Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Mol Ther Methods Clin Dev ; 32(1): 101191, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38352271

ABSTRACT

Despite the implementation of lifesaving newborn screening programs and a galactose-restricted diet, many patients with classic galactosemia develop long-term debilitating neurological deficits and primary ovarian insufficiency. Previously, we showed that the administration of human GALT mRNA predominantly expressed in the GalT gene-trapped mouse liver augmented the expression of hepatic GALT activity, which decreased not only galactose-1 phosphate (gal-1P) in the liver but also peripheral tissues. Since each peripheral tissue requires distinct methods to examine the biomarker and/or GALT effect, this highlights the necessity for alternative strategies to evaluate the overall impact of therapies. In this study, we established that whole-body galactose oxidation (WBGO) as a robust, noninvasive, and specific method to assess the in vivo pharmacokinetic and pharmacodynamic parameters of two experimental gene-based therapies that aimed to restore GALT activity in a mouse model of galactosemia. Although our results illustrated the long-lasting efficacy of AAVrh10-mediated GALT gene transfer, we found that GALT mRNA therapy that targets the liver predominantly is sufficient to sustain WBGO. The latter could have important implications in the design of novel targeted therapy to ensure optimal efficacy and safety.

2.
J Phys Chem Lett ; 14(49): 10920-10929, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38033191

ABSTRACT

Understanding and controlling carrier dynamics in two-dimensional (2D) van der Waals heterostructures through strain are crucial for their flexible applications. Here, femtosecond transient absorption spectroscopy is employed to elucidate the interlayer electron transfer and relaxation dynamics under external tensile strains in a WSe2/MoS2 heterostructure. The results show that a modest ∼1% tensile strain can significantly alter the lifetimes of electron transfer and nonradiative electron-hole recombination by >30%. Ab initio non-adiabatic molecular dynamics simulations suggest that tensile strain weakens the electron-phonon coupling, thereby suppressing the transfer and recombination dynamics. Theoretical predictions indicate that strain-induced energy difference increases along the electron transfer path could contribute to the prolongation of the transfer lifetime. A subpicosecond decay process, related to hot-electron cooling, remains almost unaffected by strain. This study demonstrates the potential of tuning interlayer carrier dynamics through external strains, offering insights into flexible optoelectronic device design with 2D materials.

3.
Front Med (Lausanne) ; 9: 799083, 2022.
Article in English | MEDLINE | ID: mdl-35814753

ABSTRACT

Background: Uveal melanoma (UM) is the most frequent primary intraocular tumor in adults. This study aims to develop a nomogram and an individualized web-based calculator to predict the overall survival (OS) of elderly patients with UM. Methods: Patients aged more than 60 years and diagnosed with UM were derived from the Surveillance, Epidemiology, and End Results (SEER) database during 2004-2015. The selected patients were randomly divided into training and validation cohorts. In the training cohort, the univariate and multivariate Cox analyses were carried out to determine the independent prognostic factors, and the predictors were integrated to establish a nomogram for predicting the 1-, 2-, and 3-year OS of elderly patients with UM. The discrimination of the nomogram was validated by receiver operating characteristic (ROC) curves and the area under the curve (AUC). The clinical practicability and accuracy of the nomogram were evaluated by the calibration curves and decision curve analysis (DCA). A web-based survival calculator was then constructed using a fitted survival prediction model (https://yuexinupup.shinyapps.io/DynNomapp/). Results: A total of 1,427 patients with UM were included in this study. Age, T stage, N stage, M stage, marital status, sex, and radiotherapy (RT) were identified as independent prognostic factors. Based on the abovementioned factors, the nomogram was then constructed. The AUC values of the nomogram predicting 1-, 2-, and 3-year OS were 0.841, 0.801, and 0.768 in the training cohort, and 0.745, 0.717, and 0.710 in the validation cohort, respectively. The calibration curves and DCA also indicated the good performance of the predictive model. Conclusion: This study established and validated a novel nomogram risk stratification model and a web-based survival rate calculator that can dynamically predict the long-term OS for elderly patients with UM.

4.
Nat Commun ; 11(1): 5339, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087718

ABSTRACT

Propionic acidemia/aciduria (PA) is an ultra-rare, life-threatening, inherited metabolic disorder caused by deficiency of the mitochondrial enzyme, propionyl-CoA carboxylase (PCC) composed of six alpha (PCCA) and six beta (PCCB) subunits. We herein report an enzyme replacement approach to treat PA using a combination of two messenger RNAs (mRNAs) (dual mRNAs) encoding both human PCCA (hPCCA) and PCCB (hPCCB) encapsulated in biodegradable lipid nanoparticles (LNPs) to produce functional PCC enzyme in liver. In patient fibroblasts, dual mRNAs encoded proteins localize in mitochondria and produce higher PCC enzyme activity vs. single (PCCA or PCCB) mRNA alone. In a hypomorphic murine model of PA, dual mRNAs normalize ammonia similarly to carglumic acid, a drug approved in Europe for the treatment of hyperammonemia due to PA. Dual mRNAs additionally restore functional PCC enzyme in liver and thus reduce primary disease-associated toxins in a dose-dependent manner in long-term 3- and 6-month repeat-dose studies in PA mice. Dual mRNAs are well-tolerated in these studies with no adverse findings. These studies demonstrate the potential of mRNA technology to chronically administer multiple mRNAs to produce large complex enzymes, with applicability to other genetic disorders.


Subject(s)
Enzyme Replacement Therapy/methods , Propionic Acidemia/therapy , RNA, Messenger/therapeutic use , Animals , Disease Models, Animal , Glutamates/therapeutic use , Humans , Kinetics , Lipids/chemistry , Liver/enzymology , Methylmalonyl-CoA Decarboxylase/chemistry , Methylmalonyl-CoA Decarboxylase/genetics , Methylmalonyl-CoA Decarboxylase/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/enzymology , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Propionic Acidemia/genetics , Propionic Acidemia/metabolism , Protein Subunits/chemistry , Protein Subunits/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics
5.
J Thorac Dis ; 11(11): 4551-4561, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31903244

ABSTRACT

BACKGROUND: To investigate the relationship between pulmonary function and brachial-ankle pulse wave velocity (baPWV). METHODS: A cross-sectional study was conducted. A total of 11,388 people with complete pulmonary function test and baPWV data and who participated in both the health examination of the Kailuan Occupational Disease Prevention and Treatment Center in 2014-2016 and the health checkup of the Kailuan Group in 2012 and 2014 were selected as subjects. The study population was divided into four groups by forced vital capacity (FVC) quartiles (group 1: FVC <3.50 L; group 2: 3.50 L ≤ FVC <3.96 L; group 3: 3.96 L ≤ FVC <4.47 L; group 4: FVC ≥4.47 L) and divided into four groups by forced expiratory volume in one second (FEV1) quartile (group 1: FEV1 <3.15 L; group 2: 3.15 L ≤ FVC <3.61 L; group 3: 3.61 L ≤ FVC <4.08 L; group 4: FVC ≥4.08 L). Linear regression analysis and multivariate logistic regression were used to analyze the effects of pulmonary function on baPWV. RESULTS: When grouped by FVC, the baPWV of the first group was significantly higher than the other groups. Similarly, the incidence of arteriosclerosis in the first group was significantly higher than the other groups. When grouped by FEV1, the baPWV of the first group was significantly higher than the other groups. The incidence of arteriosclerosis was also significantly higher in the first group than the other groups. After correcting for other confounding factors using linear regression, it was found that the effects of FVC and FEV1 on the study subject's baPWV were -23.84 and -24.65 L, respectively. Multivariate logistic regression analysis showed that when grouped by FVC quartile, the risk of arteriosclerosis was increased by 34% in group 1 compared with group 4 (OR: 1.34, 95% CI: 1.17-1.52); the risk of arteriosclerosis was increased by 16% in group 2 compared with group 4 (OR: 1.16, 95% CI: 1.03-1.31). When grouped by the FEV1 quartile, the risk of arteriosclerosis was increased by 25% in group 1 compared with group 4 (OR: 1.25, 95% CI: 1.10-1.42). CONCLUSIONS: Decreased pulmonary function is negatively correlated with baPWV and is an independent risk factor for arteriosclerosis.

6.
Radiat Res ; 188(2): 191-203, 2017 08.
Article in English | MEDLINE | ID: mdl-28613990

ABSTRACT

Deep-space travel presents risks of exposure to ionizing radiation composed of a spectrum of low-fluence protons (1H) and high-charge and energy (HZE) iron nuclei (e.g., 56Fe). When exposed to galactic cosmic rays, each cell in the body may be traversed by 1H every 3-4 days and HZE nuclei every 3-4 months. The effects of low-dose sequential fractionated 1H or HZE on the heart are unknown. In this animal model of simulated ionizing radiation, middle-aged (8-9 months old) male C57BL/6NT mice were exposed to radiation as follows: group 1, nonirradiated controls; group 2, three fractionated doses of 17 cGy 1H every other day (1H × 3); group 3, three fractionated doses of 17 cGy 1H every other day followed by a single low dose of 15 cGy 56Fe two days after the final 1H dose (1H × 3 + 56Fe); and group 4, a single low dose of 15 cGy 56Fe followed (after 2 days) by three fractionated doses of 17 cGy 1H every other day (56Fe + 1H × 3). A subgroup of mice from each group underwent myocardial infarction (MI) surgery at 28 days postirradiation. Cardiac structure and function were assessed in all animals at days 7, 14 and 28 after MI surgery was performed. Compared to the control animals, the treatments that groups 2 and 3 received did not induce negative effects on cardiac function or structure. However, compared to all other groups, the animals in group 4, showed depressed left ventricular (LV) functions at 1 month with concomitant enhancement in cardiac fibrosis and induction of cardiac hypertrophy signaling at 3 months. In the irradiated and MI surgery groups compared to the control group, the treatments received by groups 2 and 4 did not induce negative effects at 1 month postirradiation and MI surgery. However, in group 3 after MI surgery, there was a 24% increase in mortality, significant decreases in LV function and a 35% increase in post-infarction size. These changes were associated with significant decreases in the angiogenic and cell survival signaling pathways. These data suggest that fractionated doses of radiation induces cellular and molecular changes that result in depressed heart functions both under basal conditions and particularly after myocardial infarction.


Subject(s)
Dose Fractionation, Radiation , Heart/physiology , Heart/radiation effects , Iron/adverse effects , Protons/adverse effects , Animals , Cosmic Radiation/adverse effects , Dose-Response Relationship, Radiation , Heart/physiopathology , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/etiology , Myocardial Infarction/physiopathology
12.
Front Oncol ; 5: 231, 2015.
Article in English | MEDLINE | ID: mdl-26528440

ABSTRACT

Exposure of individuals to ionizing radiation (IR), as in the case of astronauts exploring space or radiotherapy cancer patients, increases their risk of developing secondary cancers and other health-related problems. Bone marrow (BM), the site in the body where hematopoietic stem cell (HSC) self-renewal and differentiation to mature blood cells occurs, is extremely sensitive to low-dose IR, including irradiation by high-charge and high-energy particles. Low-dose IR induces DNA damage and persistent oxidative stress in the BM hematopoietic cells. Inefficient DNA repair processes in HSC and early hematopoietic progenitors can lead to an accumulation of mutations whereas long-lasting oxidative stress can impair hematopoiesis itself, thereby causing long-term damage to hematopoietic cells in the BM niche. We report here that low-dose (1)H- and (56)Fe-IR significantly decreased the hematopoietic early and late multipotent progenitor (E- and L-MPP, respectively) cell numbers in mouse BM over a period of up to 10 months after exposure. Both (1)H- and (56)Fe-IR increased the expression of pluripotent stem cell markers Sox2, Nanog, and Oct4 in L-MPPs and 10 months post-IR exposure. We postulate that low doses of (1)H- and (56)Fe-IR may induce endogenous cellular reprogramming of BM hematopoietic progenitor cells to assume a more primitive pluripotent phenotype and that IR-induced oxidative DNA damage may lead to mutations in these BM progenitors. This could then be propagated to successive cell lineages. Persistent impairment of BM progenitor cell populations can disrupt hematopoietic homeostasis and lead to hematologic disorders, and these findings warrant further mechanistic studies into the effects of low-dose IR on the functional capacity of BM-derived hematopoietic cells including their self-renewal and pluripotency.

13.
Am J Physiol Heart Circ Physiol ; 309(11): H1947-63, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26408534

ABSTRACT

There are 160,000 cancer patients worldwide treated with particle radiotherapy (RT). With the advent of proton, and high (H) charge (Z) and energy (E) HZE ionizing particle RT, the cardiovascular diseases risk estimates are uncertain. In addition, future deep space exploratory-type missions will expose humans to unknown but low doses of particle irradiation (IR). We examined molecular responses using transcriptome profiling in left ventricular murine cardiomyocytes isolated from mice that were exposed to 90 cGy, 1 GeV proton ((1)H) and 15 cGy, 1 GeV/nucleon iron ((56)Fe) over 28 days after exposure. Unsupervised clustering analysis of gene expression segregated samples according to the IR response and time after exposure, with (56)Fe-IR showing the greatest level of gene modulation. (1)H-IR showed little differential transcript modulation. Network analysis categorized the major differentially expressed genes into cell cycle, oxidative responses, and transcriptional regulation functional groups. Transcriptional networks identified key nodes regulating expression. Validation of the signal transduction network by protein analysis and gel shift assay showed that particle IR clearly regulates a long-lived signaling mechanism for ERK1/2, p38 MAPK signaling and identified NFATc4, GATA4, STAT3, and NF-κB as regulators of the response at specific time points. These data suggest that the molecular responses and gene expression to (56)Fe-IR in cardiomyocytes are unique and long-lasting. Our study may have significant implications for the efforts of National Aeronautics and Space Administration to develop heart disease risk estimates for astronauts and for patients receiving conventional and particle RT via identification of specific HZE-IR molecular markers.


Subject(s)
Gene Regulatory Networks/radiation effects , Iron Radioisotopes/toxicity , Myocytes, Cardiac/radiation effects , Radiotherapy, High-Energy/adverse effects , Signal Transduction/radiation effects , Animals , Cells, Cultured , Cluster Analysis , Enzyme Activation , Fibrosis , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/radiation effects , Male , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Risk Assessment , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Time Factors , Transcription, Genetic/radiation effects , Transcriptome/radiation effects , Whole-Body Irradiation
14.
Stem Cells Int ; 2015: 496512, 2015.
Article in English | MEDLINE | ID: mdl-26074973

ABSTRACT

Bone-marrow- (BM-) derived endothelial progenitor cells (EPCs) are critical for endothelial cell maintenance and repair. During future space exploration missions astronauts will be exposed to space irradiation (IR) composed of a spectrum of low-fluence protons ((1)H) and high charge and energy (HZE) nuclei (e.g., iron-(56)Fe) for extended time. How the space-type IR affects BM-EPCs is limited. In media transfer experiments in vitro we studied nontargeted effects induced by (1)H- and (56)Fe-IR conditioned medium (CM), which showed significant increase in the number of p-H2AX foci in nonirradiated EPCs between 2 and 24 h. A 2-15-fold increase in the levels of various cytokines and chemokines was observed in both types of IR-CM at 24 h. Ex vivo analysis of BM-EPCs from single, low-dose, full-body (1)H- and (56)Fe-IR mice demonstrated a cyclical (early 5-24 h and delayed 28 days) increase in apoptosis. This early increase in BM-EPC apoptosis may be the effect of direct IR exposure, whereas late increase in apoptosis could be a result of nontargeted effects (NTE) in the cells that were not traversed by IR directly. Identifying the role of specific cytokines responsible for IR-induced NTE and inhibiting such NTE may prevent long-term and cyclical loss of stem and progenitors cells in the BM milieu.

15.
FASEB J ; 29(4): 1208-19, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25466901

ABSTRACT

We have previously shown that TNF-tumor necrosis factor receptor-2/p75 (TNFR2/p75) signaling plays a critical role in ischemia-induced neovascularization in skeletal muscle and heart tissues. To determine the role of TNF-TNFR2/p75 signaling in ischemia-induced inflammation and muscle regeneration, we subjected wild-type (WT) and TNFR2/p75 knockout (p75KO) mice to hind limb ischemia (HLI) surgery. Ischemia induced significant and long-lasting inflammation associated with considerable decrease in satellite-cell activation in p75KO muscle tissue up to 10 d after HLI surgery. To determine the possible additive negative roles of tissue aging and the absence of TNFR2/p75, either in the tissue or in the bone marrow (BM), we generated 2 chimeric BM transplantation (BMT) models where both young green fluorescent protein (GFP)-positive p75KO and WT BM-derived cells were transplanted into adult p75KO mice. HLI surgery was performed 1 mo after BMT, after confirming complete engraftment of the recipient BM with GFP donor cells. In adult p75KO with the WT-BMT, proliferative (Ki67(+)) cells were detected only by d 28 and were exclusively GFP(+), suggesting significantly delayed contribution of young WT-BM cell to adult p75KO ischemic tissue recovery. No GFP(+) young p75KO BM cells survived in adult p75KO tissue, signifying the additive negative roles of tissue aging combined with decreased/absent TNFR2/p75 signaling in postischemic recovery.


Subject(s)
Ischemia/pathology , Ischemia/physiopathology , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Satellite Cells, Skeletal Muscle/pathology , Satellite Cells, Skeletal Muscle/physiology , Animals , Apoptosis , Bone Marrow Transplantation , Cell Proliferation , Disease Models, Animal , Green Fluorescent Proteins/genetics , Hindlimb/blood supply , Inflammation/etiology , Inflammation/pathology , Inflammation/physiopathology , Ischemia/therapy , Macrophages/pathology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neovascularization, Physiologic , Neutrophil Infiltration , Receptors, Tumor Necrosis Factor, Type II/metabolism , Recombinant Proteins/genetics , Regeneration , Signal Transduction
16.
PLoS One ; 9(10): e110269, 2014.
Article in English | MEDLINE | ID: mdl-25337914

ABSTRACT

Previous epidemiologic data demonstrate that cardiovascular (CV) morbidity and mortality may occur decades after ionizing radiation exposure. With increased use of proton and carbon ion radiotherapy and concerns about space radiation exposures to astronauts on future long-duration exploration-type missions, the long-term effects and risks of low-dose charged particle irradiation on the CV system must be better appreciated. Here we report on the long-term effects of whole-body proton ((1)H; 0.5 Gy, 1 GeV) and iron ion ((56)Fe; 0.15 Gy, 1GeV/nucleon) irradiation with and without an acute myocardial ischemia (AMI) event in mice. We show that cardiac function of proton-irradiated mice initially improves at 1 month but declines by 10 months post-irradiation. In AMI-induced mice, prior proton irradiation improved cardiac function restoration and enhanced cardiac remodeling. This was associated with increased pro-survival gene expression in cardiac tissues. In contrast, cardiac function was significantly declined in (56)Fe ion-irradiated mice at 1 and 3 months but recovered at 10 months. In addition, (56)Fe ion-irradiation led to poorer cardiac function and more adverse remodeling in AMI-induced mice, and was associated with decreased angiogenesis and pro-survival factors in cardiac tissues at any time point examined up to 10 months. This is the first study reporting CV effects following low dose proton and iron ion irradiation during normal aging and post-AMI. Understanding the biological effects of charged particle radiation qualities on the CV system is necessary both for the mitigation of space exploration CV risks and for understanding of long-term CV effects following charged particle radiotherapy.


Subject(s)
Heart/radiation effects , Iron Radioisotopes/adverse effects , Myocardial Ischemia/physiopathology , Protons/adverse effects , Whole-Body Irradiation/adverse effects , Acute Disease , Animals , Astronauts , Disease Models, Animal , Heart/physiopathology , Heart Function Tests , Humans , Male , Mice , Mice, Inbred C57BL , Myocardial Ischemia/metabolism , Radiation Dosage , Radiation, Ionizing , Risk , Space Flight , Time Factors
17.
Curr Pharm Des ; 20(30): 4911-9, 2014.
Article in English | MEDLINE | ID: mdl-24898245

ABSTRACT

A significant milestone in the treatment of breast cancer is the identification of the HER2 receptor as a drug target for cancer therapies. Trastuzumab (Herceptin), a monoclonal antibody that blocks the HER2 receptor, is among the first of such drugs approved by the US Food and Drug Administration for targeted cancer therapy. Clinical studies have shown that Trastuzumab significantly improves the overall survival of breast cancer patients. However, an unforeseen significant side-effect of cardiotoxicity manifested as left ventricular dysfunction and heart failure. Concurrent studies have demonstrated the essential role of the HER2 receptor in cardiac development and maintaining the physiological function of an adult heart. The HER2 receptor, therefore, has become a critical link between the oncology and cardiology fields. In addition to Trastuzumab, new drugs targeting the HER2 receptor, such as Lapatinib, Pertuzumab and Afatinib, are either approved or being evaluated in clinical trials for cancer therapy. With the concern of cardiotoxicity caused by HER2 inhibition, it becomes clear that new therapeutic strategies for preventing such cardiac side effects need to be developed. It is the intent of this paper to review the potential cardiac impact of anti-HER2 cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Heart/drug effects , Receptor, ErbB-2/antagonists & inhibitors , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Humans
18.
J Biol Chem ; 289(20): 14178-93, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24711449

ABSTRACT

TNF-α, a pro-inflammatory cytokine, is highly expressed after being irradiated (IR) and is implicated in mediating radiobiological bystander responses (RBRs). Little is known about specific TNF receptors in regulating TNF-induced RBR in bone marrow-derived endothelial progenitor cells (BM-EPCs). Full body γ-IR WT BM-EPCs showed a biphasic response: slow decay of p-H2AX foci during the initial 24 h and increase between 24 h and 7 days post-IR, indicating a significant RBR in BM-EPCs in vivo. Individual TNF receptor (TNFR) signaling in RBR was evaluated in BM-EPCs from WT, TNFR1/p55KO, and TNFR2/p75KO mice, in vitro. Compared with WT, early RBR (1-5 h) were inhibited in p55KO and p75KO EPCs, whereas delayed RBR (3-5 days) were amplified in p55KO EPCs, suggesting a possible role for TNFR2/p75 signaling in delayed RBR. Neutralizing TNF in γ-IR conditioned media (CM) of WT and p55KO BM-EPCs largely abolished RBR in both cell types. ELISA protein profiling of WT and p55KO EPC γ-IR-CM over 5 days showed significant increases in several pro-inflammatory cytokines, including TNF-α, IL-1α (Interleukin-1 alpha), RANTES (regulated on activation, normal T cell expressed and secreted), and MCP-1. In vitro treatments with murine recombinant (rm) TNF-α and rmIL-1α, but not rmMCP-1 or rmRANTES, increased the formation of p-H2AX foci in nonirradiated p55KO EPCs. We conclude that TNF-TNFR2 signaling may induce RBR in naïve BM-EPCs and that blocking TNF-TNFR2 signaling may prevent delayed RBR in BM-EPCs, conceivably, in bone marrow milieu in general.


Subject(s)
Bone Marrow Cells/cytology , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Animals , Bystander Effect/drug effects , Bystander Effect/radiation effects , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/radiation effects , Gene Knockout Techniques , Histones/metabolism , Insulin-Like Growth Factor I/metabolism , Interleukin-1alpha/pharmacology , Ligands , Mice , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction/drug effects , Signal Transduction/radiation effects , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
19.
PLoS One ; 9(3): e92373, 2014.
Article in English | MEDLINE | ID: mdl-24664144

ABSTRACT

Tumor necrosis factor-alpha (TNF) binds to two receptors: TNFR1/p55-cytotoxic and TNFR2/p75-pro-survival. We have shown that tumor growth in p75 knockout (KO) mice was decreased more than 2-fold in Lewis lung carcinoma (LLCs). We hypothesized that selective blocking of TNFR2/p75 LLCs may sensitize them to TNF-induced apoptosis and affect the tumor growth. We implanted intact and p75 knockdown (KD)-LLCs (>90%, using shRNA) into wild type (WT) mice flanks. On day 8 post-inoculation, recombinant murine (rm) TNF-α (12.5 ng/gr of body weight) or saline was injected twice daily for 6 days. Tumor volumes (tV) were measured daily and tumor weights (tW) on day 15, when study was terminated due to large tumors in LLC+TNF group. Tubular bones, spleens and peripheral blood (PB) were examined to determine possible TNF toxicity. There was no significant difference in tV or tW between LLC minus (-) TNF and p75KD/LLC-TNF tumors. Compared to 3 control groups, p75KD/LLC+TNF showed >2-5-fold decreases in tV (p<0.001) and tW (p<0.0001). There was no difference in tV or tW end of study vs. before injections in p75KD/LLC+TNF group. In 3 other groups tV and tW were increased 2.7-4.5-fold (p<0.01, p<0.0002 and p<0.0001). Pathological examination revealed that 1/3 of p75KD/LLC+rmTNF tumors were 100% necrotic, the remaining revealed 40-60% necrosis. No toxicity was detected in bone marrow, spleen and peripheral blood. We concluded that blocking TNFR2/p75 in LLCs combined with intra-tumoral rmTNF injections inhibit LLC tumor growth. This could represent a novel and effective therapy against lung neoplasms and a new paradigm in cancer therapeutics.


Subject(s)
Carcinoma, Lewis Lung/pathology , Cell Transformation, Neoplastic , Gene Knockdown Techniques , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Tumor Necrosis Factor-alpha/pharmacology , Animals , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/genetics , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Male , Mice , Models, Biological , Necrosis/chemically induced , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...