Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Adv Drug Deliv Rev ; 188: 114442, 2022 09.
Article in English | MEDLINE | ID: mdl-35817212

ABSTRACT

The control of eukaryotic gene expression occurs at multiple levels, from transcription to messenger RNA processing, transport, localization, turnover, and translation. RNA-binding proteins control gene expression and are involved in different stages of mRNA processing, including splicing, maturation, turnover, and translation. A ubiquitously expressed RBP Human antigen R is engaged in the RNA processes mentioned above but, most importantly, controls mRNA stability and turnover. Dysregulation of HuR is linked to many diseases, including cancer and other immune-related disorders. HuR targets mRNAs containing AU-rich elements at their 3'untranslated region, which encodes proteins involved in cell growth, proliferation, tumor formation, angiogenesis, immune evasion, inflammation, invasion, and metastasis. HuR overexpression has been reported in many tumor types, which led to a poor prognosis for patients. Hence, HuR is considered an appealing drug target for cancer treatment. Therefore, multiple attempts have been made to identify small molecule inhibitors for blocking HuR functions. This article reviews the current prospects of drugs that target HuR in numerous cancer types, their mode of action, and off-target effects. Furthermore, we will summarize drugs that interfered with HuR-RNA interactions and established themselves as novel therapeutics. We will also highlight the significance of HuR overexpression in multiple cancers and discuss its role in immune functions. This review provides evidence of a new era of HuR-targeted small molecules that can be used for cancer therapeutics either as a monotherapy or in combination with other cancer treatment modalities.


Subject(s)
ELAV-Like Protein 1 , Neoplasms , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Humans , Neoplasms/pathology , Neovascularization, Pathologic , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
2.
Cell Immunol ; 312: 67-70, 2017 02.
Article in English | MEDLINE | ID: mdl-27825564

ABSTRACT

Human cytomegalovirus (CMV), a ubiquitous herpesvirus, has been implicated in the etiology of breast cancer. It is clear that not all people exposed to CMV are equally likely to develop this malignancy, implying the presence of host genetic factors that might modulate the cancer-spurring properties of the virus. CMV has evolved sophisticated strategies for evading host immunosurveillance. One strategy involves encoding decoy Fcγ receptors (FcγR) that thwart the Fcγ-mediated effector functions, such as antibody-dependent cellular cytotoxicity. In this investigation, using an enzyme-linked immunosorbent assay (ELISA), we aimed to determine whether the decoy FcγR encoded by the CMV gene RL13 binds differentially to anti-CMV antibodies expressing different immunoglobulin GM (γ marker) allotypes, genetic markers of immunoglobulin G (IgG). Results of our ELISA binding studies showed that the absorbance values for the binding of the viral FcγR to the GM 17-expressing IgG antibodies were significantly higher than for the GM 3-expressing antibodies (0.60 vs. 0.36; p=0.0019). These findings provide mechanistic insights into the modulating role played by the genetic variants of IgG in the generation of immunity to CMV in patients with breast cancer.


Subject(s)
Breast Neoplasms/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Receptors, IgG/metabolism , Viral Proteins/metabolism , Adult , Black or African American , Aged , Antibodies, Viral/metabolism , Antibody Affinity , Female , Genetic Predisposition to Disease , Genotype , Humans , Immune Evasion , Immunoglobulin G/metabolism , Immunoglobulin Gm Allotypes/metabolism , Middle Aged , Neoplasm Staging , Protein Binding , Receptors, IgG/genetics , Viral Proteins/genetics
3.
Sci Transl Med ; 7(319): 319ra206, 2015 12 23.
Article in English | MEDLINE | ID: mdl-26702094

ABSTRACT

Passive immunization with HIV-1-neutralizing monoclonal antibodies (mAbs) is being considered for prevention and treatment of HIV-1 infection. As therapeutic agents, mAbs could be used to suppress active virus replication, maintain suppression induced by antiretroviral therapy (ART), and/or decrease the size of the persistent virus reservoir. We assessed the impact of VRC01, a potent human mAb targeting the HIV-1 CD4 binding site, on ART-treated and untreated HIV-1-infected subjects. Among six ART-treated individuals with undetectable plasma viremia, two infusions of VRC01 did not reduce the peripheral blood cell-associated virus reservoir measured 4 weeks after the second infusion. In contrast, six of eight ART-untreated, viremic subjects infused with a single dose of VRC01 experienced a 1.1 to 1.8 log10 reduction in plasma viremia. The two subjects with minimal responses to VRC01 were found to have predominantly VRC01-resistant virus before treatment. Notably, two subjects with plasma virus load <1000 copies/ml demonstrated virus suppression to undetectable levels for over 20 days until VRC01 levels declined. Among the remaining four subjects with baseline virus loads between 3000 and 30,000 copies, viremia was only partially suppressed by mAb infusion, and we observed strong selection pressure for the outgrowth of less neutralization-sensitive viruses. In summary, a single infusion of mAb VRC01 significantly decreased plasma viremia and preferentially suppressed neutralization-sensitive virus strains. These data demonstrate the virological effect of this neutralizing antibody and highlight the need for combination strategies to maintain virus suppression.


Subject(s)
Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/immunology , Chronic Disease , HIV Antibodies/blood , HIV Infections/blood , HIV Infections/drug therapy , Humans , Kinetics , Middle Aged , Viral Load/immunology , Young Adult
4.
J Clin Cell Immunol ; 42013 Jun 30.
Article in English | MEDLINE | ID: mdl-24324917

ABSTRACT

Multiple Sclerosis (MS) is an incurable central nervous system (CNS) demyelinating disease affecting several million people worldwide. Due to the multifactorial and complex pathology of MS, FDA approved drugs often show limited efficacy inpatients. We earlier documented that both lovastatin (cholesterol lowering drug) and metformin (anti-diabetic drug) attenuate experimental autoimmune encephalomyelitis (EAE), a widely used model of MS via different mechanisms of action. Since combination therapy of two or more agents has advantage over monotherapy, we here assessed the therapeutic efficacy of metformin and lovastatin combination in EAE. We found that suboptimal doses of these drugs in combination had additive effect to attenuate established EAE in treated animals than their individual treatments. Histological, immunohistochemistry and western blotting analyses revealed that the observed demyelination and axonal loss as evident from reduced levels of myelin and neurofilament proteins in the spinal cords of EAE animals were attenuated by treatment with these drugs in combination. Accordingly, the observed infiltration of myelin reactive T cells (CD4 and CD8) and macrophages (CD68) as well as the increased expression of their signatory cytokines in the spinal cords of EAE animals were attenuated by this regimen as revealed by enzyme-linked immune-sorbent assay and real-time PCR analyses. In the periphery, this regimen biased the class of elicited anti-myelin basic protein immunoglobulins from IgG2a to IgG1 and IgG2b, suggesting a Th1 to Th2 shift which was further supported by the increased expression of their signatory cytokines in EAE animals. Taken together, these data imply that metformin and lovastatin combination attenuates T-cell autoimmunity and neurodegeneration in treated EAE animals thereby suggesting that the oral administration of these FDA approved drugs in combination has potential to limit MS pathogenesis.

5.
Am J Pathol ; 183(2): 526-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23759513

ABSTRACT

AMP-activated protein kinase (AMPK) signaling is reported to protect neurons under pathologic conditions; however, its effect on oligodendrocytes (OLs) remains to be elucidated. We investigated whether AMPK signaling protects OLs to restore central nervous system (CNS) functions in an experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis. Increased inflammation and demyelination in the CNS and peripheral immune responses were consistent with the observed clinical impairments in EAE animals, which were attenuated by treatment with metformin compared with vehicle. In addition, expressions of neurotrophic factors and of signatory genes of OL lineages were increased in the CNS of metformin-treated EAE animals. Likewise, metformin attenuated inflammatory response and enhanced expressions of neurotrophic factors, thereby protecting OLs via AMPK activation in mixed glial cultures stimulated with lipopolysaccharide/interferon γ in vitro, as evidenced by analysis of the expression of signatory genes of O1(+)/MBP(+) OLs and their cellular populations. Metformin also attenuated oxidative stress and malondialdehyde-containing protein levels, with corresponding induction of antioxidative defenses in OLs exposed to cytokines via AMPK activation. These effects of metformin were evident in the CNS of EAE animals. These data provide evidence that AMPK signaling is crucial to protect OLs and, thus, CNS functions in EAE animals. We conclude that AMPK activators, including metformin, have the potential to limit neurologic deficits in multiple sclerosis and related neurodegenerative disorders.


Subject(s)
AMP-Activated Protein Kinases/physiology , Central Nervous System/physiology , Encephalomyelitis, Autoimmune, Experimental/enzymology , Oligodendroglia/enzymology , Animals , Central Nervous System/enzymology , Central Nervous System Agents/pharmacology , Cytokines/drug effects , Cytokines/physiology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Metformin/pharmacology , Neuroglia/enzymology , Neuroglia/physiology , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
6.
PLoS Pathog ; 6(1): e1000742, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20126446

ABSTRACT

Upregulation of xCT, the inducible subunit of a membrane-bound amino acid transporter, replenishes intracellular glutathione stores to maintain cell viability in an environment of oxidative stress. xCT also serves as a fusion-entry receptor for the Kaposi's sarcoma-associated herpesvirus (KSHV), the causative agent of Kaposi's sarcoma (KS). Ongoing KSHV replication and infection of new cell targets is important for KS progression, but whether xCT regulation within the tumor microenvironment plays a role in KS pathogenesis has not been determined. Using gene transfer and whole virus infection experiments, we found that KSHV-encoded microRNAs (KSHV miRNAs) upregulate xCT expression by macrophages and endothelial cells, largely through miR-K12-11 suppression of BACH-1-a negative regulator of transcription recognizing antioxidant response elements within gene promoters. Correlative functional studies reveal that upregulation of xCT by KSHV miRNAs increases cell permissiveness for KSHV infection and protects infected cells from death induced by reactive nitrogen species (RNS). Interestingly, KSHV miRNAs simultaneously upregulate macrophage secretion of RNS, and biochemical inhibition of RNS secretion by macrophages significantly reduces their permissiveness for KSHV infection. The clinical relevance of these findings is supported by our demonstration of increased xCT expression within more advanced human KS tumors containing a larger number of KSHV-infected cells. Collectively, these data support a role for KSHV itself in promoting de novo KSHV infection and the survival of KSHV-infected, RNS-secreting cells in the tumor microenvironment through the induction of xCT.


Subject(s)
Amino Acid Transport System y+/biosynthesis , Gene Expression Regulation/physiology , Herpesvirus 8, Human/pathogenicity , MicroRNAs/metabolism , Oxidative Stress/physiology , Sarcoma, Kaposi/virology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Line , Endothelial Cells/metabolism , Endothelial Cells/virology , Fluorescent Antibody Technique , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Humans , Immunoblotting , Immunohistochemistry , Macrophages/metabolism , Macrophages/virology , Mice , MicroRNAs/genetics , Neoplasm Staging , Polymerase Chain Reaction , Reactive Nitrogen Species/metabolism , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/pathology , Transfection , Up-Regulation
7.
Clin Cancer Res ; 14(1): 300-8, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18172282

ABSTRACT

PURPOSE: Chemoprevention is an upcoming approach to control bladder cancer, which is one of the commonly diagnosed malignancies showing recurrence rate of 70% or even higher. Recently, we observed the in vitro efficacy of silibinin, a flavanolignan, in human bladder transitional cell papilloma RT4 cells. Here, we investigated the antitumor efficacy and associated mechanisms of silibinin in RT4 tumor xenograft. EXPERIMENTAL DESIGN: RT4 tumor xenograft was implanted s.c. in athymic nude mice, and then animals were oral gavaged with silibinin at 100 and 200 mg/kg doses, 5 days/week for 12 weeks. Tumor growth, body weight, and diet consumption were recorded, and tumors were analyzed for proliferation, apoptosis, and angiogenesis biomarkers and molecular alterations by immunohistochemistry, immunoblot analysis, and ELISA. p53 small interfering RNA was used in cell culture to examine the role of p53 in survivin expression. RESULTS: Silibinin feeding inhibited tumor xenograft growth without any gross signs of toxicity. Silibinin decreased tumor volume by 51% to 58% (P

Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Transitional Cell/drug therapy , Microtubule-Associated Proteins/drug effects , Urinary Bladder Neoplasms/drug therapy , Administration, Oral , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Transitional Cell/pathology , Cell Proliferation/drug effects , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Male , Mice , Mice, Nude , Microtubule-Associated Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Repressor Proteins , Silybin , Silymarin/administration & dosage , Survivin , Tumor Suppressor Protein p53/drug effects , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 5(8): 2121-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16928834

ABSTRACT

UVB radiation-induced DNA damage in skin activates cellular pathways involved in DNA repair, cell cycle regulation, and apoptosis, important events that prevent conversion of damaged skin cells into cancer. We reported recently the efficacy of silibinin against photocarcinogenesis along with altered molecular events in tumors (Cancer Research, 64:6349-56, 2004). The molecular and biological events modulated by silibinin in chronically UVB-irradiated skin leading to cancer prevention, however, are not known. Herein, we describe effect of silibinin on skin 15 and 25 weeks after UVB exposure and compared them with molecular alterations in skin tumors. UVB decreased E2F1 but increased E2F2 and E2F3 protein levels in skin, and these were reversed by silibinin treatment. Silibinin-induced E2F1 was accompanied by an inhibition of apoptosis and decreases in p53 and cyclin-dependent kinase inhibitors. Silibinin-caused decrease in E2F2 and E2F3 was accompanied by reduced levels of cyclin-dependent kinases, cyclins, CDC25C, and mitogen-activated protein kinases and Akt signaling and inhibition of cell proliferation. In tumorigenesis protocols, topical or dietary silibinin significantly inhibited tumor appearance and growth. As opposed to UVB-exposed skin, UVB-induced tumors showed elevated levels of E2F1, but these were reduced in silibinin-treated tumors without any effect on E2F2 and E2F3. Contrary to the inhibition of apoptosis and p53 expression in UVB-exposed skin cells, silibinin increased these variables in tumors. These differential effects of silibinin on E2F1 versus E2F2 and E2F3 and their associated molecular alterations and biological effects in chronic UVB-exposed skin suggest their role in silibinin interference with photocarcinogenesis.


Subject(s)
E2F Transcription Factors/drug effects , Skin Neoplasms/drug therapy , Skin/radiation effects , Ultraviolet Rays/adverse effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis/radiation effects , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Cycle/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/radiation effects , Cyclin-Dependent Kinase Inhibitor p27/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/radiation effects , E2F Transcription Factors/metabolism , Female , Mice , Mice, Hairless , Signal Transduction/drug effects , Signal Transduction/radiation effects , Silybin , Silymarin/pharmacology , Skin/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects
9.
Mol Cancer Ther ; 5(5): 1145-53, 2006 May.
Article in English | MEDLINE | ID: mdl-16731746

ABSTRACT

UVB radiation is the major etiologic factor in the development of nonmelanoma skin cancer. In addition to tumor-initiating effect, UVB also causes tumor promotion via mitogenic and survival signaling. Studies have shown strong preventive effects of silibinin against both UVB-induced and chemically induced tumor promotion in mouse skin models; however, mechanisms are not understood completely. Here, we used tumor promoter-sensitive JB6 mouse epithelial cell model and studied the effect of silibinin on two different mitogens [UVB and epidermal growth factor (EGF)] that induce mitogenic and cell survival signaling pathways. UVB (50-800 mJ/cm(2)) dose-dependently induced phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun-NH(2)-kinase 1/2 (JNK1/2), and p38 kinase (p38K) as well as Akt, with an optimum response at 400 mJ/cm(2) UVB dose. UVB caused a biphasic phosphorylation of ERK1/2 in a time kinetics study. Silibinin treatment before or immediately after UVB exposure, or both, resulted in a strong decrease in UVB-caused phosphorylation of ERK1/2 and Akt in both dose- and time-dependent manner, without any substantial response on JNK1/2 and p38K. Silibinin also suppressed UVB-induced activator protein-1 (AP-1) and nuclear factor-kappaB (NF-kappaB) activation, which are activated by ERK1/2 and Akt. Silibinin treatment under similar conditions also strongly inhibited EGF-induced ERK1/2, JNK1/2, and p38K as well as Akt phosphorylation, and also suppressed EGF-induced AP-1 and NF-kappaB activation. Because AP-1 and NF-kappaB are important nuclear transcription factors for tumor promotion, these results suggest that silibinin possibly prevents skin tumor promotion by inhibiting UVB- and EGF-induced mitogenic and cell survival signaling involving both AP-1 and NF-kappaB.


Subject(s)
Antioxidants/pharmacology , Cell Survival , Epidermal Growth Factor/pharmacology , Epidermis/metabolism , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Ultraviolet Rays , Animals , Antioxidants/metabolism , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Epidermal Cells , Epidermis/drug effects , Keratinocytes/metabolism , MAP Kinase Signaling System/radiation effects , Mice , Mitosis/drug effects , Mitosis/radiation effects , Models, Biological , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Silybin , Silymarin/metabolism , Silymarin/pharmacology , Ultraviolet Rays/adverse effects
10.
Carcinogenesis ; 26(8): 1404-13, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15831527

ABSTRACT

Ultraviolet B (UVB) radiation is a complete skin carcinogen causing DNA damage as a tumor-initiating event and activating signaling cascades that play a critical role in its tumor-promoting potential. Recently we reported that a naturally occurring flavonoid, silibinin, protects UVB-induced skin damages and prevents photocarcinogenesis. Here we examined silibinin efficacy on acute and chronic UVB-caused mitogen-activated protein kinases (MAPKs) and AKT activation and associated biological responses in SKH-1 hairless mouse skin. A single UVB exposure at 180 mJ/cm2 dose resulted in varying degrees of ERK1/2, JNK1/2, MAPK/p38 and AKT phosphorylation at various time-points in mouse skin; however, topical application of silibinin prior to or immediately after UVB exposure, or its dietary feeding strongly inhibited the activation of these molecules at all the time-points examined. Stronger effects of silibinin towards inhibition of UVB-caused phosphorylation of MAPKs and AKT were also observed in a chronic UVB (180 mJ/cm2/day for 5 days) exposure protocol. Immunohistochemical analysis of chronically exposed skin sections showed that silibinin treatment in all three protocols increases UVB-induced p53-positive cells and decreases UVB-caused cell proliferation, apoptotic and sunburn cells. These findings suggest that silibinin inhibits UVB-induced MAPK and AKT signaling and increases p53 in mouse skin, and that these effects of silibinin possibly lead to a decrease in UVB-caused proliferation and apoptosis, which might, in part, be responsible for its overall efficacy against photocarcinogenesis.


Subject(s)
Radiation-Protective Agents/pharmacology , Skin/radiation effects , Ultraviolet Rays , Animals , Apoptosis/radiation effects , Female , In Situ Nick-End Labeling , Mice , Mice, Hairless , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/radiation effects , Proliferating Cell Nuclear Antigen/analysis , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/radiation effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/radiation effects , Proto-Oncogene Proteins c-akt , Silybin , Silymarin/pharmacology , Tumor Suppressor Protein p53/analysis
11.
Biochem Biophys Res Commun ; 320(1): 183-9, 2004 Jul 16.
Article in English | MEDLINE | ID: mdl-15207719

ABSTRACT

Several recent studies by us have shown the strong chemopreventive efficacy of silibinin against both ultraviolet B (UVB) radiation and chemical carcinogen-induced tumorigenesis in mouse skin models. The molecular mechanisms underlying silibinin protective efficacy, however, are not completely known. Here, we examined the effect of silibinin on UVB-caused apoptosis in human epidermoid carcinoma A431 cells. Irradiation of cells with different doses of UVB (5-100 mJ/cm2) and different time periods (0.5-24h) resulted in a dose- and time-dependent increase in apoptosis (P < 0.05-0.001). Silibinin (100-200 microM) pre-treatment, however, resulted in an increase in UVB-induced apoptosis (P < 0.05-0.001); interestingly, its post-treatment caused a decrease in UVB-induced apoptosis (P < 0.05-0.001). A similar pattern in the activation of caspases-9, -3, and -7 was observed with these silibinin treatments. Further, silibinin treatment prior to or immediately after UVB exposure altered Bcl-2, Bax, Bak, and cytochrome c levels in mitochondria and cytosol in favor of or against apoptosis, respectively. Silibinin treatment prior to UVB also increased the activation of mitogen/stress activated protein kinases Erk1/2, JNK, and p38 kinase as compared to its post-treatment. Together, for the first time, our results demonstrate the role of mitochondrial apoptotic machinery and MAPK signaling cascade in silibinin-caused increase as well as protection in UVB-induced apoptosis in A431 cells, and suggest that similar mechanisms might be involved in preventive efficacy of silibinin against UVB-induced skin tumorigenesis.


Subject(s)
Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Squamous Cell/metabolism , Caspases/metabolism , Mitochondrial Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/radiation effects , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Enzyme Activation , Humans , Radiation Tolerance/genetics , Signal Transduction/drug effects , Signal Transduction/radiation effects , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...