Try a new search

Format these results:

Searched for:

person:barcem01

Total Results:

256


Autocrine TGFβ Is a Survival Factor for Monocytes and Drives Immunosuppressive Lineage Commitment

Gonzalez-Junca, Alba; Driscoll, Kyla E; Pellicciotta, Ilenia; Du, Shisuo; Lo, Chen Hao; Roy, Ritu; Parry, Renate; Tenvooren, Iliana; Marquez, Diana M; Spitzer, Matthew H; Barcellos-Hoff, Mary Helen
Transforming growth factor β (TGFβ) is an effector of immune suppression and contributes to a permissive tumor microenvironment that compromises effective immunotherapy. We identified a correlation between TGFB1 and genes expressed by myeloid cells, but not granulocytes, in The Cancer Genome Atlas lung adenocarcinoma data, in which high TGFB1 expression was associated with poor survival. To determine whether TGFβ affected cell fate decisions and lineage commitment, we studied primary cultures of CD14+ monocytes isolated from peripheral blood of healthy donors. We discovered that TGFβ was a survival factor for CD14+ monocytes, which rapidly executed an apoptotic program in its absence. Continued exposure to TGFβ in combination with granulocyte-macrophage colony stimulating factor (GM-CSF) and interleukin 6 (IL6) amplified HLA-DRlowCD14+CD11b+CD33+ myeloid-derived suppressor cells (MDSCs) at the expense of macrophage and dendritic cell (DC) differentiation. MDSCs generated in the presence of TGFβ were more effective in suppressing T-cell proliferation and promoted the T regulatory cell phenotype. In contrast, inhibition of TGFβ signaling using a small-molecule inhibitor of receptor kinase activity in CD14+ monocytes treated with GM-CSF and IL6 decreased MDSC differentiation and increased differentiation to proinflammatory macrophages and antigen-presenting DCs. The effect of autocrine and paracrine TGFβ on myeloid cell survival and lineage commitment suggests that pharmacologic inhibition of TGFβ-dependent signaling in cancer would favor antitumor immunity.
PMID: 30538091
ISSN: 2326-6074
CID: 3646732

Misrepair in Context: TGFβ Regulation of DNA Repair

Liu, Qi; Lopez, Kirsten; Murnane, John; Humphrey, Timothy; Barcellos-Hoff, Mary Helen
Repair of DNA damage protects genomic integrity, which is key to tissue functional integrity. In cancer, the type and fidelity of DNA damage response is the fundamental basis for clinical response to cytotoxic therapy. Here we consider the contribution of transforming growth factor-beta (TGFβ), a ubiquitous, pleotropic cytokine that is abundant in the tumor microenvironment, to therapeutic response. The action of TGFβ is best illustrated in head and neck squamous cell carcinoma (HNSCC). Survival of HNSCC patients with human papilloma virus (HPV) positive cancer is more than double compared to those with HPV-negative HNSCC. Notably, HPV infection profoundly impairs TGFβ signaling. HPV blockade of TGFβ signaling, or pharmaceutical TGFβ inhibition that phenocopies HPV infection, shifts cancer cells from error-free homologous-recombination DNA double-strand-break (DSB) repair to error-prone alternative end-joining (altEJ). Cells using altEJ are more sensitive to standard of care radiotherapy and cisplatin, and are sensitized to PARP inhibitors. Hence, HPV-positive HNSCC is an experiment of nature that provides a strong rationale for the use of TGFβ inhibitors for optimal therapeutic combinations that improve patient outcome.
PMCID:6736563
PMID: 31552165
ISSN: 2234-943x
CID: 4107642

Subjugation of TGFβ Signaling by Human Papilloma Virus in Head and Neck Squamous Cell Carcinoma Shifts DNA Repair from Homologous Recombination to Alternative End-Joining

Liu, Qi; Ma, Lin; Jones, Trevor; Palomero, Luis; Pujana, Miguel Angel; Martinez-Ruiz, Haydeliz; Ha, Patrick K; Murnane, John; Cuartas, Isabel; Seoane, Joan; Baumann, Michael; Linge, Annett; Barcellos-Hoff, Mary Helen
PURPOSE/OBJECTIVE:Following cytotoxic therapy, 70% of patients with human papillomavirus (HPV) positive oropharyngeal head and neck squamous cell carcinoma (HNSCC) are alive at 5 years compared to 30% of those with similar HPV-negative cancer, which is thought to be due to dysregulation of DNA repair. Loss of transforming growth factor β (TGFβ) signaling is a poorly studied consequence of HPV that could contribute to this phenotype. EXPERIMENTAL DESIGN/METHODS:Human HNSCC cell lines (n=9), patient-derived xenografts (n=9), tissue microarray (n=194), TCGA expression data and primary tumor specimens (n=10) were used to define the relationship between TGFβ competency, response to DNA damage, and type of DNA repair. RESULTS:Analysis of HNSCC specimens in situ and in vitro showed that HPV associates with loss of TGFβ signaling that increases the response to radiation or cisplatin. TGFβ suppressed miR-182 that inhibited both BRCA1, necessary for homologous recombination repair, and FOXO3, which is required for ATM kinase activity. TGFβ signaling blockade by either HPV or inhibitors released this control, compromised HRR and increased response to PARP inhibition. Antagonizing miR-182 rescued the homologous recombination deficit in HPV+ cells. Loss of TGFβ signaling unexpectedly increased error-prone, alternative end-joining repair. CONCLUSIONS:HPV-positive HNSCC cells are unresponsive to TGFβ. Abrogated TGFβ signaling compromises homologous recombination and shifts reliance on alt-EJ repair that provides a mechanistic basis for sensitivity to PARP inhibitors. The effect of HPV in HNSCC provides critical validation of TGFβ's role in DNA repair proficiency and further raises the translational potential of TGFβ inhibitors in cancer therapy.
PMID: 30087144
ISSN: 1078-0432
CID: 3226602

Evaluation of Radioresponse and Radiosensitizers in Glioblastoma Organotypic Cultures

Bayin, N Sumru; Ma, Lin; Placantonakis, Dimitris G; Barcellos-Hoff, Mary Helen
Glioblastoma (GBM), a deadly primary brain malignancy, manifests pronounced radioresistance. Identifying agents that improve the sensitivity of tumor tissue to radiotherapy is critical for improving patient outcomes. The response to ionizing radiation is regulated by both cell-intrinsic and -extrinsic mechanisms. In particular, the tumor microenvironment is known to promote radioresistance in GBM. Therefore, model systems used to test radiosensitizing agents need to take into account the tumor microenvironment. We recently showed that GBM explant cultures represent an adaptable ex vivo platform for rapid and personalized testing of radiosensitizers. These explants preserve the cellular composition and tissue architecture of parental patient tumors and therefore capture the microenvironmental context that critically determines the response to radiotherapy. This chapter focuses on the detailed protocol for testing candidate radiosensitizing agents in GBM explants.
PMID: 29392699
ISSN: 1940-6029
CID: 2933592

Notch signaling regulates metabolic heterogeneity in glioblastoma stem cells

Bayin, N Sumru; Frenster, Joshua D; Sen, Rajeev; Si, Sheng; Modrek, Aram S; Galifianakis, Nataliya; Dolgalev, Igor; Ortenzi, Valerio; Illa-Bochaca, Irineu; Khahera, Anadjeet; Serrano, Jonathan; Chiriboga, Luis; Zagzag, David; Golfinos, John G; Doyle, Werner; Tsirigos, Aristotelis; Heguy, Adriana; Chesler, Mitch; Barcellos-Hoff, Mary Helen; Snuderl, Matija; Placantonakis, Dimitris G
Glioblastoma (GBM) stem cells (GSCs) reside in both hypoxic and vascular microenvironments within tumors. The molecular mechanisms that allow GSCs to occupy such contrasting niches are not understood. We used patient-derived GBM cultures to identify GSC subtypes with differential activation of Notch signaling, which co-exist in tumors but occupy distinct niches and match their metabolism accordingly. Multipotent GSCs with Notch pathway activation reside in perivascular niches, and are unable to entrain anaerobic glycolysis during hypoxia. In contrast, most CD133-expressing GSCs do not depend on canonical Notch signaling, populate tumors regardless of local vascularity and selectively utilize anaerobic glycolysis to expand in hypoxia. Ectopic activation of Notch signaling in CD133-expressing GSCs is sufficient to suppress anaerobic glycolysis and resistance to hypoxia. These findings demonstrate a novel role for Notch signaling in regulating GSC metabolism and suggest intratumoral GSC heterogeneity ensures metabolic adaptations to support tumor growth in diverse tumor microenvironments.
PMCID:5630302
PMID: 29029402
ISSN: 1949-2553
CID: 2738172

Radiation-Induced Fibrosis: Mechanisms and Opportunities to Mitigate. Report of an NCI Workshop, September 19, 2016 [Meeting Abstract]

Citrin, Deborah E; Prasanna, Pataje G S; Walker, Amanda J; Freeman, Michael L; Eke, Iris; Barcellos-Hoff, Mary Helen; Arankalayil, Molykutty J; Cohen, Eric P; Wilkins, Ruth C; Ahmed, Mansoor M; Anscher, Mitchell S; Movsas, Benjamin; Buchsbaum, Jeffrey C; Mendonca, Marc S; Wynn, Thomas A; Coleman, C Norman
A workshop entitled "Radiation-Induced Fibrosis: Mechanisms and Opportunities to Mitigate" (held in Rockville, MD, September 19, 2016) was organized by the Radiation Research Program and Radiation Oncology Branch of the Center for Cancer Research (CCR) of the National Cancer Institute (NCI), to identify critical research areas and directions that will advance the understanding of radiation-induced fibrosis (RIF) and accelerate the development of strategies to mitigate or treat it. Experts in radiation biology, radiation oncology and related fields met to identify and prioritize the key areas for future research and clinical translation. The consensus was that several known and newly identified targets can prevent or mitigate RIF in pre-clinical models. Further, basic and translational research and focused clinical trials are needed to identify optimal agents and strategies for therapeutic use. It was felt that optimally designed preclinical models are needed to better study biomarkers that predict for development of RIF, as well as to understand when effective therapies need to be initiated in relationship to manifestation of injury. Integrating appropriate endpoints and defining efficacy in clinical trials testing treatment of RIF were felt to be critical to demonstrating efficacy. The objective of this meeting report is to (a) highlight the significance of RIF in a global context, (b) summarize recent advances in our understanding of mechanisms of RIF,
PMCID:5558616
PMID: 28489488
ISSN: 1938-5404
CID: 2979782

Stem cell-like transcriptional reprogramming mediates metastatic resistance to mTOR inhibition

Mateo, F; Arenas, E J; Aguilar, H; Serra-Musach, J; de Garibay, G Ruiz; Boni, J; Maicas, M; Du, S; Iorio, F; Herranz-Ors, C; Islam, A; Prado, X; Llorente, A; Petit, A; Vidal, A; Catala, I; Soler, T; Venturas, G; Rojo-Sebastian, A; Serra, H; Cuadras, D; Blanco, I; Lozano, J; Canals, F; Sieuwerts, A M; de Weerd, V; Look, M P; Puertas, S; Garcia, N; Perkins, A S; Bonifaci, N; Skowron, M; Gomez-Baldo, L; Hernandez, V; Martinez-Aranda, A; Martinez-Iniesta, M; Serrat, X; Ceron, J; Brunet, J; Barretina, M P; Gil, M; Falo, C; Fernandez, A; Morilla, I; Pernas, S; Pla, M J; Andreu, X; Segui, M A; Ballester, R; Castella, E; Nellist, M; Morales, S; Valls, J; Velasco, A; Matias-Guiu, X; Figueras, A; Sanchez-Mut, J V; Sanchez-Cespedes, M; Cordero, A; Gomez-Miragaya, J; Palomero, L; Gomez, A; Gajewski, T F; Cohen, E E W; Jesiotr, M; Bodnar, L; Quintela-Fandino, M; Lopez-Bigas, N; Valdes-Mas, R; Puente, X S; Vinals, F; Casanovas, O; Graupera, M; Hernandez-Losa, J; Ramon Y Cajal, S; Garcia-Alonso, L; Saez-Rodriguez, J; Esteller, M; Sierra, A; Martin-Martin, N; Matheu, A; Carracedo, A; Gonzalez-Suarez, E; Nanjundan, M; Cortes, J; Lazaro, C; Odero, M D; Martens, J W M; Moreno-Bueno, G; Barcellos-Hoff, M H; Villanueva, A; Gomis, R R; Pujana, M A
Inhibitors of the mechanistic target of rapamycin (mTOR) are currently used to treat advanced metastatic breast cancer. However, whether an aggressive phenotype is sustained through adaptation or resistance to mTOR inhibition remains unknown. Here, complementary studies in human tumors, cancer models and cell lines reveal transcriptional reprogramming that supports metastasis in response to mTOR inhibition. This cancer feature is driven by EVI1 and SOX9. EVI1 functionally cooperates with and positively regulates SOX9, and promotes the transcriptional upregulation of key mTOR pathway components (REHB and RAPTOR) and of lung metastasis mediators (FSCN1 and SPARC). The expression of EVI1 and SOX9 is associated with stem cell-like and metastasis signatures, and their depletion impairs the metastatic potential of breast cancer cells. These results establish the mechanistic link between resistance to mTOR inhibition and cancer metastatic potential, thus enhancing our understanding of mTOR targeting failure.Oncogene advance online publication, 19 December 2016; doi:10.1038/onc.2016.427.
PMCID:5442428
PMID: 27991928
ISSN: 1476-5594
CID: 2374272

Hydrogen Peroxide Enhances TGFβ-mediated Epithelial-to-Mesenchymal Transition in Human Mammary Epithelial MCF-10A Cells

Iizuka, Daisuke; Sasatani, Megumi; Barcellos-Hoff, Mary Helen; Kamiya, Kenji
AIM:This study investigated the effect of reactive oxygen species (ROS) on transforming growth factor (TGF)-β-mediated epithelial-to-mesenchymal transition (EMT) in order to clarify the influence of ROS and TGFβ on the induction of dysplasia and ultimately, tumorigenesis. MATERIALS AND METHODS:for 1 h, then reseeded at low density in the presence of TGFβ and cultured until confluence. RESULTS:enhances TGFβ-mediated EMT via SMAD and MEK/ERK signaling.
PMID: 28314256
ISSN: 1791-7530
CID: 2979772

Modification and reversal of the aggressive behavior of triple negative breast cancer by caffeic acid phenethyl ester (CAPE) [Meeting Abstract]

Omene, C; Patel, M; Bochaca, IIlla; Barcellos-Hoff, MH
ISI:000397999001148
ISSN: 1538-7445
CID: 2529382

A TGFbeta-miR-182-BRCA1 axis controls the mammary differentiation hierarchy

Martinez-Ruiz, Haydeliz; Illa-Bochaca, Irineu; Omene, Coral; Hanniford, Douglas; Liu, Qi; Hernando, Eva; Barcellos-Hoff, Mary Helen
Maintenance of mammary functional capacity during cycles of proliferation and regression depends on appropriate cell fate decisions of mammary progenitor cells to populate an epithelium consisting of secretory luminal cells and contractile myoepithelial cells. It is well established that transforming growth factor-beta (TGFbeta) restricts mammary epithelial cell proliferation and that sensitivity to TGFbeta is decreased in breast cancer. We show that TGFbeta also exerts control of mammary progenitor self-renewal and lineage commitment decisions by stringent regulation of breast cancer associated 1 (BRCA1), which controls stem cell self-renewal and lineage commitment. Either genetic depletion of Tgfb1 or transient blockade of TGFbeta increased self-renewal of mammary progenitor cells in mice, cultured primary mammary epithelial cells, and also skewed lineage commitment toward the myoepithelial fate. TGFbeta stabilized the abundance of BRCA1 by reducing the abundance of microRNA-182 (miR-182). Ectopic expression of BRCA1 or antagonism of miR-182 in cultured TGFbeta-deficient mammary epithelial cells restored luminal lineage commitment. These findings reveal that TGFbeta modulation of BRCA1 directs mammary epithelial cell fate and, because stem or progenitor cells are thought to be the cell of origin for aggressive breast cancer subtypes, suggest that TGFbeta dysregulation during tumorigenesis may promote distinct breast cancer subtypes.
PMCID:5619986
PMID: 27923913
ISSN: 1937-9145
CID: 2353502