Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Endocrinology ; 158(9): 2754-2773, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28911166

ABSTRACT

We discovered that pigment epithelium-derived factor (PEDF)-null mice have endometrial hyperplasia, the precursor to human type I endometrial cancer (ECA), which is etiologically linked to unopposed estrogen (E2), suggesting that this potent antiangiogenic factor might contribute to dysregulated growth and the development of type I ECA. Treatment of both ECA cell lines and primary ECA cells with recombinant PEDF dose dependently decreased cellular proliferation via an autocrine mechanism by blocking cells in G1 and G2 phases of the cell cycle. Consistent with the known opposing effects of E2 and progesterone (Pg) on endometrial proliferation, Pg increases PEDF protein synthesis and release, whereas E2 has the converse effect. Using PEDF luciferase promoter constructs containing two Pg and one E2 response elements, E2 reduced and Pg increased promoter activity due to distal response elements. Furthermore, E2 decreases and Pg increases PEDF secretion into conditioned media (CM) by both normal endometrial stromal fibroblasts (ESFs) and cancer-associated fibroblasts (CAFs), but only CM from ESFs mediated growth-inhibitory activity of primary endometrial epithelial cells (EECs). In addition, in cocultures with primary EECs, Pg-induced growth inhibition is mediated by ESFs, but not CAFs. This is consistent with reduced levels of Pg receptors on CAFs surrounding human malignant glands in vivo. Taken together, the data suggest that PEDF is a hormone-regulated negative autocrine mediator of endometrial proliferation, and that paracrine growth inhibition by soluble factors, possibly PEDF, released by ESFs in response to Pg, but not CAFs, exemplifies a tumor microenvironment that contributes to the pathogenesis of ECA.


Subject(s)
Carcinoma, Endometrioid/pathology , Cell Proliferation , Endometrial Neoplasms/pathology , Endometrium , Epithelial Cells/physiology , Eye Proteins/physiology , Hormones/pharmacology , Nerve Growth Factors/physiology , Serpins/physiology , Stromal Cells/physiology , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Endometrium/cytology , Endometrium/drug effects , Endometrium/metabolism , Endometrium/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Estradiol/pharmacology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Progesterone/pharmacology , Stromal Cells/drug effects , Stromal Cells/pathology , Tumor Cells, Cultured
2.
Cell Cycle ; 15(7): 931-47, 2016.
Article in English | MEDLINE | ID: mdl-26963853

ABSTRACT

We previously reported that aberrant TGF-ß/Smad2/3 signaling in endometrial cancer (ECA) leads to continuous ubiquitylation of p27(kip1)(p27) by the E3 ligase SCF-Skp2/Cks1 causing its degradation, as a putative mechanism involved in the pathogenesis of this cancer. In contrast, normal intact TGF-ß signaling prevents degradation of nuclear p27 by SCF-Skp2/Cks1 thereby accumulating p27 to block Cdk2 for growth arrest. Here we show that in ECA cell lines and normal primary endometrial epithelial cells, TGF-ß increases Cdh1 and its binding to APC/C to form the E3 ligase complex that ubiquitylates Cks1 and Skp2 prompting their proteasomal degradation and thus, leaving p27 intact. Knocking-down Cdh1 in ECA cell lines increased Skp2/Cks1 E3 ligase activity, completely diminished nuclear and cytoplasmic p27, and obviated TGF-ß-mediated inhibition of proliferation. Protein synthesis was not required for TGF-ß-induced increase in nuclear p27 and decrease in Cks1 and Skp2. Moreover, half-lives of Cks1 and Skp2 were extended in the Cdh1-depleted cells. These results suggest that the levels of p27, Skp2 and Cks1 are strongly or solely regulated by proteasomal degradation. Finally, an inverse relationship of low p27 and high Cks1 in the nucleus was shown in patients in normal proliferative endometrium and grade I-III ECAs whereas differentiated secretory endometrium showed the reverse. These studies implicate Cdh1 as the master regulator of TGF-ß-induced preservation of p27 tumor suppressor activity. Thus, Cdh1 is a potential therapeutic target for ECA and other human cancers showing an inverse relationship between Cks1/Skp2 and p27 and/or dysregulated TGF-ß signaling.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , CDC2-CDC28 Kinases/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endometrial Neoplasms/enzymology , S-Phase Kinase-Associated Proteins/metabolism , Transforming Growth Factor beta/physiology , Cdh1 Proteins/biosynthesis , Cdh1 Proteins/genetics , Cdh1 Proteins/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Nucleus/enzymology , Cell Nucleus/genetics , Cell Proliferation , Endometrial Neoplasms/metabolism , Endometrium/enzymology , Endometrium/growth & development , Endometrium/metabolism , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Female , Humans , Proteasome Endopeptidase Complex/metabolism
3.
Microb Cell Fact ; 14: 165, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26471510

ABSTRACT

BACKGROUND: Calreticulin (CRT) resides in the endoplasmic reticulum (ER) and functions to chaperone proteins, ensuring proper folding, and intracellular Ca(2+) homeostasis. Emerging evidence shows that CRT is a multifunctional protein with significant roles in physiological and pathological processes with presence both inside and outside of the ER, including the cell surface and extracellular space. These recent findings suggest the possible use of this ER chaperone in development of new therapeutic pharmaceuticals. Our study was focused on human CRT production in two yeast species, Saccharomyces cerevisiae and Pichia pastoris. RESULTS: Expression of a full-length human CRT precursor including its native signal sequence resulted in high-level secretion of mature recombinant protein into the culture medium by both S. cerevisiae and P. pastoris. To ensure the structural and functional quality of the yeast-derived CRTs, we compared yeast-secreted human recombinant CRT with native CRT isolated from human placenta. In ESI-MS (electrospray ionization mass spectrometry), both native and recombinant full-length CRT showed an identical molecular weight (mass) of 46,466 Da and were monomeric by non-denaturing PAGE. Moreover, limited trypsin digestion yielded identical fragment patterns of calcium-binding recombinant and native CRT suggesting that the yeast-derived CRT was correctly folded. Furthermore, both native and recombinant CRT induced cellular proliferation (MTS assay) and migration of human dermal fibroblasts (in vitro wound healing assay) with the same specific activities (peak responses at 1-10 ng/ml) indicating that the functional integrity of yeast-derived CRT was completely preserved. Simple one-step purification of CRT from shake-flask cultures resulted in highly pure recombinant CRT protein with yields reaching 75 % of total secreted protein and with production levels of 60 and 200 mg/l from S. cerevisiae and P. pastoris, respectively. Finally, cultivation of P. pastoris in a bioreactor yielded CRT secretion titer to exceed 1.5 g/l of culture medium. CONCLUSIONS: Yeasts are able to correctly process and secrete large amounts of mature recombinant human CRT equally and fully biologically active as native human CRT. This allows efficient production of high-quality CRT protein in grams per liter scale.


Subject(s)
Calreticulin/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Bioreactors , Calreticulin/chemistry , Calreticulin/genetics , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Molecular Sequence Data , Molecular Weight , Native Polyacrylamide Gel Electrophoresis , Pichia/metabolism , Placenta/metabolism , Plasmids/genetics , Plasmids/metabolism , Pregnancy , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Spectrometry, Mass, Electrospray Ionization
4.
Endocrinology ; 154(11): 4030-45, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24035998

ABSTRACT

In many human cancers, the tumor suppressor, p27(kip1) (p27), a cyclin-dependent kinase inhibitor critical to cell cycle arrest, undergoes perpetual ubiquitin-mediated proteasomal degradation by the E3 ligase complex SCF-Skp2/Cks1 and/or cytoplasmic mislocalization. Lack of nuclear p27 causes aberrant cell cycle progression, and cytoplasmic p27 mediates cell migration/metastasis. We previously showed that mitogenic 17-ß-estradiol (E2) induces degradation of p27 by the E3 ligase Skp1-Cullin1-F-Box- S phase kinase-associated protein2/cyclin dependent kinase regulatory subunit 1 in primary endometrial epithelial cells and endometrial carcinoma (ECA) cell lines, suggesting a pathogenic mechanism for type I ECA, an E2-induced cancer. The current studies show that treatment of endometrial carcinoma cells-1 (ECC-1) with small molecule inhibitors of Skp2/Cks1 E3 ligase activity (Skp2E3LIs) stabilizes p27 in the nucleus, decreases p27 in the cytoplasm, and prevents E2-induced proliferation and degradation of p27 in endometrial carcinoma cells-1 and primary ECA cells. Furthermore, Skp2E3LIs increase p27 half-life by 6 hours, inhibit cell proliferation (IC50, 14.3µM), block retinoblastoma protein (pRB) phosphorylation, induce G1 phase block, and are not cytotoxic. Similarly, using super resolution fluorescence localization microscopy and quantification, Skp2E3LIs increase p27 protein in the nucleus by 1.8-fold. In vivo, injection of Skp2E3LIs significantly increases nuclear p27 and reduces proliferation of endometrial epithelial cells by 42%-62% in ovariectomized E2-primed mice. Skp2E3LIs are specific inhibitors of proteolytic degradation that pharmacologically target the binding interaction between the E3 ligase, SCF-Skp2/Cks1, and p27 to stabilize nuclear p27 and prevent cell cycle progression. These targeted inhibitors have the potential to be an important therapeutic advance over general proteasome inhibitors for cancers characterized by SCF-Skp2/Cks1-mediated destruction of nuclear p27.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endometrial Neoplasms/drug therapy , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Antineoplastic Agents/therapeutic use , CDC2-CDC28 Kinases/genetics , CDC2-CDC28 Kinases/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cullin Proteins/genetics , Cullin Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Female , Humans , Mice , Mice, Inbred C57BL , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Transport , RNA Interference , S-Phase Kinase-Associated Proteins/genetics , Ubiquitin-Protein Ligases/genetics
5.
PLoS One ; 7(9): e46072, 2012.
Article in English | MEDLINE | ID: mdl-23029392

ABSTRACT

The levels of proteins that control the cell cycle are regulated by ubiquitin-mediated degradation via the ubiquitin-proteasome system (UPS) by substrate-specific E3 ubiquitin ligases. The cyclin-dependent kinase inhibitor, p27kip1 (p27), that blocks the cell cycle in G1, is ubiquitylated by the E3 ligase SCF-Skp2/Cks1 for degradation by the UPS. In turn, Skp2 and Cks1 are ubiquitylated by the E3 ligase complex APC/Cdh1 for destruction thereby maintaining abundant levels of nuclear p27. We previously showed that perpetual proteasomal degradation of p27 is an early event in Type I endometrial carcinogenesis (ECA), an estrogen (E2)-induced cancer. The present studies demonstrate that E2 stimulates growth of ECA cell lines and normal primary endometrial epithelial cells (EECs) and induces MAPK-ERK1/2-dependent phosphorylation of p27 on Thr187, a prerequisite for p27 ubiquitylation by nuclear SCF-Skp2/Cks1 and subsequent degradation. In addition, E2 decreases the E3 ligase [APC]Cdh1 leaving Skp2 and Cks1 intact to cause p27 degradation. Furthermore, knocking-down Skp2 prevents E2-induced p27 degradation and growth stimulation suggesting that the pathogenesis of E2-induced ECA is dependent on Skp2-mediated degradation of p27. Conversely, progesterone (Pg) as an inhibitor of endometrial proliferation increases nuclear p27 and Cdh1 in primary EECs and ECA cells. Pg, also increases Cdh1 binding to APC to form the active E3ligase. Knocking-down Cdh1 obviates Pg-induced stabilization of p27 and growth inhibition. Notably, neither E2 nor Pg affected transcription of Cdh1, Skp2, Cks1 nor p27. These studies provide new insights into hormone regulation of cell proliferation through the UPS. The data implicates that preventing nuclear p27 degradation by blocking Skp2/Cks1-mediated degradation of p27 or increasing Cdh1 to mediate degradation of Skp2-Cks1 are potential strategies for the prevention and treatment of ECA.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Estrogens/metabolism , Progesterone/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , CDC2-CDC28 Kinases/genetics , CDC2-CDC28 Kinases/metabolism , Cell Line, Tumor , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrium/cytology , Endometrium/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Knockdown Techniques , Humans , Phosphorylation , RNA, Messenger/genetics , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
6.
Wound Repair Regen ; 20(5): 715-30, 2012.
Article in English | MEDLINE | ID: mdl-22985041

ABSTRACT

A serious consequence of diabetes mellitus is impaired wound healing, which largely resists treatment. We previously reported that topical application of calreticulin (CRT), an endoplasmic reticulum chaperone protein, markedly enhanced the rate and quality of wound healing in an experimental porcine model of cutaneous repair. Consistent with these in vivo effects, in vitro CRT induced the migration and proliferation of normal human cells critical to the wound healing process. These functions are particularly deficient in poor healing diabetic wounds. Using a genetically engineered diabetic mouse (db/db) in a full-thickness excisional wound healing model, we now show that topical application of CRT induces a statistically significant decrease in the time to complete wound closure compared with untreated wounds by 5.6 days (17.6 vs. 23.2). Quantitative analysis of the wounds shows that CRT increases the rate of reepithelialization at days 7 and 10 and increases the amount of granulation tissue at day 7 persisting to day 14. Furthermore, CRT treatment induces the regrowth of pigmented hair follicles observed on day 28. In vitro, fibroblasts isolated from diabetic compared with wild-type mouse skin and human fibroblasts cultured under hyperglycemic compared with normal glucose conditions proliferate and strongly migrate in response to CRT compared with untreated controls. The in vitro effects of CRT on these functions are consistent with CRT's potent effects on wound healing in the diabetic mouse. These studies implicate CRT as a potential powerful topical therapeutic agent for the treatment of diabetic and other chronic wounds.


Subject(s)
Calreticulin/pharmacology , Diabetes Mellitus/metabolism , Fibroblasts/metabolism , Granulation Tissue/metabolism , Macrophages/metabolism , Wound Healing , Animals , Cell Proliferation/drug effects , Cells, Cultured , Diabetes Mellitus/drug therapy , Diabetes Mellitus/physiopathology , Disease Models, Animal , Female , Fibroblasts/drug effects , Granulation Tissue/drug effects , Humans , Macrophages/drug effects , Mice , Mice, Inbred NOD , Wound Healing/drug effects
7.
Cell Immunol ; 280(2): 148-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23399841

ABSTRACT

Intestinal epithelial cells (IEC) play a role in mucosal inflammation by producing pro-inflammatory chemokines that may initiate or amplify local responses. IL-1 is a potent activator of IEC and its receptor localizes to focal adhesions. Since the Rho-associated kinase, ROCK, also localizes to focal adhesions, we examined the role of ROCK in IL-1-induced chemokine responses in IEC cell lines. Suppressing ROCK with the Y27632 inhibitor suppressed IL-1-stimulated Caco-2 cell CXCL8/IL-8 and IEC-6 cell CCL2/MCP-1 secretion and mRNA levels. ROCK inhibition also suppressed IL-1-induced JNK phosphorylation in both cell lines, but high levels of the inhibitor had no significant effect on IL-1-stimulated Caco-2 IκBα phosphorylation and degradation or IKK phosphorylation and kinase activity. Therefore, ROCK may exert an effect on IL-1-stimulated JNK signaling to AP-1 activation, with little effect on IKK/IκBα signaling, defining a potentially important mechanism for regulating IL-1 signaling in IEC that may be essential for optimal cytokine responses.


Subject(s)
Epithelial Cells/drug effects , Interleukin-1/pharmacology , Intestinal Mucosa/drug effects , rho-Associated Kinases/physiology , Caco-2 Cells , Epithelial Cells/metabolism , Humans , Intestinal Mucosa/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/physiology , Phosphorylation , Signal Transduction , rho-Associated Kinases/antagonists & inhibitors
8.
J Insect Physiol ; 57(5): 635-44, 2011 May.
Article in English | MEDLINE | ID: mdl-21501620

ABSTRACT

Diapause is an alternate developmental pathway that is regulated by the neuroendocrine system in insects. To date, much of the information that has been published regarding the possible molecular events associated with diapause have been at the level of transcription. However, since transcription and translation are not linked in eukaryotic systems, a proteomics approach may represent a better tool to identify the gene products that regulate this period of developmental arrest. In this study, we performed gel-based proteomic and phospho-proteomic analyses to identify proteins that are differentially expressed or differentially phosphorylated in the brain during the initiation of pupal diapause in the flesh fly, Sarcophaga crassipalpis. A total of 27 proteins and phosphoproteins were identified by LC-MS/MS, including 16 that were either upregulated or phosphorylated during diapause, including proteins that function in cellular defense, cell cycle inhibition and neuronal protection. Of equal importance, 11 proteins were identified that were either downregulated at the total protein level, or from nuclear fractions. These included proteins involved in cell proliferation, adult development and aging. These data provide potentially valuable insight into the regulation of insect dormancy as well as the general phenomenon of aging in eukaryotic systems.


Subject(s)
Insect Proteins/analysis , Phosphoproteins/analysis , Proteome/analysis , Sarcophagidae/metabolism , Animals , Brain/growth & development , Brain/metabolism , Down-Regulation , Electrophoresis, Gel, Two-Dimensional , Metamorphosis, Biological , Pupa/genetics , Pupa/growth & development , Pupa/metabolism , Sarcophagidae/genetics , Sarcophagidae/growth & development , Tandem Mass Spectrometry , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...