Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Toxicol Sci ; 193(2): 175-191, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37074955

ABSTRACT

Exposure to traffic-related air pollution consisting of particulate matter (PM) is associated with cognitive decline leading to Alzheimer's disease (AD). In this study, we sought to examine the neurotoxic effects of exposure to ultrafine PM and how it exacerbates neuronal loss and AD-like neuropathology in wildtype (WT) mice and a knock-in mouse model of AD (AppNL-G-F/+-KI) when the exposure occurs at a prepathologic stage or at a later age with the presence of neuropathology. AppNL-G-F/+-KI and WT mice were exposed to concentrated ultrafine PM from local ambient air in Irvine, California, for 12 weeks, starting at 3 or 9 months of age. Particulate matter-exposed animals received concentrated ultrafine PM up to 8 times above the ambient levels, whereas control animals were exposed to purified air. Particulate matter exposure resulted in a marked impairment of memory tasks in prepathologic AppNL-G-F/+-KI mice without measurable changes in amyloid-ß pathology, synaptic degeneration, and neuroinflammation. At aged, both WT and AppNL-G-F/+-KI mice exposed to PM showed a significant memory impairment along with neuronal loss. In AppNL-G-F/+-KI mice, we also detected an increased amyloid-ß buildup and potentially harmful glial activation including ferritin-positive microglia and C3-positive astrocytes. Such glial activation could promote the cascade of degenerative consequences in the brain. Our results suggest that exposure to PM impairs cognitive function at both ages while exacerbation of AD-related pathology and neuronal loss may depend on the stage of pathology, aging, and/or state of glial activation. Further studies will be required to unveil the neurotoxic role of glial activation activated by PM exposure.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Particulate Matter/toxicity , Amyloid beta-Peptides/metabolism , Disease Models, Animal , Brain/metabolism , Memory Disorders/chemically induced , Mice, Transgenic
2.
Neuroscience ; 453: 69-80, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33246059

ABSTRACT

Effective clearance of neurotoxic amyloid-beta (Aß) from the brain is a critical process to prevent Alzheimer's disease (AD). One major clearance mechanism is Aß transcytosis mediated by low-density lipoprotein receptor-related protein 1 (LRP1) in capillary endothelial cells. A marked loss of endothelial LRP1 is found in AD brains and is believed to significantly impair Aß clearance. Recently, we demonstrated that pro-inflammatory cytokines IL-1ß, IL-6 and TNF-α, significantly down-regulated LRP1 in human primary microvascular endothelial cells (MVECs). In this study, we sought to determine the underlying molecular mechanism by which IL-1ß led to LRP1 loss in MVECs. Reduced LRP1 protein and transcript were detected up to 24 h post-exposure and returned to the baseline levels after 48 h post-exposure with 1 ng/ml IL-1ß. This reduction was in part mediated by microRNA-205-5p, -200b-3p, and -200c-3p, as these microRNAs were concomitantly upregulated in MVECs exposed to IL-1ß. Synthetic microRNA-205-5p, -200b-3p, and -200c-3p mimics recapitulated LRP1 loss in MVECs without IL-1ß, and their synthetic antagomirs effectively reversed IL-1ß-mediated LRP1 loss. Importantly, we found that the expression of these three microRNAs was controlled by NF-κB as pharmacological NF-κB inhibitor, BMS-345541, inhibited the IL-1ß-mediated upregulation of these microRNAs and rescued LRP1 expression. siRNA-mediated silencing of IκB in MVECs elevated microRNA-200b-3p and decreased LRP1 transcript, partially confirming our overall findings. In conclusion, our study provides a mechanism by which pro-inflammatory IL-1ß instigates the suppression of LRP1 expression in MVECs. Our findings could implicate spatiotemporal loss of LRP1 and impairment of the LRP1-mediated clearance mechanism by endothelial cells.


Subject(s)
Endothelial Cells , Gene Silencing , Interleukin-1beta/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1/genetics , MicroRNAs , Amyloid beta-Peptides/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Humans , MicroRNAs/genetics
3.
J Trace Elem Med Biol ; 62: 126578, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32599538

ABSTRACT

BACKGROUND: Copper (Cu) is an essential metal mediating a variety of vital biological reactions with its redox property. Its dyshomeostasis has been associated with accelerated cognitive decline and neurodegenerative disorders, such as Alzheimer's disease (AD). However, underlying neurotoxic mechanisms elicited by dysregulated Cu remain largely elusive. We and others previously demonstrated that exposure to Cu in drinking water significantly exacerbated pathological hallmarks of AD and pro-inflammatory activation of microglia, coupled with impaired phagocytic capacity, in mouse models of AD. METHODS: In the present study, we extended our investigation to evaluate whether chronic Cu exposure to wild-type (WT) and J20 mouse model of AD perturbs homeostatic dynamics of microglia and contributes to accelerated transformation of microglia towards degenerative phenotypes that are closely associated with neurodegeneration. We further looked for evidence of alterations in the microglial morphology and spatial memory of the Cu-exposed mice to assess the extent of the Cu toxicity. RESULTS: We find that chronic Cu exposure to pre-pathological J20 mice upregulates the translation of degenerative genes and represses homeostatic genes within microglia even in the absence amyloid-beta plaques. We also observe similar expression signatures in Cu-exposed WT mice, suggesting that excess Cu exposure alone could lead to perturbed microglial homeostatic phenotypes and contribute to accelerated cognitive decline. CONCLUSION: Our findings highlight the risk of chronic Cu exposure on cognitive decline and altered microglia activation towards degenerative phenotypes. These changes may represent one of the key mechanisms linking Cu exposure or its dyshomeostasis to an increased risk for AD.


Subject(s)
Alzheimer Disease/etiology , Cognition Disorders/chemically induced , Copper/toxicity , Microglia/drug effects , Microglia/pathology , Alzheimer Disease/chemically induced , Alzheimer Disease/genetics , Animals , Cognition Disorders/pathology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Inflammation/chemically induced , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Tamoxifen/pharmacology , Toxicity Tests, Chronic
4.
Toxicol Sci ; 170(1): 144-156, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30923833

ABSTRACT

Chronic exposure to copper and its dyshomeostasis have been linked to accelerated cognitive decline and potentially increasing risk for Alzheimer's disease (AD). We and others have previously demonstrated that exposure to copper through drinking water significantly increased parenchymal amyloid-beta (Aß) plaques and decreased endothelial low-density lipoprotein receptor-related protein 1 (LRP1) in mouse models of AD. In this study, we determined the underlying mechanisms that microRNA critically mediated the copper-induced loss of endothelial LRP1. In human primary microvascular endothelial cells (MVECs), microRNA-200b-3p, -200c-3p, and -205-5p were significantly elevated within the 24-h exposure to copper and returned to baseline after 48-h postexposure, which corresponded with the temporal change of LRP1 expression in these cells. Transient expression of synthetic microRNA-200b-3p, -200c-3p, or -205-5p on MVECs significantly decreased endothelial LRP1, and cotreatment of synthetic antagomirs effectively prevented the loss of LRP1 during copper exposure, collectively supporting the key regulatory role of these microRNAs in copper-induced loss of LRP1. In mice, a significant reduction of LRP1 in cortical vasculature was evident following 9 months exposure to 1.3 ppm copper in drinking water, although the levels of cortical microRNA-205-5p, -200b-3p, and -200c-3p were only marginally elevated. This, however, correlated with increased vascular accumulation of Aß and impairment of spatial memory, indicating that copper exposure has the pivotal role in the vascular damage and development of cognitive decline.


Subject(s)
Alzheimer Disease/chemically induced , Brain/drug effects , Copper/toxicity , Endothelial Cells/drug effects , Low Density Lipoprotein Receptor-Related Protein-1/antagonists & inhibitors , MicroRNAs/genetics , Alzheimer Disease/metabolism , Animals , Brain/blood supply , Cell Survival/drug effects , Disease Models, Animal , Endothelial Cells/metabolism , Female , Humans , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microvessels/drug effects , Microvessels/metabolism , Spatial Memory/drug effects , Transfection , Up-Regulation
5.
Toxicol Sci ; 163(2): 338-345, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29409005

ABSTRACT

Metals are commonly found in the environment, household, and workplaces in various forms, and a significant segment of the population is routinely exposed to the trace amount of metals from variety of sources. Exposure to metals, such as aluminum, lead, iron, and copper, from environment has long been debated as a potential environmental risk factor for Alzheimer's disease (AD) for decades, yet results from in vitro, in vivo, and human population remain controversial. In the case of copper, the neurotoxic mechanism of action was classically viewed as its strong affinity to amyloid-beta (Aß) to help its aggregation and increase oxidative stress via Fenton reaction. Thus, it has been thought that accumulation of copper mediates neurotoxicity, and removing it from the brain prevents or reverse Aß plaque burden. Recent evidence, however, suggests dyshomeostasis of copper and its valency in the body, instead of the accumulation and interaction with Aß, are major determinants of its beneficial effects as an essential metal or its neurotoxic counterpart. This notion is also supported by the fact that genetic loss-of-function mutations on copper transporters lead to severe neurological symptoms. Along with its altered distribution, recent studies have also proposed novel mechanisms of copper neurotoxicity mediated by nonneuronal cell lineages in the brain, such as capillary endothelial cells, leading to development of AD neuropathology. This review covers recent findings of multifactorial toxic mechanisms of copper and discusses the risk of environmental exposure as a potential factor in accounting for the variability of AD incidence.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Copper/toxicity , Dietary Exposure/adverse effects , Environmental Pollutants/toxicity , Protein Aggregation, Pathological/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Biomarkers/metabolism , Copper/metabolism , Dietary Exposure/analysis , Environmental Pollutants/metabolism , Humans , Oxidative Stress/drug effects , Risk Factors
6.
Neuroscience ; 348: 191-200, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28215745

ABSTRACT

Abnormal buildup of the microtubule associated protein tau is a major pathological hallmark of Alzheimer's disease (AD) and various tauopathies. The mechanisms by which pathological tau accumulates and spreads throughout the brain remain largely unknown. Previously, we demonstrated that a restoration of the major astrocytic glutamate transporter, GLT1, ameliorated a buildup of tau pathology and rescued cognition in a mouse model of AD. We hypothesized that aberrant extracellular glutamate and abnormal neuronal excitatory activities promoted tau pathology. In the present study, we investigated genetic interactions between tau and the GLT1 homolog dEaat1 in Drosophila melanogaster. Neuronal-specific overexpression of human wildtype tau markedly shortened lifespan and impaired motor behavior. RNAi depletion of dEaat1 in astrocytes worsened these phenotypes, whereas overexpression of dEaat1 improved them. However, the synaptic neuropil appeared unaffected, and we failed to detect any major neuronal loss with tau overexpression in combination with dEaat1 depletion. To mimic glutamate-induced aberrant excitatory input in neurons, repeated depolarization of neurons via transgenic TrpA1 was applied to the adult Drosophila optic nerves, and we examined the change of tau deposits. Repeated depolarization significantly increased the accumulation of tau in these neurons. We propose that increased neuronal excitatory activity exacerbates tau-mediated neuronal toxicity and behavioral deficits.


Subject(s)
Astrocytes/metabolism , Drosophila Proteins/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/metabolism , Neurons/metabolism , tau Proteins/metabolism , Animals , Biological Transport , Brain/metabolism , Disease Models, Animal , Drosophila melanogaster
7.
Toxicol Sci ; 152(1): 194-204, 2016 07.
Article in English | MEDLINE | ID: mdl-27122238

ABSTRACT

Copper promotes a toxic buildup of amyloid-beta (Aß) and neurofibrillary tangle pathology in the brain, and its exposure may increase the risk for Alzheimer's disease (AD). However, underlying molecular mechanisms by which copper triggers such pathological changes remain largely unknown. We hypothesized that the copper exposure perturbs brain inflammatory responses, leading to impairment of Aß clearance from the brain parenchyma. Here, we investigated whether copper attenuated Aß clearance by microglial phagocytosis or by low-density lipoprotein-related receptor protein-1 (LRP1) dependent transcytosis in both in vitro and in vivo When murine monocyte BV2 cells were exposed to copper, their phagocytic activation induced by fibrillar Aß or LPS was significantly reduced, while the secretion of pro-inflammatory cytokines, such as IL-1ß, TNF-α, and IL-6, were increased. Interestingly, not only copper itself but also IL-1ß, IL-6, or TNF-α were capable of markedly reducing the expression of LRP1 in human microvascular endothelial cells (MVECs) in a concentration-dependent manner. While copper-mediated downregulation of LRP1 was proteasome-dependent, the cytokine-induced loss of LRP1 was proteasome- or lysosome-independent. In the mouse model, copper exposure also significantly elevated neuroinflammation and downregulated LRP1 in the brain, consistent with our in vitro results. Taken together, our findings support the pathological impact of copper on inflammatory responses and Aß clearance in the brain, which could serve as key mechanisms to explain, in part, the copper exposure as an environmental risk factor for AD.


Subject(s)
Alzheimer Disease/chemically induced , Amyloid beta-Peptides/metabolism , Brain/drug effects , Copper Sulfate/toxicity , Cytokines/metabolism , Endothelial Cells/drug effects , Inflammation Mediators/metabolism , Microglia/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Phagocytosis/drug effects , Proteasome Endopeptidase Complex/metabolism , Receptors, LDL/metabolism , Transcytosis/drug effects , Tumor Suppressor Proteins/metabolism
8.
Cancer Lett ; 366(1): 44-51, 2015 Sep 28.
Article in English | MEDLINE | ID: mdl-26118775

ABSTRACT

HL60 and U937 (acute myeloid leukemia (AML) cell lines) were assessed for sensitivity to YM155, and found to have distinct sensitive and resistant phenotypes, respectively. In HL60 cells, YM155 inhibition of growth proliferation was due to apoptosis which was measured by annexin V/PI staining. YM155 induced apoptosis through activation of intrinsic and extrinsic pathways that also culminated in caspase-3 activity and PARP cleavage. YM155 sensitivity was partially associated with this compound's ability to down-regulate survivin transcription since this was more pronounced in the HL60 cell line. However, marked differences were also observed in XIAP, Bcl-2, and Mcl-1L, and Mcl-1s. Furthermore, YM155 treatment completely inhibited production of total Akt protein in HL60, but not U937 cells. Importantly, Akt activity (pAkt-Ser473) levels were maintained in YM155 treated U937 cells which may help stabilize other anti-apoptotic proteins. Combination treatments with an Akt inhibitor, MK-2206, reduced levels of pAkt-Ser473 in U937 cells and synergistically sensitized them to YM155 cytotoxicity. Collectively our results indicate that Akt signaling may be an important factor mediating YM155 response in AML, and combinatorial therapies with Akt inhibitors could improve treatment efficacy in YM155-resistant cells.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/physiology , Naphthoquinones/pharmacology , Proto-Oncogene Proteins c-akt/physiology , Apoptosis/drug effects , Cell Cycle/drug effects , Dose-Response Relationship, Drug , HL-60 Cells , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins c-bcl-2/analysis , Signal Transduction/physiology , Survivin , U937 Cells
9.
Oncotarget ; 5(14): 5523-34, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25015118

ABSTRACT

Cytolethal distending toxin (CDT) produced by Campylobacter jejuni is a genotoxin that induces cell-cycle arrest and apoptosis in mammalian cells. Recent studies have demonstrated that prostate cancer (PCa) cells can acquire radio-resistance when DOC-2/DAB2 interactive protein (DAB2IP) is downregulated. In this study, we showed that CDT could induce cell death in DAB2IP-deficient PCa cells. A combination of CDT and radiotherapy significantly elicited cell death in DAB2IP-deficient PCa cells by inhibiting the repair of ionizing radiation (IR)-induced DNA double-strand break (DSB) during G2/M arrest, which is triggered by ataxia telangiectasia mutated (ATM)-dependent DNA damage checkpoint responses. We also found that CDT administration significantly increased the efficacy of radiotherapy in a xenograft mouse model. These results indicate that CDT can be a potent therapeutic agent for radio-resistant PCa.


Subject(s)
Bacterial Toxins/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred BALB C , Prostatic Neoplasms/pathology , Radiation, Ionizing , Xenograft Model Antitumor Assays , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
10.
Oral Oncol ; 50(7): 662-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24735547

ABSTRACT

OBJECTIVES: PARP inhibitors (PARPi) may provide an opportunity to gain selective killing of tumor cells which have deficiencies in cellular DNA repair systems. We previously demonstrated linifanib (ABT-869), a multi-receptor tyrosine kinase inhibitor of VEGF and PDGF receptor families, radiosensitized Head and Neck Squamous Cell Carcinoma cells (HNSCC) via inhibiting STAT3 activation. Given that STAT3 can modulate DNA damage response (DDR) pathway, in this study, we evaluate the effects of linifanib to enhance cytotoxicity with the PARPi, veliparib (ABT-888), in HNSCC. MATERIALS AND METHODS: UMSCC-22A and UMSCC-22B cells were treated with linifanib (ABT-869) and veliparib (ABT-888). Cell viability, cytotoxicity, apoptosis induction, DNA single strand break (SSB) and double strand break (DSB) damages were examined by MTT assay, colony formation assay, flow cytometry and comet assay. In addition, the expression of DNA homologous recombination repair protein Rad51, γH2AX, a double strand break marker and cleaved PARP, an apoptotic cell death marker, were assessed using western immunoblotting. RESULTS: Combination treatment resulted in more cell growth inhibition, induction of apoptosis, DNA damages and double strand breaks, lower expression of Rad51, increase γH2AX expression and PARP cleavage. CONCLUSION: These data suggest the possibility of combining targeted therapeutic such as linifanib with veliparib to augment the inhibition of cell growth and apoptosis via synthetic lethality in HNSCC cells. Thus, it may provide a novel therapeutic strategy and improve efficacy and outcome in HNSCC.


Subject(s)
Benzimidazoles/pharmacology , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Indazoles/pharmacology , Phenylurea Compounds/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA Breaks/drug effects , Histones/metabolism , Humans , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Rad51 Recombinase/metabolism
11.
Oral Oncol ; 50(1): 19-26, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24269532

ABSTRACT

Tumor angiogenesis is a hallmark of advanced cancers and promotes invasion and metastasis. Over 90% of head and neck squamous cell carcinomas (HNSCC) express angiogenic factors such as vascular endothelial growth factor (VEGF). Several preclinical studies support the prognostic implications of angiogenic markers for HNSCC and currently this is an attractive treatment target in solid tumors. Since radiotherapy is one of the most commonly used treatments for HNSCC, it is imperative to identify the interactions between antiangiogenic therapy and radiotherapy, and to develop combination therapy to improve clinical outcome. The mechanisms between antiangiogenic agents and ionizing radiation are complicated and involve many interactions between the vasculature, tumor stroma and tumor cells. The proliferation and metastasis of tumor cells rely on angiogenesis/blood vessel formation. Rapid growing tumors will cause hypoxia, which up-regulates tumor cell survival factors, such as hypoxia-inducing factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF), giving rise to more tumor proliferation, angiogenesis and increased radioresistance. Thus, agents that target tumor vasculature and new tumor vessel formation can modulate the tumor microenvironment to improve tumor blood flow and oxygenation, leading to enhanced radiosensitivity. In this review, we discuss the mechanisms of how antiangiogenic therapies improve tumor response to radiation and data that support this combination strategy as a promising method for the treatment of HNSCC in the future.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/therapy , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Combined Modality Therapy , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Humans
12.
Oral Oncol ; 49(6): 591-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23490884

ABSTRACT

OBJECTIVES: Novel targeted therapeutic strategies to overcome radio-resistance of cancer cells traditionally treated with radiation may improve patient survival with the added benefit of reduced systemic toxicity. Herein, we tested the feasibility of Linifanib (ABT-869), a multi-receptor tyrosine kinase inhibitor of members of vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF) receptor families, on radio-sensitization of Head and Neck Squamous Cell Carcinoma (HNSCC). MATERIALS AND METHODS: UMSCC-22A and UMSCC-22B cells were treated with Linifanib and γ-radiation response was determined. Cell viability, cytotoxicity, apoptosis induction and cell cycle distribution were examined by MTT assay, colony formation assay and flow cytometry. In addition, expression of STAT3 and downstream signaling proteins were assessed using western immunoblotting. RESULTS: Treatment with Linifanib resulted in cell growth inhibition, G2/M cell cycle arrest, induction of cell death via apoptosis, reduced phosphorylation of STAT3, which has been linked to radio-resistance, lower expression of cyclin D1, survivin and increased PARP cleavage. In addition, Linifanib overcame the radio-resistance of the cell lines and significantly enhanced radiation-induced cytotoxicity (p<0.05). CONCLUSION: These data suggest the possibility of combining targeted therapeutic such as Linifanib with radiation to enhance inhibition of cell growth and apoptosis in HNSCC cells. Thus, it may provide a novel therapeutic strategy and improve efficacy of radiation against HNSCC in the future.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Head and Neck Neoplasms/radiotherapy , Indazoles/pharmacology , Phenylurea Compounds/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Head and Neck Neoplasms/pathology , Humans , STAT3 Transcription Factor/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...