Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cancers (Basel) ; 16(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38730612

ABSTRACT

High-risk human papillomaviruses (HPVs) are the main cause of cervical, oropharyngeal, and anogenital cancers, which are all treated with definitive chemoradiation therapy when locally advanced. HPV proteins are known to exploit the host DNA damage response to enable viral replication and the epithelial differentiation protocol. This has far-reaching consequences for the host genome, as the DNA damage response is critical for the maintenance of genomic stability. HPV+ cells therefore have increased DNA damage, leading to widespread genomic instability, a hallmark of cancer, which can contribute to tumorigenesis. Following transformation, high-risk HPV oncoproteins induce chromosomal instability, or chromosome missegregation during mitosis, which is associated with a further increase in DNA damage, particularly due to micronuclei and double-strand break formation. Thus, HPV induces significant DNA damage and activation of the DNA damage response in multiple contexts, which likely affects radiation sensitivity and efficacy. Here, we review how HPV activates the DNA damage response, how it induces chromosome missegregation and micronuclei formation, and discuss how these factors may affect radiation response. Understanding how HPV affects the DNA damage response in the context of radiation therapy may help determine potential mechanisms to improve therapeutic response.

2.
Viruses ; 16(4)2024 03 25.
Article in English | MEDLINE | ID: mdl-38675844

ABSTRACT

Chromosomal instability (CIN) and aneuploidy are hallmarks of cancer. CIN is defined as a continuous rate of chromosome missegregation events over the course of multiple cell divisions. CIN causes aneuploidy, a state of abnormal chromosome content differing from a multiple of the haploid. Human papillomavirus (HPV) is a well-known cause of squamous cancers of the oropharynx, cervix, and anus. The HPV E6 and E7 oncogenes have well-known roles in carcinogenesis, but additional genomic events, such as CIN and aneuploidy, are often required for tumor formation. HPV+ squamous cancers have an increased frequency of specific types of CIN, including polar chromosomes. CIN leads to chromosome gains and losses (aneuploidies) specific to HPV+ cancers, which are distinct from HPV- cancers. HPV-specific CIN and aneuploidy may have implications for prognosis and therapeutic response and may provide insight into novel therapeutic vulnerabilities. Here, we review HPV-specific types of CIN and patterns of aneuploidy in squamous cancers, as well as how this impacts patient prognosis and treatment.


Subject(s)
Aneuploidy , Chromosomal Instability , Papillomavirus Infections , Humans , Papillomavirus Infections/virology , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Papillomaviridae/genetics , Papillomaviridae/pathogenicity , Carcinoma, Squamous Cell/virology , Carcinoma, Squamous Cell/genetics , Neoplasms, Squamous Cell/virology , Neoplasms, Squamous Cell/genetics , Neoplasms, Squamous Cell/pathology , Female , Alphapapillomavirus/genetics , Alphapapillomavirus/pathogenicity , Human Papillomavirus Viruses
3.
Radiat Res ; 199(5): 429-438, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37014873

ABSTRACT

Low-energy X-ray sources that operate in the kilovoltage energy range have been shown to induce more cellular damage when compared to their megavoltage counterparts. However, low-energy X-ray sources are more susceptible to the effects of filtration on the beam spectrum. This work sought to characterize the biological effects of the Xoft Axxent® source, a low-energy therapeutic X-ray source, both with and without the titanium vaginal applicator in place. It was hypothesized that there would be an increase in relative biological effectiveness (RBE) of the Axxent® source compared to 60Co and that the source in the titanium vaginal applicator (SIA) would have decreased biological effects compared to the bare source (BS). This hypothesis was drawn from linear energy transfer (LET) simulations performed using the TOPAS Monte Carlo user code as well a reduction in dose rate of the SIA compared to the BS. A HeLa cell line was maintained and used to evaluate these effects. Clonogenic survival assays were performed to evaluate differences in the RBE between the BS and SIA using 60Co as the reference beam quality. Neutral comet assay was used to assess induction of DNA strand damage by each beam to estimate differences in RBE. Quantification of mitotic errors was used to evaluate differences in chromosomal instability (CIN) induced by the three beam qualities. The BS was responsible for the greatest quantity of cell death due to a greater number of DNA double strand breaks (DSB) and CIN observed in the cells. The differences observed in the BS and SIA surviving fractions and RBE values were consistent with the 13% difference in LET as well as the factor of 3.5 reduction in dose rate of the SIA. Results from the comet and CIN assays were consistent with these results as well. The use of the titanium applicator results in a reduction in the biological effects observed with these sources, but still provides an advantage over megavoltage beam qualities. © 2023 by Radiation Research Society.


Subject(s)
Brachytherapy , Uterine Cervical Neoplasms , Female , Humans , Brachytherapy/methods , HeLa Cells , Uterine Cervical Neoplasms/radiotherapy , Titanium/pharmacology , Relative Biological Effectiveness , DNA , Monte Carlo Method
4.
Proc Natl Acad Sci U S A ; 120(14): e2216700120, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36989302

ABSTRACT

Chromosome segregation during mitosis is highly regulated to ensure production of genetically identical progeny. Recurrent mitotic errors cause chromosomal instability (CIN), a hallmark of tumors. The E6 and E7 oncoproteins of high-risk human papillomavirus (HPV), which causes cervical, anal, and head and neck cancers (HNC), cause mitotic defects consistent with CIN in models of anogenital cancers, but this has not been studied in the context of HNC. Here, we show that HPV16 induces a specific type of CIN in patient HNC tumors, patient-derived xenografts, and cell lines, which is due to defects in chromosome congression. These defects are specifically induced by the HPV16 oncogene E6 rather than E7. We show that HPV16 E6 expression causes degradation of the mitotic kinesin CENP-E, whose depletion produces chromosomes that are chronically misaligned near spindle poles (polar chromosomes) and fail to congress. Though the canonical oncogenic role of E6 is the degradation of the tumor suppressor p53, CENP-E degradation and polar chromosomes occur independently of p53. Instead, E6 directs CENP-E degradation in a proteasome-dependent manner via the E6-associated ubiquitin protein ligase E6AP/UBE3A. This study reveals a mechanism by which HPV induces CIN, which may impact HPV-mediated tumor initiation, progression, and therapeutic response.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Humans , Chromosomal Instability , Chromosomes/metabolism , Human papillomavirus 16/genetics , Kinesins/genetics , Kinesins/metabolism , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism
5.
Semin Radiat Oncol ; 32(1): 54-63, 2022 01.
Article in English | MEDLINE | ID: mdl-34861996

ABSTRACT

Chromosome missegregation over the course of multiple cell divisions, termed chromosomal instability (CIN), is a hallmark of cancer. Multiple causes of CIN have been identified, including defects in the mitotic checkpoint, altered kinetochore-microtubule dynamics, centrosome amplification, and ionizing radiation. Here we review the types, mechanisms, and cellular implications of CIN. We discuss the evidence that CIN can promote tumors, suppress them, or do neither, depending on the rates of chromosome missegregration and the cellular context. Very high rates of chromosome missegregation lead to cell death due to loss of essential chromosomes; thus elevating CIN above a tolerable threshold provides a mechanistic opportunity to promote cancer cell death. Lethal rates of CIN can be achieved by a single insult or through a combination of insults. Because ionizing radiation induces CIN, additional therapies that increase CIN may serve as useful modulators of radiation sensitivity. Ultimately, quantifying the intrinsic CIN in a tumor and modulating this level pharmacologically as well as with radiation may allow for a more rational, personalized radiation therapy prescription, thereby decreasing side effects and increasing local control.


Subject(s)
Chromosome Segregation , Neoplasms , Centrosome/metabolism , Centrosome/pathology , Chromosomal Instability/genetics , Humans , Kinetochores/metabolism , Kinetochores/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/radiotherapy , Radiation Tolerance/genetics
6.
Semin Radiat Oncol ; 31(4): 265-273, 2021 10.
Article in English | MEDLINE | ID: mdl-34455982

ABSTRACT

Human papillomavirus (HPV) is a ubiquitous DNA virus that infects squamous epithelia. Though HPV only encodes 8 genes, it is capable of causing cellular transformation and ultimately cancer in host cells. In this article we review the classification of HPV viruses, their genetic structure and life cycle, viral gene biology, and provide an overview of the role of HPV in cancer. We explain how the viral life cycle can lead to integration of viral DNA into the host genome leading to increased cell cycle progression, decreased apoptosis, altered DNA repair, and chromosomal instability. We describe the multifaceted roles of the canonical oncogenes E6 and E7 in promoting tumorigenesis and the important role of other viral genes in regulating cancer development. We also review how the virus actively suppresses innate and adaptive immunity to evade immune detection and promote a pro-tumorigenic microenvironment. The biology presented here will serve as a foundation to the other chapters in this edition and we hope it will incite enthusiasm for continued research on this fascinating virus that causes significant morbidity and mortality worldwide.


Subject(s)
Alphapapillomavirus , Neoplasms , Oncogene Proteins, Viral , Papillomavirus Infections , Alphapapillomavirus/metabolism , Biology , Carcinogenesis/genetics , Humans , Neoplasms/genetics , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomaviridae/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Tumor Microenvironment
8.
J Clin Oncol ; 39(24): 2735-2736, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34043438
9.
Int J Radiat Oncol Biol Phys ; 109(5): 1483-1494, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33253820

ABSTRACT

PURPOSE: Cervical cancer represents the fourth most frequent malignancy in the world among women, and mortality has remained stable for the past 4 decades. Intravenous cisplatin with concurrent radiation therapy is the standard-of-care for patients with local and regional cervical cancer. However, cisplatin induces serious dose-limiting systemic toxicities and recurrence frequently occurs. In this study, we aimed to develop an intracervical drug delivery system that allows cisplatin release directly into the tumor and minimize systemic side effects. METHODS AND MATERIALS: Twenty patient biopsies and 5 cell lines treated with cisplatin were analyzed for platinum content using inductively coupled plasma mass spectrometry. Polymeric implants loaded with cisplatin were developed and evaluated for degradation and drug release. The effect of local or systemic cisplatin delivery on drug biodistribution as well as tumor burden were evaluated in vivo, in combination with radiation therapy. RESULTS: Platinum levels in patient biopsies were 6-fold lower than the levels needed for efficacy and radiosensitization in vitro. Cisplatin local delivery implant remarkably improved drug specificity to the tumor and significantly decreased accumulation in the blood, kidney, and other distant normal organs, compared with traditional systemic delivery. The localized treatment further resulted in complete inhibition of tumor growth. CONCLUSIONS: The current standard-of-care systemic administration of cisplatin provides a subtherapeutic dose. We developed a polymeric drug delivery system that delivered high doses of cisplatin directly into the cervical tumor, while lowering drug accumulation and consequent side effects in normal tissues. Moving forward, these data will be used as the basis of a future first-in-human clinical trial to test the efficacy of localized cisplatin as adjuvant or neoadjuvant chemotherapy in local and regional cervical cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Injections, Intralesional/methods , Radiation-Sensitizing Agents/administration & dosage , Uterine Cervical Neoplasms/drug therapy , Adult , Aged , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Biopsy , Cell Line, Tumor , Chemoradiotherapy/methods , Cisplatin/adverse effects , Cisplatin/analysis , Cisplatin/pharmacokinetics , Drug Implants , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Polymers/administration & dosage , Radiation-Sensitizing Agents/adverse effects , Radiation-Sensitizing Agents/pharmacokinetics , Tissue Distribution , Tumor Burden , Uterine Cervical Neoplasms/chemistry , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
10.
Cancers (Basel) ; 12(2)2020 Feb 12.
Article in English | MEDLINE | ID: mdl-32059418

ABSTRACT

Patient-derived model systems are important tools for studying novel anti-cancer therapies. Patient-derived xenografts (PDXs) have gained favor over the last 10 years as newer mouse strains have improved the success rate of establishing PDXs from patient biopsies. PDXs can be engrafted from head and neck cancer (HNC) samples across a wide range of cancer stages, retain the genetic features of their human source, and can be treated with both chemotherapy and radiation, allowing for clinically relevant studies. Not only do PDXs allow for the study of patient tissues in an in vivo model, they can also provide a renewable source of cancer cells for organoid cultures. Herein, we review the uses of HNC patient-derived models for radiation research, including approaches to establishing both orthotopic and heterotopic PDXs, approaches and potential pitfalls to delivering chemotherapy and radiation to these animal models, biological advantages and limitations, and alternatives to animal studies that still use patient-derived tissues.

11.
Int J Cancer ; 146(7): 2047-2058, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31732968

ABSTRACT

More than one-third of patients with locally advanced cervical cancer do not respond to chemoradiation therapy (CRT). We aimed to characterize the transcriptional landscape of paired human cervical tumors before and during CRT in order to gain insight into the evolution of treatment response and to elucidate mechanisms of treatment resistance. We prospectively collected cervical tumor biopsies from 115 patients both before and 3 weeks into CRT. RNA-sequencing, Gene Set Enrichment Analysis and HPV gene expression were performed on 20 paired samples that had adequate neoplastic tissue mid-treatment. Tumors from patients with no evidence of disease (NED) at last follow-up had enrichment in pathways related to the immune response both pretreatment and mid-treatment, while tumors from patients dead of disease (DOD) demonstrated enrichment in biosynthetic and mitotic pathways but not in immune-related pathways. Patients DOD had decreased expression of T-cell and cytolytic genes and increased expression of PD-L2 mid-treatment compared to patients NED. Histological and immunohistochemical analysis revealed a decrease in tumor-associated lymphocytes (TAL) during CRT in all patients but tumors from patients DOD had a significantly more pronounced decrease in TALs and CD8+ cells mid-treatment, which was validated in a larger mid-treatment cohort. Finally, patients DOD retained more HPV E6/E7 gene expression during CRT and this was associated with increased expression of genes driving mitosis, which was corroborated in vitro. Our results suggest that decreased local immune response and retained HPV gene expression may be acting together to promote treatment resistance during CRT in patients with cervical cancer.


Subject(s)
Drug Resistance, Neoplasm , Gene Expression Regulation, Viral , Immunomodulation/drug effects , Papillomaviridae/genetics , Papillomavirus Infections/complications , Papillomavirus Infections/virology , Uterine Cervical Neoplasms/etiology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , Combined Modality Therapy , Drug Resistance, Neoplasm/immunology , Female , Gene Expression Profiling , Humans , Lymphatic Metastasis , Neoplasm Staging , Papillomaviridae/classification , Papillomaviridae/immunology , Papillomavirus Infections/immunology , Prognosis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/mortality
12.
Radiat Oncol ; 12(1): 198, 2017 Dec 08.
Article in English | MEDLINE | ID: mdl-29216884

ABSTRACT

BACKGROUND: Peri-operative radiation of retroperitoneal sarcomas (RPS) is an important component of multidisciplinary treatment. All retrospective series thus far included patients treated with older radiation therapy (RT) techniques including 2D and 3DRT. Intensity modulated radiation therapy (IMRT) allows for selective dose escalation while sparing adjacent organs. We therefore report the first series of patients with RPS treated solely with IMRT, surgery and chemotherapy. We hypothesized that IMRT would permit safe dose escalation and superior rates of local control (LC) in this high-risk patient population. METHODS: Thirty patients with RPS treated with curative intent between 2006 and 2015 were included in this retrospective study. RT was administered either pre- or post-operatively and IMRT was used in all patients. Statistical comparisons, LC, distant metastasis (DM), and overall survival (OS) were calculated by Kaplan-Meier analysis and univariate Cox regression. RESULTS: Median follow-up time after completion of RT was 36 months (range 1.4-112). Median tumor size was 14 cm (range 3.6 - 28 cm). The most prevalent histologies were liposarcoma in 10 (33%) patients and leiomyosarcoma in 10 (33%) with 21 patients (70%) having high-grade disease. Twenty-eight (93%) patients had surgical resection with 47% having positive margins. Chemotherapy was administered in 9 (30%) patients. RT was delivered pre-operatively in 11 (37%) patients, and post-operatively in 19 (63%) with 60% of patients receiving a simultaneous integrated boost. Pre-operative median RT dose to the high-risk area was 55 Gy (range, 43-66 Gy) while median post-operative dose was 60.4 Gy (range, 45-66.6 Gy). There was one acute grade 3 and one late grade 3 toxicity and no grade 4 or 5 toxicities. Three year actuarial LC, freedom from DM, and OS rates were 84%, 64%, and 68% respectively. Positive surgical margins were associated with a higher risk of local recurrence (p = 0.02) and decreased OS (p = 0.04). Pre-operative RT was associated with improved LC (p = 0.1) with a 5-year actuarial LC of 100%. Administration of chemotherapy, timing of RT, histology or grade was not predictive of OS. CONCLUSIONS: Patients with RPS treated with peri-operative IMRT at our institution had excellent local control and low incidences of toxicity.


Subject(s)
Neoplasm Recurrence, Local/pathology , Radiotherapy, Intensity-Modulated/methods , Retroperitoneal Neoplasms/pathology , Sarcoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/radiotherapy , Neoplasm Recurrence, Local/surgery , Prognosis , Radiotherapy Dosage , Retroperitoneal Neoplasms/radiotherapy , Retroperitoneal Neoplasms/surgery , Retrospective Studies , Sarcoma/radiotherapy , Sarcoma/surgery , Survival Rate , Young Adult
13.
Sarcoma ; 2017: 3761292, 2017.
Article in English | MEDLINE | ID: mdl-28546782

ABSTRACT

Background and Objectives. Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft tissue sarcomas with poor overall survival. Response to chemotherapy has been debated for these tumors. Methods. We performed a retrospective analysis of the patients at our institution with a biopsy-proven diagnosis of MPNST that underwent neoadjuvant chemotherapy prior to surgery. Results. We retrospectively identified five patients who received neoadjuvant chemotherapy with epirubicin and ifosfamide that demonstrated a 30% reduction in tumor growth and a 60% response rate by RECIST criteria. Additionally, a metabolic response was observed in all three patients who received serial PET scans during neoadjuvant treatment. The clinical benefit rate, which includes stable disease, was 100%. Conclusions. Our data suggest that MPNSTs do respond to epirubicin and ifosfamide based chemotherapy and prospective studies are warranted to further define the clinical benefit.

14.
Am J Pathol ; 181(6): 2038-46, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23058369

ABSTRACT

Interferon-γ (IFN-γ), a proinflammatory cytokine, has been implicated in the pathogenesis of a number of forms of heart disease including myocarditis and congestive heart failure. In fact, overexpression of IFN-γ in mice causes dilated cardiomyopathy. However, the direct effects of IFN-γ on cardiac myocytes and the mechanism by which it causes cardiac dysfunction have not been described. Here, we present the molecular pathology of IFN-γ exposure and its effect on myofibrillar proteins in isolated neonatal rat ventricular myocytes. Treatment with IFN-γ caused cardiac myocyte atrophy attributable to a specific decrease in myosin heavy chain protein. This selective degradation of myosin heavy chain was not accompanied by a decrease in total protein synthesis or by an increase in total protein degradation. IFN-γ increased both proteasome and immunoproteasome activity in cardiac myocytes and their inhibition blocked myosin heavy chain loss and myocyte atrophy, whereas inhibition of the lysosome or autophagosome did not. Collectively, these results provide a mechanism by which IFN-γ causes cardiac pathology in the setting of chronic inflammatory diseases.


Subject(s)
Interferon-gamma/adverse effects , Myocarditis/metabolism , Myocarditis/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myosin Heavy Chains/metabolism , Proteolysis , Animals , Animals, Newborn , Atrophy/metabolism , Atrophy/pathology , Chronic Disease , Disease Models, Animal , Heart Ventricles/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Myocytes, Cardiac/drug effects , Phagosomes/drug effects , Phagosomes/metabolism , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis/drug effects , Proteolysis/drug effects , Rats , Ubiquitin/metabolism
15.
Int J Cancer ; 130(11): 2722-7, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-21796617

ABSTRACT

Cachexia is a severe muscle-wasting syndrome associated with several chronic diseases such as cancer and AIDS. Muscle mass loss significantly decreases prognosis and survival. The mechanisms of muscle atrophy and the specific proteins targeted for degradation have been intensely studied and are potential therapeutic targets. Published reports that myosin heavy chain (MyHC), the most abundant protein by mass in skeletal muscle, is selectively targeted for degradation in cancer cachexia remain controversial. Here we show that the results of previous studies showing a selective decrease in MyHC are likely an artifact resulting from muscle lysis methods which do not solubilize myosin out of myofibrils. We show that MyHC decreases in parallel with other myofibrillar proteins in cachectic skeletal muscle, which has mechanistic and therapeutic implications. These findings should lead to mechanistic insight into the stoichiometry of sarcomeric disassembly and degradation during cancer cachexia.


Subject(s)
Cachexia/metabolism , Myosin Heavy Chains/metabolism , Neoplasms, Experimental/metabolism , Animals , Cachexia/etiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Muscle, Skeletal/metabolism , Neoplasms, Experimental/complications
16.
Cancer Res ; 71(5): 1710-20, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21163868

ABSTRACT

Approximately one-third of cancer deaths are caused by cachexia, a severe form of skeletal muscle and adipose tissue wasting that affects men more than women. The heart also undergoes atrophy in cancer patients, but the mechanisms and the basis for apparent sex differences are unclear. In a mouse colon-adenocarcinoma model, cancer causes a loss of cardiac mass due to a decrease in cardiac myocyte size that is associated with reduced levels of all sarcomeric proteins. Unlike skeletal muscle cachexia, atrophic hearts do not upregulate the ubiquitin-proteasome system or its activity but increase autophagy. Thus, cancer causes cardiac atrophy by a mechanism distinct from that in skeletal muscle. Male tumor-bearing mice have a more severe phenotype than females, including greater cardiac mass loss and mortality, a more robust pro-inflammatory response to the tumor, and greater cardiac autophagy. In females, estrogen protects against cancer-induced cardiac atrophy and body weight loss by signaling through its receptor. Sex differences in cardiac atrophy need to be considered during the treatment of patients suffering from chemotherapy-induced cardiomyopathy to prevent exacerbation of cardiac dysfunction.


Subject(s)
Autophagy/physiology , Heart Diseases/etiology , Heart Diseases/pathology , Neoplasms, Experimental/complications , Sex Characteristics , Adenocarcinoma/complications , Animals , Atrophy/etiology , Blotting, Western , Colonic Neoplasms/complications , Estrogens/metabolism , Female , Heart Diseases/physiopathology , Immunohistochemistry , Male , Mice , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
Mol Carcinog ; 41(4): 187-96, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15390079

ABSTRACT

Pre-mRNA processing is an important mechanism for globally modifying cellular protein composition during tumorigenesis. To understand this process during lung cancer, expression of two key pre-mRNA alternative splicing factors was compared in a mouse model of early lung carcinogenesis and during regenerative growth following reversible lung injury. Heterogeneous nuclear ribonucleoprotein (hnRNP) A1 and alternative splicing factor/splicing factor 2 (ASF/SF2) act antagonistically to modulate splice site selection. Both hnRNP A1 and ASF/SF2 contents rose in adenomas and during injury-induced hyperplasia compared to control lungs, as measured by immunoblotting. While both proteins increased similarly during compensatory hyperplasia, hnRNP A1 increased to a much greater extent than ASF/SF2 in tumors, resulting in a 6-fold increase of the hnRNP A1 to ASF/SF2 ratio. Immunohistochemical analysis showed that hnRNP A1 localized exclusively within tumor nuclei, while ASF/SF2 appeared in cytoplasm and/or nuclei, depending on the growth pattern of the tumor cells. We also demonstrated cancer-associated changes in the pre-mRNA alternative splicing of CD44, a membrane glycoprotein involved in cell-cell and cell-extracellular matrix interactions. hnRNP A1 and ASF/SF2 expression is thus differentially altered in neoplastic lung cells by mechanisms that do not strictly arise from increased cell division. These changes are influenced by tumor histology and may be associated with production of variant CD44 mRNA isoforms.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , RNA Precursors/metabolism , RNA Splicing , RNA-Binding Proteins/metabolism , Alternative Splicing/genetics , Animals , Butylated Hydroxytoluene/pharmacology , Cell Proliferation , Cell Transformation, Neoplastic/chemically induced , Heterogeneous Nuclear Ribonucleoprotein A1 , Hyaluronan Receptors/genetics , Hyperplasia/chemically induced , Hyperplasia/metabolism , Lung Neoplasms/chemically induced , Male , Mice , Mice, Inbred BALB C , Neoplasm Staging , Protein Isoforms/genetics , Protein Transport , RNA Precursors/genetics , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine-Arginine Splicing Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...