Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
1.
Life Sci Space Res (Amst) ; 41: 202-209, 2024 May.
Article in English | MEDLINE | ID: mdl-38670648

ABSTRACT

Explorations of the Moon and Mars are planned as future manned space missions, during which humans will be exposed to both radiation and microgravity. We do not, however, know the health effects for such combined exposures. In a ground-based experiment, we evaluated the combined effects of radiation and simulated microgravity on tumorigenesis by performing X-irradiation and tail suspension in C3B6F1 ApcMin/+ mice, a well-established model for intestinal tumorigenesis. Mice were irradiated at 2 weeks of age and underwent tail suspension for 3 or 11 weeks using a special device that avoids damage to the tail. The tail suspension treatment significantly reduced the thymus weight after 3 weeks but not 11 weeks, suggesting a transient stress response. The combination of irradiation and tail suspension significantly increased the number of small intestinal tumors less than 2 mm in diameter as compared with either treatment alone. The combined treatment also increased the fraction of malignant tumors among all small intestinal tumors as compared with the radiation-only treatment. Thus, the C3B6F1 ApcMin/+ mouse is a useful model for assessing cancer risk in a simulated space environment, in which simulated microgravity accelerates tumor progression when combined with radiation exposure.


Subject(s)
Intestinal Neoplasms , Weightlessness Simulation , Animals , Mice , Intestinal Neoplasms/pathology , Intestinal Neoplasms/etiology , Carcinogenesis/radiation effects , Mice, Inbred C57BL , Hindlimb Suspension , Male , X-Rays , Disease Models, Animal , Female , Intestine, Small/radiation effects , Intestine, Small/pathology , Thymus Gland/radiation effects , Thymus Gland/pathology , Neoplasms, Radiation-Induced/pathology , Neoplasms, Radiation-Induced/etiology
2.
Radiother Oncol ; 195: 110267, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38614282

ABSTRACT

BACKGROUND AND PURPOSE: Medulloblastoma (MB) is a common primary brain cancer in children. Proton therapy in pediatric MB is intensively studied and widely adopted. Compared to photon, proton radiations offer potential for reduced toxicity due to the characteristic Bragg Peak at the end of their path in tissue. The aim of this study was to compare the effects of irradiation with the same dose of protons or photons in Patched1 heterozygous knockout mice, a murine model predisposed to cancer and non-cancer radiogenic pathologies, including MB and lens opacity. MATERIALS AND METHODS: TOP-IMPLART is a pulsed linear proton accelerator for proton therapy applications. We compared the long-term health effects of 3 Gy of protons or photons in neonatal mice exposed at postnatal day 2, during a peculiarly susceptible developmental phase of the cerebellum, lens, and hippocampus, to genotoxic stress. RESULTS: Experimental testing of the 5 mm Spread-Out Bragg Peak (SOBP) proton beam, through evaluation of apoptotic response, confirmed that both cerebellum and hippocampus were within the SOBP irradiation field. While no differences in MB induction were observed after irradiation with protons or photons, lens opacity examination confirmed sparing of the lens after proton exposure. Marked differences in expression of neurogenesis-related genes and in neuroinflammation, but not in hippocampal neurogenesis, were observed after irradiation of wild-type mice with both radiation types. CONCLUSION: In-vivo experiments with radiosensitive mouse models improve our mechanistic understanding of the dependence of brain damage on radiation quality, thus having important implications in translational research.


Subject(s)
Animals, Newborn , Apoptosis , Hippocampus , Photons , Proton Therapy , Animals , Mice , Apoptosis/radiation effects , Proton Therapy/adverse effects , Hippocampus/radiation effects , Medulloblastoma/radiotherapy , Medulloblastoma/pathology , Carcinogenesis/radiation effects , Mice, Knockout , Cerebellar Neoplasms/radiotherapy , Cerebellar Neoplasms/pathology , Brain/radiation effects , Patched-1 Receptor/genetics , Disease Models, Animal , Protons/adverse effects
3.
Photochem Photobiol Sci ; 23(3): 517-526, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38337129

ABSTRACT

Squamous cell carcinoma represents the second most common type of keratinocyte carcinoma with ultraviolet radiation (UVR) making up the primary risk factor. Oral photoprotection aims to reduce incidence rates through oral intake of photoprotective compounds. Recently, drug repurposing has gained traction as an interesting source of chemoprevention. Because of their reported photoprotective properties, we investigated the potential of bucillamine, carvedilol, metformin, and phenformin as photoprotective compounds following oral intake in UVR-exposed hairless mice. Tumour development was observed in all groups in response to UVR, with only the positive control (Nicotinamide) demonstrating a reduction in tumour incidence (23.8%). No change in tumour development was observed in the four repurposed drug groups compared to the UV control group, whereas nicotinamide significantly reduced carcinogenesis (P = 0.00012). Metformin treatment significantly reduced UVR-induced erythema (P = 0.012), bucillamine and phenformin increased dorsal pigmentation (P = 0.0013, and P = 0.0005), but no other photoprotective effect was observed across the repurposed groups. This study demonstrates that oral supplementation with bucillamine, carvedilol, metformin, or phenformin does not affect UVR-induced carcinogenesis in hairless mice.


Subject(s)
Carcinoma, Squamous Cell , Cysteine/analogs & derivatives , Skin Neoplasms , Mice , Animals , Ultraviolet Rays , Carvedilol/pharmacology , Mice, Hairless , Phenformin/pharmacology , Carcinoma, Squamous Cell/prevention & control , Carcinoma, Squamous Cell/etiology , Carcinogenesis/radiation effects , Niacinamide/pharmacology , Skin Neoplasms/etiology , Skin Neoplasms/prevention & control , Skin Neoplasms/pathology , Skin/radiation effects
4.
J Biol Chem ; 299(7): 104900, 2023 07.
Article in English | MEDLINE | ID: mdl-37301510

ABSTRACT

Nucleotide excision repair (NER) eliminates highly genotoxic solar UV-induced DNA photoproducts that otherwise stimulate malignant melanoma development. Here, a genome-wide loss-of-function screen, coupling CRISPR/Cas9 technology with a flow cytometry-based DNA repair assay, was used to identify novel genes required for efficient NER in primary human fibroblasts. Interestingly, the screen revealed multiple genes encoding proteins, with no previously known involvement in UV damage repair, that significantly modulate NER uniquely during S phase of the cell cycle. Among these, we further characterized Dyrk1A, a dual specificity kinase that phosphorylates the proto-oncoprotein cyclin D1 on threonine 286 (T286), thereby stimulating its timely cytoplasmic relocalization and proteasomal degradation, which is required for proper regulation of the G1-S phase transition and control of cellular proliferation. We demonstrate that in UV-irradiated HeLa cells, depletion of Dyrk1A leading to overexpression of cyclin D1 causes inhibition of NER uniquely during S phase and reduced cell survival. Consistently, expression/nuclear accumulation of nonphosphorylatable cyclin D1 (T286A) in melanoma cells strongly interferes with S phase NER and enhances cytotoxicity post-UV. Moreover, the negative impact of cyclin D1 (T286A) overexpression on repair is independent of cyclin-dependent kinase activity but requires cyclin D1-dependent upregulation of p21 expression. Our data indicate that inhibition of NER during S phase might represent a previously unappreciated noncanonical mechanism by which oncogenic cyclin D1 fosters melanomagenesis.


Subject(s)
Cyclin D1 , Cyclin-Dependent Kinase Inhibitor p21 , DNA Repair , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Humans , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/radiation effects , HeLa Cells , Protein-Tyrosine Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/radiation effects , S Phase , G1 Phase , Melanoma/genetics , Melanoma/pathology , Cells, Cultured , Ultraviolet Rays/adverse effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Dyrk Kinases
5.
Sci Rep ; 11(1): 23467, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34873209

ABSTRACT

Ionizing radiations encountered by astronauts on deep space missions produce biological damage by two main mechanisms: (1) Targeted effects (TE) due to direct traversals of cells by ionizing tracks. (2) Non-targeted effects (NTE) caused by release of signals from directly hit cells. The combination of these mechanisms generates non-linear dose response shapes, which need to be modeled quantitatively to predict health risks from space exploration. Here we used a TE + NTE model to analyze data on APC(1638N/+) mouse tumorigenesis induced by space-relevant doses of protons, 4He, 12C, 16O, 28Si or 56Fe ions, or γ rays. A customized weighted Negative Binomial distribution was used to describe the radiation type- and dose-dependent data variability. This approach allowed detailed quantification of dose-response shapes, NTE- and TE-related model parameters, and radiation quality metrics (relative biological effectiveness, RBE, and radiation effects ratio, RER, relative to γ rays) for each radiation type. Based on the modeled responses for each radiation type, we predicted the tumor yield for a Mars-mission-relevant mixture of these radiations, using the recently-developed incremental effect additivity (IEA) synergy theory. The proposed modeling approach can enhance current knowledge about quantification of space radiation quality effects, dose response shapes, and ultimately the health risks for astronauts.


Subject(s)
Carcinogenesis/radiation effects , Cell Transformation, Neoplastic/radiation effects , Cosmic Radiation/adverse effects , Animals , Gamma Rays/adverse effects , Humans , Linear Energy Transfer/radiation effects , Male , Mice , Neoplasms, Radiation-Induced/etiology , Protons/adverse effects , Relative Biological Effectiveness , Space Flight
6.
Curr Probl Dermatol ; 55: 339-353, 2021.
Article in English | MEDLINE | ID: mdl-34698023

ABSTRACT

Cutaneous malignant melanoma (CMM) and nonmelanoma skin cancers (NMSC), squamous cell and basal cell carcinomas, have been increasing at exponential rates for as long as the International Agency for Research on Cancer (IARC) have been collecting data starting from 1955 in some northern European countries and 1960 in most other European countries. Different strains of the human papilloma virus (HPV) have been found in CMM and NMSC biopsies and implicated in the carcinogenic process as a "hit-and-run" mechanism and can spread at exponential rates, especially since the 1960s' sexual revolution. This chapter covers only IARC data for CMM in the European countries from 1960 to 2018, plotted by regions (northern, middle, and southern latitudes and eastern versus western longitudes), countries latitudes, and each country over time, which shows that about half have linear and the other half have exponential increases in CMM. From this analyzed data and published data in the literature, the major risk factors of CMM appear to be light hair color, especially red and white hair (reactive oxygen species and UVA; 320-400 nm), low cutaneous vitamin D3 levels, and HPV after 1960, while there was no apparent risk from exposure to UVB (290-320 nm) or sunburns.


Subject(s)
Alphapapillomavirus/radiation effects , Papillomavirus Infections/etiology , Skin Neoplasms/etiology , Ultraviolet Rays/adverse effects , Alphapapillomavirus/pathogenicity , Carcinogenesis/radiation effects , Humans , Papillomavirus Infections/pathology , Reactive Oxygen Species/metabolism , Risk Factors , Skin/metabolism , Skin/pathology , Skin/radiation effects , Skin/virology , Skin Neoplasms/pathology
7.
Oxid Med Cell Longev ; 2021: 8457521, 2021.
Article in English | MEDLINE | ID: mdl-34616505

ABSTRACT

Ferroptosis is a new type of regulatory cell death that differs from autophagy, apoptosis, necrosis, and pyroptosis; it is caused primarily by the accumulation of iron and lipid peroxides in the cell. Studies have shown that many classical signaling pathways and biological processes are involved in the process of ferroptosis. In recent years, investigations have revealed that ferroptosis plays a crucial role in the progression of tumors, especially lung cancer. In particular, inducing ferroptosis in cells can inhibit the growth of tumor cells, thereby reversing tumorigenesis. In this review, we summarize the characteristics of ferroptosis from its underlying basis and role in lung cancer and provide possible applications for it in lung cancer therapies.


Subject(s)
Carcinogenesis/metabolism , Ferroptosis/drug effects , Immunotherapy/methods , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/radiation effects , Ferroptosis/immunology , Ferroptosis/radiation effects , Humans , Iron/metabolism , Lipid Peroxidation/drug effects , Lung Neoplasms/radiotherapy , Signal Transduction/immunology , Signal Transduction/radiation effects , Treatment Outcome
8.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34504008

ABSTRACT

Chronic ultraviolet (UV) radiation exposure is the greatest risk factor for cutaneous squamous cell carcinoma (cSCC) development, and compromised immunity accelerates this risk. Having previously identified that epidermal Langerhans cells (LC) facilitate the expansion of UV-induced mutant keratinocytes (KC), we sought to more fully elucidate the immune pathways critical to cutaneous carcinogenesis and to identify potential targets of intervention. Herein, we reveal that chronic UV induces and LC enhance a local immune shift toward RORγt+ interleukin (IL)-22/IL-17A-producing cells that occurs in the presence or absence of T cells while identifying a distinct RORγt+ Sca-1+ CD103+ ICOS+ CD2+/- CCR6+ intracellular CD3+ cutaneous innate lymphoid cell type-3 (ILC3) population (uvILC3) that is associated with UV-induced mutant KC growth. We further show that mutant KC clone size is markedly reduced in the absence of RORγt+ lymphocytes or IL-22, both observed in association with expanding KC clones, and find that topical application of a RORγ/γt inhibitor during chronic UV exposure reduces local expression of IL-22 and IL-17A while markedly limiting mutant p53 KC clonal expansion. We implicate upstream Toll-like receptor signaling in driving this immune response to chronic UV exposure, as MyD88/Trif double-deficient mice also show substantially reduced p53 island number and size. These data elucidate key immune components of chronic UV-induced cutaneous carcinogenesis that might represent targets for skin cancer prevention.


Subject(s)
Interleukins/metabolism , Keratinocytes/pathology , Lymphocytes/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Skin Neoplasms/pathology , Skin/pathology , Ultraviolet Rays/adverse effects , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Cells, Cultured , Immunity, Innate/immunology , Interleukins/genetics , Keratinocytes/metabolism , Keratinocytes/radiation effects , Langerhans Cells/immunology , Langerhans Cells/metabolism , Langerhans Cells/pathology , Langerhans Cells/radiation effects , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/radiation effects , Mice , Mutation , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Skin/metabolism , Skin/radiation effects , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Interleukin-22
9.
Sci Rep ; 11(1): 19096, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34580369

ABSTRACT

Childhood radiation exposure is a known thyroid cancer risk factor. This study evaluated the effects of age on radiation-induced thyroid carcinogenesis in rats irradiated with 8 Gy X-rays. We analyzed cell proliferation, cell death, DNA damage response, and autophagy-related markers in 4-week-old (4W) and 7-month-old (7M) rats and the incidence of thyroid tumors in 4W, 4-month-old (4M), and 7M rats 18 months after irradiation. Cell death and DNA damage response were increased in 4W rats compared to those in controls at 1 month post-irradiation. More Ki-67-positive cells were observed in 4W rats at 12 months post-irradiation. Thyroid tumors were confirmed in 61.9% (13/21), 63.6% (7/11), and 33.3% (2/6) of irradiated 4W, 4M, and 7M rats, respectively, compared to 0%, 14.3% (1/7), and 16.7% (1/6) in the respective nonirradiated controls. There were 29, 9, and 2 tumors in irradiated 4W, 4M, and 7M rats, respectively. The expression of several autophagy components was downregulated in the area surrounding radiation-induced thyroid carcinomas in 4W and 7M rats. LC3 and p62 expression levels decreased in radiation-induced follicular carcinoma in 4W rats. Radiosensitive cells causing thyroid tumors may be more prevalent in young rats, and abrogation of autophagy may be associated with radiation-induced thyroid carcinogenesis.


Subject(s)
Carcinogenesis/radiation effects , Neoplasms, Radiation-Induced/epidemiology , Radiation Injuries, Experimental/epidemiology , Thyroid Neoplasms/epidemiology , Adult , Age Factors , Animals , Child , Dose-Response Relationship, Radiation , Humans , Incidence , Male , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/pathology , Radiation Tolerance , Rats , Risk Factors , Thyroid Gland/pathology , Thyroid Neoplasms/etiology , Thyroid Neoplasms/pathology , X-Rays/adverse effects
10.
Int J Mol Sci ; 22(11)2021 Jun 06.
Article in English | MEDLINE | ID: mdl-34204077

ABSTRACT

Skin cancers are growing in incidence worldwide and are primarily caused by exposures to ultraviolet (UV) wavelengths of sunlight. UV radiation induces the formation of photoproducts and other lesions in DNA that if not removed by DNA repair may lead to mutagenesis and carcinogenesis. Though the factors that cause skin carcinogenesis are reasonably well understood, studies over the past 10-15 years have linked the timing of UV exposure to DNA repair and skin carcinogenesis and implicate a role for the body's circadian clock in UV response and disease risk. Here we review what is known about the skin circadian clock, how it affects various aspects of skin physiology, and the factors that affect circadian rhythms in the skin. Furthermore, the molecular understanding of the circadian clock has led to the development of small molecules that target clock proteins; thus, we discuss the potential use of such compounds for manipulating circadian clock-controlled processes in the skin to modulate responses to UV radiation and mitigate cancer risk.


Subject(s)
Carcinogenesis/pathology , Circadian Clocks/physiology , Skin Neoplasms/physiopathology , Skin Physiological Phenomena , Skin/pathology , Skin/physiopathology , Animals , Carcinogenesis/radiation effects , Circadian Clocks/radiation effects , Humans , Risk Factors , Skin/radiation effects , Skin Physiological Phenomena/radiation effects
11.
J Dermatol Sci ; 103(3): 130-134, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34238637

ABSTRACT

Human skin is a highly efficient self-renewing barrier that is critical to withstanding environmental insults. Undifferentiated keratinocyte stem cells reside in the basal layer of the epidermis and in hair follicles that continuously give rise to progenies ensuring epidermal turnover and renewal. Ultraviolet (UV) radiation is a proven cause of skin keratinocyte cancers, which preferentially occur at sun-exposed areas of the skin. Fortunately, melanocytes produce melanin that is packaged in specific organelles (termed melanosomes) that are then delivered to nearby keratinocytes, endowing the recipient cells with photoprotection. It has long been thought that melanosome transfer takes place stochastically from melanocytes to keratinocytes via an as-yet-unrecognized manner. However, recent studies have indicated that melanosomes are distributed regionally in the basal layer of the skin, affording localized intensive photoprotection for progenitor keratinocytes and stem cells that reside in the microenvironment of the basal epidermis. In this review, we summarize current knowledge about molecular and cellular mechanisms that are responsible for the selective transfer and exclusive degradation of melanosomes in the epidermis, emphasizing implications for skin carcinogenesis.


Subject(s)
Epidermis/radiation effects , Melanosomes/metabolism , Stem Cells/cytology , Ultraviolet Rays/adverse effects , Carcinogenesis/radiation effects , Cells, Cultured , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/radiation effects , Skin Aging/radiation effects , Stem Cells/metabolism , Stem Cells/radiation effects
12.
FASEB J ; 35(7): e21671, 2021 07.
Article in English | MEDLINE | ID: mdl-34105803

ABSTRACT

Ultraviolet irradiation (UV) exposure is the leading factor underlying the development of skin malignancies. D-dopachrome tautomerase (D-DT), a functional homolog of macrophage migration inhibitory factor (MIF), has functional similarities to MIF. However, its role, unlike the role of MIF in photocarcinogenesis, is unknown. We therefore explored the role of D-DT in photocarcinogenesis by developing D-DT transgenic (D-DT Tg) mice and provided a research model for future studies targeting D-DT. Chronic UVB exposure accelerated tumor development in D-DT Tg mice compared with wild-type (WT) mice, with a higher incidence of tumors observed in D-DT Tg mice than in WT mice. In D-DT Tg irradiated mouse keratinocytes, the p53, PUMA, and Bax expression was lower than that in WT mice. These results indicate that D-DT Tg overexpression confers prevention against UVB-induced apoptosis in keratinocytes. Taken together, these findings support D-DT as a functionally important cytokine in photocarcinogenesis and potential therapeutic target for the prevention of photocarcinogenesis.


Subject(s)
Carcinogenesis/pathology , Intramolecular Oxidoreductases/metabolism , Keratinocytes/pathology , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects , Animals , Apoptosis , Carcinogenesis/metabolism , Carcinogenesis/radiation effects , Cell Proliferation , Female , Intramolecular Oxidoreductases/genetics , Keratinocytes/metabolism , Keratinocytes/radiation effects , Mice , Mice, Transgenic , Skin Neoplasms/etiology , Skin Neoplasms/metabolism
13.
Radiat Res ; 195(6): 561-567, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33826740

ABSTRACT

The genomic landscape neighboring large deletions including the hypoxanthine-guanine phosphoribosyl transferase (HPRT) locus on human X chromosome in 6-thioguanine-resistant mutants originating from immortalized human fibroblast cells exposed to X rays was characterized by real-time quantitative PCR (qPCR)-based analyses. Among the 13 mutant clones with large deletions extending over several Mb, including the HPRT locus, revealed by 10 conventional sequence-tagged site (STS) markers, three clones bearing the largest deletions were selected for further qPCR analysis using another 21 STS markers and 15 newly designed PCR primer pairs. The results indicated that the major deletions were in very specific regions between the 130-Mb and 140-Mb positions containing the HPRT locus on the X chromosome and, contrary to our initial expectations, additional minor deletions were distributed in a patchwork pattern. These findings strongly indicate that the complex deletion patterns in the affected chromosome are related to the radiation track structure with spatially heterogeneous energy deposition and the specific structure of the chromatin-nuclear membrane complex. The uncovered complex deletion patterns are in agreement with the idea of complex chromatin damage, which is frequently associated with carcinogenesis.


Subject(s)
Chromosome Deletion , Genome, Human/genetics , Carcinogenesis/genetics , Carcinogenesis/radiation effects , Genetic Loci/genetics , Humans , Polymerase Chain Reaction , X-Rays/adverse effects
14.
Eur J Immunol ; 51(7): 1799-1808, 2021 07.
Article in English | MEDLINE | ID: mdl-33759186

ABSTRACT

Mononuclear phagocytes consisting of monocytes, macrophages, and DCs play a complex role in tumor development by either promoting or restricting tumor growth. Cutaneous squamous cell carcinoma (cSCC) is the second most common nonmelanoma skin cancer arising from transformed epidermal keratinocytes. While present at high numbers, the role of tumor-infiltrating and resident myeloid cells in the formation of cSCC is largely unknown. Using transgenic mice and depleting antibodies to eliminate specific myeloid cell types in the skin, we investigated the involvement of mononuclear phagocytes in the development of UV-induced cSCC in K14-HPV8-E6 transgenic mice. Although resident Langerhans cells were enriched in the tumor, their contribution to tumor formation was negligible. Equally, dermal macrophages were dispensable for the development of cSCC. In contrast, mice lacking circulating monocytes were completely resistant to UV-induced cSCC, indicating that monocytes promote tumor development. Collectively, these results demonstrate a critical role for classical monocytes in the initiation of skin cancer.


Subject(s)
Carcinogenesis/pathology , Epidermis/pathology , Monocytes/pathology , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects , Animals , Carcinogenesis/radiation effects , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epidermis/radiation effects , Female , Keratinocytes/pathology , Keratinocytes/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/radiation effects , Skin/pathology , Skin/radiation effects
15.
Signal Transduct Target Ther ; 6(1): 129, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33785736

ABSTRACT

Maintenance of genetic stability via proper DNA repair in stem and progenitor cells is essential for the tissue repair and regeneration, while preventing cell transformation after damage. Loss of PUMA dramatically increases the survival of mice after exposure to a lethal dose of ionizing radiation (IR), while without promoting tumorigenesis in the long-term survivors. This finding suggests that PUMA (p53 upregulated modulator of apoptosis) may have a function other than regulates apoptosis. Here, we identify a novel role of PUMA in regulation of DNA repair in embryonic or induced pluripotent stem cells (PSCs) and immortalized hematopoietic progenitor cells (HPCs) after IR. We found that PUMA-deficient PSCs and HPCs exhibited a significant higher double-strand break (DSB) DNA repair activity via Rad51-mediated homologous recombination (HR). This is because PUMA can be associated with early mitotic inhibitor 1 (EMI1) and Rad51 in the cytoplasm to facilitate EMI1-mediated cytoplasmic Rad51 ubiquitination and degradation, thereby inhibiting Rad51 nuclear translocation and HR DNA repair. Our data demonstrate that PUMA acts as a repressor for DSB DNA repair and thus offers a new rationale for therapeutic targeting of PUMA in regenerative cells in the context of DNA damage.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Embryonic Stem Cells/metabolism , Hematopoietic Stem Cells/metabolism , Proteins/genetics , Rad51 Recombinase/genetics , Tumor Suppressor Proteins/genetics , Animals , Carcinogenesis/radiation effects , Cell Line, Tumor , Cytoplasm/genetics , Cytoplasm/radiation effects , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/genetics , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Embryonic Stem Cells/pathology , Embryonic Stem Cells/radiation effects , Gene Expression Regulation, Developmental/radiation effects , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/radiation effects , Mice , Radiation, Ionizing , Recombinational DNA Repair/radiation effects , Regeneration/genetics , Ubiquitination/genetics
16.
Radiat Res ; 195(5): 427-440, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33760917

ABSTRACT

Ionizing radiation induces DNA damage to cycling cells which, if left unrepaired or misrepaired, can cause cell inactivation or heritable, viable mutations. The latter can lead to cell transformation, which is thought to be an initial step of cancer formation. Consequently, the study of radiation-induced cell transformation promises to offer insights into the general properties of radiation carcinogenesis. As for other end points, the effectiveness in inducing cell transformation is elevated for radiation qualities with high linear energy transfer (LET), and the same is true for cancer induction. In considering DNA damage as a common cause of both cell death and transformations, a worthwhile approach is to apply mathematical models for the relative biological effectiveness (RBE) of cell killing to also assess the carcinogenic potential of high-LET radiation. In this work we used an established RBE model for cell survival and clinical end points, the local effect model (LEM), to estimate the transformation probability and the carcinogenic potential of ion radiation. The provided method consists of accounting for the competing processes of cell inactivation and induction of transformations or carcinogenic events after radiation exposure by a dual use of the LEM. Correlations between both processes inferred by the number of particle impacts to individual cells were considered by summing over the distribution of hits that individual cells receive. RBE values for cell transformation in vitro were simulated for three independent data sets, which were also used to gauge the approach. The simulations reflect the general RBE systematics both in magnitude and in energy and LET dependence. To challenge the developed method, in vivo carcinogenesis was investigated using the same concepts, where the probability for cancer induction within an irradiated organ was derived from the probability of finding carcinogenic events in individual cells. The predictions were compared with experimental data of carcinogenesis in Harderian glands of mice. Again, the developed method shows the same characteristics as the experimental data. We conclude that the presented method is helpful to predictively assess RBE for both neoplastic cell transformation and tumor induction after ion exposure within a wide range of LET values. The theoretical concept requires a non-linear component in the photon dose response for carcinogenic end points as a precondition for the observed enhanced effects after ion exposure, thus contributing to a long debate in epidemiology. Future work will use the method for assessing cancer induction in radiation therapy and exposure scenarios frequently discussed in radiation protection.


Subject(s)
Cell Transformation, Neoplastic/radiation effects , Models, Biological , Animals , Carcinogenesis/radiation effects , Cell Line, Tumor , Humans , Mice , Relative Biological Effectiveness
17.
J Dermatol Sci ; 102(1): 25-35, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33642112

ABSTRACT

BACKGROUND: Excessive inflammation and cell death induced by ultraviolet (UV) cause skin photodamage. Metformin possesses anti-inflammatory and cytoprotective effects. However, whether metformin inhibits inflammation and cell death in UVB-induced acute skin damage is unclear. OBJECTIVE: To evaluate the anti-inflammatory and cytoprotective effects of metformin in vitro and in vivo. Furthermore, its potential mechanism has been explored. METHODS: Transcriptome sequencing and multiplex cytokines analysis were used to evaluate the validity of in vitro UVB-induced acute damage keratinocyte model and anti-inflammatory effects of metformin. We also determined the expression and nuclear translocation of CCAAT/enhancer-binding protein beta (C/EBPß), an important transcriptional factor of Interleukin-1beta (IL-1ß). Cell viability and cell death of keratinocytes were evaluated upon UVB irradiation in the presence or absence of metformin. 0.6% metformin cream was applied on UVB-irradiated mice to explore its pharmacological effects in vivo. RESULTS: Transcriptional landscape of 50 mJ/cm2 UVB-irradiated HaCaT cells is typical of UVB-induced acute damage keratinocyte model in vitro. Metformin alleviated transcription and secretion of IL-1ß, Tumor Necrosis Factor-alpha, and Fibroblast Growth Factor 2, expression and nuclear translocation of C/EBPß in this model. Metformin also protected keratinocytes from cell death caused by UVB-induced cellular secretions, which contributed to its cytoprotective effects. Topical administration of 0.6% metformin cream alleviated UVB-induced skin damage in mice. CONCLUSION: We proved the protective roles of metformin in UVB-challenged keratinocytes and UVB-irradiated mice, which indicated the potential value of metformin in topical therapy against skin photodamage.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Keratinocytes/drug effects , Metformin/pharmacology , Skin/drug effects , Ultraviolet Rays/adverse effects , Administration, Cutaneous , Animals , Anti-Inflammatory Agents/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Disease Models, Animal , Drug Evaluation, Preclinical , HaCaT Cells , Humans , Keratinocytes/pathology , Keratinocytes/radiation effects , Metformin/therapeutic use , Mice , Reactive Oxygen Species , Skin/pathology , Skin/radiation effects , Skin Aging/drug effects , Skin Aging/radiation effects , Skin Neoplasms/etiology , Skin Neoplasms/pathology , Skin Neoplasms/prevention & control , Sunburn/etiology , Sunburn/pathology , Sunburn/prevention & control
18.
Cancer Prev Res (Phila) ; 14(5): 527-540, 2021 05.
Article in English | MEDLINE | ID: mdl-33648941

ABSTRACT

Skin cancer is the most common malignancy worldwide and is rapidly rising in incidence, representing a significant public health challenge. The ß-blocker, carvedilol, has shown promising effects in preventing skin cancer. However, as a potent ß-blocker, repurposing carvedilol to an anticancer agent is limited by cardiovascular effects. Carvedilol is a racemic mixture consisting of equimolar S- and R-carvedilol, whereas the R-carvedilol enantiomer does not possess ß-blocking activity. Because previous studies suggest that carvedilol's cancer preventive activity is independent of ß-blockade, we examined the skin cancer preventive activity of R-carvedilol compared with S-carvedilol and the racemic carvedilol. R- and S-carvedilol were equally effective in preventing EGF-induced neoplastic transformation of the mouse epidermal JB6 Cl 41-5a (JB6 P+) cells and displayed similar attenuation of EGF-induced ELK-1 activity. R-carvedilol appeared slightly better than S-carvedilol against UV-induced intracellular oxidative stress and release of prostaglandin E2 from the JB6 P+ cells. In an acute UV-induced skin damage and inflammation mouse model using a single irradiation of 300 mJ/cm2 UV, topical treatment with R-carvedilol dose dependently attenuated skin edema and reduced epidermal thickening, Ki-67 staining, COX-2 protein, and IL6 and IL1ß mRNA levels similar to carvedilol. In a chronic UV (50-150 mJ/cm2) induced skin carcinogenesis model in mice with pretreatment of test agents, topical treatment with R-carvedilol, but not racemic carvedilol, significantly delayed and reduced skin squamous cell carcinoma development. Therefore, as an enantiomer present in an FDA-approved agent, R-carvedilol may be a better option for developing a safer and more effective preventive agent for skin carcinogenesis. PREVENTION RELEVANCE: In this study, we demonstrated the skin cancer preventive activity of R-carvedilol, the non-ß-blocking enantiomer present in the racemic ß-blocker, carvedilol. As R-carvedilol does not have ß-blocking activity, such a preventive treatment would not lead to common cardiovascular side effects of ß-blockers.


Subject(s)
Carcinogenesis/drug effects , Carvedilol/administration & dosage , Epidermis/drug effects , Neoplasms, Experimental/prevention & control , Skin Neoplasms/prevention & control , Animals , Carcinogenesis/chemically induced , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Carvedilol/chemistry , Epidermal Cells , Epidermal Growth Factor/toxicity , Epidermis/pathology , Epidermis/radiation effects , Female , HEK293 Cells , Humans , Mice , Neoplasms, Experimental/etiology , Neoplasms, Experimental/pathology , Skin Neoplasms/etiology , Skin Neoplasms/pathology , Stereoisomerism , Ultraviolet Rays/adverse effects
19.
Cancer Sci ; 112(6): 2233-2244, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33743547

ABSTRACT

According to TCGA database, mutations in PPP6C (encoding phosphatase PP6) are found in c. 10% of tumors from melanoma patients, in which they coexist with BRAF and NRAS mutations. To assess PP6 function in melanoma carcinogenesis, we generated mice in which we could specifically induce BRAF(V600E) expression and delete Ppp6c in melanocytes. In these mice, melanoma susceptibility following UVB irradiation exhibited the following pattern: Ppp6c semi-deficient (heterozygous) > Ppp6c wild-type > Ppp6c-deficient (homozygous) tumor types. Next-generation sequencing of Ppp6c heterozygous and wild-type melanoma tumors revealed that all harbored Trp53 mutations. However, Ppp6c heterozygous tumors showed a higher Signature 1 (mitotic/mitotic clock) mutation index compared with Ppp6c wild-type tumors, suggesting increased cell division. Analysis of cell lines derived from either Ppp6c heterozygous or wild-type melanoma tissues showed that both formed tumors in nude mice, but Ppp6c heterozygous tumors grew faster compared with those from the wild-type line. Ppp6c knockdown via siRNA in the Ppp6c heterozygous line promoted the accumulation of genomic damage and enhanced apoptosis relative to siRNA controls. We conclude that in the presence of BRAF(V600E) expression and UV-induced Trp53 mutation, Ppp6c haploinsufficiency promotes tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Melanoma/genetics , Phosphoprotein Phosphatases/genetics , Proto-Oncogene Proteins B-raf/genetics , Ultraviolet Rays/adverse effects , Animals , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Exome/genetics , Exome/radiation effects , Genotype , Haploinsufficiency , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanocytes/radiation effects , Melanoma/pathology , Mice , Mice, Nude , Mice, Transgenic , Mutation/radiation effects , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
20.
In Vitro Cell Dev Biol Anim ; 57(3): 300-314, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33537930

ABSTRACT

Cancer metastasis and recurrence are potentially lethal. A small number of cancer cell groups called cancer stem cells (CSCs) have both stem cell capacity and cancer-forming ability and are reported to play important roles in cancer metastasis and recurrence. These CSCs are considered to be radiation-resistant (RR). Therefore, understanding the biological effects of radiation on squamous cell carcinoma (SCC) cell lines in vitro and in vivo might be worthwhile to circumvent radiation resistance. Currently, there are no reports on the establishment of RR-SCC cells in serum-free defined culture, which mimics biological mechanisms and prevents instability of using serum in the culture medium. We isolated radiation-resistant strains, designated A431-LDR and A431-HDR, from A431 cells derived from vulval SCC and irradiated them with a total dose of 60 Gy at a low-dose rate (2.2 Gy/d) (RM1000) and a high-dose rate (5 Gy/5.75min) in serum-free defined culture. These cells exhibited high sphere-forming and migration ability in vitro and high tumor-forming ability in nude mice xenografts. Overexpression of KRT13 in A431-RR cells might play a role in its radiation-resistant characteristics. These cells might be useful not only to study cancer stem cells but also to study the circumvention of radiation resistance by novel cancer treatment modalities.


Subject(s)
Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Keratin-13/genetics , Radiation Tolerance/genetics , Vulvar Neoplasms/genetics , Animals , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Culture Media, Serum-Free , Dose-Response Relationship, Radiation , Female , Humans , Keratin-13/metabolism , Mice, Nude , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , RNA, Small Interfering/metabolism , Tumor Stem Cell Assay , Vulvar Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...