Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Sci Adv ; 10(14): eadj7540, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38579004

ABSTRACT

Fewer than 20% of triple-negative breast cancer patients experience long-term responses to mainstay chemotherapy. Resistant tumor subpopulations use alternative metabolic pathways to escape therapy, survive, and eventually recur. Here, we show in vivo, longitudinal metabolic reprogramming in residual disease and recurrence of triple-negative breast cancer xenografts with varying sensitivities to the chemotherapeutic drug paclitaxel. Optical imaging coupled with metabolomics reported an increase in non-glucose-driven mitochondrial metabolism and an increase in intratumoral metabolic heterogeneity during regression and residual disease in resistant MDA-MB-231 tumors. Conversely, sensitive HCC-1806 tumors were primarily reliant on glucose uptake and minimal changes in metabolism or heterogeneity were observed over the tumors' therapeutic life cycles. Further, day-matched resistant HCC-1806 tumors revealed a higher reliance on mitochondrial metabolism and elevated metabolic heterogeneity compared to sensitive HCC-1806 tumors. Together, metabolic flexibility, increased reliance on mitochondrial metabolism, and increased metabolic heterogeneity are defining characteristics of persistent residual disease, features that will inform the appropriate type and timing of therapies.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Triple Negative Breast Neoplasms , Humans , Metabolic Reprogramming , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Optical Imaging , Cell Line, Tumor
2.
Contemp Clin Trials Commun ; 38: 101267, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38419810

ABSTRACT

Background: The use of a second informant (co-respondent) is a common method of identifying potential bias in outcome data (e.g., parent-report child outcomes). There is, however, limited evidence regarding methods of increasing response rates from co-respondents. The use of financial incentives is associated with higher levels of engagement and follow-up data collection in online surveys. This study investigated whether financial incentives paid to index participants in an online trial of a parenting-focused intervention, would lead to higher levels of co-respondent data collection. Methods: A study within a trial (SWAT) using a parallel group RCT design. Participants in the host study (an RCT of an online intervention) were randomised into one of two SWAT arms: received/did not receive a £15 voucher when referred co-respondent completed baseline measures. Primary outcome was completion (No/Yes) of Spence Children's Anxiety Scale (SCAS or SCAS-Pre) at baseline. Additional analysis explored impact of incentives on data quality. Results: Intention to treat analysis of 899 parents (183 co-respondents) in the no-incentive arm, and 911 parents (199 co-respondents) in incentive arm. Nomination of co-respondents was similar between incentive arms. The RR for the incentive arm compared to the no incentive arm was 1.13 (95% CI: 0.91 to 1.41, p = 0.264) indicating that incentives did not impact completion of outcomes by consented co-respondents. There were no indications of different data quality between arms. Discussion: The finding that payment of financial incentives to index participant does not lead to greater levels of co-respondent outcome completion suggests that careful consideration should be made before allocating resources in this way in future trials. Trial registration: The host study was registered at Study Record | ClinicalTrials.gov and the SWAT study was registered in the SWAT Store | The Northern Ireland Network for Trials Methodology Research (qub.ac.uk): SWAT number 143: Filetoupload,1099612,en.pdf (qub.ac.uk).

3.
bioRxiv ; 2023 Jul 03.
Article in English | MEDLINE | ID: mdl-37461590

ABSTRACT

APOBEC mutagenesis is one of the most common endogenous sources of mutations in human cancer and is a major source of genetic intratumor heterogeneity. High levels of APOBEC mutagenesis are associated with poor prognosis and aggressive disease across diverse cancers, but the mechanistic and functional impacts of APOBEC mutagenesis on tumor evolution and therapy resistance remain relatively unexplored. To address this, we investigated the contribution of APOBEC mutagenesis to acquired therapy resistance in a model of EGFR-mutant non-small cell lung cancer. We find that inhibition of EGFR in lung cancer cells leads to a rapid and pronounced induction of APOBEC3 expression and activity. Functionally, APOBEC expression promotes the survival of drug-tolerant persister cells (DTPs) following EGFR inhibition. Constitutive expression of APOBEC3B alters the evolutionary trajectory of acquired resistance to the EGFR inhibitor gefitinib, making it more likely that resistance arises through de novo acquisition of the T790M gatekeeper mutation and squamous transdifferentiation during the DTP state. APOBEC3B expression is associated with increased expression of the squamous cell transcription factor ΔNp63 and squamous cell transdifferentiation in gefitinib-resistant cells. Knockout of ΔNp63 in gefitinibresistant cells reduces the expression of the p63 target genes IL1a/b and sensitizes these cells to the thirdgeneration EGFR inhibitor osimertinib. These results suggest that APOBEC activity promotes acquired resistance by facilitating evolution and transdifferentiation in DTPs, and suggest that approaches to target ΔNp63 in gefitinib-resistant lung cancers may have therapeutic benefit.

4.
Trends Cancer ; 9(7): 554-565, 2023 07.
Article in English | MEDLINE | ID: mdl-37150627

ABSTRACT

Tumor recurrence following potentially curative therapy constitutes a major obstacle to achieving cures in patients with cancer. Recurrent tumors frequently arise from a population of residual cancer cells - also referred to as minimal residual disease (RD) or persister cells - that survive therapy and persist for prolonged periods prior to tumor relapse. While there has been significant recent progress in deciphering tumor-cell-intrinsic pathways that regulate residual cancer cell survival and recurrence, much less is known about how the tumor microenvironment (TME) of residual tumors impacts persister cancer cells or tumor recurrence. In this review, we highlight recent studies exploring the regulation and function of immune cells in RD and discuss therapeutic opportunities to target immune cells in residual tumors.


Subject(s)
Neoplasm Recurrence, Local , Tumor Microenvironment , Humans , Neoplasm, Residual , Neoplasm Recurrence, Local/pathology , Cell Line, Tumor , Cell Survival
5.
Contemp Clin Trials Commun ; 32: 101090, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36865678

ABSTRACT

Background: Parent-report questionnaires are a common method of generating data on child outcomes in mental health studies. A second report from another person who knows the child (co-respondent) is implemented to reduce bias and increase objectivity. The success of this approach is dependent on the engagement of co-respondents, which can be difficult. Financial incentives are used to increase data return in clinical trials, and to promote referral rates in online marketing. This protocol describes the use of an embedded randomised controlled trial (RCT) to investigate the effect of financial incentives on rates of co-respondent data completion. In the host RCT (of an online intervention designed to reduce the impact of a parent's anxiety on their child) index participants (i.e. parents) are asked to invite a co-respondent to complete measures on the index child. This study will test the hypothesis that providing monetary incentives to index participants will increase the outcome measure completion rate of co-respondents. Methods: Embedded RCT of two parallel groups. Participants in the intervention arm will be sent a £10 voucher if their chosen co-respondent completes online baseline measures. Participants in the control arm will not be offered payment regardless of their chosen co-respondent's behaviour. 1754 participants will take part. Analysis will compare co-respondent outcome measure completion rates between the two arms at baseline and follow-up. Conclusion: Findings from this study will provide evidence on the impact of offering payment to index participants on return rates of co-respondent data. This will inform resource allocation within future clinical trials.

6.
JMIR Res Protoc ; 11(11): e40707, 2022 Nov 10.
Article in English | MEDLINE | ID: mdl-36355406

ABSTRACT

BACKGROUND: Anxiety is the most common childhood mental health condition and is associated with impaired child outcomes, including increased risk of mental health difficulties in adulthood. Anxiety runs in families: when a parent has anxiety, their child has a 50% higher chance of developing it themselves. Environmental factors are predominant in the intergenerational transmission of anxiety and, of these, parenting processes play a major role. Interventions that target parents to support them to limit the impact of any anxiogenic parenting behaviors are associated with reduced anxiety in their children. A brief UK-based group intervention delivered to parents within the UK National Health Service led to a 16% reduction in children meeting the criteria for an anxiety disorder. However, this intervention is not widely accessible. To widen access, a 9-module web-based version of this intervention has been developed. This course comprises psychoeducation and home practice delivered through text, video, animations, and practice tasks. OBJECTIVE: This study seeks to evaluate the feasibility of delivering this web-based intervention and assess its effectiveness in reducing child anxiety symptoms. METHODS:  This is the protocol for a randomized controlled trial (RCT) of a community sample of 1754 parents with self-identified high levels of anxiety with a child aged 2-11 years. Parents in the intervention arm will receive access to the web-based course, which they undertake at a self-determined rate. The control arm receives no intervention. Follow-up data collection is at months 6 and months 9-21. Intention-to-treat analysis will be conducted on outcomes including child anxiety, child mental health symptoms, and well-being; parental anxiety and well-being; and parenting behaviors. RESULTS: Funding was received in April 2020, and recruitment started in February 2021 and is projected to end in October 2022. A total of 1350 participants have been recruited as of May 2022. CONCLUSIONS: The results of this RCT will provide evidence on the utility of a web-based course in preventing intergenerational transmission of anxiety and increase the understanding of familial anxiety. TRIAL REGISTRATION: ClinicalTrials.gov NCT04755933; https://clinicaltrials.gov/ct2/show/NCT04755933. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): DERR1-10.2196/40707.

7.
NPJ Breast Cancer ; 8(1): 111, 2022 Sep 26.
Article in English | MEDLINE | ID: mdl-36163365

ABSTRACT

Recurrent cancer cells that evade therapy is a leading cause of death in breast cancer patients. This risk is high for women showing an overexpression of human epidermal growth factor receptor 2 (Her2). Cells that persist can rely on different substrates for energy production relative to their primary tumor counterpart. Here, we characterize metabolic reprogramming related to tumor dormancy and recurrence in a doxycycline-induced Her2+/Neu model of breast cancer with varying times to recurrence using longitudinal fluorescence microscopy. Glucose uptake (2-NBDG) and mitochondrial membrane potential (TMRE) imaging metabolically phenotype mammary tumors as they transition to regression, dormancy, and recurrence. "Fast-recurrence" tumors (time to recurrence ~55 days), transition from glycolysis to mitochondrial metabolism during regression and this persists upon recurrence. "Slow-recurrence" tumors (time to recurrence ~100 days) rely on both glycolysis and mitochondrial metabolism during recurrence. The increase in mitochondrial activity in fast-recurrence tumors is attributed to a switch from glucose to fatty acids as the primary energy source for mitochondrial metabolism. Consequently, when fast-recurrence tumors receive treatment with a fatty acid inhibitor, Etomoxir, tumors report an increase in glucose uptake and lipid synthesis during regression. Treatment with Etomoxir ultimately prolongs survival. We show that metabolic reprogramming reports on tumor recurrence characteristics, particularly at time points that are essential for actionable targets. The temporal characteristics of metabolic reprogramming will be critical in determining the use of an appropriate timing for potential therapies; namely, the notion that metabolic-targeted inhibition during regression reports long-term therapeutic benefit.

8.
Behav Res Methods ; 54(3): 1148-1160, 2022 06.
Article in English | MEDLINE | ID: mdl-34463952

ABSTRACT

There is a need for a straightforward, accessible and accurate pediatric test for color vision deficiency (CVD). We present and evaluate ColourSpot, a self-administered, gamified and color calibrated tablet-based app, which diagnoses CVD from age 4. Children tap colored targets with saturations that are altered adaptively along the three dichromatic confusion lines. Two cohorts (Total, N = 772; Discovery, N = 236; Validation, N = 536) of 4-7-year-old boys were screened using the Ishihara test for Unlettered Persons and the Neitz Test of Color Vision. ColourSpot was evaluated by testing any child who made an error on the Ishihara Unlettered test alongside a randomly selected control group who made no errors. Psychometric functions were fit to the data and "threshold ratios" were calculated as the ratio of tritan to protan or deutan thresholds. Based on the threshold ratios derived using an optimal fitting procedure that best categorized children in the discovery cohort, ColourSpot showed a sensitivity of 1.00 and a specificity of 0.97 for classifying CVD against the Ishihara Unlettered in the independent validation cohort. ColourSpot was also able to categorize individuals with ambiguous results on the Ishihara Unlettered. Compared to the Ishihara Unlettered, the Neitz Test generated an unacceptably high level of false positives. ColourSpot is an accurate test for CVD, which could be used by anyone to diagnose CVD in children from the start of their education. ColourSpot could also have a wider impact: its interface could be adapted for measuring other aspects of children's visual performance.


Subject(s)
Cardiovascular Diseases , Color Vision Defects , Color Vision , Child , Child, Preschool , Color Perception Tests/methods , Color Vision Defects/diagnosis , Humans , Male
9.
Cancer Immunol Res ; 10(1): 70-86, 2022 01.
Article in English | MEDLINE | ID: mdl-34795033

ABSTRACT

The APOBEC family of cytidine deaminases is one of the most common endogenous sources of mutations in human cancer. Genomic studies of tumors have found that APOBEC mutational signatures are enriched in the HER2 subtype of breast cancer and are associated with immunotherapy response in diverse cancer types. However, the direct consequences of APOBEC mutagenesis on the tumor immune microenvironment have not been thoroughly investigated. To address this, we developed syngeneic murine mammary tumor models with inducible expression of APOBEC3B. We found that APOBEC activity induced antitumor adaptive immune responses and CD4+ T cell-mediated, antigen-specific tumor growth inhibition. Although polyclonal APOBEC tumors had a moderate growth defect, clonal APOBEC tumors were almost completely rejected, suggesting that APOBEC-mediated genetic heterogeneity limits antitumor adaptive immune responses. Consistent with the observed immune infiltration in APOBEC tumors, APOBEC activity sensitized HER2-driven breast tumors to anti-CTLA-4 checkpoint inhibition and led to a complete response to combination anti-CTLA-4 and anti-HER2 therapy. In human breast cancers, the relationship between APOBEC mutagenesis and immunogenicity varied by breast cancer subtype and the frequency of subclonal mutations. This work provides a mechanistic basis for the sensitivity of APOBEC tumors to checkpoint inhibitors and suggests a rationale for using APOBEC mutational signatures and clonality as biomarkers predicting immunotherapy response in HER2-positive (HER2+) breast cancers.


Subject(s)
APOBEC Deaminases/genetics , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Immunotherapy/methods , T-Lymphocytes/immunology , APOBEC Deaminases/immunology , Animals , Antigens, Neoplasm , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mutagenesis/immunology , Mutation , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
10.
Sci Rep ; 11(1): 14932, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294755

ABSTRACT

Whole-genome duplication (WGD) generates polyploid cells possessing more than two copies of the genome and is among the most common genetic abnormalities in cancer. The frequency of WGD increases in advanced and metastatic tumors, and WGD is associated with poor prognosis in diverse tumor types, suggesting a functional role for polyploidy in tumor progression. Experimental evidence suggests that polyploidy has both tumor-promoting and suppressing effects, but how polyploidy regulates tumor progression remains unclear. Using a genetically engineered mouse model of Her2-driven breast cancer, we explored the prevalence and consequences of whole-genome duplication during tumor growth and recurrence. While primary tumors in this model are invariably diploid, nearly 40% of recurrent tumors undergo WGD. WGD in recurrent tumors was associated with increased chromosomal instability, decreased proliferation and increased survival in stress conditions. The effects of WGD on tumor growth were dependent on tumor stage. Surprisingly, in recurrent tumor cells WGD slowed tumor formation, growth rate and opposed the process of recurrence, while WGD promoted the growth of primary tumors. These findings highlight the importance of identifying conditions that promote the growth of polyploid tumors, including the cooperating genetic mutations that allow cells to overcome the barriers to WGD tumor cell growth and proliferation.


Subject(s)
Gene Duplication , In Situ Hybridization, Fluorescence/methods , Karyotyping/methods , Mammary Neoplasms, Experimental/genetics , Receptor, ErbB-2/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Chromosomal Instability , Evolution, Molecular , Female , Humans , Mice , Mice, Transgenic , Polyploidy
12.
Oncogene ; 40(11): 2018-2034, 2021 03.
Article in English | MEDLINE | ID: mdl-33603168

ABSTRACT

Recurrent breast cancer presents significant challenges with aggressive phenotypes and treatment resistance. Therefore, novel therapeutics are urgently needed. Here, we report that murine recurrent breast tumor cells, when compared with primary tumor cells, are highly sensitive to ferroptosis. Discoidin Domain Receptor Tyrosine Kinase 2 (DDR2), the receptor for collagen I, is highly expressed in ferroptosis-sensitive recurrent tumor cells and human mesenchymal breast cancer cells. EMT regulators, TWIST and SNAIL, significantly induce DDR2 expression and sensitize ferroptosis in a DDR2-dependent manner. Erastin treatment induces DDR2 upregulation and phosphorylation, independent of collagen I. Furthermore, DDR2 knockdown in recurrent tumor cells reduces clonogenic proliferation. Importantly, both the ferroptosis protection and reduced clonogenic growth may be compatible with the compromised YAP/TAZ upon DDR2 inhibition. Collectively, these findings identify the important role of EMT-driven DDR2 upregulation in recurrent tumors in maintaining growth advantage but activating YAP/TAZ-mediated ferroptosis susceptibility, providing potential strategies to eradicate recurrent breast cancer cells with mesenchymal features.


Subject(s)
Breast Neoplasms/genetics , Discoidin Domain Receptor 2/genetics , Ferroptosis/genetics , Neoplasm Recurrence, Local/genetics , Animals , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic/genetics , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Neoplasm Recurrence, Local/pathology , Nuclear Proteins/genetics , Phosphorylation , Piperazines/pharmacology , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Snail Family Transcription Factors/genetics , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Twist-Related Protein 1/genetics
13.
Cell Rep ; 33(5): 108341, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33147463

ABSTRACT

Dysregulated gene expression is a common feature of cancer and may underlie some aspects of tumor progression, including tumor relapse. Here, we show that recurrent mammary tumors exhibit global changes in gene expression and histone modifications and acquire dependence on the G9a histone methyltransferase. Genetic ablation of G9a delays tumor recurrence, and pharmacologic inhibition of G9a slows the growth of recurrent tumors. Mechanistically, G9a activity is required to silence pro-inflammatory cytokines, including tumor necrosis factor (TNF), through H3K9 methylation at gene promoters. G9a inhibition induces re-expression of these cytokines, leading to p53 activation and necroptosis. Recurrent tumors upregulate receptor interacting protein kinase-3 (RIPK3) expression and are dependent upon RIPK3 activity. High RIPK3 expression renders recurrent tumors sensitive to necroptosis following G9a inhibition. These findings demonstrate that G9a-mediated silencing of pro-necroptotic proteins is a critical step in tumor recurrence and suggest that G9a is a targetable dependency in recurrent breast cancer.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Inflammation/pathology , Mammary Neoplasms, Animal/enzymology , Mammary Neoplasms, Animal/pathology , Neoplasm Recurrence, Local/pathology , Animals , Cell Death , Cell Survival , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Mammary Neoplasms, Animal/genetics , Mice, Nude , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Risk Factors , Transcription, Genetic , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism
14.
Nat Commun ; 11(1): 5017, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33024122

ABSTRACT

The survival and recurrence of residual tumor cells following therapy constitutes one of the biggest obstacles to obtaining cures in breast cancer, but it remains unclear how the clonal composition of tumors changes during relapse. We use cellular barcoding to monitor clonal dynamics during tumor recurrence in vivo. We find that clonal diversity decreases during tumor regression, residual disease, and recurrence. The recurrence of dormant residual cells follows several distinct routes. Approximately half of the recurrent tumors exhibit clonal dominance with a small number of subclones comprising the vast majority of the tumor; these clonal recurrences are frequently dependent upon Met gene amplification. A second group of recurrent tumors comprises thousands of subclones, has a clonal architecture similar to primary tumors, and is dependent upon the Jak/Stat pathway. Thus the regrowth of dormant tumors proceeds via multiple routes, producing recurrent tumors with distinct clonal composition, genetic alterations, and drug sensitivities.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Animals , Cell Line, Tumor , Crizotinib/pharmacology , Doxycycline/pharmacology , Epithelial-Mesenchymal Transition/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice, Nude , Neoplasm Recurrence, Local/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Receptor, ErbB-2/genetics , Single-Cell Analysis , Xenograft Model Antitumor Assays
15.
Nat Metab ; 2(4): 318-334, 2020 04.
Article in English | MEDLINE | ID: mdl-32691018

ABSTRACT

The survival and recurrence of dormant tumour cells following therapy is a leading cause of death in cancer patients. The metabolic properties of these cells are likely distinct from those of rapidly growing tumours. Here we show that Her2 down-regulation in breast cancer cells promotes changes in cellular metabolism, culminating in oxidative stress and compensatory upregulation of the antioxidant transcription factor, NRF2. NRF2 is activated during dormancy and in recurrent tumours in animal models and breast cancer patients with poor prognosis. Constitutive activation of NRF2 accelerates recurrence, while suppression of NRF2 impairs it. In recurrent tumours, NRF2 signalling induces a transcriptional metabolic reprogramming to re-establish redox homeostasis and upregulate de novo nucleotide synthesis. The NRF2-driven metabolic state renders recurrent tumour cells sensitive to glutaminase inhibition, which prevents reactivation of dormant tumour cells in vitro, suggesting that NRF2-high dormant and recurrent tumours may be targeted. These data provide evidence that NRF2-driven metabolic reprogramming promotes the recurrence of dormant breast cancer.


Subject(s)
NF-E2-Related Factor 2/metabolism , Nucleotides/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Death , Cell Line, Tumor , Down-Regulation , Female , Homeostasis , Humans , Mice , Neoplasm Recurrence, Local , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Transcription, Genetic
16.
Cell Death Differ ; 27(7): 2234-2247, 2020 07.
Article in English | MEDLINE | ID: mdl-31988496

ABSTRACT

The molecular and genetic basis of tumor recurrence is complex and poorly understood. RIPK3 is a key effector in programmed necrotic cell death and, therefore, its expression is frequently suppressed in primary tumors. In a transcriptome profiling between primary and recurrent breast tumor cells from a murine model of breast cancer recurrence, we found that RIPK3, while absent in primary tumor cells, is dramatically reexpressed in recurrent breast tumor cells by an epigenetic mechanism. Unexpectedly, we found that RIPK3 knockdown in recurrent tumor cells reduced clonogenic growth, causing cytokinesis failure, p53 stabilization, and repressed the activities of YAP/TAZ. These data uncover a surprising role of the pro-necroptotic RIPK3 kinase in enabling productive cell cycle during tumor recurrence. Remarkably, high RIPK3 expression also rendered recurrent tumor cells exquisitely dependent on extracellular cystine and undergo necroptosis upon cystine deprivation. The induction of RIPK3 in recurrent tumors unravels an unexpected mechanism that paradoxically confers on tumors both growth advantage and necrotic vulnerability, providing potential strategies to eradicate recurrent tumors.


Subject(s)
Cystine/metabolism , Mammary Neoplasms, Animal/pathology , Neoplasm Recurrence, Local/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Up-Regulation/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Proliferation/drug effects , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Mammary Neoplasms, Animal/genetics , Mitosis/drug effects , Neoplasm Recurrence, Local/genetics , Piperazines/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Transcriptome/genetics , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , YAP-Signaling Proteins
17.
Child Dev ; 91(4): e799-e813, 2020 07.
Article in English | MEDLINE | ID: mdl-31580499

ABSTRACT

This study examined how colored educational tools improve children's numerosity ("number sense") and/or mathematics. We tested children 6-10 years (n = 3,236) who had been exposed to colored numbers from the educational tools Numicon (Oxford University Press, 2018) or Numberjacks (Ellis, 2006), which map colors to magnitudes or Arabic numerals, respectively. In a free association task pairing numbers with colors, a subset of children spontaneously provided colors matching one of these schemas. These children, who had internalized Numicon (colored magnitude), showed significantly better numerosity but not mathematics compared to peers. There was no similar benefit from internalizing Numberjacks (colored numerals). These data support a model in which colored number tools provide benefits at different levels of numerical cognition, according to their different levels of cross-modal mappings.


Subject(s)
Color , Mathematics , Child , Child, Preschool , Cognition , Educational Status , Female , Humans , Male
19.
Elife ; 82019 04 16.
Article in English | MEDLINE | ID: mdl-30990165

ABSTRACT

Over half of breast-cancer-related deaths are due to recurrence 5 or more years after initial diagnosis and treatment. This latency suggests that a population of residual tumor cells can survive treatment and persist in a dormant state for many years. The role of the microenvironment in regulating the survival and proliferation of residual cells following therapy remains unexplored. Using a conditional mouse model for Her2-driven breast cancer, we identify interactions between residual tumor cells and their microenvironment as critical for promoting tumor recurrence. Her2 downregulation leads to an inflammatory program driven by TNFα/NFκB signaling, which promotes immune cell infiltration in regressing and residual tumors. The cytokine CCL5 is elevated following Her2 downregulation and remains high in residual tumors. CCL5 promotes tumor recurrence by recruiting CCR5-expressing macrophages, which may contribute to collagen deposition in residual tumors. Blocking this TNFα-CCL5-macrophage axis may be efficacious in preventing breast cancer recurrence.


Subject(s)
Breast Neoplasms/physiopathology , Chemokine CCL5/metabolism , Animals , Disease Models, Animal , Macrophages/immunology , Mice , Neoplasm, Residual/physiopathology , Receptor, ErbB-2/metabolism , Recurrence , Tumor Necrosis Factor-alpha/metabolism
20.
Mol Cancer Res ; 17(7): 1545-1555, 2019 07.
Article in English | MEDLINE | ID: mdl-30902832

ABSTRACT

With the large number of women diagnosed and treated for breast cancer each year, the importance of studying recurrence has become evident due to most deaths from breast cancer resulting from tumor recurrence following therapy. To mitigate this, cellular and molecular pathways used by residual disease prior to recurrence must be studied. An altered metabolism has long been considered a hallmark of cancer, and several recent studies have gone further to report metabolic dysfunction and alterations as key to understanding the underlying behavior of dormant and recurrent cancer cells. Our group has used two probes, 2-[N-(7-nitrobenz-2-oxa-1, 3-diaxol-4-yl) amino]-2-deoxyglucose (2-NBDG) and tetramethyl rhodamine ethyl ester (TMRE), to image glucose uptake and mitochondrial membrane potential, respectively, to report changes in metabolism between primary tumors, regression, residual disease, and after regrowth in genetically engineered mouse (GEM)-derived mammospheres. Imaging revealed unique metabolic phenotypes across the stages of tumor development. Although primary mammospheres overexpressing Her2 maintained increased glucose uptake ("Warburg effect"), after Her2 downregulation, during regression and residual disease, mammospheres appeared to switch to oxidative phosphorylation. Interestingly, in mammospheres where Her2 overexpression was turned back on to model recurrence, glucose uptake was lowest, indicating a potential change in substrate preference following the reactivation of Her2, reeliciting growth. Our findings highlight the importance of imaging metabolic adaptions to gain insight into the fundamental behaviors of residual and recurrent disease. IMPLICATIONS: This study demonstrates these functional fluorescent probes' ability to report metabolic adaptations during primary tumor growth, regression, residual disease, and regrowth in Her2 breast tumors.


Subject(s)
Breast Neoplasms/genetics , Glucose/metabolism , Neoplasm Recurrence, Local/genetics , Receptor, ErbB-2/genetics , 4-Chloro-7-nitrobenzofurazan/analogs & derivatives , 4-Chloro-7-nitrobenzofurazan/pharmacology , Animals , Animals, Genetically Modified , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Deoxyglucose/analogs & derivatives , Deoxyglucose/pharmacology , Female , Gene Expression Regulation, Neoplastic , Glucose/genetics , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Membrane Potential, Mitochondrial/drug effects , Neoplasm Recurrence, Local/metabolism , Organometallic Compounds/pharmacology , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...