Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.949
Filter
2.
Sci Rep ; 14(1): 9876, 2024 04 30.
Article in English | MEDLINE | ID: mdl-38684776

ABSTRACT

The purpose of this study is to investigate the association between handgrip strength (HGS) and health-related quality of life (HRQoL), demonstrating HGS as an effective indicator for evaluating HRQoL of patients with cancer. Analyzing 1657 Korean adult cancer patients (644 males, 1013 females) aged ≥ 20 years from the Korea National Health and Nutrition Examination Survey (2014-2019), HGS was standardized based on body mass index and categorized by sex. HRQoL was assessed using the Euro Quality of Life-5-Dimension 3-Level version (EQ-5D-3L) Index. Lower relative HGS was associated with decreased HRQoL in female patients, while no significant association was found in male patients. The lowest quartile of relative HGS exhibited a 2.5-fold decrease in HRQoL compared to the highest quartile (OR 2.50, 95% CI 1.59-3.95, p < 0.001). Both male and female patients with cancer were affected by age, subjective health perception, and stress recognition regarding HRQoL. This study suggests that HGS may be associated with the HRQoL of female patients with cancer, emphasizing that the HGS measurement can be effectively utilized as a pivotal tool for evaluating HRQoL in female patients with cancer.


Subject(s)
Hand Strength , Neoplasms , Quality of Life , Humans , Female , Male , Hand Strength/physiology , Neoplasms/physiopathology , Neoplasms/psychology , Middle Aged , Adult , Republic of Korea , Aged , Sex Factors , Nutrition Surveys , Young Adult , Sex Characteristics
3.
Rev. Asoc. Méd. Argent ; 137(1): 4-10, mar. 2024.
Article in Spanish | LILACS | ID: biblio-1552830

ABSTRACT

Se exponen los hallazgos históricos y la importancia biológica de los telómeros en la vida celular y en los aspectos genéticos del ADN humano. (AU)


The discovery and the biological importance of the telomeres are exposed. (AU)


Subject(s)
Humans , DNA/genetics , Telomere/physiology , Telomere/genetics , Telomerase/physiology , Telomerase/genetics , Aging/physiology , DNA/metabolism , Cellular Senescence , Telomerase/metabolism , DNA Replication/physiology , Telomere Shortening , Neoplasms/physiopathology
4.
Curr Mol Pharmacol ; 17: e18761429273223, 2024.
Article in English | MEDLINE | ID: mdl-38389419

ABSTRACT

Mitosis of somatic cells produces a daughter cell. Apoptosis, a naturally programmed cellular death mechanism, kills abnormal cells produced by mitosis. Cancer can develop when this equilibrium is disrupted, either by an upsurge in cell propagation or a reduction in tissue demise. Cancer therapy aims to cause cancer cells to die while inflicting little harm to healthy cells. This review of apoptotic mechanism processes improves our understanding of how certain malignancies begin and develop. The current cancer treatments can operate either by inducing apoptosis or causing direct cell damage. An insight into the resistance to apoptosis may explicate why malignancy treatments fail in some situations. New therapies grounded on our understanding of apoptotic processes are being developed to induce apoptosis of cancer cells while limiting the simultaneous death of normal cells. Various biological activities require redox equilibrium to function properly. Antineoplastic medications that cause oxidative stress by raising ROS and blocking antioxidant mechanisms have recently attracted much interest. The rapid accumulation of ROS impairs redox balance and damages cancer cells severely. Here, we discuss ROS-instigating malignancy therapy and the antineoplastic mechanism used by prooxidative drugs.


Subject(s)
Antineoplastic Agents , Apoptosis , Neoplasms , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Cell Death , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/physiopathology , Reactive Oxygen Species/metabolism
5.
IEEE J Biomed Health Inform ; 28(5): 2636-2649, 2024 May.
Article in English | MEDLINE | ID: mdl-38381641

ABSTRACT

Monitoring vital signs is a key part of standard medical care for cancer patients. However, the traditional methods have instability especially when big fluctuations of signals happen, while the deep-learning-based methods lack pertinence to the sensors. A dual-path micro-bend optical fiber sensor and a targeted model based on the Divided-Frequency-CNN (DFC) are developed in this paper to measure the heart rate (HR) and respiratory rate (RR). For each path, features of frequency division based on the mechanism of signal periodicity cooperate with the operation of stable phase extraction to reduce the interference of body movements for monitoring. Then, the DFC model is designed to learn the inner information from the features robustly. Lastly, a weighted strategy is used to estimate the HR and RR via dual paths to increase the anti-interference for errors from one source. The experiments were carried out on the actual clinical data of cancer patients by a hospital. The results show that the proposed method has good performance in error (3.51 (4.51 %) and 2.53 (3.28 %) beats per minute (bpm) for cancer patients with pain and without pain respectively), relevance, and consistency with the values from hospital equipment. Besides, the proposed method significantly improved the ability in the report time interval (30 to 9 min), and mean / confidential interval (3.60/[-22.61,29.81] to -0.64 / [-9.21,7.92] for patients with pain and 1.87 / [-5.49,9.23] to -0.16 / [-6.21,5.89] for patients without pain) compared with our previous work.


Subject(s)
Heart Rate , Neoplasms , Respiratory Rate , Signal Processing, Computer-Assisted , Vital Signs , Humans , Neoplasms/physiopathology , Monitoring, Physiologic/methods , Monitoring, Physiologic/instrumentation , Vital Signs/physiology , Heart Rate/physiology , Respiratory Rate/physiology , Neural Networks, Computer , Male , Deep Learning , Female , Middle Aged , Adult
7.
Nucleic Acids Res ; 52(D1): D1246-D1252, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37956338

ABSTRACT

Advancements in high-throughput technology offer researchers an extensive range of multi-omics data that provide deep insights into the complex landscape of cancer biology. However, traditional statistical models and databases are inadequate to interpret these high-dimensional data within a multi-omics framework. To address this limitation, we introduce DriverDBv4, an updated iteration of the DriverDB cancer driver gene database (http://driverdb.bioinfomics.org/). This updated version offers several significant enhancements: (i) an increase in the number of cohorts from 33 to 70, encompassing approximately 24 000 samples; (ii) inclusion of proteomics data, augmenting the existing types of omics data and thus expanding the analytical scope; (iii) implementation of multiple multi-omics algorithms for identification of cancer drivers; (iv) new visualization features designed to succinctly summarize high-context data and redesigned existing sections to accommodate the increased volume of datasets and (v) two new functions in Customized Analysis, specifically designed for multi-omics driver identification and subgroup expression analysis. DriverDBv4 facilitates comprehensive interpretation of multi-omics data across diverse cancer types, thereby enriching the understanding of cancer heterogeneity and aiding in the development of personalized clinical approaches. The database is designed to foster a more nuanced understanding of the multi-faceted nature of cancer.


Subject(s)
Databases, Genetic , Multiomics , Neoplasms , Humans , Algorithms , Databases, Genetic/standards , Neoplasms/genetics , Neoplasms/physiopathology
8.
J Biol Chem ; 300(1): 105492, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38000655

ABSTRACT

Homozygous 5'-methylthioadenosine phosphorylase (MTAP) deletions occur in approximately 15% of human cancers. Co-deletion of MTAP and methionine adenosyltransferase 2 alpha (MAT2a) induces a synthetic lethal phenotype involving protein arginine methyltransferase 5 (PRMT5) inhibition. MAT2a inhibitors are now in clinical trials for genotypic MTAP-/- cancers, however the MTAP-/- genotype represents fewer than 2% of human colorectal cancers (CRCs), limiting the utility of MAT2a inhibitors in these and other MTAP+/+ cancers. Methylthio-DADMe-immucillin-A (MTDIA) is a picomolar transition state analog inhibitor of MTAP that renders cells enzymatically MTAP-deficient to induce the MTAP-/- phenotype. Here, we demonstrate that MTDIA and MAT2a inhibitor AG-270 combination therapy mimics synthetic lethality in MTAP+/+ CRC cell lines with similar effects in mouse xenografts and without adverse histology on normal tissues. Combination treatment is synergistic with a 104-fold increase in drug potency for inhibition of CRC cell growth in culture. Combined MTDIA and AG-270 decreases S-adenosyl-L-methionine and increases 5'-methylthioadenosine in cells. The increased intracellular methylthioadenosine:S-adenosyl-L-methionine ratio inhibits PRMT5 activity, leading to cellular arrest and apoptotic cell death by causing MDM4 alternative splicing and p53 activation. Combination MTDIA and AG-270 treatment differs from direct inhibition of PRMT5 by GSK3326595 by avoiding toxicity caused by cell death in the normal gut epithelium induced by the PRMT5 inhibitor. The combination of MTAP and MAT2a inhibitors expands this synthetic lethal approach to include MTAP+/+ cancers, especially the remaining 98% of CRCs without the MTAP-/- genotype.


Subject(s)
Deoxyadenosines , Methionine Adenosyltransferase , Neoplasms , Protein-Arginine N-Methyltransferases , Purine-Nucleoside Phosphorylase , S-Adenosylmethionine , Animals , Humans , Mice , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxyadenosines/antagonists & inhibitors , Deoxyadenosines/genetics , Deoxyadenosines/metabolism , Drug Synergism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Methionine Adenosyltransferase/antagonists & inhibitors , Methionine Adenosyltransferase/genetics , Methionine Adenosyltransferase/metabolism , Neoplasms/genetics , Neoplasms/physiopathology , Neoplasms/therapy , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Purine-Nucleoside Phosphorylase/genetics , Purine-Nucleoside Phosphorylase/metabolism , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , S-Adenosylmethionine/metabolism
9.
J Biol Chem ; 300(1): 105465, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37979915

ABSTRACT

Calreticulin (CRT) was originally identified as a key calcium-binding protein of the endoplasmic reticulum. Subsequently, CRT was shown to possess multiple intracellular functions, including roles in calcium homeostasis and protein folding. Recently, several extracellular functions have been identified for CRT, including roles in cancer cell invasion and phagocytosis of apoptotic and cancer cells by macrophages. In the current report, we uncover a novel function for extracellular CRT and report that CRT functions as a plasminogen-binding receptor that regulates the conversion of plasminogen to plasmin. We show that human recombinant or bovine tissue-derived CRT dramatically stimulated the conversion of plasminogen to plasmin by tissue plasminogen activator or urokinase-type plasminogen activator. Surface plasmon resonance analysis revealed that CRT-bound plasminogen (KD = 1.8 µM) with moderate affinity. Plasminogen binding and activation by CRT were inhibited by ε-aminocaproic acid, suggesting that an internal lysine residue of CRT interacts with plasminogen. We subsequently show that clinically relevant CRT variants (lacking four or eight lysines in carboxyl-terminal region) exhibited decreased plasminogen activation. Furthermore, CRT-deficient fibroblasts generated 90% less plasmin and CRT-depleted MDA MB 231 cells also demonstrated a significant reduction in plasmin generation. Moreover, treatment of fibroblasts with mitoxantrone dramatically stimulated plasmin generation by WT but not CRT-deficient fibroblasts. Our results suggest that CRT is an important cellular plasminogen regulatory protein. Given that CRT can empower cells with plasmin proteolytic activity, this discovery may provide new mechanistic insight into the established role of CRT in cancer.


Subject(s)
Calreticulin , Plasminogen , Animals , Cattle , Humans , Calreticulin/genetics , Calreticulin/isolation & purification , Calreticulin/metabolism , Fibrinolysin/metabolism , Plasminogen/genetics , Plasminogen/metabolism , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Protein Domains/genetics , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Gene Knockout Techniques , Cell Line, Tumor , Neoplasms/physiopathology
10.
BMC Womens Health ; 23(1): 448, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37620873

ABSTRACT

BACKGROUND: African American (AA) women navigate the world with multiple intersecting marginalized identities. Accordingly, AA women have higher cumulative stress burden or allostatic load (AL) compared to other women. Studies suggest that AA women with a college degree or higher have lower AL than AA women with less than a high school diploma. We examined the joint effect of educational attainment and AL status with long-term risk of cancer mortality, and whether education moderated the association between AL and cancer mortality. METHODS: We performed a retrospective analysis among 4,677 AA women within the National Health and Nutrition Examination Survey (NHANES) from 1988 to 2010 with follow-up data through December 31, 2019. We fit weighted Cox proportional hazards models to estimate adjusted hazard ratios (aHRs) of cancer death between educational attainment/AL (adjusted for age, income, and smoking status). RESULTS: AA women with less than a high school diploma living with high AL had nearly a 3-fold increased risk (unadjusted HR: 2.98; 95%C CI: 1.24-7.15) of cancer death compared to AA college graduates living with low AL. However, after adjusting for age, this effect attenuated (age-adjusted HR: 1.11; 95% CI: 0.45-2.74). AA women with high AL had 2.3-fold increased risk of cancer death (fully adjusted HR: 2.26; 95% CI: 1.10-4.57) when compared to AA with low AL, specifically among women with high school diploma or equivalent and without history of cancer. CONCLUSIONS: Our findings suggest that high allostatic load is associated with a higher risk of cancer mortality among AA women with lower educational attainment, while no such association was observed among AA women with higher educational attainment. Thus, educational attainment plays a modifying role in the relationship between allostatic load and the risk of cancer death for AA women. Higher education can bring several benefits, including improved access to medical care and enhanced medical literacy, which in turn may help mitigate the adverse impact of AL and the heightened risk of cancer mortality among AA women.


Subject(s)
Allostasis , Black or African American , Educational Status , Neoplasms , Female , Humans , Allostasis/physiology , Black or African American/psychology , Neoplasms/ethnology , Neoplasms/mortality , Neoplasms/physiopathology , Neoplasms/psychology , Nutrition Surveys , Retrospective Studies , Stress, Physiological , Stress, Psychological , Risk
11.
J Biol Chem ; 299(9): 105062, 2023 09.
Article in English | MEDLINE | ID: mdl-37468105

ABSTRACT

SUMOylation is a post-translational modification frequently found on nuclear proteins, including transcription factors (TFs) and coactivators. By controlling the activity of several TFs, SUMOylation may have far-reaching effects. MYB is an example of a developmental TF subjected to SUMO-mediated regulation, through both SUMO conjugation and SUMO binding. How SUMO affects MYB target genes is unknown. Here, we explored the global effect of reduced SUMOylation of MYB on its downstream gene programs. RNA-Seq in K562 cells after MYB knockdown and rescue with mutants having an altered SUMO status revealed a number of differentially regulated genes and distinct gene ontology term enrichments. Clearly, the SUMO status of MYB both quantitatively and qualitatively affects its regulome. The transcriptome data further revealed that MYB upregulates the SUMO protease SENP1, a key enzyme that removes SUMO conjugation from SUMOylated proteins. Given this role of SENP1 in the MYB regulome, we expanded the analysis, mapped interaction partners of SENP1, and identified UXT as a novel player affecting the SUMO system by acting as a repressor of SENP1. MYB inhibits the expression of UXT suggesting that MYB is able not only to control a specific gene program directly but also indirectly by affecting the SUMO landscape through SENP1 and UXT. These findings suggest an autoactivation loop whereby MYB, through enhancing SENP1 and reducing UXT, is itself being activated by a reduced level of repressive SUMOylation. We propose that overexpressed MYB, seen in multiple cancers, may drive this autoactivation loop and contribute to oncogenic activation of MYB.


Subject(s)
Cell Cycle Proteins , Gene Expression Regulation , Genes, myb , Peptide Hydrolases , Humans , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Gene Expression Regulation/genetics , Gene Knockdown Techniques , K562 Cells , Neoplasms/physiopathology , Peptide Hydrolases/metabolism , Protein Binding , Sumoylation , Transcriptional Activation
12.
Nature ; 618(7965): 467-479, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37316719

ABSTRACT

The nervous system regulates tissue stem and precursor populations throughout life. Parallel to roles in development, the nervous system is emerging as a critical regulator of cancer, from oncogenesis to malignant growth and metastatic spread. Various preclinical models in a range of malignancies have demonstrated that nervous system activity can control cancer initiation and powerfully influence cancer progression and metastasis. Just as the nervous system can regulate cancer progression, cancer also remodels and hijacks nervous system structure and function. Interactions between the nervous system and cancer occur both in the local tumour microenvironment and systemically. Neurons and glial cells communicate directly with malignant cells in the tumour microenvironment through paracrine factors and, in some cases, through neuron-to-cancer cell synapses. Additionally, indirect interactions occur at a distance through circulating signals and through influences on immune cell trafficking and function. Such cross-talk among the nervous system, immune system and cancer-both systemically and in the local tumour microenvironment-regulates pro-tumour inflammation and anti-cancer immunity. Elucidating the neuroscience of cancer, which calls for interdisciplinary collaboration among the fields of neuroscience, developmental biology, immunology and cancer biology, may advance effective therapies for many of the most difficult to treat malignancies.


Subject(s)
Neoplasms , Neuroimmunomodulation , Neurosciences , Humans , Carcinogenesis , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/physiopathology , Neoplasms/therapy , Neuroglia , Tumor Microenvironment , Neoplasm Metastasis , Disease Progression
13.
Adv Healthc Mater ; 12(26): e2300871, 2023 10.
Article in English | MEDLINE | ID: mdl-37204046

ABSTRACT

Ferroptosis is a form of programmed cell death and plays an important role in many diseases. Dihydroorotate dehydrogenase (DHODH) and glutathione peroxidase 4 (GPX4) play major roles in cell resistance to ferroptosis. Therefore, inactivation of these proteins provides an excellent opportunity for efficient ferroptosis-based synergistic cancer therapy. In this study, a multifunctional nanoagent (BPNpro ) containing a GPX4 targeting boron dipyrromethene (Bodipy) probe (BP) and a DHODH targeting proteolysis targeting chimera (PROTAC) is reported. BPNpro is prepared using a nanoprecipitation method in the presence of a thermoresponsive liposome, where BP is encapsulated inside and the cathepsin B (CatB)-cleavable PROTAC peptide (DPCP) is modified on the outer surface. In the presence of near-infrared (NIR) photoirradiation, BPNpro is melted and BP is released in tumor cells. Subsequently, BP inhibits the activity of GPX4 by covalently bonding with the selenocysteine at the enzyme active site. In addition, DPCP achieves sustained degradation of DHODH upon activation by CatB overexpressed in the tumor. The synergistic deactivation of GPX4 and DHODH induces extensive ferroptosis and subsequent cell death. In vivo and in vitro studies clearly show that the proposed ferroptosis therapy provides excellent antitumor effect.


Subject(s)
Dihydroorotate Dehydrogenase , Ferroptosis , Neoplasms , Humans , Boron , Ferroptosis/genetics , Ferroptosis/physiology , Neoplasms/drug therapy , Neoplasms/physiopathology
14.
Clin. transl. oncol. (Print) ; 25(5): 1135-1144, mayo 2023. ilus
Article in English | IBECS | ID: ibc-219502

ABSTRACT

During the initial stages of gastrulation during embryonic differentiation and wound healing, Cripto-1 is a critical protein for human growth. The epithelial adhesion molecules’ downregulation, the mesenchymal overexpression, and mobile proteins are important mechanisms by which Cripto-1 initiates epithelial to mesenchymal transition (EMT). As a result, the function of Cripto-1 for inducing EMT to increase cell migration is advantageous during embryogenesis; however, it is deleterious during the formation, growth, and malignant tumor metastasis. The majority of malignancies are reported to have elevated levels of Cripto-1. Cripto-1 can modify cancerous cells through its function in EMT, which enables these cells to migrate via the extracellular matrix, bloodstream, and lymphatic vessels, on their way for metastasizing to other organs. The goal of this review is to explain what role Cripto-1 plays in common cancers and to summarize how therapeutic strategies are used to interfere with this molecule to target cancers (AU)


Subject(s)
Humans , Epithelial-Mesenchymal Transition/physiology , Neoplasms/pathology , Neoplasms/physiopathology , Cell Differentiation , Epidermal Growth Factor/metabolism , Neoplasm Proteins/metabolism
15.
J Biol Chem ; 299(4): 104601, 2023 04.
Article in English | MEDLINE | ID: mdl-36907441

ABSTRACT

Plant homeodomain (PHD) fingers are structurally conserved zinc fingers that selectively bind unmodified or methylated at lysine 4 histone H3 tails. This binding stabilizes transcription factors and chromatin-modifying proteins at specific genomic sites, which is required for vital cellular processes, including gene expression and DNA repair. Several PHD fingers have recently been shown to recognize other regions of H3 or histone H4. In this review, we detail molecular mechanisms and structural features of the noncanonical histone recognition, discuss biological implications of the atypical interactions, highlight therapeutic potential of PHD fingers, and compare inhibition strategies.


Subject(s)
Histones , PHD Zinc Fingers , DNA-Binding Proteins/metabolism , Histones/chemistry , Histones/metabolism , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Mice , Neoplasms/genetics , Neoplasms/physiopathology
16.
Cancer ; 129(10): 1557-1568, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36935617

ABSTRACT

BACKGROUND: Discrimination can adversely affect health and accelerate aging, but little is known about these relationships in cancer survivors. This study examines associations of discrimination and aging among self-identified African American survivors. METHODS: A population-based sample of 2232 survivors 20-79 years old at diagnosis were enrolled within 5 years of breast (n = 787), colorectal (n = 227), lung (n = 223), or prostate (n = 995) cancer between 2017 and 2022. Surveys were completed post-active therapy. A deficit accumulation index measured aging-related disease and function (score range, 0-1, where <0.20 is robust, 0.20 to <0.35 is pre-frail, and 0.35+ is frail; 0.06 is a large clinically meaningful difference). The discrimination scale assessed ever experiencing major discrimination and seven types of events (score, 0-7). Linear regression tested the association of discrimination and deficit accumulation, controlling for age, time from diagnosis, cancer type, stage and therapy, and sociodemographic variables. RESULTS: Survivors were an average of 62 years old (SD, 9.6), 63.2% reported ever experiencing major discrimination, with an average of 2.4 (SD, 1.7) types of discrimination events. Only 24.4% had deficit accumulation scores considered robust (mean score, 0.30 [SD, 0.13]). Among those who reported ever experiencing major discrimination, survivors with four to seven types of discrimination events (vs. 0-1) had a large, clinically meaningful increase in adjusted deficits (0.062, p < .001) and this pattern was consistent across cancer types. CONCLUSION: African American cancer survivors have high deficit accumulated index scores, and experiences of major discrimination were positively associated with these deficits. Future studies are needed to understand the intersectionality between aging, discrimination, and cancer survivorship among diverse populations.


Subject(s)
Aging , Black or African American , Cancer Survivors , Neoplasms , Racism , Social Determinants of Health , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Aging/ethnology , Aging/physiology , Black or African American/statistics & numerical data , Breast Neoplasms/epidemiology , Breast Neoplasms/ethnology , Breast Neoplasms/physiopathology , Neoplasms/epidemiology , Neoplasms/ethnology , Neoplasms/physiopathology , Racism/ethnology , Racism/statistics & numerical data , Social Determinants of Health/ethnology , Social Determinants of Health/statistics & numerical data , Surveys and Questionnaires , Michigan/epidemiology
17.
J Virol ; 97(2): e0187222, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36715516

ABSTRACT

The expression of human papillomavirus (HPV) oncoproteins perturbed multiple cellular events of the host cells, leading to the formation of cancer phenotypes. Our current and previous studies indicated that Aurora kinase A (AurA), a mitotic regulator that is often aberrantly expressed in human cancers, is preferentially bound to E6-encoded by cancer-causing HPV. AurA is believed to be important for the proliferation and survival of HPV-positive cells. Nonetheless, the interaction between AurA and E6, and the mechanism of how this association is involved in carcinogenesis, have not been elucidated clearly. Hence, we performed a series of biochemical assays to characterize the AurA-E6 association and complex formation. We found the C-terminus of E6, upstream of the PDZ binding motif of E6, is important to forming the AurA-E6 complex in the nucleus. We also showed that the expression level of E6 corresponded positively with AurA expression. Meanwhile, the functional consequences of the AurA-E6 association to AurA kinase function and host cellular events were also delineated. Intriguingly, we revealed that AurA-E6 association regulated the expression of cyclin E and phosphor-Histone H3, which are involved in G1/S and mitotic phases of the cell cycle, respectively. Depletion of AurA also reduced the invasive ability of HPV-positive cells. AurA inhibition may not be sufficient to reduce the oncogenic potential exerted by E6. Altogether, our study unleashed the mechanism of how HPVE6 deploy AurA to promote cancer phenotypes, particularly through dysregulation of cell cycle checkpoints and suggests that the AurA-E6 complex possesses a therapeutic value. IMPORTANCE We unveiled the mechanism of how HPV employs Aurora kinase A (AurA) of host cells to exert its oncogenic capability synergistically. We systematically characterized the mode of interaction between E6-encoded by cancer-causing HPV and AurA. Then, we delineated the consequences of AurA-E6 complex formation on AurA kinase function and changes to cellular events at molecular levels. Using a cell-based approach, we unleashed that disruption of AurA-E6 association can halt cancer phenotype exhibited by HPV-positive cancer cells. Our findings are vital for the designing of state-of-the-art therapies for HPV-associated cancers.


Subject(s)
Aurora Kinase A , Human Papillomavirus Viruses , Neoplasms , Papillomavirus Infections , Viral Envelope Proteins , Humans , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Carcinogenesis/pathology , Human Papillomavirus Viruses/genetics , Human Papillomavirus Viruses/metabolism , Papillomavirus Infections/complications , Papillomavirus Infections/virology , Viral Envelope Proteins/metabolism , Gene Expression Regulation, Viral , Neoplasms/etiology , Neoplasms/physiopathology , Neoplasms/virology
18.
J Biol Chem ; 299(3): 102935, 2023 03.
Article in English | MEDLINE | ID: mdl-36693448

ABSTRACT

Although cancer is a genetic disease, physical changes such as stiffening of the extracellular matrix also commonly occur in cancer. Cancer cells sense and respond to extracellular matrix stiffening through the process of mechanotransduction. Cancer cell mechanotransduction can enhance cancer-promoting cell behaviors such as survival signaling, proliferation, and migration. Glycans, carbohydrate-based polymers, have recently emerged as important mediators and/or modulators of cancer cell mechanotransduction. Stiffer tumors are characterized by increased glycan content on cancer cells and their associated extracellular matrix. Here we review the role of cancer-associated glycans in coupled mechanical and biochemical alterations during cancer progression. We discuss the recent evidence on how increased expression of different glycans, in the form of glycoproteins and proteoglycans, contributes to both mechanical changes in tumors and corresponding cancer cell responses. We conclude with a summary of emerging tools that can be used to modify glycans for future studies in cancer mechanobiology.


Subject(s)
Mechanotransduction, Cellular , Neoplasms , Polysaccharides , Humans , Biophysics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Mechanotransduction, Cellular/physiology , Neoplasms/metabolism , Neoplasms/physiopathology , Polysaccharides/metabolism
19.
Cell Death Dis ; 14(1): 57, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36693836

ABSTRACT

There is an urgent need to identify reliable genetic biomarkers for accurate diagnosis, prognosis, and treatment of different tumor types. Described as a prognostic marker for many tumors is the neuronal protein carnitine palmitoyltransferase 1 C (CPT1C). Several studies report that CPT1C is involved in cancer cell adaptation to nutrient depletion and hypoxia. However, the molecular role played by CPT1C in cancer cells is controversial. Most published studies assume that, like canonical CPT1 isoforms, CPT1C is a mediator of fatty acid transport to mitochondria for beta-oxidation, despite the fact that CPT1C has inefficient catalytic activity and is located in the endoplasmic reticulum. In this review, we collate existing evidence on CPT1C in neurons, showing that CPT1C is a sensor of nutrients that interacts with and regulates other proteins involved in lipid metabolism and transport, lysosome motility, and the secretory pathway. We argue, therefore, that CPT1C expression in cancer cells is not a direct regulator of fat burn, but rather is a regulator of lipid metabolic reprograming and cell adaptation to environmental stressors. We also review the clinical relevance of CPT1C as a prognostic indicator and its contribution to tumor growth, cancer invasiveness, and cell senescence. This new and integrated vision of CPT1C function can help better understand the metabolic plasticity of cancer cells and improve the design of therapeutic strategies.


Subject(s)
Carnitine O-Palmitoyltransferase , Neoplasms , Humans , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Hypoxia/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/physiopathology , Neurons/metabolism , Oxidation-Reduction
20.
J Biol Chem ; 299(1): 102766, 2023 01.
Article in English | MEDLINE | ID: mdl-36470425

ABSTRACT

Epidermal growth factor receptor (EGFR) signaling is frequently dysregulated in various cancers. The ubiquitin ligase Casitas B-lineage lymphoma proto-oncogene (Cbl) regulates degradation of activated EGFR through ubiquitination and acts as an adaptor to recruit proteins required for trafficking. Here, we used stable isotope labeling with amino acids in cell culture mass spectrometry to compare Cbl complexes with or without epidermal growth factor (EGF) stimulation. We identified over a hundred novel Cbl interactors, and a secondary siRNA screen found that knockdown of Flotillin-2 (FLOT2) led to increased phosphorylation and degradation of EGFR upon EGF stimulation in HeLa cells. In PC9 and H441 cells, FLOT2 knockdown increased EGF-stimulated EGFR phosphorylation, ubiquitination, and downstream signaling, reversible by EGFR inhibitor erlotinib. CRISPR knockout (KO) of FLOT2 in HeLa cells confirmed EGFR downregulation, increased signaling, and increased dimerization and endosomal trafficking. Furthermore, we determined that FLOT2 interacted with both Cbl and EGFR. EGFR downregulation upon FLOT2 loss was Cbl dependent, as coknockdown of Cbl and Cbl-b restored EGFR levels. In addition, FLOT2 overexpression decreased EGFR signaling and growth. Overexpression of wildtype (WT) FLOT2, but not the soluble G2A FLOT2 mutant, inhibited EGFR phosphorylation upon EGF stimulation in HEK293T cells. FLOT2 loss induced EGFR-dependent proliferation and anchorage-independent growth. Lastly, FLOT2 KO increased tumor formation and tumor volume in nude mice and NSG mice, respectively. Together, these data demonstrated that FLOT2 negatively regulated EGFR activation and dimerization, as well as its subsequent ubiquitination, endosomal trafficking, and degradation, leading to reduced proliferation in vitro and in vivo.


Subject(s)
ErbB Receptors , Neoplasms , Proto-Oncogene Proteins c-cbl , Animals , Humans , Mice , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , HEK293 Cells , HeLa Cells , Mice, Nude , Neoplasms/genetics , Neoplasms/physiopathology , Phosphorylation , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism , Ubiquitination , Membrane Proteins/metabolism , Proteolysis , Gene Expression Regulation, Neoplastic
SELECTION OF CITATIONS
SEARCH DETAIL
...