Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Development ; 151(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38276965

ABSTRACT

The varying pathways of mammary gland development across species and evolutionary history are underexplored, largely due to a lack of model systems. Recent progress in organoid technology holds the promise of enabling in-depth studies of the developmental adaptations that have occurred throughout the evolution of different species, fostering beneficial phenotypes. The practical application of this technology for mammary glands has been mostly confined to rodents and humans. In the current study, we have successfully created next-generation 3D mammary gland organoids from eight eutherian mammals and the first branched organoid of a marsupial mammary gland. Using mammary organoids, we identified a role for ROCK protein in regulating branching morphogenesis, a role that manifests differently in organoids from different mammals. This finding demonstrates the utility of the 3D organoid model for understanding the evolution and adaptations of signaling pathways. These achievements highlight the potential for organoid models to expand our understanding of mammary gland biology and evolution, and their potential utility in studies of lactation or breast cancer.


Subject(s)
Mammary Glands, Human , Marsupialia , Humans , Female , Animals , Marsupialia/genetics , Organoids/metabolism , Lactation , Eutheria , Mammary Glands, Animal/metabolism
2.
J Immunother Cancer ; 12(1)2024 01 25.
Article in English | MEDLINE | ID: mdl-38272561

ABSTRACT

BACKGROUND: Recent trials suggest that programmed cell death 1 (PD-1)-directed immunotherapy may be beneficial for some patients with anal squamous cell carcinoma and biomarkers predictive of response are greatly needed. METHODS: This multicenter phase II clinical trial (NCT02919969) enrolled patients with metastatic or locally advanced incurable anal squamous cell carcinoma (n=32). Patients received pembrolizumab 200 mg every 3 weeks. The primary endpoint of the trial was objective response rate (ORR). Exploratory objectives included analysis of potential predictive biomarkers including assessment of tumor-associated immune cell populations with multichannel immunofluorescence and analysis of circulating tumor tissue modified viral-human papillomavirus DNA (TTMV-HPV DNA) using serially collected blood samples. To characterize the clinical features of long-term responders, we combined data from our prospective trial with a retrospective cohort of patients with anal cancer treated with anti-PD-1 immunotherapy (n=18). RESULTS: In the phase II study, the ORR to pembrolizumab monotherapy was 9.4% and the median progression-free survival was 2.2 months. Despite the high level of HPV positivity observed with circulating TTMV-HPV DNA testing, the majority of patients had low levels of tumor-associated CD8+PD-1+ T cells on pretreatment biopsy. Patients who benefited from pembrolizumab had decreasing TTMV-HPV DNA scores and a complete responder's TTMV-HPV DNA became undetectable. Long-term pembrolizumab responses were observed in one patient from the trial (5.3 years) and three patients (2.5, 6, and 8 years) from the retrospective cohort. Long-term responders had HPV-positive tumors, lacked liver metastases, and achieved a radiological complete response. CONCLUSIONS: Pembrolizumab has durable efficacy in a rare subset of anal cancers. However, despite persistence of HPV infection, indicated by circulating HPV DNA, most advanced anal cancers have low numbers of tumor-associated CD8+PD-1+ T cells and are resistant to pembrolizumab.


Subject(s)
Antibodies, Monoclonal, Humanized , Anus Neoplasms , Carcinoma, Squamous Cell , Papillomavirus Infections , Humans , Retrospective Studies , Prospective Studies , Programmed Cell Death 1 Receptor , Carcinoma, Squamous Cell/drug therapy , Anus Neoplasms/drug therapy , DNA
3.
bioRxiv ; 2023 Oct 11.
Article in English | MEDLINE | ID: mdl-37873154

ABSTRACT

Organoids have been widely used for studying tissue growth and modeling diseases, but achieving physiologically relevant architecture, size, and function has remained a challenge. Here, we develop a next-generation organotypic culture method that enables the formation of a highly patterned, complex, branched tissue that is spatially organized to accurately recapitulate the morphology, scale, cellular, transcriptional, and tissue-level heterogeneity of human breast tissue. Hormone responsiveness of organoids is also a feature allowing for examination of androgen therapy or post-menopausal changes to breast tissue development and regeneration. Live imaging allows for studying stem cell dynamics during organoid formation and is adaptable to a high throughput setting. Real-time imaging of organoid formation reveals activation of latent epithelial organogenesis programs and inductive cellular dynamics that drive formation of a miniature breast tissue along with its mesenchyme akin to tissue stroma. By advancing human breast organoid technology, this model can elucidate cell- and tissue-level consequences to hormonal changes and therapy. In addition, this method can lead to new insights into the cellular, molecular, and tissue-level processes involved in organogenesis and regeneration, as well as disease.

4.
Oral Oncol ; 147: 106584, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37837735

ABSTRACT

OBJECTIVES: While survival outcomes are favorable for Human Papillomavirus (HPV)-positive oropharyngeal squamous cell carcinomas (OPSCCs), early diagnosis may minimize treatment-related morbidity and mortality. This study evaluated circulating tumor tissue-modified viral (TTMV)-HPV DNA plasma testing to facilitate early diagnosis of HPV-positive OPSCCs. METHODS: In this prospective exploratory cohort study, patients presenting to an Otolaryngology-Head and Neck Surgery clinic with unexplained signs or symptoms considered high-risk for HPV-positive OPSCC were recruited between March 2021-October 2022. Circulating TTMV-HPV DNA testing was performed, and results were shared with subjects and treating clinicians. Clinicians were surveyed regarding the perceived clinical utility of the test. RESULTS: Thirty-nine subjects were included. Most subjects were women (N = 23, 59 %), white (N = 32, 82 %) and never-smokers (N = 20, 51 %) with median age 60 years. Circulating TTMV-HPV DNA was detected in 2/39 subjects, both subsequently diagnosed with HPV-positive OPSCC. Both were white men aged 70-80 years with a neck mass. One subject with undetectable TTMV-HPV DNA was also diagnosed with HPV-positive OPSCC through excisional neck mass biopsy. Other eventual diagnoses included 3 HPV-negative head and neck squamous cell carcinomas and 4 other malignancies. Testing was perceived as helpful in clinical decision-making for 26/38 (68 %) subjects, and useful for similar future patients for 32/37 (86 %) subjects. CONCLUSION: Circulating TTMV-HPV DNA testing is feasible and holds potential as a diagnostic aid for HPV-positive OPSCC alongside standard clinical workup. Clinicians should be cognizant of its limitations, as a negative test does not necessarily indicate the absence of disease. Further studies to evaluate its utility are warranted.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Oropharyngeal Neoplasms , Papillomavirus Infections , Male , Humans , Female , Middle Aged , Carcinoma, Squamous Cell/genetics , Papillomavirus Infections/complications , Papillomavirus Infections/diagnosis , Cohort Studies , Prospective Studies , Prognosis , Oropharyngeal Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/diagnosis , DNA, Viral/genetics , Head and Neck Neoplasms/diagnosis , Papillomaviridae/genetics
5.
Clin Cancer Res ; 29(20): 4306-4313, 2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37566241

ABSTRACT

PURPOSE: Human papillomavirus (HPV) is causally linked to oropharyngeal squamous cell carcinoma (OPSCC). Consensus guidelines recommend clinical exams and imaging in decreasing frequency as part of posttreatment surveillance for recurrence. Plasma tumor tissue modified viral (TTMV)-HPV DNA testing has emerged as a biomarker which can inform disease status during surveillance. EXPERIMENTAL DESIGN: This retrospective observational cohort study involved 543 patients who completed curative-intent therapy for HPV-associated OPSCC between February 2020 and January 2022 at eight U.S. cancer care institutions. We determined the negative predictive value (NPV) of TTMV-HPV DNA for recurrence when matched to physician-reported clinical outcome data (median follow-up time: 27.9 months; range: 4.5-154). RESULTS: The cohort included mostly men with a median age of 61 who had locoregionally advanced disease. HPV status was determined by p16 positivity in 87% of patients, with a positive HPV PCR/ISH among 55%; while pretreatment TTMV-HPV DNA status was unknown for most (79%) patients. Patients had a mean of 2.6 tests and almost half had three or more TTMV-HPV DNA results during surveillance. The per-test and per-patient sensitivity of the assay was 92.5% [95% confidence interval (CI): 87.5-97.5] and 87.3% (95% CI: 79.1-95.5), respectively. The NPV for the assay was 99.4% (95% CI: 98.9-99.8) and 98.4% (95% CI: 97.3-99.5), respectively. CONCLUSIONS: TTMV-HPV DNA surveillance testing yields few false negative results and few missed recurrences. These data could inform decisions on when to pursue reimaging following first disease restaging and could inform future surveillance practice. Additional study of how pretreatment TTMV-HPV DNA status impacts sensitivity for recurrence is needed.

7.
Diagnostics (Basel) ; 13(4)2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36832208

ABSTRACT

The NavDx® blood test analyzes tumor tissue modified viral (TTMV)-HPV DNA to provide a reliable means of detecting and monitoring HPV-driven cancers. The test has been clinically validated in a large number of independent studies and has been integrated into clinical practice by over 1000 healthcare providers at over 400 medical sites in the US. This Clinical Laboratory Improvement Amendments (CLIA), high complexity laboratory developed test, has also been accredited by the College of American Pathologists (CAP) and the New York State Department of Health. Here, we report a detailed analytical validation of the NavDx assay, including sample stability, specificity as measured by limits of blank (LOBs), and sensitivity illustrated via limits of detection and quantitation (LODs and LOQs). LOBs were 0-0.32 copies/µL, LODs were 0-1.10 copies/µL, and LOQs were <1.20-4.11 copies/µL, demonstrating the high sensitivity and specificity of data provided by NavDx. In-depth evaluations including accuracy and intra- and inter-assay precision studies were shown to be well within acceptable ranges. Regression analysis revealed a high degree of correlation between expected and effective concentrations, demonstrating excellent linearity (R2 = 1) across a broad range of analyte concentrations. These results demonstrate that NavDx accurately and reproducibly detects circulating TTMV-HPV DNA, which has been shown to aid in the diagnosis and surveillance of HPV-driven cancers.

8.
Clin Cancer Res ; 28(19): 4292-4301, 2022 10 03.
Article in English | MEDLINE | ID: mdl-35576437

ABSTRACT

PURPOSE: Despite generally favorable outcomes, 15% to 25% of patients with human papillomavirus (HPV)-driven oropharyngeal squamous cell carcinoma (OPSCC) will have recurrence. Current posttreatment surveillance practices rely on physical examinations and imaging and are inconsistently applied. We assessed circulating tumor tissue modified viral (TTMV)-HPV DNA obtained during routine posttreatment surveillance among a large population of real-world patients. EXPERIMENTAL DESIGN: This retrospective clinical case series included 1,076 consecutive patients across 108 U.S. sites who were ≥ 3 months posttreatment for HPV-driven OPSCC and who had one or more TTMV-HPV DNA tests (NavDx, Naveris Laboratories) obtained during surveillance between February 6, 2020, and June 29, 2021. Test results were compared with subsequent clinical evaluations. RESULTS: Circulating TTMV-HPV DNA was positive in 80 of 1,076 (7.4%) patients, with follow-up available on all. At first positive surveillance testing, 21 of 80 (26%) patients had known recurrence while 59 of 80 (74%) patients were not known to have recurrent disease. Among these 59 patients, 55 (93%) subsequently had a confirmed recurrence, 2 patients had clinically suspicious lesions, and 2 had clinically "no evidence of disease" (NED) at last follow-up. To date, the overall positive predictive value of TTMV-HPV DNA testing for recurrent disease is 95% (N = 76/80). In addition, the point-in-time negative predictive value is 95% (N = 1,198/1,256). CONCLUSIONS: These findings highlight the clinical potential for circulating TTMV-HPV DNA testing in routine practice. As a surveillance tool, TTMV-HPV DNA positivity was the first indication of recurrence in the majority of cases, pre-dating identification by routine clinical and imaging exams. These data may inform future clinical and guideline-endorsed strategies for HPV-driven malignancy surveillance. See related commentary by Colevas, p. 4171.


Subject(s)
Alphapapillomavirus , Head and Neck Neoplasms , Oropharyngeal Neoplasms , Papillomavirus Infections , Alphapapillomavirus/genetics , Biomarkers , DNA, Viral/genetics , Humans , Oropharyngeal Neoplasms/diagnosis , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/therapy , Papillomaviridae/genetics , Papillomavirus Infections/complications , Papillomavirus Infections/diagnosis , Papillomavirus Infections/pathology , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/therapy
9.
Int J Cancer ; 151(7): 1081-1085, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35262203

ABSTRACT

Human papillomavirus (HPV), most commonly HPV16, causes a growing subset of head and neck squamous cell carcinomas (HNSCCs), including the overwhelming majority of oropharynx squamous cell carcinomas in many developed countries. Circulating biomarkers for HPV-positive HNSCC may allow for earlier diagnosis, with potential to decrease morbidity and mortality. This case-control study evaluated whether circulating tumor HPV DNA (ctHPVDNA) is detectable in prediagnostic plasma from individuals later diagnosed with HPV-positive HNSCC. Cases were participants in a hospital-based research biobank with archived plasma collected ≥6 months before HNSCC diagnosis, and available archival tumor tissue for HPV testing. Controls were biobank participants without cancer or HPV-related diagnoses, matched 10:1 to cases by sex, race, age and year of plasma collection. HPV DNA was detected in plasma and tumor tissue using a previously validated digital droplet PCR-based assay that quantifies tumor-tissue-modified viral (TTMV) HPV DNA. Twelve HNSCC patients with median age of 68.5 years (range, 51-87 years) were included. Ten (83.3%) had HPV16 DNA-positive tumors. ctHPV16DNA was detected in prediagnostic plasma from 3 of 10 (30%) patients with HPV16-positive tumors, including 3 of 7 (43%) patients with HPV16-positive oropharynx tumors. The timing of the plasma collection was 19, 34 and 43 months before cancer diagnosis. None of the 100 matched controls had detectable ctHPV16DNA. This is the first report that ctHPV16 DNA is detectable at least several years before diagnosis of HPV16-positive HNSCC for a subset of patients. Further investigation of ctHPV16DNA as a biomarker for early diagnosis of HPV16-positive HNSCC is warranted.


Subject(s)
Alphapapillomavirus , Carcinoma, Squamous Cell , Circulating Tumor DNA , Head and Neck Neoplasms , Papillomavirus Infections , Aged , Aged, 80 and over , Alphapapillomavirus/genetics , Carcinoma, Squamous Cell/pathology , Case-Control Studies , DNA, Viral/genetics , Head and Neck Neoplasms/diagnosis , Humans , Middle Aged , Papillomaviridae/genetics , Papillomavirus Infections/complications , Papillomavirus Infections/diagnosis , Papillomavirus Infections/pathology , Squamous Cell Carcinoma of Head and Neck/diagnosis
10.
Clin Cancer Res ; 28(11): 2329-2338, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35344035

ABSTRACT

PURPOSE: A phase II multi-institutional clinical trial was conducted to determine overall survival (OS) in patients with recurrent and/or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) treated with a combination of cetuximab and nivolumab. PATIENTS AND METHODS: Patients with R/M HNSCC were treated with cetuximab 500 mg/m2 i.v. on day 14 as a lead-in followed by cetuximab 500 mg/m2 i.v. and nivolumab 240 mg i.v. on day 1 and day 15 of each 28-day cycle. Expression of p16 and programmed cell death-ligand 1 (PD-L1) in archived tumors were determined. Tumor-tissue-modified human papillomavirus (TTMV) DNA was quantified in plasma. RESULTS: Ninety-five patients were enrolled, and 88 patients were evaluable for OS with a median follow-up of 15.9 months. Median OS in the 45 patients who had prior therapy for R/M HNSCC (cohort A) was 11.4 months, with a 1 year OS 50% [90% confidence interval (CI), 0.43-0.57]. Median OS in the 43 patients who had no prior therapy (cohort B) was 20.2 months, with a 1-year OS 66% (90% CI, 0.59-0.71). In the combined cohorts, the p16-negative immunostaining was associated with higher response rate (RR; P = 0.02) but did not impact survival while higher PD-L1 combined positive score was associated with higher RR (P = 0.03) and longer OS (log-rank P = 0.04). In the p16-positive patients, lower median (1,230 copies/mL) TTMV DNA counts were associated with higher RR (P = 0.01) and longer OS compared with higher median (log-rank P = 0.05). CONCLUSIONS: The combination of cetuximab and nivolumab is effective in patients with both previously treated and untreated R/M HNSCC and warrants further evaluation.


Subject(s)
Head and Neck Neoplasms , Nivolumab , B7-H1 Antigen/metabolism , Cetuximab , Head and Neck Neoplasms/drug therapy , Humans , Neoplasm Recurrence, Local/pathology , Squamous Cell Carcinoma of Head and Neck/drug therapy
11.
Nat Commun ; 12(1): 7116, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34893587

ABSTRACT

Mammary morphogenesis is an orchestrated process involving differentiation, proliferation and organization of cells to form a bi-layered epithelial network of ducts and lobules embedded in stromal tissue. We have engineered a 3D biomimetic human breast that makes it possible to study how stem cell fate decisions translate to tissue-level structure and function. Using this advancement, we describe the mechanism by which breast epithelial cells build a complex three-dimensional, multi-lineage tissue by signaling through a collagen receptor. Discoidin domain receptor tyrosine kinase 1 induces stem cells to differentiate into basal cells, which in turn stimulate luminal progenitor cells via Notch signaling to differentiate and form lobules. These findings demonstrate how human breast tissue regeneration is triggered by transmission of signals from the extracellular matrix through an epithelial bilayer to coordinate structural changes that lead to formation of a complex ductal-lobular network.


Subject(s)
Breast/cytology , Breast/physiology , Cell Communication/physiology , Cell Differentiation/physiology , Discoidin Domain Receptor 1/metabolism , Biocompatible Materials , Biomedical Engineering , Cell Line , Discoidin Domain Receptor 1/genetics , Epithelial Cells/cytology , Extracellular Matrix , Humans , Regeneration , Signal Transduction , Stem Cells/cytology
12.
Dev Cell ; 56(13): 1875-1883, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34256927

ABSTRACT

Cell fate decisions are critical for adequate tissue development, maintenance and regeneration. In the mammary gland, epithelial cell fates are tightly controlled by the microenvironment. Here, we review how cell fate decisions are regulated by components of the microenvironment during mammary gland development and how pathological changes in the microenvironment can alter cell fates, leading to malignancy. Specifically, we describe the current understanding of how mammary cell fate is controlled and directed by three elements: the extracellular matrix, the immune microenvironment, and hormones-and how these elements can converge to create microenvironments that promote a fourth element: DNA damage.


Subject(s)
Cellular Microenvironment/genetics , Extracellular Matrix/genetics , Mammary Glands, Animal/growth & development , Mammary Glands, Human/growth & development , Animals , Breast/growth & development , Breast/pathology , Cell Differentiation/genetics , Cell Lineage/genetics , Female , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Neoplasms/genetics , Neoplasms/pathology , Tumor Microenvironment
13.
Dev Cell ; 56(8): 1077-1079, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33878297

ABSTRACT

Selective pressure and signals from the tissue microenvironment drive metastasis and determine the survival of metastatic tumor cells at distant organs. Zhang et al. and Bado et al. apply CRISPR-mediated evolving barcode technology to elucidate the role of the bone microenvironment in the evolution of breast cancer metastasis.


Subject(s)
Breast Neoplasms , Tumor Microenvironment , Bone and Bones , Female , Humans
14.
J Cell Sci ; 132(23)2019 12 02.
Article in English | MEDLINE | ID: mdl-31792043

ABSTRACT

The transcription factor Snai2, encoded by the SNAI2 gene, is an evolutionarily conserved C2H2 zinc finger protein that orchestrates biological processes critical to tissue development and tumorigenesis. Initially characterized as a prototypical epithelial-to-mesenchymal transition (EMT) transcription factor, Snai2 has been shown more recently to participate in a wider variety of biological processes, including tumor metastasis, stem and/or progenitor cell biology, cellular differentiation, vascular remodeling and DNA damage repair. The main role of Snai2 in controlling such processes involves facilitating the epigenetic regulation of transcriptional programs, and, as such, its dysregulation manifests in developmental defects, disruption of tissue homeostasis, and other disease conditions. Here, we discuss our current understanding of the molecular mechanisms regulating Snai2 expression, abundance and activity. In addition, we outline how these mechanisms contribute to disease phenotypes or how they may impact rational therapeutic targeting of Snai2 dysregulation in human disease.


Subject(s)
Snail Family Transcription Factors/metabolism , Animals , Epigenesis, Genetic/genetics , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/genetics , Humans , Models, Biological , Signal Transduction/genetics , Signal Transduction/physiology , Snail Family Transcription Factors/genetics
15.
Cell Rep ; 28(13): 3435-3449.e5, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31553912

ABSTRACT

Comprehensive sequencing approaches have allowed for the identification of the most frequent contributors to cancer, known as drivers. They have also revealed a class of mutations in understudied, infrequently altered genes, referred to as "long tail" (LT) drivers. A key challenge has been to find clinically relevant LT drivers and to understand how they cooperate to drive disease. Here, we identified far upstream binding protein 1 (FUBP1) as an LT driver using an in vivo CRISPR screen. FUBP1 cooperates with other tumor suppressor genes to transform mammary epithelial cells by disrupting cellular differentiation and tissue architecture. Mechanistically, FUBP1 participates in regulating N6-methyladenosine (m6A) RNA methylation, and its loss leads to global changes in RNA splicing and widespread expression of aberrant driver isoforms. These findings suggest that somatic alteration of a single gene involved in RNA splicing and m6A methylation can produce the necessary panoply of contributors for neoplastic transformation.


Subject(s)
Alternative Splicing/genetics , DNA-Binding Proteins/genetics , Neoplasms/genetics , Oncogenes/genetics , RNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Humans
16.
Cell Rep ; 28(2): 394-407.e6, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31291576

ABSTRACT

DNA damage activates checkpoints that limit the replicative potential of stem cells, including differentiation. These checkpoints protect against cancer development but also promote tissue aging. Because mice lacking Slug/Snai2 exhibit limited stem cell activity, including luminobasal differentiation, and are protected from mammary cancer, we reasoned that Slug might regulate DNA damage checkpoints in mammary epithelial cells. Here, we show that Slug facilitates efficient execution of RPA32-mediated DNA damage response (DDR) signaling. Slug deficiency leads to delayed phosphorylation of ataxia telangiectasia mutated and Rad3-related protein (ATR) and its effectors RPA32 and CHK1. This leads to impaired RAD51 recruitment to DNA damage sites and persistence of unresolved DNA damage. In vivo, Slug/Snai2 loss leads to increased DNA damage and premature aging of mammary epithelium. Collectively, our work demonstrates that the mammary stem cell regulator Slug controls DDR checkpoints by dually inhibiting differentiation and facilitating DDR repair, and its loss causes unresolved DNA damage and accelerated aging.


Subject(s)
DNA Damage , DNA Repair , Mammary Glands, Animal/cytology , Mammary Glands, Human/cytology , Snail Family Transcription Factors/deficiency , Animals , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Cellular Senescence/physiology , HEK293 Cells , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
17.
Cell Stem Cell ; 24(1): 65-78, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30554963

ABSTRACT

Our traditional understanding of phenotypic plasticity in adult somatic cells comprises dedifferentiation and transdifferentiation in the context of tissue regeneration or wound healing. Although dedifferentiation is central to tissue repair and stemness, this process inherently carries the risk of cancer initiation. Consequently, recent research suggests phenotypic plasticity as a new paradigm for understanding cancer initiation, progression, and resistance to therapy. Here, we discuss how cells acquire plasticity and the role of plasticity in initiating cancer, cancer progression, and metastasis and in developing therapy resistance. We also highlight the epithelial-to-mesenchymal transition (EMT) and known molecular mechanisms underlying plasticity and we consider potential therapeutic avenues.


Subject(s)
Adaptation, Physiological , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Disease Progression , Humans , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects
18.
Retrovirology ; 15(1): 57, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30126415

ABSTRACT

BACKGROUND: Increased transcription of the human endogenous retrovirus group HERV-K (HML-2) is often seen during disease. Although the mechanism of its tissue-specific activation is unclear, research shows that LTR CpG hypomethylation alone is not sufficient to induce its promoter activity and that the transcriptional milieu of a malignant cell contributes, at least partly, to differential HML-2 expression. RESULTS: We analyzed the relationship between LTR sequence variation and promoter expression patterns in human breast cancer cell lines, finding them to be positively correlated. In particular, two proviruses (3q12.3 and 11p15.4) displayed increased activity in almost all tumorigenic cell lines sampled. Using a transcription factor binding site prediction algorithm, we identified two unique binding sites in each 5' LTR that appeared to be associated with inducing promoter activity during neoplasia. Genomic analysis of the homologous proviruses in several non-human primates indicated post-integration genetic drift in two transcription factor binding sites, away from the ancestral sequence and towards the active form. Based on the sequences of 2504 individuals from the 1000 Genomes Project, the active form of the 11p15.4 site was found to be polymorphic within the human population, with an allele frequency of 51%, whereas the activating mutation in the 3q12.3 provirus was fixed in humans but not present in the orthologous provirus in chimpanzees or gorillas. CONCLUSIONS: These data suggest that stage-specific transcription factors at least partly contribute to LTR promoter activity during transformation and that, in some cases, transcription factor binding site polymorphisms may be responsible for the differential HML-2 expression often seen between individuals.


Subject(s)
Endogenous Retroviruses/genetics , Gene Expression , Promoter Regions, Genetic/genetics , Proviruses/genetics , Terminal Repeat Sequences/genetics , Transcription Factors/metabolism , Binding Sites/genetics , Cell Line, Tumor , Endogenous Retroviruses/classification , Genetic Drift , Genetic Variation , Genome, Viral/genetics , Humans , Mutation , Polymorphism, Genetic , Proviruses/classification
19.
Stem Cell Reports ; 10(3): 1131-1145, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29503088

ABSTRACT

The epithelial compartment of the mammary gland contains basal and luminal cell lineages, as well as stem and progenitor cells that reside upstream in the differentiation hierarchy. Stem and progenitor cell differentiation is regulated to maintain adult tissue and mediate expansion during pregnancy and lactation. The genetic factors that regulate the transition of cells between differentiation states remain incompletely understood. Here, we present a genome-scale method to discover genes driving cell-state specification. Applying this method, we identify a transcription factor, BCL11B, which drives stem cell self-renewal in vitro, by inhibiting differentiation into the basal lineage. To validate BCL11B's functional role, we use two-dimensional colony-forming and three-dimensional tissue differentiation assays to assess the lineage differentiation potential and functional abilities of primary human mammary cells. These findings show that BCL11B regulates mammary cell differentiation and demonstrate the utility of our proposed genome-scale strategy for identifying lineage regulators in mammalian tissues.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Self Renewal/genetics , Cell Self Renewal/physiology , Mammary Glands, Human/physiology , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , Cell Lineage/genetics , Cell Lineage/physiology , Epithelial Cells/physiology , Female , Humans , Stem Cells/physiology , Transcription Factors/genetics
20.
Sci Rep ; 8(1): 1415, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29362392

ABSTRACT

In cancer, tumor suppressor genes (TSGs) are frequently truncated, causing their encoded products to be non-functional or dominant-negative. We previously showed that premature polyadenylation (pPA) of MAGI3 truncates the gene, switching its functional role from a TSG to a dominant-negative oncogene. Here we report that MAGI3 undergoes pPA at the intron immediately downstream of its large internal exon, which is normally highly modified by N6-methyladenosine (m6A). In breast cancer cells that upregulate MAGI3 pPA , m6A levels in the large internal exon of MAGI3 are significantly reduced compared to cells that do not express MAGI3 pPA . We further find that MAGI3 pPA transcripts are significantly depleted of m6A modifications, in contrast to highly m6A-modified full-length MAGI3 mRNA. Finally, we analyze public expression data and find that other TSGs, including LATS1 and BRCA1, also undergo intronic pPA following large internal exons, and that m6A levels in these exons are reduced in pPA-activated breast cancer cells relative to untransformed mammary cells. Our study suggests that m6A may play a role in regulating intronic pPA of MAGI3 and possibly other TSGs, warranting further investigation.


Subject(s)
Adenosine/analogs & derivatives , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Membrane Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Adenosine/metabolism , BRCA1 Protein/metabolism , Cell Line, Tumor , Exons , Female , Gene Expression Regulation, Neoplastic , Humans , Introns , Membrane Proteins/metabolism , Polyadenylation , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...