differentiation. The signaling pathways orchestrated by these nuclear receptors are complex, require the participation of many nuclear proteins serving as key binding partners or targets, and involve a range of paracrine and autocrine regulatory circuits. The members of the Krüppel-like factor (KLF) family of transcription factors are ubiquitously expressed in reproductive tissues and have been increasingly implicated as critical co-regulators and integrators of steroid hormone actions. Herein, we explore the involvement of KLF family members in uterine pathology, describe their currently known molecular mechanisms, and discuss their potential as targets for therapeutic intervention.


Subject(s)
Genitalia, Female/metabolism , Genitalia, Female/pathology , Kruppel-Like Transcription Factors/metabolism , Amino Acid Sequence , Female , Gene Expression Regulation , Humans , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/genetics , Molecular Sequence Data , Ovary/metabolism , Signal Transduction/genetics , Uterus/metabolism , Uterus/pathology
9.
Endocrinology ; 155(4): 1532-46, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24476135

ABSTRACT

Endometriosis, a steroid hormone-dependent disease characterized by aberrant activation of estrogen receptor signaling and progesterone resistance, remains intractable because of the complexity of the pathways underlying its manifestation. We previously showed that eutopic endometria of women with endometriosis exhibit lower expression of Krüppel-like factor 9 (KLF9), a progesterone receptor coregulator in the uterus, relative to that of women without disease. Here we examined whether loss of endometrial KLF9 expression causes ectopic lesion establishment using syngeneic wild-type (WT) mice as recipients of endometrial fragments from WT and Klf9 null donors. We found significantly higher incidence of ectopic lesions with Klf9 null than WT endometria 8 weeks after tissue injection into the intraperitoneal cavity. The increased incidence of lesion establishment with Klf9 null endometria was associated with a higher expression ratio of estrogen receptor 2 isoform relative to that of estrogen receptor 1 and attenuated progesterone receptor levels in endometriotic stromal cells. PCR array analyses of Notch and Hedgehog signaling components in ectopic lesions demonstrated up-regulated expression of select genes (Jag 2, Shh, Gli1, and Stil 1) in Klf9 null lesions relative to that in WT lesions. Immunohistochemical analyses showed increased levels of Notch intracellular domain and Sonic Hedgehog proteins in Klf9 null lesions relative to that in WT lesions, confirming pathway activation. WT recipients with Klf9 null lesions displayed lower systemic levels of TNFα and IL-6 and higher soluble TNF receptor 1 than corresponding recipients with WT lesions. Our results suggest that endometrial KLF9 deficiency promotes endometriotic lesion establishment by the coincident deregulation of Notch-, Hedgehog-, and steroid receptor-regulated pathways.


Subject(s)
Endometriosis/metabolism , Endometrium/metabolism , Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/physiology , Receptors, Estrogen/metabolism , Receptors, Notch/metabolism , Animals , Apoptosis , Ascitic Fluid/metabolism , Cell Proliferation , Disease Models, Animal , Endometriosis/genetics , Endometrium/cytology , Female , Gene Expression Regulation , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Receptors, Steroid/metabolism , Signal Transduction
10.
Stem Cell Res ; 11(3): 1149-62, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24012543

ABSTRACT

Diet is highly linked to breast cancer risk, yet little is known about its influence on mammary epithelial populations with distinct regenerative and hence, tumorigenic potential. To investigate this, we evaluated the relative frequency of lineage-negative CD29(hi)CD24(+), CD29(lo)CD24(+) and CD29(hi)Thy1(+)CD24(+) epithelial subpopulations in pre-neoplastic mammary tissue of adult virgin MMTV-Wnt1-transgenic mice fed either control (Casein) or soy-based diets. We found that mammary epithelial cells exposed to soy diet exhibited a lower percentage of CD29(hi)CD24(+)Lin(-) population, decreased ability to form mammospheres in culture, lower mammary outgrowth potential when transplanted into cleared fat pads, and reduced appearance of tumor-initiating CD29(hi)Thy1(+)CD24(+) cells, than in those of control diet-fed mice. Diet had no comparable influence on the percentage of the CD29(lo)CD24(+)Lin(-) population. Global gene expression profiling of the CD29(hi)CD24(+)subpopulation revealed markedly altered expression of genes important to inflammation, cytokine and chemokine signaling, and proliferation. Soy-fed relative to casein-fed mice showed lower mammary tumor incidence, shorter tumor latency, and reduced systemic levels of estradiol 17-ß, progesterone and interleukin-6. Our results provide evidence for the functional impact of diet on specific epithelial subpopulations that may relate to breast cancer risk and suggest that diet-regulated cues can be further explored for breast cancer risk assessment and prevention.


Subject(s)
CD24 Antigen/metabolism , Cytokines/genetics , Diet , Epithelial Cells/cytology , Integrin beta1/metabolism , Mammary Neoplasms, Animal/pathology , Animals , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Cytokines/metabolism , Down-Regulation , Epithelial Cells/metabolism , Female , Humans , Male , Mammary Glands, Human/metabolism , Mammary Neoplasms, Animal/prevention & control , Mice , Mice, Transgenic , Receptors, Virus/deficiency , Receptors, Virus/genetics , Receptors, Virus/metabolism , Risk Factors , Transcriptome , Wnt1 Protein/deficiency , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL