Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Cell ; 173(1): 276-276.e1, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29570996

ABSTRACT

CGAS responds to cytosolic DNA by initiating a STING-dependent response that ultimately engages innate immune effectors to ensure the preservation of organismal homeostasis.


Subject(s)
Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Cellular Microenvironment , Humans , Immunity, Innate , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , NF-kappaB-Inducing Kinase
2.
EMBO J ; 42(7): e111961, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36574362

ABSTRACT

Cytosolic DNA promotes inflammatory responses upon detection by the cyclic GMP-AMP (cGAMP) synthase (cGAS). It has been suggested that cGAS downregulation is an immune escape strategy harnessed by tumor cells. Here, we used glioblastoma cells that show undetectable cGAS levels to address if alternative DNA detection pathways can promote pro-inflammatory signaling. We show that the DNA-PK DNA repair complex (i) drives cGAS-independent IRF3-mediated type I Interferon responses and (ii) that its catalytic activity is required for cGAS-dependent cGAMP production and optimal downstream signaling. We further show that the cooperation between DNA-PK and cGAS favors the expression of chemokines that promote macrophage recruitment in the tumor microenvironment in a glioblastoma model, a process that impairs early tumorigenesis but correlates with poor outcome in glioblastoma patients. Thus, our study supports that cGAS-dependent signaling is acquired during tumorigenesis and that cGAS and DNA-PK activities should be analyzed concertedly to predict the impact of strategies aiming to boost tumor immunogenicity.


Subject(s)
DNA-Activated Protein Kinase , Glioblastoma , Nucleotidyltransferases , Humans , Carcinogenesis , DNA/metabolism , DNA Damage , DNA Repair , Glioblastoma/genetics , Immunity, Innate , Inflammation , Nucleotidyltransferases/metabolism , Tumor Microenvironment , DNA-Activated Protein Kinase/metabolism
3.
Trends Immunol ; 44(4): 245-247, 2023 04.
Article in English | MEDLINE | ID: mdl-36933950

ABSTRACT

Mitochondrial outer membrane permeabilization (MOMP) is crucial for the cytosolic accumulation of mitochondrial DNA (mtDNA) species that are required to jumpstart innate and adaptive immunity. Recent data reported by Ghosh et al. suggest that tumor protein p53 regulates MOMP-dependent type I interferon (IFN) production, not only via MOMP-promoting effects, but also by directing mtDNA-degrading exonucleases to proteasomal processing.


Subject(s)
Mitochondria , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Mitochondria/metabolism , Apoptosis , DNA, Mitochondrial/genetics
4.
Semin Immunol ; 52: 101474, 2021 02.
Article in English | MEDLINE | ID: mdl-33741223

ABSTRACT

The ability of radiotherapy to enhance antigenicity and adjuvanticity of an irradiated tumor has stimulated the interest for its combination with immuno-oncology agents. However, radiotherapy often generates multiple layers of host responses which likely depends on the tumor biology, the immune cell infiltration and the induction of immunosuppressive signals post radiotherapy. Consequently, translation of preclinical findings to the clinic is more convoluted than anticipated which underscore the need to decipher molecular and cellular mechanisms elicited by radiotherapy. Here we review pro-inflammatory and immunosuppressive mechanisms triggered by radiotherapy that impact the outcome of antigen specific T cell killing and discuss how radiation-induced immunostimulatory mechanisms can be exploited to reactivate the host's immune system, especially in the context of immunotherapy.


Subject(s)
Neoplasms , Humans , Immunity , Immunotherapy , Neoplasms/drug therapy , Neoplasms/radiotherapy , T-Lymphocytes
5.
Cancer Immunol Immunother ; 71(4): 839-850, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34435232

ABSTRACT

The expression of immune-related genes in cancer cells can alter the anti-tumor immune response and thereby impact patient outcomes. Radiotherapy has been shown to modulate immune-related genes dependent on the fractionation regimen. To identify long-term changes in gene expression after irradiation, PC3 (p53 deleted) and LNCaP (p53 wildtype) prostate cancer cells were irradiated with either a single dose (SD, 10 Gy) or a fractionated regimen (MF) of 10 fractions (1 Gy per fraction). Whole human genome arrays were used to determine gene expression at 24 h and 2 months after irradiation. Immune pathway activation was analyzed with Ingenuity Pathway Analysis software. Additionally, 3D colony formation assays and T-cell cytotoxicity assays were performed. LNCaP had a higher basal expression of immunogenic genes and was more efficiently killed by cytotoxic T-cells compared to PC3. In both cell lines, MF irradiation resulted in an increase in multiple immune-related genes immediately after irradiation, while at 2 months, SD irradiation had a more pronounced effect on radiation-induced gene expression. Both immunogenic and immunosuppressive genes were upregulated in the long term in PC3 cells by a 10 Gy SD irradiation but not in LNCaP. T-cell-mediated cytotoxicity was significantly increased in 10 Gy SD PC3 cells compared to the unirradiated control and could be further enhanced by treatment with immune checkpoint inhibitors. Irradiation impacts the expression of immune-related genes in cancer cells in a fractionation-dependent manner. Understanding and targeting these changes may be a promising strategy for primary prostate cancer and recurrent tumors.


Subject(s)
Neoplasm Recurrence, Local , Prostatic Neoplasms , Apoptosis , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/radiotherapy
6.
J Transl Med ; 19(1): 336, 2021 08 07.
Article in English | MEDLINE | ID: mdl-34364390

ABSTRACT

BACKGROUND: Radiation therapy is integral to effective thoracic cancer treatments, but its application is limited by sensitivity of critical organs such as the heart. The impacts of acute radiation-induced damage and its chronic effects on normal heart cells are highly relevant in radiotherapy with increasing lifespans of patients. Biomarkers for normal tissue damage after radiation exposure, whether accidental or therapeutic, are being studied as indicators of both acute and delayed effects. Recent research has highlighted the potential importance of RNAs, including messenger RNAs (mRNAs), microRNAs (miRNAs), and long non-coding RNAs (lncRNAs) as biomarkers to assess radiation damage. Understanding changes in mRNA and non-coding RNA expression will elucidate biological pathway changes after radiation. METHODS: To identify significant expression changes in mRNAs, lncRNAs, and miRNAs, we performed whole transcriptome microarray analysis of mouse heart tissue at 48 h after whole-body irradiation with 1, 2, 4, 8, and 12 Gray (Gy). We also validated changes in specific lncRNAs through RT-qPCR. Ingenuity Pathway Analysis (IPA) was used to identify pathways associated with gene expression changes. RESULTS: We observed sustained increases in lncRNAs and mRNAs, across all doses of radiation. Alas2, Aplnr, and Cxc3r1 were the most significantly downregulated mRNAs across all doses. Among the significantly upregulated mRNAs were cell-cycle arrest biomarkers Gdf15, Cdkn1a, and Ckap2. Additionally, IPA identified significant changes in gene expression relevant to senescence, apoptosis, hemoglobin synthesis, inflammation, and metabolism. LncRNAs Abhd11os, Pvt1, Trp53cor1, and Dino showed increased expression with increasing doses of radiation. We did not observe any miRNAs with sustained up- or downregulation across all doses, but miR-149-3p, miR-6538, miR-8101, miR-7118-5p, miR-211-3p, and miR-3960 were significantly upregulated after 12 Gy. CONCLUSIONS: Radiation-induced RNA expression changes may be predictive of normal tissue toxicities and may indicate targetable pathways for radiation countermeasure development and improved radiotherapy treatment plans.


Subject(s)
MicroRNAs , RNA, Long Noncoding , 5-Aminolevulinate Synthetase , Animals , Gene Regulatory Networks , Humans , Mice , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Whole-Body Irradiation
7.
Trends Immunol ; 39(6): 435-437, 2018 06.
Article in English | MEDLINE | ID: mdl-29598848

ABSTRACT

Tumors that elude infiltration by CD8+ T lymphocytes are particularly resistant to multiple forms of treatment, including immune checkpoint blockade. Stromal transforming growth factor (TGF)-ß appears to play a key role in this process, potentially constituting a target for novel combinatorial regimens tackling immune-excluded neoplasms.


Subject(s)
Immune Evasion , Transforming Growth Factor beta , CD8-Positive T-Lymphocytes , Colonic Neoplasms , Humans , Transforming Growth Factors
8.
Trends Immunol ; 39(8): 644-655, 2018 08.
Article in English | MEDLINE | ID: mdl-30001871

ABSTRACT

Radiotherapy has been used for more than a hundred years as a local tumor treatment. The occurrence of systemic antitumor effects manifesting as regression of tumors outside of the irradiated field (abscopal effect) was occasionally observed but deemed too rare and unpredictable to be a therapeutic goal. This has changed with the advent of immunotherapy. Remarkable systemic effects have been observed in patients receiving radiotherapy to control tumors that were progressing during immune checkpoint blockade, stimulating interest in using radiation to overcome primary and acquired cancer resistance to immunotherapy. Here, we review the immunological mechanisms that are responsible for the ability of focal radiation to promote antitumor T cell responses that mediate tumor rejection and, in some cases, result in systemic effects.


Subject(s)
Immunotherapy/methods , Neoplasms/radiotherapy , Radiotherapy/methods , T-Lymphocytes/immunology , Animals , Apoptosis , Combined Modality Therapy , Humans , Lymphocyte Activation , Neoplasm Metastasis , Tumor Burden
9.
Immunol Rev ; 280(1): 220-230, 2017 11.
Article in English | MEDLINE | ID: mdl-29027232

ABSTRACT

Ionizing irradiation has been extensively employed for the clinical management of solid tumors, with therapeutic or palliative intents, for decades. Until recently, radiation therapy (RT) was believed to mediate antineoplastic activity mostly (if not only) as a consequence of cancer cell-intrinsic effects. Indeed, the macromolecular damage imposed to malignant cells by RT initiates one or multiple signal transduction cascades that drive a permanent proliferative arrest (cellular senescence) or regulated cell death. Both these phenomena show a rather linear dose-response correlation. However, RT also mediates consistent immunological activity, not only as an "on-target effect" originating within irradiated cancer cells, but also as an "off-target effect" depending on the interaction between RT and stromal, endothelial, and immune components of the tumor microenvironment. Interestingly, the immunological activity of RT does not exhibit linear dose-response correlation. Here, we discuss the mechanisms whereby RT alters the capacity of the immune system to recognize and eliminate irradiated cancer cells, either as an "on-target" or as on "off-target" effect. In particular, we discuss the antagonism between the immunostimulatory and immunosuppressive effects of RT as we delineate combinatorial strategies to boost the former at the expenses of the latter.


Subject(s)
Cell Death , Cytotoxicity, Immunologic , Immunity , Neoplasms/radiotherapy , Animals , Antigens, Neoplasm/immunology , Autophagy , Combined Modality Therapy , Humans , Neoplasms/immunology , Radiation, Ionizing , Signal Transduction , Tumor Microenvironment
10.
Cancer Treat Res ; 180: 281-296, 2020.
Article in English | MEDLINE | ID: mdl-32215874

ABSTRACT

Immunogenic cell death (ICD) is a particular form of cell death that can initiate adaptive immunity against antigens expressed by dying cells in the absence of exogenous adjuvants. This implies that cells undergoing ICD not only express antigens that are not covered by thymic tolerance, but also deliver adjuvant-like signals that enable the recruitment and maturation of antigen-presenting cells toward an immunostimulatory phenotype, culminating with robust cross-priming of antigen-specific CD8+ T cells. Such damage-associated molecular patterns (DAMPs), which encompass cellular proteins, small metabolites and cytokines, are emitted in a spatiotemporally defined manner in the context of failing adaptation to stress. Radiation therapy (RT) is a bona fide inducer of ICD, at least when employed according to specific doses and fractionation schedules. Here, we discuss the mechanisms whereby DAMPs emitted by cancer cells undergoing RT-driven ICD alter the functional configuration of the tumor microenvironment.


Subject(s)
Alarmins , Immunogenic Cell Death , Neoplasms/immunology , Tumor Microenvironment/immunology , CD8-Positive T-Lymphocytes , Cytokines , Humans
11.
Semin Cancer Biol ; 52(Pt 2): 125-134, 2018 10.
Article in English | MEDLINE | ID: mdl-29258856

ABSTRACT

Over the past few years, multiple immune checkpoint blockers (ICBs) have achieved unprecedented clinical success and have been approved by regulatory agencies for the treatment of an increasing number of malignancies. However, only a limited fraction of patients responds to ICBs employed as a standalone intervention, calling for the development of combinatorial regimens. Radiation therapy (RT) stands out as a very promising candidate for this purpose. Indeed, RT mediates antineoplastic effects not only by cytotoxic and cytostatic mechanisms, but also by modulating immunological functions, both locally (within the irradiated field) and systemically. As combinatorial regimens involving RT and ICBs are being developed and clinically tested at an accelerating pace, it is paramount to identify biomarkers that reliably predict the likelihood of individual patients to respond. Here, we discuss emerging biomarkers that may potentially predict the response of cancer patients to RT plus ICBs.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/immunology , Neoplasms/radiotherapy , Neoplasms/therapy , Animals , Antineoplastic Agents/immunology , Humans , Neoplasms/immunology , Radiotherapy/methods
12.
Biomarkers ; 23(7): 689-703, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29799276

ABSTRACT

CONTEXT: Accidental exposure to life-threatening radiation in a nuclear event is a major concern; there is an enormous need for identifying biomarkers for radiation biodosimetry to triage populations and treat critically exposed individuals. OBJECTIVE: To identify dose-differentiating miRNA signatures from whole blood samples of whole body irradiated mice. METHODS: Mice were whole body irradiated with X-rays (2 Gy-15 Gy); blood was collected at various time-points post-exposure; total RNA was isolated; miRNA microarrays were performed; miRNAs differentially expressed in irradiated vs. unirradiated controls were identified; feature extraction and classification models were applied to predict dose-differentiating miRNA signature. RESULTS: We observed a time and dose responsive alteration in the expression levels of miRNAs. Maximum number of miRNAs were altered at 24-h and 48-h time-points post-irradiation. A 23-miRNA signature was identified using feature selection algorithms and classifier models. An inverse correlation in the expression level changes of miR-17 members, and their targets were observed in whole body irradiated mice and non-human primates. CONCLUSION: Whole blood-based miRNA expression signatures might be used for predicting radiation exposures in a mass casualty nuclear incident.


Subject(s)
MicroRNAs/blood , Microarray Analysis/methods , Whole-Body Irradiation/adverse effects , Animals , Dose-Response Relationship, Radiation , Gene Expression Profiling , Mice , Radiation Exposure/adverse effects , Time Factors
13.
J Transl Med ; 19(1): 255, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112189

Subject(s)
Neoplasms , Radiobiology , Humans
14.
Trends Cancer ; 10(3): 177-179, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38355355

ABSTRACT

Mammalian cells react to the accumulation of double-stranded (ds)DNA in the cytosol by secreting antiviral and proinflammatory cytokines, notably type I interferon (IFN). Recent data reported by Tani et al. demonstrate that overactivation of this pathway is prevented by an adaptive feedback mechanism elicited by type I IFN receptors and executed by the exonuclease three prime repair exonuclease 1 (TREX1).


Subject(s)
Cytokines , Exodeoxyribonucleases , Phosphoproteins , Animals , DNA , Mammals/genetics , Mammals/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism
15.
Trends Cancer ; 10(7): 573-575, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38821853

ABSTRACT

Prostaglandin E2 (PGE2) is well known to promote tumor progression by boosting cancer cell proliferation while inhibiting anticancer immunity. Recent data from Lacher et al. and Morotti et al. demonstrate that one of the mechanisms through which PGE2 suppresses tumor-targeting immune responses involves downregulation of interleukin 2 (IL2) receptors and consequent inhibition of mitochondrial metabolism in T cells.


Subject(s)
Cyclooxygenase 2 , Dinoprostone , Mitochondria , Neoplasms , Humans , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/drug therapy , Dinoprostone/metabolism , Dinoprostone/immunology , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/immunology , Animals , Mitochondria/metabolism , Mitochondria/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Receptors, Interleukin-2/metabolism , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/immunology
16.
Nat Rev Immunol ; 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649722

ABSTRACT

Accumulating evidence suggests that metabolic rewiring in malignant cells supports tumour progression not only by providing cancer cells with increased proliferative potential and an improved ability to adapt to adverse microenvironmental conditions but also by favouring the evasion of natural and therapy-driven antitumour immune responses. Here, we review cancer cell-intrinsic and cancer cell-extrinsic mechanisms through which alterations of metabolism in malignant cells interfere with innate and adaptive immune functions in support of accelerated disease progression. Further, we discuss the potential of targeting such alterations to enhance anticancer immunity for therapeutic purposes.

17.
Cancers (Basel) ; 16(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38893165

ABSTRACT

Malignant central nervous system (CNS) cancers include a group of heterogeneous dis-eases characterized by a relative resistance to treatments and distinguished as either primary tumors arising in the CNS or secondary tumors that spread from other organs into the brain. Despite therapeutic efforts, they often cause significant mortality and morbidity across all ages. Radiotherapy (RT) remains the main treatment for brain cancers, improving associated symptoms, improving tumor control, and inducing a cure in some. However, the ultimate goal of cancer treatment, to improve a patient's survival, remains elusive for many CNS cancers, especially primary tumors. Over the years, there have thus been many preclinical studies and clinical trials designed to identify and overcome mechanisms of resistance to improve outcomes after RT and other therapies. For example, immunotherapy delivered concurrent with RT, especially hypo-fractionated stereotactic RT, is synergistic and has revolutionized the clinical management and outcome of some brain tumors, in particular brain metastases (secondary brain tumors). However, its impact on gliomas, the most common primary malignant CNS tumors, remains limited. In this review, we provide an overview of radioresistance mechanisms, the emerging strategies to overcome radioresistance, the role of the tumor microenviroment (TME), and the selection of the most significant results of radiation-immuno-oncological investigations. We also identify novel therapeutic opportunities in primary and secondary brain tumors with the purpose of elucidating current knowledge and stimulating further research to improve tumor control and patients' survival.

18.
Int J Radiat Oncol Biol Phys ; 119(4): 1248-1260, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38364947

ABSTRACT

PURPOSE: Diffuse midline glioma (DMG) is a fatal tumor traditionally treated with radiation therapy (RT) and previously characterized as having a noninflammatory tumor immune microenvironment (TIME). FLASH is a novel RT technique using ultra-high dose rate that is associated with decreased toxicity and effective tumor control. However, the effect of FLASH and conventional (CONV) RT on the DMG TIME has not yet been explored. METHODS AND MATERIALS: Here, we performed single-cell RNA sequencing (scRNA-seq) and flow cytometry on immune cells isolated from an orthotopic syngeneic murine model of brainstem DMG after the use of FLASH (90 Gy/sec) or CONV (2 Gy/min) dose-rate RT and compared to unirradiated tumor (SHAM). RESULTS: At day 4 post-RT, FLASH exerted similar effects as CONV in the predominant microglial (MG) population, including the presence of two activated subtypes. However, at day 10 post-RT, we observed a significant increase in the type 1 interferon α/ß receptor (IFNAR+) in MG in CONV and SHAM compared to FLASH. In the non-resident myeloid clusters of macrophages (MACs) and dendritic cells (DCs), we found increased type 1 interferon (IFN1) pathway enrichment for CONV compared to FLASH and SHAM by scRNA-seq. We observed this trend by flow cytometry at day 4 post-RT in IFNAR+ MACs and DCs, which equalized by day 10 post-RT. DMG control and murine survival were equivalent between RT dose rates. CONCLUSIONS: Our work is the first to map CONV and FLASH immune alterations of the DMG TIME with single-cell resolution. Although DMG tumor control and survival were similar between CONV and FLASH, we found that changes in immune compartments differed over time. Importantly, although both RT modalities increased IFN1, we found that the timing of this response was cell-type and dose-rate dependent. These temporal differences, particularly in the context of tumor control, warrant further study.


Subject(s)
Glioma , Microglia , Animals , Glioma/radiotherapy , Glioma/immunology , Glioma/pathology , Mice , Microglia/radiation effects , Microglia/immunology , Tumor Microenvironment/immunology , Brain Neoplasms/radiotherapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Receptor, Interferon alpha-beta/genetics , Mice, Inbred C57BL , Single-Cell Analysis/methods , Dendritic Cells/immunology , Dendritic Cells/radiation effects , Macrophages/immunology
19.
Int Rev Cell Mol Biol ; 376: 121-141, 2023.
Article in English | MEDLINE | ID: mdl-36997267

ABSTRACT

Fatty acid metabolic reprogramming has emerged as a major regulator of anti-tumor immune responses with large body of evidence that demonstrate its ability to impact the differentiation and function of immune cells. Therefore, depending on the metabolic cues that stem in the tumor microenvironment, the tumor fatty acid metabolism can tilt the balance of inflammatory signals to either promote or impair anti-tumor immune responses. Oxidative stressors such as reactive oxygen species generated from radiation therapy can rewire the tumor energy supply, suggesting that radiation therapy can further perturb the energy metabolism of a tumor by promoting fatty acid production. In this review, we critically discuss the network of fatty acid metabolism and how it regulates immune response especially in the context of radiation therapy.


Subject(s)
Energy Metabolism , Neoplasms , Humans , Neoplasms/metabolism , Tumor Microenvironment , Lipid Metabolism , Fatty Acids/metabolism
20.
Sci Rep ; 13(1): 200, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36604457

ABSTRACT

Radiation injury from medical, accidental, or intentional sources can induce acute and long-term hepatic dysregulation, fibrosis, and cancer. This long-term hepatic dysregulation decreases quality of life and may lead to death. Our goal in this study is to determine acute changes in biological pathways and discover potential RNA biomarkers predictive of radiation injury. We performed whole transcriptome microarray analysis of mouse liver tissue (C57BL/6 J) 48 h after whole-body irradiation with 1, 2, 4, 8, and 12 Gray to identify significant expression changes in mRNAs, lncRNAs, and miRNAs, We also validated changes in specific RNAs through qRT-PCR. We used Ingenuity Pathway Analysis (IPA) to identify pathways associated with gene expression changes. We observed significant dysregulation of multiple mRNAs across all doses. In contrast, miRNA dysregulation was observed upwards of 2 Gray. The most significantly upregulated mRNAs function as tumor suppressors: Cdkn1a, Phlda3, and Eda2r. The most significantly downregulated mRNAs were involved in hemoglobin synthesis, inflammation, and mitochondrial function including multiple members of Hbb and Hba. The most significantly upregulated miRNA included: miR-34a-5p, miR-3102-5p, and miR-3960, while miR-342-3p, miR-142a-3p, and miR-223-3p were most significantly downregulated. IPA predicted activation of cell cycle checkpoint control pathways and inhibition of pathways relevant to inflammation and erythropoietin. Clarifying expression of mRNA, miRNA and lncRNA at a short time point (48 h) offers insight into potential biomarkers, including radiation markers shared across organs and animal models. This information, once validated in human models, can aid in development of bio-dosimetry biomarkers, and furthers our understanding of acute pathway dysregulation.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Animals , Mice , Inflammation , Liver/metabolism , Mice, Inbred C57BL , Microarray Analysis , MicroRNAs/genetics , MicroRNAs/metabolism , Quality of Life , RNA, Long Noncoding/genetics , Xedar Receptor
SELECTION OF CITATIONS
SEARCH DETAIL