Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Publication year range
1.
Oncotarget ; 8(7): 11302-11315, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28076321

ABSTRACT

PURPOSE: Discovery and development of a novel anticancer PEG-SMR-Clu peptide to prevent breast cancer metastasis. How breast cancer cells and primary mammary epithelial cells interact and communicate with each other to promote tumorigenesis and how to prevent tumor metastasis has long been a concern of researchers. Cancer cells secrete exosomes containing proteins and RNA. These factors can influence tumor development by directly targeting cancer cells and tumor stroma. In this study, we determined the effects of a peptide as an inhibitor of exosome secretion on breast tumors. We developed a peptide derived from the Secretion Modification Region (SMR) of HIV-1 Nef protein that was modified with PEG on the N-terminus and with a Clusterin (Clu)-binding peptide on the C-terminus. Attachment of PEG to the SMR peptide, termed PEGylation, offers improved water solubility and stability as well as reduced clearance through the kidneys, leading to a longer circulation time. The 12-mer Clu-binding peptide plays multiple roles in tumor development and metastasis. The Clu peptide can be detected by antibody in vivo, thus it has the potential to be used to monitor tumor status and treatment efficacy in animal studies and eventually in cancer patients. RESULTS: PEG-SMRwt-Clu and PEG-SMRwt peptides inhibited the growth of both of MCF-7 (estrogen responsive, ER+) and MDA-MD-231 (estrogen non-responsive, ER-) human breast cancer cells in a dose and time-dependent manner, without inducing cytotoxic effects. The SMRwt peptide, combined with paclitaxel, induced G2/M phase cell cycle arrest on MCF-7 and MDA-MB-231 cells but did not promote apoptosis. PEG-SMRwt-Clu peptide treatment blocked exosome release from both MCF-7 and MDA-MB-231 cells. This effect was blocked by knockdown of the chaperone protein mortalin by either antibody or siRNA. MATERIALS AND METHODS: MCF-7 and MDA-MB-231 breast tumor cells were treated with PEG-SMR-Clu peptide alone and in combination with paclitaxel and cisplatin. Cell proliferation and viabilty were determined via cell cycle analysis using Cellometer imaging cytometry, Annexin V and MTT assays. The effects of the PEG-SMR-Clu peptide on tumor exosome release were determined by testing isolated exosome fractions, for (i) expression of CD63 and Alix proteins by Western blotting, (ii) NanoSight nanoparticle tracking analysis (NTA 10) to measure exosomes size and concentration, and (iii) measurement of acetylcholinesterase (AchE) for exosome specific enzyme activity. CONCLUSIONS: PEG-SMRwt-CLU peptides inhibited the growth of human breast cancer cells and blocked tumor exosome release in vitro. The peptide alone did not cause increased cytotoxicity or apoptosis induction, but did cause cell cycle G2/M phase arrest in both estrogen responsive and non-responsive breast cancer cells. These data suggest a potential therapeutic value of SMR to prevent breast cancer metastasis and as an adjuvant for the chemotherapeutic treatment of human breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Clusterin/pharmacology , Exosomes/metabolism , Peptides/pharmacology , nef Gene Products, Human Immunodeficiency Virus/pharmacology , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Polyethylene Glycols/pharmacology
2.
ACS Infect Dis ; 1(12): 604-14, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26771003

ABSTRACT

Antifolates are widely used to treat several diseases but are not currently used in the first-line treatment of tuberculosis, despite evidence that some of these molecules can target Mycobacterium tuberculosis (Mtb) bacilli in vitro. To identify new antifolate candidates for animal-model efficacy studies of tuberculosis, we paired knowledge and tools developed in academia with the infrastructure and chemistry resources of a large pharmaceutical company. Together we curated a focused library of 2508 potential antifolates, which were then tested for activity against live Mtb. We identified 210 primary hits, confirmed the on-target activity of potent compounds, and now report the identification and characterization of 5 hit compounds, representative of 5 different chemical scaffolds. These antifolates have potent activity against Mtb and represent good starting points for improvement that could lead to in vivo efficacy studies.

3.
Int J Cancer ; 123(12): 2782-90, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18798554

ABSTRACT

Several proangiogenic/proinflammatory factors involved in endometrial cancer are regulated by leptin, but the signaling mechanisms responsible for these leptin-induced actions are largely unknown. Here, we report that in benign (primary and HES) and cancerous-endometrial epithelial cells (EEC) (An3Ca, SK-UT2 and Ishikawa), leptin in a dose-dependent manner regulates vascular endothelial growth factor, (VEGF); interleukin-1 beta, (IL-1beta); leukemia inhibitory factor, (LIF) and their respective receptors, VEGFR2, IL-1R tI and LIFR. Remarkably, leptin induces a greater increase in VEGF/VEGFR2 and LIF levels in cancer than in benign cells. However, IL-1beta was only increased by leptin in benign primary-EEC. Cancer-EEC expressed higher levels of leptin receptor (full-length OB-Rb and short isoforms) in contrast to benign primary-EEC. Leptin-mediated activation of JAK2 (janus kinase 2) was upstream to the activation of PI-3K (phosphatidylinositol-3 kinase) and/or MAPK (mitogen-activated protein kinase) signaling pathways. Leptin induction of cytokines/receptors generally involved JAK2 and MAPK activation, but PI-3K phosphorylation was required for leptin increase of LIF, IL-1/IL-1R tI. Leptin-mediated activation of mTOR (mammalian target of Rapamycin), mainly linked to MAPK, played a central role in leptin regulation of all cytokines and receptors. These results suggest that leptin's effects are cell-specific and could confer a proliferative or cell survival advantage or possibly promote endometrial thickness. Leptin's effects on proangiogenic molecules were more evident in malignant versus benign cells and may imply that there is an underlying shift in leptin-induced cell signaling pathways in endometrial cancer cells.


Subject(s)
Adenocarcinoma/metabolism , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Leptin/metabolism , Neovascularization, Pathologic/metabolism , Protein Kinases/metabolism , Adenocarcinoma/blood supply , Blotting, Western , Endometrial Neoplasms/blood supply , Endometrium/blood supply , Female , Humans , Interleukin-1beta/metabolism , Leukemia Inhibitory Factor/metabolism , Receptors, Interleukin-1/metabolism , Receptors, OSM-LIF/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
4.
Endocrinology ; 149(2): 506-14, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17962343

ABSTRACT

Leptin, a 16-kDa cytokine, has been implicated in several reproductive processes and disorders. Notably, elevated leptin levels in the peritoneal fluid of women with mild endometriosis has been demonstrated, suggesting a role for this cytokine in the early stages of disease establishment. To gain insight into the functional significance of leptin during the initial requisite proliferative and neovascularization events involved in endometriosis, we investigated the effect of disruption of in vivo leptin signaling on the establishment and/or maintenance of an endometriosis-like lesion in a syngeneic immunocompetent mouse model of endometriosis. Findings of this study show that the disruption of leptin signaling by ip injection of the pegylated leptin peptide receptor antagonist (LPrA) impairs the establishment of endometriosis-like lesions (derived from uteri of C57BL/6 female siblings) and results in a reduction of viable organized glandular epithelium, vascular endothelial growth factor-A expression, and mitotic activity. LPrA treatment resulted in a significant reduction of microvascular density in endometriosis-like lesions after continuous and acute courses. Endometriosis-like lesions (derived from tissue with functional leptin receptor) of Lepr(db) hosts (nonfunctional leptin receptor) were phenotypically similar to those of LPrA-treated mice. Our results confirm that leptin signaling is a necessary component in lesion proliferation, early vascular recruitment, and maintenance of neoangiogenesis in a murine model of endometriosis.


Subject(s)
Endometriosis/metabolism , Endometriosis/pathology , Leptin/genetics , Leptin/metabolism , Signal Transduction/physiology , Animals , Disease Models, Animal , Endometriosis/physiopathology , Female , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology , Phenotype , Uterus/pathology , Uterus/transplantation
5.
J Biol Chem ; 281(36): 26320-8, 2006 Sep 08.
Article in English | MEDLINE | ID: mdl-16825198

ABSTRACT

To gain insight into the mechanism(s) by which leptin contributes to mammary tumor (MT) development we investigated the effects of leptin, kinase inhibitors, and/or leptin receptor antagonists (LPrA2) on 4T1 mouse mammary cancer cells in vitro and LPrA2 on 4T1-MT development in vivo. Leptin increases the expression of vascular endothelial growth factor (VEGF), its receptor (VEGF-R2), and cyclin D1 through phosphoinositide 3-kinase, Janus kinase 2/signal transducer and activator of transcription 3, and/or extracellular signal-activated kinase 1/2 signaling pathways. In contrast to leptin-induced levels of cyclin D1 the changes in VEGF or VEGF-R2 were more dependent on specific signaling pathways. Incubation of 4T1 cells with anti-VEGF-R2 antibody increased leptin-mediated VEGF expression suggesting an autocrine/paracrine loop. Pretreatment of syngeneic mice with LPrA2 prior to inoculation with 4T1 cells delayed the development and slowed the growth of MT (up to 90%) compared with controls. Serum VEGF levels and VEGF/VEGF-R2 expression in MT were significantly lower in mice treated with LPrA2. Interestingly, LPrA2-induced effects were more pronounced in vivo than in vitro suggesting paracrine actions in stromal, endothelial, and/or inflammatory cells that may impact the growth of MT. Although all the mechanism(s) by which leptin contributes to tumor development are unknown, it appears leptin stimulates an increase in cell numbers, and the expression of VEGF/VEGF-R2. Together, these results provide further evidence suggesting leptin is a MT growth-promoting factor. The inhibition of leptin signaling could serve as a potential adjuvant therapy for treatment of breast cancer and/or provide a new target for the designing strategies to prevent MT development.


Subject(s)
Leptin/metabolism , Mammary Neoplasms, Animal/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Line, Tumor , Cyclin D1/metabolism , Female , Humans , Leptin/genetics , Mice , Mice, Inbred BALB C , Peptides/genetics , Peptides/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Receptors, Leptin , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
6.
Rev. cuba. endocrinol ; 3(2): 96-109, jul.-dic. 1992. ilus, tab
Article in Spanish | LILACS | ID: lil-140787

ABSTRACT

Se realizò el montaje de un método inmunoenzimático (ELISA) para la detección de anticuerpos capaces de reconocer la molécula de insulina y se aplicó a un grupo de 130 sujetos normales; se escogió el valor límite de negatividad para el ensayo y se aplicó a la población estudiada de diabéticos. Los coeficientes de variación obtenidos se hallan dentro del rango de valores normales para este procedimiento. No se observó correspondencia total entre los resultados de las muestras evaluadas con este método y el isotópico convencional empleado en el Instituto Nacional de Endocrinología. Se llegó a la conclusión de que estos 2 métodos detectan diferentes tipos de anticuerpos antiinsulina


Subject(s)
Child , Adolescent , Adult , Middle Aged , Humans , Enzyme-Linked Immunosorbent Assay , Insulin Antibodies , Blood Donors , Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2
7.
Rev. cuba. endocrinol ; 3(2): 96-109, jul.-dic. 1992. ilus, tab
Article in Spanish | CUMED | ID: cum-967

ABSTRACT

Se realizò el montaje de un método inmunoenzimático (ELISA) para la detección de anticuerpos capaces de reconocer la molécula de insulina y se aplicó a un grupo de 130 sujetos normales; se escogió el valor límite de negatividad para el ensayo y se aplicó a la población estudiada de diabéticos. Los coeficientes de variación obtenidos se hallan dentro del rango de valores normales para este procedimiento. No se observó correspondencia total entre los resultados de las muestras evaluadas con este método y el isotópico convencional empleado en el Instituto Nacional de Endocrinología. Se llegó a la conclusión de que estos 2 métodos detectan diferentes tipos de anticuerpos antiinsulina


Subject(s)
Child , Adolescent , Adult , Middle Aged , Aged , Humans , Insulin Antibodies , Enzyme-Linked Immunosorbent Assay , Blood Donors , Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2
SELECTION OF CITATIONS
SEARCH DETAIL
...