105 results on '"Aguirre-Ghiso JA"'
Search Results
2. Guidelines for the use and interpretation of assays for monitoring autophagy
- Author
-
Klionsky, DJ, Abdalla, FC, Abeliovich, H, Abraham, RT, Acevedo-Arozena, A, Adeli, K, Agholme, L, Agnello, M, Agostinis, P, Aguirre-Ghiso, JA, Ahn, HJ, Ait-Mohamed, O, Ait-Si-Ali, S, Akematsu, T, Akira, S, Al-Younes, HM, Al-Zeer, MA, Albert, ML, Albin, RL, Alegre-Abarrategui, J, Aleo, MF, Alirezaei, M, Almasan, A, Almonte-Becerril, M, Amano, A, Amaravadi, R, Amarnath, S, Amer, AO, Andrieu-Abadie, N, Anantharam, V, Ann, DK, Anoopkumar-Dukie, S, Aoki, H, Apostolova, N, Arancia, G, Aris, JP, Asanuma, K, Asare, NYO, Ashida, H, Askanas, V, Askew, DS, Auberger, P, Baba, M, Backues, SK, Baehrecke, EH, Bahr, BA, Bai, XY, Bailly, Y, Baiocchi, R, Baldini, G, Balduini, W, Ballabio, A, Bamber, BA, Bampton, ETW, Bánhegyi, G, Bartholomew, CR, Bassham, DC, Bast, RC, Batoko, H, Bay, BH, Beau, I, Béchet, DM, Begley, TJ, Behl, C, Behrends, C, Bekri, S, Bellaire, B, Bendall, LJ, Benetti, L, Berliocchi, L, Bernardi, H, Bernassola, F, Besteiro, S, Bhatia-Kissova, I, Bi, X, Biard-Piechaczyk, M, Blum, JS, Boise, LH, Bonaldo, P, Boone, DL, Bornhauser, BC, Bortoluci, KR, Bossis, I, Bost, F, Bourquin, JP, Boya, P, Boyer-Guittaut, M, Bozhkov, PV, Brady, NR, Brancolini, C, Brech, A, and Brenman, JE
- Abstract
In 2008 we published the first set of guidelines for standardizing research in autophagy. Since then, research on this topic has continued to accelerate, and many new scientists have entered the field. Our knowledge base and relevant new technologies have also been expanding. Accordingly, it is important to update these guidelines for monitoring autophagy in different organisms. Various reviews have described the range of assays that have been used for this purpose. Nevertheless, there continues to be confusion regarding acceptable methods to measure autophagy, especially in multicellular eukaryotes. A key point that needs to be emphasized is that there is a difference between measurements that monitor the numbers or volume of autophagic elements (e.g., autophagosomes or autolysosomes) at any stage of the autophagic process vs. those that measure flux through the autophagy pathway (i.e., the complete process); thus, a block in macroautophagy that results in autophagosome accumulation needs to be differentiated from stimuli that result in increased autophagic activity, defined as increased autophagy induction coupled with increased delivery to, and degradation within, lysosomes (in most higher eukaryotes and some protists such as Dictyostelium) or the vacuole (in plants and fungi). In other words, it is especially important that investigators new to the field understand that the appearance of more autophagosomes does not necessarily equate with more autophagy. In fact, in many cases, autophagosomes accumulate because of a block in trafficking to lysosomes without a concomitant change in autophagosome biogenesis, whereas an increase in autolysosomes may reflect a reduction in degradative activity. Here, we present a set of guidelines for the selection and interpretation of methods for use by investigators who aim to examine macroautophagy and related processes, as well as for reviewers who need to provide realistic and reasonable critiques of papers that are focused on these processes. These guidelines are not meant to be a formulaic set of rules, because the appropriate assays depend in part on the question being asked and the system being used. In addition, we emphasize that no individual assay is guaranteed to be the most appropriate one in every situation, and we strongly recommend the use of multiple assays to monitor autophagy. In these guidelines, we consider these various methods of assessing autophagy and what information can, or cannot, be obtained from them. Finally, by discussing the merits and limits of particular autophagy assays, we hope to encourage technical innovation in the field. © 2012 Landes Bioscience.
- Published
- 2012
- Full Text
- View/download PDF
3. Regulating the activity of p38α/β as a potential therapy against childhood acute lymphoblastic leukemia
- Author
-
Alsadeq, A, primary, Strube, S, additional, Krause, S, additional, Carlet, M, additional, Jeremias, I, additional, Vokuhl, C, additional, Loges, S, additional, Aguirre-Ghiso, JA, additional, Cario, G, additional, Stanulla, M, additional, Schrappe, M, additional, and Schewe, DM, additional
- Published
- 2014
- Full Text
- View/download PDF
4. Lung-resident alveolar macrophages regulate the timing of breast cancer metastasis.
- Author
-
Dalla E, Papanicolaou M, Park MD, Barth N, Hou R, Segura-Villalobos D, Valencia Salazar L, Sun D, Forrest ARR, Casanova-Acebes M, Entenberg D, Merad M, and Aguirre-Ghiso JA
- Subjects
- Animals, Female, Mice, Humans, Cell Line, Tumor, Receptor, Transforming Growth Factor-beta Type II metabolism, Receptor, Transforming Growth Factor-beta Type II genetics, Neoplasm Metastasis, Receptors, Transforming Growth Factor beta metabolism, Signal Transduction, Lung pathology, Lung metabolism, Macrophages, Alveolar metabolism, Breast Neoplasms pathology, Breast Neoplasms metabolism, Lung Neoplasms pathology, Lung Neoplasms secondary, Lung Neoplasms metabolism, Transforming Growth Factor beta2 metabolism
- Abstract
Breast disseminated cancer cells (DCCs) can remain dormant in the lungs for extended periods, but the mechanisms limiting their expansion are not well understood. Research indicates that tissue-resident alveolar macrophages suppress breast cancer metastasis in lung alveoli by inducing dormancy. Through ligand-receptor mapping and intravital imaging, it was found that alveolar macrophages express transforming growth factor (TGF)-β2. This expression, along with persistent macrophage-cancer cell interactions via the TGF-βRIII receptor, maintains cancer cells in a dormant state. Depleting alveolar macrophages or losing the TGF-β2 receptor in cancer cells triggers metastatic awakening. Aggressive breast cancer cells are either suppressed by alveolar macrophages or evade this suppression by avoiding interaction and downregulating the TGF-β2 receptor. Restoring TGF-βRIII in aggressive cells reinstates TGF-β2-mediated macrophage growth suppression. Thus, alveolar macrophages act as a metastasis immune barrier, and downregulation of TGF-β2 signaling allows cancer cells to overcome macrophage-mediated growth suppression., Competing Interests: Declaration of interests J.A.A.-G. is a scientific co-founder and scientific advisory board member of and an equity owner in HiberCell and receives financial compensation as a consultant for HiberCell, a Mount Sinai spin-off company focused on the research and development of therapeutics that prevent or delay the recurrence of cancer. J.A.A.-G. is also a consultant for Astrin Biosciences and Chief Mission Advisor for the Samuel Waxman Cancer Research Foundation. M.M. serves on the scientific advisory board of and holds stock from Compugen Inc., Myeloid Therapeutics Inc., Morphic Therapeutic Inc., Asher Bio Inc., Dren Bio Inc., Nirogy Inc., Oncoresponse Inc., Owkin Inc., Pionyr Inc., OSE Inc., and Larkspur Inc. M.M. serves on the scientific advisory board of Innate Pharma Inc., DBV Inc., and Genenta Inc. M.M. receives funding for contracted research from Regeneron Inc. and Boerhinger Ingelheim Inc. M.M. is a named co-inventor on an issued patent for multiplex immunohistochemistry to characterize tumors and treatment responses. The technology is filed through Icahn School of Medicine at Mount Sinai (ISMMS) and is currently unlicensed. This technology was used to evaluate tissue in this study, and the results could impact the value of this technology. M.M. has ownership interest (<5%) in Compugen Inc., Morphic Therapeutic Inc., Myeloid Therapeutics Inc., Asher Bio Inc., Dren Bio Inc., Nirogy Inc., Owkin Inc., Pionyr Inc., and Larkspur Inc. M.M. and J.A.A.-G. declare no ownership interest greater than or equal to 5%., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Targeting the dependence on PIK3C3-mTORC1 signaling in dormancy-prone breast cancer cells blunts metastasis initiation.
- Author
-
Elkholi IE, Robert A, Malouf C, Kuasne H, Drapela S, Macleod G, Hébert S, Pacis A, Calderon V, Kleinman CL, Gomes AP, Aguirre-Ghiso JA, Park M, Angers S, and Côté JF
- Abstract
Halting breast cancer metastatic relapses following primary tumor removal and the clinical dormant phase, remains challenging, due to a lack of specific vulnerabilities to target during dormancy. To address this, we conducted genome-wide CRISPR screens on two breast cancer cell lines with distinct dormancy properties: 4T1 (short-term dormancy) and 4T07 (prolonged dormancy). We discovered that loss of class-III PI3K, Pik3c3, revealed a unique vulnerability in 4T07 cells. Surprisingly, dormancy-prone 4T07 cells exhibited higher mTORC1 activity than 4T1 cells, due to lysosome-dependent signaling occurring at the cell periphery. Pharmacological inhibition of Pik3c3 counteracted this phenotype in 4T07 cells, and selectively reduced metastasis burden only in the 4T07 dormancy-prone model. This mechanism was also detected in human breast cancer cell lines in addition to a breast cancer patient-derived xenograft supporting that it may be relevant in humans. Our findings suggest dormant cancer cell-initiated metastasis may be prevented in patients carrying tumor cells that display PIK3C3-peripheral lysosomal signaling to mTORC1., Statement of Significance: We reveal that dormancy-prone breast cancer cells depend on the class III PI3K to mediate a constant peripheral lysosomal positioning and mTORC1 hyperactivity. Targeting this pathway might blunt breast cancer metastasis.
- Published
- 2024
- Full Text
- View/download PDF
6. How can we integrate the biology of breast cancer cell dormancy into clinical practice?
- Author
-
Elkholi IE, Rose AAN, Aguirre-Ghiso JA, and Côté JF
- Subjects
- Female, Humans, Neoplasm Metastasis, Neoplasm Recurrence, Local pathology, Neoplasm Recurrence, Local genetics, Breast Neoplasms pathology, Breast Neoplasms genetics, Breast Neoplasms metabolism
- Abstract
Clinical practice and clinical research heavily rely on primary tumors, circulating tumor DNA, and/or overt metastases as sources of material for predicting or investigating breast cancer metastatic relapses. However, these approaches do not consider emerging fundamentals in the biology of metastatic dormancy and relapse. Conversely, the field of metastatic dormancy often discounts key clinical factors influencing relapse dynamics (e.g., patient's age and overall health condition). Here, we delineate these disparities into four gaps and propose a framework to bridge them., Competing Interests: Declaration of interests A.A.N.R. reports personal fees from Advanced Accelerator Applications/Novartis and EMD Serono in addition to institutional research funding for clinical research from: Bayer, Merck, Pfizer, AstraZeneca, and Seagen. She also reports personal/speaker fees from AstraZeneca. These financial relationships are unrelated to the current manuscript. J.A.A.-G. is a scientific co-founder, scientific advisory board member, and equity owner in HiberCell and receives financial compensation as a consultant for HiberCell, a Mount Sinai spin-off company focused on the research and development of therapeutics that prevent or delay the recurrence of cancer. He is a consultant for Astrin Biosciences, Chief Mission Officer for the Samuel Waxman Cancer Research Foundation, and a member of the advisory board of Cancer Cell., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Slow proliferation of BAP1-deficient uveal melanoma cells is associated with reduced S6 signaling and resistance to nutrient stress.
- Author
-
Chua V, Lopez-Anton M, Mizue Terai, Ryota Tanaka, Baqai U, Purwin TJ, Haj JI, Waltrich FJ Jr, Trachtenberg I, Luo K, Tudi R, Jeon A, Han A, Chervoneva I, Davies MA, Aguirre-Ghiso JA, Sato T, and Aplin AE
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Mutation, Phosphorylation, Ribosomal Protein S6 metabolism, Ribosomal Protein S6 genetics, Stress, Physiological, Female, Cell Proliferation, Melanoma genetics, Melanoma metabolism, Melanoma pathology, Signal Transduction, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Ubiquitin Thiolesterase genetics, Ubiquitin Thiolesterase metabolism, Uveal Neoplasms genetics, Uveal Neoplasms metabolism, Uveal Neoplasms pathology
- Abstract
Uveal melanoma (UM) is the deadliest form of eye cancer in adults. Inactivating mutations and/or loss of expression of the gene encoding BRCA1-associated protein 1 (BAP1) in UM tumors are associated with an increased risk of metastasis. To investigate the mechanisms underlying this risk, we explored the functional consequences of BAP1 deficiency. UM cell lines expressing mutant BAP1 grew more slowly than those expressing wild-type BAP1 in culture and in vivo. The ability of BAP1 reconstitution to restore cell proliferation in BAP1-deficient cells required its deubiquitylase activity. Proteomic analysis showed that BAP1-deficient cells had decreased phosphorylation of ribosomal S6 and its upstream regulator, p70S6K1, compared with both wild-type and BAP1 reconstituted cells. In turn, expression of p70S6K1 increased S6 phosphorylation and proliferation of BAP1-deficient UM cells. Consistent with these findings, BAP1 mutant primary UM tumors expressed lower amounts of p70S6K1 target genes, and S6 phosphorylation was decreased in BAP1 mutant patient-derived xenografts (PDXs), which grew more slowly than wild-type PDXs in the liver (the main metastatic site of UM) in mice. BAP1-deficient UM cells were also more resistant to amino acid starvation, which was associated with diminished phosphorylation of S6. These studies demonstrate that BAP1 deficiency slows the proliferation of UM cells through regulation of S6 phosphorylation. These characteristics may be associated with metastasis by ensuring survival during amino acid starvation.
- Published
- 2024
- Full Text
- View/download PDF
8. Respiratory viral infection promotes the awakening and outgrowth of dormant metastatic breast cancer cells in lungs.
- Author
-
Chia SB, Johnson BJ, Hu J, Vermeulen R, Chadeau-Hyam M, Guntoro F, Montgomery H, Boorgula MP, Sreekanth V, Goodspeed A, Davenport B, Pereira FV, Zaberezhnyy V, Schleicher WE, Gao D, Cadar AN, Papanicolaou M, Beheshti A, Baylin SB, Costello J, Bartley JM, Morrison TE, Aguirre-Ghiso JA, Rincon M, and DeGregori J
- Abstract
Breast cancer is the second most common cancer globally. Most deaths from breast cancer are due to metastatic disease which often follows long periods of clinical dormancy
1 . Understanding the mechanisms that disrupt the quiescence of dormant disseminated cancer cells (DCC) is crucial for addressing metastatic progression. Infection with respiratory viruses (e.g. influenza or SARS-CoV-2) is common and triggers an inflammatory response locally and systemically2,3 . Here we show that influenza virus infection leads to loss of the pro-dormancy mesenchymal phenotype in breast DCC in the lung, causing DCC proliferation within days of infection, and a greater than 100-fold expansion of carcinoma cells into metastatic lesions within two weeks. Such DCC phenotypic change and expansion is interleukin-6 (IL-6)-dependent. We further show that CD4 T cells are required for the maintenance of pulmonary metastatic burden post-influenza virus infection, in part through attenuation of CD8 cell responses in the lungs. Single-cell RNA-seq analyses reveal DCC-dependent impairment of T-cell activation in the lungs of infected mice. SARS-CoV-2 infected mice also showed increased breast DCC expansion in lungs post-infection. Expanding our findings to human observational data, we observed that cancer survivors contracting a SARS-CoV-2 infection have substantially increased risks of lung metastatic progression and cancer-related death compared to cancer survivors who did not. These discoveries underscore the significant impact of respiratory viral infections on the resurgence of metastatic cancer, offering novel insights into the interconnection between infectious diseases and cancer metastasis., Competing Interests: Additional Declarations: Yes there is potential Competing Interest. HM has consulted Astra Zeneca relating to the use of monoclonal antibodies in the prevention and treatment of SARS-CoV-2 infection. J.A.A-G is a scientific co-founder, scientific advisory board member and equity owner in HiberCell and receives financial compensation as a consultant for HiberCell, a Mount Sinai spin-off company focused on the research and development of therapeutics that prevent or delay the recurrence of cancer. J.A.A-G is also a consultant for Astrin Biosciences, Inc and chief mission officer of Samuel Waxman Cancer Research Foundation.- Published
- 2024
- Full Text
- View/download PDF
9. Targeting cancer cell dormancy.
- Author
-
Agudo J, Aguirre-Ghiso JA, Bhatia M, Chodosh LA, Correia AL, and Klein CA
- Subjects
- Humans, Cell Proliferation, Cell Line, Tumor, Neoplasms
- Published
- 2024
- Full Text
- View/download PDF
10. Immune evasion by dormant disseminated cancer cells: A Fermi paradox?
- Author
-
Adam-Artigues A, Valencia Salazar LE, and Aguirre-Ghiso JA
- Subjects
- Humans, Immune Evasion, Neoplasms
- Abstract
Rare disseminated tumor cells (DTCs) can persist after treatment in patients for years, and the immune system does not eliminate them. Goddard et al. propose that immune evasion by rare dormant DTCs is due to an improbability of contact imposed by large distances separating effector T cells and DTCs., Competing Interests: Declaration of interests J.A.A.-G is a scientific co-founder of and scientific advisory board member and equity owner in HiberCell and receives financial compensation as a consultant for HiberCell, a Mount Sinai spin-off company focused on the research and development of therapeutics that prevent or delay the recurrence of cancer., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. A PERK-Specific Inhibitor Blocks Metastatic Progression by Limiting Integrated Stress Response-Dependent Survival of Quiescent Cancer Cells.
- Author
-
Calvo V, Zheng W, Adam-Artigues A, Staschke KA, Huang X, Cheung JF, Nobre AR, Fujisawa S, Liu D, Fumagalli M, Surguladze D, Stokes ME, Nowacek A, Mulvihill M, Farias EF, and Aguirre-Ghiso JA
- Subjects
- Humans, Animals, Mice, Female, Cell Line, Tumor, Cell Cycle, Cell Proliferation, Cell Death, eIF-2 Kinase genetics, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms metabolism
- Abstract
Purpose: The integrated stress response (ISR) kinase PERK serves as a survival factor for both proliferative and dormant cancer cells. We aim to validate PERK inhibition as a new strategy to specifically eliminate solitary disseminated cancer cells (DCC) in secondary sites that eventually reawake and originate metastasis., Experimental Design: A novel clinical-grade PERK inhibitor (HC4) was tested in mouse syngeneic and PDX models that present quiescent/dormant DCCs or growth-arrested cancer cells in micro-metastatic lesions that upregulate ISR., Results: HC4 significantly blocks metastasis, by killing quiescent/slow-cycling ISRhigh, but not proliferative ISRlow DCCs. HC4 blocked expansion of established micro-metastasis that contained ISRhigh slow-cycling cells. Single-cell gene expression profiling and imaging revealed that a significant proportion of solitary DCCs in lungs were indeed dormant and displayed an unresolved ER stress as revealed by high expression of a PERK-regulated signature. In human breast cancer metastasis biopsies, GADD34 expression (PERK-regulated gene) and quiescence were positively correlated. HC4 effectively eradicated dormant bone marrow DCCs, which usually persist after rounds of therapies. Importantly, treatment with CDK4/6 inhibitors (to force a quiescent state) followed by HC4 further reduced metastatic burden. In HNSCC and HER2+ cancers HC4 caused cell death in dormant DCCs. In HER2+ tumors, PERK inhibition caused killing by reducing HER2 activity because of sub-optimal HER2 trafficking and phosphorylation in response to EGF., Conclusions: Our data identify PERK as a unique vulnerability in quiescent or slow-cycling ISRhigh DCCs. The use of PERK inhibitors may allow targeting of pre-existing or therapy-induced growth arrested "persister" cells that escape anti-proliferative therapies., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
12. Dormancy in Breast Cancer.
- Author
-
Dalla E, Sreekumar A, Aguirre-Ghiso JA, and Chodosh LA
- Subjects
- Animals, Mice, Humans, Female, Neoplasm Recurrence, Local, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
The pattern of delayed recurrence in a subset of breast cancer patients has long been explained by a model that incorporates a variable period of cellular or tumor mass dormancy prior to disease relapse. In this review, we critically evaluate existing data to develop a framework for inferring the existence of dormancy in clinical contexts of breast cancer. We integrate these clinical data with rapidly evolving mechanistic insights into breast cancer dormancy derived from a broad array of genetically engineered mouse models as well as experimental models of metastasis. Finally, we propose actionable interventions and discuss ongoing clinical trials that translate the wealth of knowledge gained in the laboratory to the long-term clinical management of patients at a high risk of developing recurrence., (Copyright © 2023 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
13. 5-Azacytidine- and retinoic-acid-induced reprogramming of DCCs into dormancy suppresses metastasis via restored TGF-β-SMAD4 signaling.
- Author
-
Singh DK, Carcamo S, Farias EF, Hasson D, Zheng W, Sun D, Huang X, Cheung J, Nobre AR, Kale N, Sosa MS, Bernstein E, and Aguirre-Ghiso JA
- Subjects
- Humans, Azacitidine pharmacology, Head and Neck Neoplasms drug therapy, Transforming Growth Factor beta metabolism, Cell Line, Tumor drug effects, Smad4 Protein, Squamous Cell Carcinoma of Head and Neck drug therapy, Tretinoin pharmacology, Breast Neoplasms drug therapy, Signal Transduction drug effects
- Abstract
Disseminated cancer cells (DCCs) in secondary organs can remain dormant for years to decades before reactivating into overt metastasis. Microenvironmental signals leading to cancer cell chromatin remodeling and transcriptional reprogramming appear to control onset and escape from dormancy. Here, we reveal that the therapeutic combination of the DNA methylation inhibitor 5-azacytidine (AZA) and the retinoic acid receptor ligands all-trans retinoic acid (atRA) or AM80, an RARα-specific agonist, promotes stable dormancy in cancer cells. Treatment of head and neck squamous cell carcinoma (HNSCC) or breast cancer cells with AZA+atRA induces a SMAD2/3/4-dependent transcriptional program that restores transforming growth factor β (TGF-β)-signaling and anti-proliferative function. Significantly, either combination, AZA+atRA or AZA+AM80, strongly suppresses HNSCC lung metastasis formation by inducing and maintaining solitary DCCs in a SMAD4
+ /NR2F1+ non-proliferative state. Notably, SMAD4 knockdown is sufficient to drive resistance to AZA+atRA-induced dormancy. We conclude that therapeutic doses of AZA and RAR agonists may induce and/or maintain dormancy and significantly limit metastasis development., Competing Interests: Declaration of interests J.A.A.-G. is a scientific co-founder of, scientific advisory board member at, and equity owner in HiberCell and receives financial compensation as a consultant for HiberCell, a Mount Sinai spin-off company focused on therapeutics that prevent or delay cancer recurrence., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
14. IGF1R Inhibition Enhances the Therapeutic Effects of Gq/11 Inhibition in Metastatic Uveal Melanoma Progression.
- Author
-
Lapadula D, Lam B, Terai M, Sugase T, Tanaka R, Farias E, Kadamb R, Lopez-Anton M, Heine CC, Modasia B, Aguirre-Ghiso JA, Aplin AE, Sato T, and Benovic JL
- Subjects
- Mice, Animals, Signal Transduction, GTP-Binding Protein alpha Subunits, Gq-G11 genetics, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Cell Line, Tumor, Melanoma drug therapy, Melanoma genetics, Melanoma pathology, Uveal Neoplasms genetics
- Abstract
Uveal melanoma (UM) is the most common intraocular tumor in adults, and up to 50% of patients develop metastatic disease, which remains uncurable. Because patients with metastatic UM have an average survival of less than 1 year after diagnosis, there is an urgent need to develop new treatment strategies. Although activating mutations in Gαq or Gα11 proteins are major drivers of pathogenesis, the therapeutic intervention of downstream Gαq/11 targets has been unsuccessful in treating UM, possibly due to alternative signaling pathways and/or resistance mechanisms. Activation of the insulin-like growth factor 1 (IGF1) signaling pathway promotes cell growth, metastasis, and drug resistance in many types of cancers, including UM, where expression of the IGF1 receptor (IGF1R) correlates with a poor prognosis. In this article, we show that direct inhibition of Gαq/11 by the cyclic depsipeptide YM-254890 in combination with inhibition of IGF1R by linsitinib cooperatively inhibits downstream signaling and proliferation of UM cells. We further demonstrate that a 2-week combination treatment of 0.3 to 0.4 mg/kg of YM-254890 administered by intraperitoneal injection and 25 to 40 mg/kg linsitinib administered by oral gavage effectively inhibits the growth of metastatic UM tumors in immunodeficient NOD scid gamma (NSG) mice and identifies the IGF1 pathway as a potential resistance mechanism in response to Gαq/11 inhibition in UM. These data suggest that the combination of Gαq/11 and IGF1R inhibition provides a promising therapeutic strategy to treat metastatic UM., (©2022 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
15. MacroH2A impedes metastatic growth by enforcing a discrete dormancy program in disseminated cancer cells.
- Author
-
Sun D, Singh DK, Carcamo S, Filipescu D, Khalil B, Huang X, Miles BA, Westra W, Sproll KC, Hasson D, Bernstein E, and Aguirre-Ghiso JA
- Subjects
- Humans, Carcinogenesis, Cell Division, Cell Cycle, Histones, Head and Neck Neoplasms genetics
- Abstract
MacroH2A variants have been linked to inhibition of metastasis through incompletely understood mechanisms. Here, we reveal that solitary dormant disseminated cancer cells (DCCs) display increased levels of macroH2A variants in head and neck squamous cell carcinoma PDX in vivo models and patient samples compared to proliferating primary or metastatic lesions. We demonstrate that dormancy-inducing transforming growth factor-β2 and p38α/β pathways up-regulate macroH2A expression and that macroH2A variant overexpression is sufficient to induce DCC dormancy and suppress metastasis in vivo. Notably, inducible expression of the macroH2A2 variant in vivo suppresses metastasis via a reversible growth arrest of DCCs. This state does not require the dormancy-regulating transcription factors DEC2 and NR2F1; instead, transcriptomic analysis reveals that macroH2A2 overexpression inhibits cell cycle and oncogenic signaling programs, while up-regulating dormancy and senescence-associated inflammatory cytokines. We conclude that the macroH2A2-enforced dormant phenotype results from tapping into transcriptional programs of both quiescence and senescence to limit metastatic outgrowth.
- Published
- 2022
- Full Text
- View/download PDF
16. ZFP281 drives a mesenchymal-like dormancy program in early disseminated breast cancer cells that prevents metastatic outgrowth in the lung.
- Author
-
Nobre AR, Dalla E, Yang J, Huang X, Wullkopf L, Risson E, Razghandi P, Anton ML, Zheng W, Seoane JA, Curtis C, Kenigsberg E, Wang J, and Aguirre-Ghiso JA
- Subjects
- Humans, Transcription Factors genetics, Lung, Fibroblast Growth Factor 2, Neoplasms
- Abstract
Increasing evidence shows that cancer cells can disseminate from early evolved primary lesions much earlier than the classical metastasis models predicted. Here, we reveal at a single-cell resolution that mesenchymal-like (M-like) and pluripotency-like programs coordinate dissemination and a long-lived dormancy program of early disseminated cancer cells (DCCs). The transcription factor ZFP281 induces a permissive state for heterogeneous M-like transcriptional programs, which associate with a dormancy signature and phenotype in vivo. Downregulation of ZFP281 leads to a loss of an invasive, M-like dormancy phenotype and a switch to lung metastatic outgrowth. We also show that FGF2 and TWIST1 induce ZFP281 expression to induce the M-like state, which is linked to CDH1 downregulation and upregulation of CDH11. We found that ZFP281 not only controls the early dissemination of cancer cells but also locks early DCCs in a dormant state by preventing the acquisition of an epithelial-like proliferative program and consequent metastases outgrowth., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
17. Metastasis.
- Author
-
Lyden D, Ghajar CM, Correia AL, Aguirre-Ghiso JA, Cai S, Rescigno M, Zhang P, Hu G, Fendt SM, Boire A, Weichselbaum RR, and Katipally RR
- Subjects
- Humans, Immune System pathology, Neoplasm Metastasis pathology, Neoplasms drug therapy, Neoplasms pathology, Tumor Microenvironment
- Abstract
Metastasis, the major cause of cancer death, represents one of the major challenges in oncology. Scientists are still trying to understand the biological basis underlying the dissemination and outgrowth of tumor cells, why these cells can remain dormant for years, how they become resistant to the immune system or cytotoxic effects of systemic therapy, and how they interact with their new microenvironment. We asked experts to discuss some of the unknowns, advances, and areas of opportunity related to cancer metastasis., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
18. Stromal changes in the aged lung induce an emergence from melanoma dormancy.
- Author
-
Fane ME, Chhabra Y, Alicea GM, Maranto DA, Douglass SM, Webster MR, Rebecca VW, Marino GE, Almeida F, Ecker BL, Zabransky DJ, Hüser L, Beer T, Tang HY, Kossenkov A, Herlyn M, Speicher DW, Xu W, Xu X, Jaffee EM, Aguirre-Ghiso JA, and Weeraratna AT
- Subjects
- Aged, Fibroblasts pathology, Humans, Neoplasm Invasiveness pathology, Neoplasm, Residual, Proto-Oncogene Proteins, Receptor Protein-Tyrosine Kinases, Skin pathology, Wnt-5a Protein, c-Mer Tyrosine Kinase, Axl Receptor Tyrosine Kinase, Aging pathology, Lung pathology, Melanoma pathology, Neoplasm Metastasis pathology, Stromal Cells pathology, Tumor Microenvironment
- Abstract
Disseminated cancer cells from primary tumours can seed in distal tissues, but may take several years to form overt metastases, a phenomenon that is termed tumour dormancy. Despite its importance in metastasis and residual disease, few studies have been able to successfully characterize dormancy within melanoma. Here we show that the aged lung microenvironment facilitates a permissive niche for efficient outgrowth of dormant disseminated cancer cells-in contrast to the aged skin, in which age-related changes suppress melanoma growth but drive dissemination. These microenvironmental complexities can be explained by the phenotype switching model, which argues that melanoma cells switch between a proliferative cell state and a slower-cycling, invasive state
1-3 . It was previously shown that dermal fibroblasts promote phenotype switching in melanoma during ageing4-8 . We now identify WNT5A as an activator of dormancy in melanoma disseminated cancer cells within the lung, which initially enables the efficient dissemination and seeding of melanoma cells in metastatic niches. Age-induced reprogramming of lung fibroblasts increases their secretion of the soluble WNT antagonist sFRP1, which inhibits WNT5A in melanoma cells and thereby enables efficient metastatic outgrowth. We also identify the tyrosine kinase receptors AXL and MER as promoting a dormancy-to-reactivation axis within melanoma cells. Overall, we find that age-induced changes in distal metastatic microenvironments promote the efficient reactivation of dormant melanoma cells in the lung., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
19. Altered BAF occupancy and transcription factor dynamics in PBAF-deficient melanoma.
- Author
-
Carcamo S, Nguyen CB, Grossi E, Filipescu D, Alpsoy A, Dhiman A, Sun D, Narang S, Imig J, Martin TC, Parsons R, Aifantis I, Tsirigos A, Aguirre-Ghiso JA, Dykhuizen EC, Hasson D, and Bernstein E
- Subjects
- Animals, Chromatin, Chromatin Assembly and Disassembly, Gene Expression Regulation, Humans, Chromosomal Proteins, Non-Histone, Melanoma genetics, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
ARID2 is the most recurrently mutated SWI/SNF complex member in melanoma; however, its tumor-suppressive mechanisms in the context of the chromatin landscape remain to be elucidated. Here, we model ARID2 deficiency in melanoma cells, which results in defective PBAF complex assembly with a concomitant genomic redistribution of the BAF complex. Upon ARID2 depletion, a subset of PBAF and shared BAF-PBAF-occupied regions displays diminished chromatin accessibility and associated gene expression, while BAF-occupied enhancers gain chromatin accessibility and expression of genes linked to the process of invasion. As a function of altered accessibility, the genomic occupancy of melanoma-relevant transcription factors is affected and significantly correlates with the observed transcriptional changes. We further demonstrate that ARID2-deficient cells acquire the ability to colonize distal organs in multiple animal models. Taken together, our results reveal a role for ARID2 in mediating BAF and PBAF subcomplex chromatin dynamics with consequences for melanoma metastasis., Competing Interests: Declaration of interests J.A.A.-G. is a scientific co-founder, scientific advisory board member, and equity owner of HiberCell and receives financial compensation as a consultant., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
20. Primary tumor associated macrophages activate programs of invasion and dormancy in disseminating tumor cells.
- Author
-
Borriello L, Coste A, Traub B, Sharma VP, Karagiannis GS, Lin Y, Wang Y, Ye X, Duran CL, Chen X, Friedman M, Sosa MS, Sun D, Dalla E, Singh DK, Oktay MH, Aguirre-Ghiso JA, Condeelis JS, and Entenberg D
- Subjects
- Animals, Breast Neoplasms genetics, Cell Line, Tumor, Female, Humans, Mice, Mice, Inbred C57BL, Neoplasms, Experimental, Neoplastic Stem Cells, Phenotype, Tumor Microenvironment physiology, Tumor-Associated Macrophages physiology
- Abstract
Metastases are initiated by disseminated tumor cells (DTCs) that colonize distant organs. Growing evidence suggests that the microenvironment of the primary tumor primes DTCs for dormant or proliferative fates. However, the manner in which this occurs remains poorly understood. Here, using the Window for High-Resolution Intravital Imaging of the Lung (WHRIL), we study the live lung longitudinally and follow the fate of individual DTCs that spontaneously disseminate from orthotopic breast tumors. We find that spontaneously DTCs have increased levels of retention, increased speed of extravasation, and greater survival after extravasation, compared to experimentally metastasized tumor cells. Detailed analysis reveals that a subset of macrophages within the primary tumor induces a pro-dissemination and pro-dormancy DTC phenotype. Our work provides insight into how specific primary tumor microenvironments prime a subpopulation of cells for expression of proteins associated with dissemination and dormancy., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
21. An NR2F1-specific agonist suppresses metastasis by inducing cancer cell dormancy.
- Author
-
Khalil BD, Sanchez R, Rahman T, Rodriguez-Tirado C, Moritsch S, Martinez AR, Miles B, Farias E, Mezei M, Nobre AR, Singh D, Kale N, Sproll KC, Sosa MS, and Aguirre-Ghiso JA
- Subjects
- Animals, COUP Transcription Factor I genetics, COUP Transcription Factor I metabolism, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell pathology, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic drug effects, HEK293 Cells, Head and Neck Neoplasms genetics, Head and Neck Neoplasms pathology, Humans, Kaplan-Meier Estimate, Lung Neoplasms genetics, Lung Neoplasms secondary, Mice, Inbred BALB C, Mice, Nude, Molecular Structure, RNA-Seq methods, Small Molecule Libraries chemistry, Small Molecule Libraries metabolism, Xenograft Model Antitumor Assays methods, Mice, COUP Transcription Factor I agonists, Carcinoma, Squamous Cell drug therapy, Head and Neck Neoplasms drug therapy, Lung Neoplasms prevention & control, Small Molecule Libraries pharmacology
- Abstract
We describe the discovery of an agonist of the nuclear receptor NR2F1 that specifically activates dormancy programs in malignant cells. The agonist led to a self-regulated increase in NR2F1 mRNA and protein and downstream transcription of a novel dormancy program. This program led to growth arrest of an HNSCC PDX line, human cell lines, and patient-derived organoids in 3D cultures and in vivo. This effect was lost when NR2F1 was knocked out by CRISPR-Cas9. RNA sequencing revealed that agonist treatment induces transcriptional changes associated with inhibition of cell cycle progression and mTOR signaling, metastasis suppression, and induction of a neural crest lineage program. In mice, agonist treatment resulted in inhibition of lung HNSCC metastasis, even after cessation of the treatment, where disseminated tumor cells displayed an NR2F1hi/p27hi/Ki-67lo/p-S6lo phenotype and remained in a dormant single-cell state. Our work provides proof of principle supporting the use of NR2F1 agonists to induce dormancy as a therapeutic strategy to prevent metastasis., Competing Interests: Disclosures: J.A. Aguirre-Ghiso reported grants from HiberCell LLC during the conduct of the study; grants from HiberCell LLC outside the submitted work; and is a HiberCell LLC co-founder, consultant, and scientific advisory board member. No other disclosures were reported., (© 2021 Khalil et al.)
- Published
- 2022
- Full Text
- View/download PDF
22. A tumor-derived type III collagen-rich ECM niche regulates tumor cell dormancy.
- Author
-
Di Martino JS, Nobre AR, Mondal C, Taha I, Farias EF, Fertig EJ, Naba A, Aguirre-Ghiso JA, and Bravo-Cordero JJ
- Subjects
- Cell Proliferation, Extracellular Matrix, Humans, Squamous Cell Carcinoma of Head and Neck, Collagen Type III, Head and Neck Neoplasms
- Abstract
Cancer cells disseminate and seed in distant organs, where they can remain dormant for many years before forming clinically detectable metastases. Here we studied how disseminated tumor cells sense and remodel the extracellular matrix (ECM) to sustain dormancy. ECM proteomics revealed that dormant cancer cells assemble a type III collagen-enriched ECM niche. Tumor-derived type III collagen is required to sustain tumor dormancy, as its disruption restores tumor cell proliferation through DDR1-mediated STAT1 signaling. Second-harmonic generation two-photon microscopy further revealed that the dormancy-to-reactivation transition is accompanied by changes in type III collagen architecture and abundance. Analysis of clinical samples revealed that type III collagen levels were increased in tumors from patients with lymph node-negative head and neck squamous cell carcinoma compared to patients who were positive for lymph node colonization. Our data support the idea that the manipulation of these mechanisms could serve as a barrier to metastasis through disseminated tumor cell dormancy induction., (© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
23. Translating the Science of Cancer Dormancy to the Clinic.
- Author
-
Aguirre-Ghiso JA
- Subjects
- Animals, Biomarkers, Tumor, Clinical Trials as Topic, Disease Management, Humans, Molecular Targeted Therapy methods, Neoplasms diagnosis, Neoplasms therapy, Recurrence, Research Design, Treatment Outcome, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Disease Susceptibility immunology, Neoplasms etiology, Neoplasms metabolism, Translational Research, Biomedical
- Abstract
The paradigm of metastasis has been significantly remodeled by the incorporation of cancer dormancy as a mechanism to explain long-term remission intervals followed by relapse. There is overall consensus on the potential impact of better understanding dormancy. Key cancer-cell autonomous and microenvironmental mechanisms might explain this biology and, in turn, the timing of metastasis. However, the approach and feasibility to apply this biology to clinical trials has been controversial. The discussion here provides insight into how these controversies are being resolved by the development of active clinical trials, thus bringing to reality opportunities to target cancer dormancy., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
24. Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells.
- Author
-
Casanova-Acebes M, Dalla E, Leader AM, LeBerichel J, Nikolic J, Morales BM, Brown M, Chang C, Troncoso L, Chen ST, Sastre-Perona A, Park MD, Tabachnikova A, Dhainaut M, Hamon P, Maier B, Sawai CM, Agulló-Pascual E, Schober M, Brown BD, Reizis B, Marron T, Kenigsberg E, Moussion C, Benaroch P, Aguirre-Ghiso JA, and Merad M
- Subjects
- Animals, CD8-Positive T-Lymphocytes immunology, Epithelial-Mesenchymal Transition, Female, Humans, Male, Mice, Mice, Inbred C57BL, Neoplasm Invasiveness, T-Lymphocytes, Regulatory immunology, Carcinogenesis, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms pathology, Macrophages immunology, Tumor Microenvironment
- Abstract
Macrophages have a key role in shaping the tumour microenvironment (TME), tumour immunity and response to immunotherapy, which makes them an important target for cancer treatment
1,2 . However, modulating macrophages has proved extremely difficult, as we still lack a complete understanding of the molecular and functional diversity of the tumour macrophage compartment. Macrophages arise from two distinct lineages. Tissue-resident macrophages self-renew locally, independent of adult haematopoiesis3-5 , whereas short-lived monocyte-derived macrophages arise from adult haematopoietic stem cells, and accumulate mostly in inflamed lesions1 . How these macrophage lineages contribute to the TME and cancer progression remains unclear. To explore the diversity of the macrophage compartment in human non-small cell lung carcinoma (NSCLC) lesions, here we performed single-cell RNA sequencing of tumour-associated leukocytes. We identified distinct populations of macrophages that were enriched in human and mouse lung tumours. Using lineage tracing, we discovered that these macrophage populations differ in origin and have a distinct temporal and spatial distribution in the TME. Tissue-resident macrophages accumulate close to tumour cells early during tumour formation to promote epithelial-mesenchymal transition and invasiveness in tumour cells, and they also induce a potent regulatory T cell response that protects tumour cells from adaptive immunity. Depletion of tissue-resident macrophages reduced the numbers and altered the phenotype of regulatory T cells, promoted the accumulation of CD8+ T cells and reduced tumour invasiveness and growth. During tumour growth, tissue-resident macrophages became redistributed at the periphery of the TME, which becomes dominated by monocyte-derived macrophages in both mouse and human NSCLC. This study identifies the contribution of tissue-resident macrophages to early lung cancer and establishes them as a target for the prevention and treatment of early lung cancer lesions., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
25. Prostate Cancer Dormancy and Reactivation in Bone Marrow.
- Author
-
Singh DK, Patel VG, Oh WK, and Aguirre-Ghiso JA
- Abstract
Prostate cancer has a variable clinical course, ranging from curable local disease to lethal metastatic spread. Eradicating metastatic cells is a unique challenge that is rarely met with the available therapies. Thus, targeting prostate cancer cells in earlier disease states is a crucial window of opportunity. Interestingly, cancer cells migrate from their primary site during pre-cancerous and malignant phases to seed secondary organs. These cells, known as disseminated cancer cells (DCCs), may remain dormant for months or decades before activating to form metastases. Bone marrow, a dormancy-permissive site, is the major organ for housed DCCs and eventual metastases in prostate cancer. The dynamic interplay between DCCs and the primary tumor microenvironment (TME), as well as that between DCCs and the secondary organ niche, controls the conversion between states of dormancy and activation. Here, we discuss recent discoveries that have improved our understanding of dormancy signaling and the role of the TME in modulating the epigenetic reprogramming of DCCs. We offer potential strategies to target DCCs in prostate cancer.
- Published
- 2021
- Full Text
- View/download PDF
26. The State of Melanoma: Emergent Challenges and Opportunities.
- Author
-
Atkins MB, Curiel-Lewandrowski C, Fisher DE, Swetter SM, Tsao H, Aguirre-Ghiso JA, Soengas MS, Weeraratna AT, Flaherty KT, Herlyn M, Sosman JA, Tawbi HA, Pavlick AC, Cassidy PB, Chandra S, Chapman PB, Daud A, Eroglu Z, Ferris LK, Fox BA, Gershenwald JE, Gibney GT, Grossman D, Hanks BA, Hanniford D, Hernando E, Jeter JM, Johnson DB, Khleif SN, Kirkwood JM, Leachman SA, Mays D, Nelson KC, Sondak VK, Sullivan RJ, and Merlino G
- Subjects
- Biomedical Research methods, Biomedical Research trends, COVID-19 epidemiology, COVID-19 virology, Humans, Medical Oncology organization & administration, Medical Oncology trends, Melanoma diagnosis, SARS-CoV-2 physiology, Skin Neoplasms diagnosis, COVID-19 prevention & control, Medical Oncology methods, Melanoma therapy, Practice Guidelines as Topic, SARS-CoV-2 isolation & purification, Skin Neoplasms therapy
- Abstract
Five years ago, the Melanoma Research Foundation (MRF) conducted an assessment of the challenges and opportunities facing the melanoma research community and patients with melanoma. Since then, remarkable progress has been made on both the basic and clinical research fronts. However, the incidence, recurrence, and death rates for melanoma remain unacceptably high and significant challenges remain. Hence, the MRF Scientific Advisory Council and Breakthrough Consortium, a group that includes clinicians and scientists, reconvened to facilitate intensive discussions on thematic areas essential to melanoma researchers and patients alike, prevention, detection, diagnosis, metastatic dormancy and progression, response and resistance to targeted and immune-based therapy, and the clinical consequences of COVID-19 for patients with melanoma and providers. These extensive discussions helped to crystalize our understanding of the challenges and opportunities facing the broader melanoma community today. In this report, we discuss the progress made since the last MRF assessment, comment on what remains to be overcome, and offer recommendations for the best path forward., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
27. Bone marrow NG2 + /Nestin + mesenchymal stem cells drive DTC dormancy via TGFβ2.
- Author
-
Nobre AR, Risson E, Singh DK, Di Martino JS, Cheung JF, Wang J, Johnson J, Russnes HG, Bravo-Cordero JJ, Birbrair A, Naume B, Azhar M, Frenette PS, and Aguirre-Ghiso JA
- Subjects
- Bone Marrow metabolism, Female, Humans, Neoplasm Recurrence, Local metabolism, Nestin metabolism, Tumor Microenvironment, Breast Neoplasms genetics, Mesenchymal Stem Cells metabolism
- Abstract
In the bone marrow (BM) microenvironment, where breast cancer (BC) disseminated tumour cells (DTCs) can remain dormant for decades, NG2
+ /Nestin+ mesenchymal stem cells (MSCs) promote hematopoietic stem cell quiescence. Here, we reveal that periarteriolar BM-resident NG2+ /Nestin+ MSCs can also instruct BC DTCs to enter dormancy. NG2+ /Nestin+ MSCs produce TGFβ2 and BMP7 and activate a quiescence pathway dependent on TGFBRIII and BMPRII, which via p38-kinase result in p27 induction. Genetic depletion of MSCs or conditional knock-out of TGFβ2 in MSCs using an NG2-CreER driver led to bone metastatic outgrowth of otherwise dormant p27+ /Ki67- DTCs. Also ER+ BC patients without systemic recurrence displayed higher frequency of TGFβ2 and BMP7 detection in the BM. Our results provide a direct proof that HSC dormancy niches control BC DTC dormancy and suggest that aging or extrinsic factors that affect the NG2+ /Nestin+ MSC niche homeostasis may result in a break from dormancy and BC bone relapse.- Published
- 2021
- Full Text
- View/download PDF
28. Immobilization rapidly selects for chemoresistant ovarian cancer cells with enhanced ability to enter dormancy.
- Author
-
Lam T, Aguirre-Ghiso JA, Geller MA, Aksan A, and Azarin SM
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents, Phytogenic pharmacology, Cell Line, Tumor, Cell Proliferation, Female, Humans, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Signal Transduction, Biological Assay methods, Cell Survival, Cisplatin pharmacology, Drug Resistance, Neoplasm, Ovarian Neoplasms drug therapy, Paclitaxel pharmacology, Silica Gel chemistry
- Abstract
Around 20-30% of ovarian cancer patients exhibit chemoresistance, but there are currently no methods to predict whether a patient will respond to chemotherapy. Here, we discovered that chemoresistant ovarian cancer cells exhibit enhanced survival in a quiescent state upon experiencing the stress of physical confinement. When immobilized in stiff silica gels, most ovarian cancer cells die within days, but surviving cells exhibit hallmarks of single-cell dormancy. Upon extraction from gels, the cells resume proliferation but demonstrate enhanced viability upon reimmobilization, indicating that initial immobilization selects for cells with a higher propensity to enter dormancy. RNA-seq analysis of the extracted cells shows they have signaling responses similar to cells surviving cisplatin treatment, and in comparison to chemoresistant patient cohorts, they share differentially expressed genes that are associated with platinum-resistance pathways. Furthermore, these extracted cells demonstrate greater resistance to cisplatin and paclitaxel, despite being proliferative. In contrast, serum starvation and hypoxia could not effectively select for chemoresistant cells upon removal of the environmental stress. These findings demonstrate that ovarian cancer chemoresistance and the ability to enter dormancy are linked, and immobilization rapidly distinguishes chemoresistant cells. This platform could be suitable for mechanistic studies, drug development, or as a clinical diagnostic tool., (© 2020 Wiley Periodicals LLC.)
- Published
- 2020
- Full Text
- View/download PDF
29. The current paradigm and challenges ahead for the dormancy of disseminated tumor cells.
- Author
-
Risson E, Nobre AR, Maguer-Satta V, and Aguirre-Ghiso JA
- Subjects
- Disease Progression, Humans, Neoplasm, Residual pathology, Neoplasm Recurrence, Local, Tumor Microenvironment
- Abstract
Disseminated tumor cells (DTCs) are known to enter a state of dormancy that is achieved via growth arrest of DTCs and/or a form of population equilibrium state, strongly influenced by the organ microenvironment. During this time, expansion of residual disseminated cancer is paused and DTCs survive to fuel relapse, sometimes decades later. This notion has opened a new window of opportunity for intervening and preventing relapse. Here we review recent data that have further augmented the understanding of cancer dormancy and discuss how this is leading to new strategies for monitoring and targeting dormant cancer.
- Published
- 2020
- Full Text
- View/download PDF
30. The importance of developing therapies targeting the biological spectrum of metastatic disease.
- Author
-
Zijlstra A, Von Lersner A, Yu D, Borrello L, Oudin M, Kang Y, Sahai E, Fingleton B, Stein U, Cox TR, Price JT, Kato Y, Welm AL, and Aguirre-Ghiso JA
- Subjects
- Animals, Drug Development, Humans, Neoplasm Metastasis prevention & control, Neoplasm Metastasis drug therapy
- Abstract
Great progress has been made in cancer therapeutics. However, metastasis remains the predominant cause of death from cancer. Importantly, metastasis can manifest many years after initial treatment of the primary cancer. This is because cancer cells can remain dormant before forming symptomatic metastasis. An important question is whether metastasis research should focus on the early treatment of metastases, before they are clinically evident ("overt"), or on developing treatments to stop overt metastasis (stage IV cancer). In this commentary we want to clarify why it is important that all avenues of treatment for stage IV patients are developed. Indeed, future treatments are expected to go beyond the mere shrinkage of overt metastases and will include strategies that prevent disseminated tumor cells from emerging from dormancy.
- Published
- 2019
- Full Text
- View/download PDF
31. An IRAK1-PIN1 signalling axis drives intrinsic tumour resistance to radiation therapy.
- Author
-
Liu PH, Shah RB, Li Y, Arora A, Ung PM, Raman R, Gorbatenko A, Kozono S, Zhou XZ, Brechin V, Barbaro JM, Thompson R, White RM, Aguirre-Ghiso JA, Heymach JV, Lu KP, Silva JM, Panageas KS, Schlessinger A, Maki RG, Skinner HD, de Stanchina E, and Sidi S
- Subjects
- Animals, Cell Line, Tumor, HCT116 Cells, HEK293 Cells, HeLa Cells, Humans, Interleukin-1 Receptor-Associated Kinases antagonists & inhibitors, Interleukin-1 Receptor-Associated Kinases genetics, MCF-7 Cells, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Mutation, NIMA-Interacting Peptidylprolyl Isomerase antagonists & inhibitors, NIMA-Interacting Peptidylprolyl Isomerase genetics, Neoplasms genetics, Neoplasms metabolism, Radiation Tolerance drug effects, Radiation Tolerance genetics, Tumor Suppressor Protein p53 genetics, Zebrafish, Interleukin-1 Receptor-Associated Kinases metabolism, NIMA-Interacting Peptidylprolyl Isomerase metabolism, Neoplasms radiotherapy, Signal Transduction, Xenograft Model Antitumor Assays methods
- Abstract
Drug-based strategies to overcome tumour resistance to radiotherapy (R-RT) remain limited by the single-agent toxicity of traditional radiosensitizers (for example, platinums) and a lack of targeted alternatives. In a screen for compounds that restore radiosensitivity in p53 mutant zebrafish while tolerated in non-irradiated wild-type animals, we identified the benzimidazole anthelmintic oxfendazole. Surprisingly, oxfendazole acts via the inhibition of IRAK1, a kinase thus far implicated in interleukin-1 receptor (IL-1R) and Toll-like receptor (TLR) immune responses. IRAK1 drives R-RT in a pathway involving IRAK4 and TRAF6 but not the IL-1R/TLR-IRAK adaptor MyD88. Rather than stimulating nuclear factor-κB, radiation-activated IRAK1 prevented apoptosis mediated by the PIDDosome complex (comprising PIDD, RAIDD and caspase-2). Countering this pathway with IRAK1 inhibitors suppressed R-RT in tumour models derived from cancers in which TP53 mutations predict R-RT. Moreover, IRAK1 inhibitors synergized with inhibitors of PIN1, a prolyl isomerase essential for IRAK1 activation in response to pathogens and, as shown here, in response to ionizing radiation. These data identify an IRAK1 radiation-response pathway as a rational chemoradiation therapy target.
- Published
- 2019
- Full Text
- View/download PDF
32. The Different Routes to Metastasis via Hypoxia-Regulated Programs.
- Author
-
Nobre AR, Entenberg D, Wang Y, Condeelis J, and Aguirre-Ghiso JA
- Subjects
- Animals, Humans, Hypoxia metabolism, Neoplasm Metastasis pathology, Neoplasms metabolism, Neoplasms pathology
- Abstract
Hypoxia is linked to metastasis; however, how it affects metastatic progression is not clear due to limited consensus in the literature. We posit that this lack of consensus is due to hypoxia being studied using different approaches, such as in vitro, primary tumor, or metastasis assays in an isolated manner. Here, we review the pros and cons of in vitro hypoxia assays, highlight in vivo studies that inform on physiological hypoxia, and review the evidence that primary tumor hypoxia might influence the fate of disseminated tumor cells (DTCs) in secondary organs. Our analysis suggests that consensus can be reached by using in vivo methods of study, which also allow better modeling of how hypoxia affects DTC fate and metastasis., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
33. NR2F1 stratifies dormant disseminated tumor cells in breast cancer patients.
- Author
-
Borgen E, Rypdal MC, Sosa MS, Renolen A, Schlichting E, Lønning PE, Synnestvedt M, Aguirre-Ghiso JA, and Naume B
- Subjects
- Biomarkers, Tumor blood, Breast Neoplasms blood, Breast Neoplasms drug therapy, Cell Line, Tumor, Chemotherapy, Adjuvant, Female, Humans, Ki-67 Antigen blood, Leukocytes, Mononuclear metabolism, Neoplasm Recurrence, Local, Prognosis, Survival Analysis, Bone Marrow Cells metabolism, Breast Neoplasms metabolism, COUP Transcription Factor I metabolism, Neoplastic Cells, Circulating metabolism
- Abstract
Background: The presence of disseminated tumor cells (DTCs) in bone marrow (BM) is an independent prognostic factor in early breast cancer but does not uniformly predict outcome. Tumor cells can persist in a quiescent state over time, but clinical studies of markers predicting the awakening potential of DTCs are lacking. Recently, experiments have shown that NR2F1 (COUP-TF1) plays a key role in dormancy signaling., Methods: We analyzed the NR2F1 expression in DTCs by double immunofluorescence (DIF) staining of extra cytospins prepared from 114 BM samples from 86 selected DTC-positive breast cancer patients. Samples collected at two or more time points were available for 24 patients. Fifteen samples were also analyzed for the proliferation marker Ki67., Results: Of the patients with detectable DTCs by DIF, 27% had ≥ 50% NR2F1
high DTCs, chosen a priori as the cut-off for "dormant profile" classification. All patients with systemic relapse within 12 months after BM aspiration carried ≤ 1% NR2F1high DTCs, including patients who transitioned from having NR2F1high -expressing DTCs in previous BM samples. Of the patients with serial samples, half of those with no relapse at follow-up had ≥ 50% NR2F1high DTCs in the last BM aspiration analyzed. Among the 18 relapse-free patients at the time of the last DTC-positive BM aspiration with no subsequent BM analysis performed, distant disease-free intervals were favorable for patients carrying ≥ 50% NR2F1high DTCs compared with those with predominantly NR2F1low DTCs (p = 0.007, log-rank). No survival difference was observed by classification according to Ki67-expressing DTCs (p = 0.520)., Conclusions: Our study translates findings from basic biological analysis of DTC dormancy to the clinical situation and supports further clinical studies of NR2F1 as a marker of dormancy.- Published
- 2018
- Full Text
- View/download PDF
34. How dormant cancer persists and reawakens.
- Author
-
Aguirre-Ghiso JA
- Subjects
- Humans, Neoplasms
- Published
- 2018
- Full Text
- View/download PDF
35. Corrigendum: Mechanism of early dissemination and metastasis in Her2 + mammary cancer.
- Author
-
Harper KL, Sosa MS, Entenberg D, Hosseini H, Cheung JF, Nobre R, Avivar-Valderas A, Nagi C, Girnius N, Davis RJ, Farias EF, Condeelis J, Klein CA, and Aguirre-Ghiso JA
- Abstract
This corrects the article DOI: 10.1038/nature20609.
- Published
- 2018
- Full Text
- View/download PDF
36. Macrophages orchestrate breast cancer early dissemination and metastasis.
- Author
-
Linde N, Casanova-Acebes M, Sosa MS, Mortha A, Rahman A, Farias E, Harper K, Tardio E, Reyes Torres I, Jones J, Condeelis J, Merad M, and Aguirre-Ghiso JA
- Subjects
- Animals, Disease Progression, Female, Mice, Neoplasm Metastasis, RAW 264.7 Cells, Receptor, ErbB-2 genetics, Wnt Signaling Pathway, Breast Neoplasms pathology, Macrophages pathology
- Abstract
Cancer cell dissemination during very early stages of breast cancer proceeds through poorly understood mechanisms. Here we show, in a mouse model of HER2
+ breast cancer, that a previously described sub-population of early-evolved cancer cells requires macrophages for early dissemination. Depletion of macrophages specifically during pre-malignant stages reduces early dissemination and also results in reduced metastatic burden at end stages of cancer progression. Mechanistically, we show that, in pre-malignant lesions, CCL2 produced by cancer cells and myeloid cells attracts CD206+ /Tie2+ macrophages and induces Wnt-1 upregulation that in turn downregulates E-cadherin junctions in the HER2+ early cancer cells. We also observe macrophage-containing tumor microenvironments of metastasis structures in the pre-malignant lesions that can operate as portals for intravasation. These data support a causal role for macrophages in early dissemination that affects long-term metastasis development much later in cancer progression. A pilot analysis on human specimens revealed intra-epithelial macrophages and loss of E-cadherin junctions in ductal carcinoma in situ, supporting a potential clinical relevance.- Published
- 2018
- Full Text
- View/download PDF
37. Mer Tyrosine Kinase Regulates Disseminated Prostate Cancer Cellular Dormancy.
- Author
-
Cackowski FC, Eber MR, Rhee J, Decker AM, Yumoto K, Berry JE, Lee E, Shiozawa Y, Jung Y, Aguirre-Ghiso JA, and Taichman RS
- Subjects
- Animals, Cell Cycle, Cell Line, Tumor, Cell Survival, Gene Knockdown Techniques, Heterografts, Histones metabolism, Humans, MAP Kinase Signaling System, Male, Mice, Mice, SCID, Neoplasm Recurrence, Local enzymology, Neoplasm Recurrence, Local pathology, Prostatic Neoplasms secondary, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins genetics, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Receptor Protein-Tyrosine Kinases genetics, Transcription Factors genetics, Transcription Factors metabolism, Tumor Escape, Tumor Microenvironment, c-Mer Tyrosine Kinase, Prostatic Neoplasms enzymology, Prostatic Neoplasms pathology, Proto-Oncogene Proteins metabolism, Receptor Protein-Tyrosine Kinases metabolism
- Abstract
Many prostate cancer (PCa) recurrences are thought to be due to reactivation of disseminated tumor cells (DTCs). We previously found a role of the TAM family of receptor tyrosine kinases TYRO3, AXL, and MERTK in PCa dormancy regulation. However, the mechanism and contributions of the individual TAM receptors is largely unknown. Knockdown of MERTK, but not AXL or TYRO3 by shRNA in PCa cells induced a decreased ratio of P-Erk1/2 to P-p38, increased expression of p27, NR2F1, SOX2, and NANOG, induced higher levels of histone H3K9me3 and H3K27me3, and induced a G1/G0 arrest, all of which are associated with dormancy. Similar effects were also observed with siRNA. Most importantly, knockdown of MERTK in PCa cells increased metastasis free survival in an intra-cardiac injection mouse xenograft model. MERTK knockdown also failed to inhibit PCa growth in vitro and subcutaneous growth in vivo, which suggests that MERTK has specificity for dormancy regulation or requires a signal from the PCa microenvironment. The effects of MERTK on the cell cycle and histone methylation were reversed by p38 inhibitor SB203580, which indicates the importance of MAP kinases for MERTK dormancy regulation. Overall, this study shows that MERTK stimulates PCa dormancy escape through a MAP kinase dependent mechanism, also involving p27, pluripotency transcription factors, and histone methylation. J. Cell. Biochem. 118: 891-902, 2017. © 2016 Wiley Periodicals, Inc., (© 2016 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
38. Phenotypic heterogeneity of disseminated tumour cells is preset by primary tumour hypoxic microenvironments.
- Author
-
Fluegen G, Avivar-Valderas A, Wang Y, Padgen MR, Williams JK, Nobre AR, Calvo V, Cheung JF, Bravo-Cordero JJ, Entenberg D, Castracane J, Verkhusha V, Keely PJ, Condeelis J, and Aguirre-Ghiso JA
- Subjects
- Animals, Breast Neoplasms pathology, COUP Transcription Factor I metabolism, Cell Hypoxia, Cell Line, Tumor, Cell Separation methods, Humans, Mice, Neoplasm Metastasis, Phenotype, Bone Marrow metabolism, Breast Neoplasms metabolism, Tumor Microenvironment
- Abstract
Hypoxia is a poor-prognosis microenvironmental hallmark of solid tumours, but it is unclear how it influences the fate of disseminated tumour cells (DTCs) in target organs. Here we report that hypoxic HNSCC and breast primary tumour microenvironments displayed upregulation of key dormancy (NR2F1, DEC2, p27) and hypoxia (GLUT1, HIF1α) genes. Analysis of solitary DTCs in PDX and transgenic mice revealed that post-hypoxic DTCs were frequently NR2F1
hi /DEC2hi /p27hi /TGFβ2hi and dormant. NR2F1 and HIF1α were required for p27 induction in post-hypoxic dormant DTCs, but these DTCs did not display GLUT1hi expression. Post-hypoxic DTCs evaded chemotherapy and, unlike ER- breast cancer cells, post-hypoxic ER+ breast cancer cells were more prone to enter NR2F1-dependent dormancy. We propose that primary tumour hypoxic microenvironments give rise to a subpopulation of dormant DTCs that evade therapy. These post-hypoxic dormant DTCs may be the source of disease relapse and poor prognosis associated with hypoxia.- Published
- 2017
- Full Text
- View/download PDF
39. Mechanism of early dissemination and metastasis in Her2 + mammary cancer.
- Author
-
Harper KL, Sosa MS, Entenberg D, Hosseini H, Cheung JF, Nobre R, Avivar-Valderas A, Nagi C, Girnius N, Davis RJ, Farias EF, Condeelis J, Klein CA, and Aguirre-Ghiso JA
- Abstract
Metastasis is the leading cause of cancer-related deaths; metastatic lesions develop from disseminated cancer cells (DCCs) that can remain dormant. Metastasis-initiating cells are thought to originate from a subpopulation present in progressed, invasive tumours. However, DCCs detected in patients before the manifestation of breast-cancer metastasis contain fewer genetic abnormalities than primary tumours or than DCCs from patients with metastases. These findings, and those in pancreatic cancer and melanoma models, indicate that dissemination might occur during the early stages of tumour evolution. However, the mechanisms that might allow early disseminated cancer cells (eDCCs) to complete all steps of metastasis are unknown. Here we show that, in early lesions in mice and before any apparent primary tumour masses are detected, there is a sub-population of Her2
+ p-p38lo p-Atf2lo Twist1hi E-cadlo early cancer cells that is invasive and can spread to target organs. Intra-vital imaging and organoid studies of early lesions showed that Her2+ eDCC precursors invaded locally, intravasated and lodged in target organs. Her2+ eDCCs activated a Wnt-dependent epithelial-mesenchymal transition (EMT)-like dissemination program but without complete loss of the epithelial phenotype, which was reversed by Her2 or Wnt inhibition. Notably, although the majority of eDCCs were Twist1hi E-cadlo and dormant, they eventually initiated metastasis. Our work identifies a mechanism for early dissemination in which Her2 aberrantly activates a program similar to mammary ductal branching that generates eDCCs that are capable of forming metastasis after a dormancy phase.- Published
- 2016
- Full Text
- View/download PDF
40. Early dissemination seeds metastasis in breast cancer.
- Author
-
Hosseini H, Obradović MMS, Hoffmann M, Harper KL, Sosa MS, Werner-Klein M, Nanduri LK, Werno C, Ehrl C, Maneck M, Patwary N, Haunschild G, Gužvić M, Reimelt C, Grauvogl M, Eichner N, Weber F, Hartkopf AD, Taran FA, Brucker SY, Fehm T, Rack B, Buchholz S, Spang R, Meister G, Aguirre-Ghiso JA, and Klein CA
- Abstract
Accumulating data suggest that metastatic dissemination often occurs early during tumour formation, but the mechanisms of early metastatic spread have not yet been addressed. Here, by studying metastasis in a HER2-driven mouse breast cancer model, we show that progesterone-induced signalling triggers migration of cancer cells from early lesions shortly after HER2 activation, but promotes proliferation in advanced primary tumour cells. The switch from migration to proliferation was regulated by increased HER2 expression and tumour-cell density involving microRNA-mediated progesterone receptor downregulation, and was reversible. Cells from early, low-density lesions displayed more stemness features, migrated more and founded more metastases than cells from dense, advanced tumours. Notably, we found that at least 80% of metastases were derived from early disseminated cancer cells. Karyotypic and phenotypic analysis of human disseminated cancer cells and primary tumours corroborated the relevance of these findings for human metastatic dissemination.
- Published
- 2016
- Full Text
- View/download PDF
41. Axl is required for TGF-β2-induced dormancy of prostate cancer cells in the bone marrow.
- Author
-
Yumoto K, Eber MR, Wang J, Cackowski FC, Decker AM, Lee E, Nobre AR, Aguirre-Ghiso JA, Jung Y, and Taichman RS
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation physiology, Humans, Male, Mice, Osteoblasts metabolism, Prostate metabolism, Signal Transduction physiology, Bone Marrow metabolism, Intercellular Signaling Peptides and Proteins metabolism, Prostatic Neoplasms metabolism, Receptor Protein-Tyrosine Kinases metabolism, Transforming Growth Factor beta2 metabolism
- Abstract
Disseminated prostate cancer (PCa) cells in the marrow survive for years without evidence of proliferation, while maintaining the capacity to develop into metastatic lesions. These dormant disseminated tumor cells (DTCs) may reside in close proximity to osteoblasts, while expressing high levels of Axl, one of the tyrosine kinase receptors for growth arrest specific 6 (Gas6). Yet how Axl regulates DTC proliferation in marrow remains undefined. Here, we explored the impact of the loss of Axl in PCa cells (PC3 and DU145) on the induction of their dormancy when they are co-cultured with a pre-osteoblastic cell line, MC3T3-E1. MC3T3-E1 cells dramatically decrease the proliferation of PCa cells, however this suppressive effect of osteoblasts is significantly reduced by the reduction of Axl expression in PCa cells. Interestingly, expression of both TGF-β and its receptors were regulated by Axl expression in PCa cells, while specific blockade of TGF-β signaling limited the ability of the osteoblasts to induce dormancy of PCa cells. Finally, we found that both Gas6 and Axl are required for TGF-β2-mediated cell growth suppression. Taken together, these data suggest that a loop between the Gas6/Axl axis and TGF-β2 signaling plays a significant role in the induction of PCa cell dormancy.
- Published
- 2016
- Full Text
- View/download PDF
42. Collagen Matrix Density Drives the Metabolic Shift in Breast Cancer Cells.
- Author
-
Morris BA, Burkel B, Ponik SM, Fan J, Condeelis JS, Aguirre-Ghiso JA, Castracane J, Denu JM, and Keely PJ
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation, Cell Survival, Citric Acid Cycle, Female, Gene Expression Regulation, Neoplastic, Glucose metabolism, Glutamine metabolism, Humans, Mitochondria metabolism, Models, Biological, Oxygen metabolism, Reactive Oxygen Species metabolism, Tumor Microenvironment, Breast Neoplasms metabolism, Collagen metabolism, Energy Metabolism, Extracellular Matrix metabolism
- Abstract
Increased breast density attributed to collagen I deposition is associated with a 4-6 fold increased risk of developing breast cancer. Here, we assessed cellular metabolic reprogramming of mammary carcinoma cells in response to increased collagen matrix density using an in vitro 3D model. Our initial observations demonstrated changes in functional metabolism in both normal mammary epithelial cells and mammary carcinoma cells in response to changes in matrix density. Further, mammary carcinoma cells grown in high density collagen matrices displayed decreased oxygen consumption and glucose metabolism via the tricarboxylic acid (TCA) cycle compared to cells cultured in low density matrices. Despite decreased glucose entry into the TCA cycle, levels of glucose uptake, cell viability, and ROS were not different between high and low density matrices. Interestingly, under high density conditions the contribution of glutamine as a fuel source to drive the TCA cycle was significantly enhanced. These alterations in functional metabolism mirrored significant changes in the expression of metabolic genes involved in glycolysis, oxidative phosphorylation, and the serine synthesis pathway. This study highlights the broad importance of the collagen microenvironment to cellular expression profiles, and shows that changes in density of the collagen microenvironment can modulate metabolic shifts of cancer cells., (Copyright © 2016 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
43. Cbx8 Acts Non-canonically with Wdr5 to Promote Mammary Tumorigenesis.
- Author
-
Chung CY, Sun Z, Mullokandov G, Bosch A, Qadeer ZA, Cihan E, Rapp Z, Parsons R, Aguirre-Ghiso JA, Farias EF, Brown BD, Gaspar-Maia A, and Bernstein E
- Subjects
- Animals, Carcinogenesis metabolism, Cell Line, Tumor, Epigenesis, Genetic, Epithelial Cells metabolism, Female, Gene Expression, Gene Expression Regulation, Neoplastic, Genetic Loci, Histones metabolism, Humans, Intracellular Signaling Peptides and Proteins, Mammary Neoplasms, Animal genetics, Mammary Neoplasms, Animal pathology, Mice, Transgenic, Neoplastic Stem Cells metabolism, Polycomb Repressive Complex 1, Protein Processing, Post-Translational, Receptors, Notch genetics, Receptors, Notch metabolism, Signal Transduction, Spheroids, Cellular metabolism, Tumor Burden, Mammary Neoplasms, Animal metabolism, Mitochondrial Membrane Transport Proteins physiology, Polycomb-Group Proteins physiology, Proteins physiology
- Abstract
Chromatin-mediated processes influence the development and progression of breast cancer. Using murine mammary carcinoma-derived tumorspheres as a functional readout for an aggressive breast cancer phenotype, we performed a loss-of-function screen targeting 60 epigenetic regulators. We identified the Polycomb protein Cbx8 as a key regulator of mammary carcinoma both in vitro and in vivo. Accordingly, Cbx8 is overexpressed in human breast cancer and correlates with poor survival. Our genomic analyses revealed that Cbx8 positively regulates Notch signaling by maintaining H3K4me3 levels on Notch-network gene promoters. Ectopic expression of Notch1 partially rescues tumorsphere formation in Cbx8-depleted cells. We find that Cbx8 associates with non-PRC1 complexes containing the H3K4 methyltransferase complex component WDR5, which together regulate Notch gene expression. Thus, our study implicates a key non-canonical role for Cbx8 in promoting breast tumorigenesis., (Copyright © 2016 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
44. Validation of a device for the active manipulation of the tumor microenvironment during intravital imaging.
- Author
-
Williams JK, Entenberg D, Wang Y, Avivar-Valderas A, Padgen M, Clark A, Aguirre-Ghiso JA, Castracane J, and Condeelis JS
- Abstract
The tumor microenvironment is recognized as playing a significant role in the behavior of tumor cells and their progression to metastasis. However, tools to manipulate the tumor microenvironment directly, and image the consequences of this manipulation with single cell resolution in real time in vivo , are lacking. We describe here a method for the direct, local manipulation of microenvironmental parameters through the use of an implantable Induction Nano Intravital Device (iNANIVID) and simultaneous in vivo visualization of the results at single-cell resolution. As a proof of concept, we deliver both a sustained dose of EGF to tumor cells while intravital imaging their chemotactic response as well as locally induce hypoxia in defined microenvironments in solid tumors., Competing Interests: of potential conflicts of interest No potential conflicts of interest were disclosed.
- Published
- 2016
- Full Text
- View/download PDF
45. The Relationship Between Dormant Cancer Cells and Their Microenvironment.
- Author
-
Linde N, Fluegen G, and Aguirre-Ghiso JA
- Subjects
- Animals, Humans, Neoplasm, Residual pathology, Neoplasms pathology, Tumor Microenvironment
- Abstract
The majority of cancer deaths are due to metastases that can occur years or decades after primary tumor diagnosis and treatment. Disseminated tumor cells (DTCs) surviving in a dormant state in target organs appear to explain the timing of this phenomenon. Knowledge on this process is important as it might provide a window of opportunity to prevent recurrences by eradicating dormant DTCs and/or by maintaining DTCs in a dormant state. Importantly, this research might offer markers of dormancy for early monitoring of metastatic relapse. However, our understanding of the mechanisms underlying the regulation of entry into and exit from dormancy is still limited and crippling any therapeutic opportunity. While cancer cell-intrinsic signaling pathways have been linked to dormancy regulation, it is likely that these pathways and the switch controlling reactivation from dormancy are regulated by microenvironmental cues. Here we review and discuss recent findings on how the microenvironment regulates cancer dormancy and raise new questions that may help advance the field., (© 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
46. Effects of p38α/β inhibition on acute lymphoblastic leukemia proliferation and survival in vivo.
- Author
-
Alsadeq A, Strube S, Krause S, Carlet M, Jeremias I, Vokuhl C, Loges S, Aguirre-Ghiso JA, Trauzold A, Cario G, Stanulla M, Schrappe M, and Schewe DM
- Subjects
- Adolescent, Animals, Cell Proliferation, Child, Child, Preschool, Female, Humans, Male, Mice, Phosphorylation, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, p38 Mitogen-Activated Protein Kinases physiology
- Abstract
P38α/β has been described as a tumor-suppressor controlling cell cycle checkpoints and senescence in epithelial malignancies. However, p38α/β also regulates other cellular processes. Here, we describe a role of p38α/β as a regulator of acute lymphoblastic leukemia (ALL) proliferation and survival in experimental ALL models. We also report first evidence that p38α/β phosphorylation is associated with the occurrence of relapses in TEL-AML1-positive leukemia. First, in vitro experiments show that p38α/β signaling is induced in a cyclical manner upon initiation of proliferation and remains activated during log-phase of cell growth. Next, we provide evidence that growth-permissive signals in the bone marrow activate p38α/β in a novel avian ALL model, in which therapeutic targeting can be tested. We further demonstrate that p38α/β inhibition by small molecules can suppress leukemic expansion and prolong survival of mice bearing ALL cell lines and primary cells. Knockdown of p38α strongly delays leukemogenesis in mice xenografted with cell lines. Finally, we show that in xenografted TEL-AML1 patients, ex vivo p38α/β phosphorylation is associated with an inferior long-term relapse-free survival. We propose p38α/β as a mediator of proliferation and survival in ALL and show first preclinical evidence for p38α/β inhibition as an adjunct approach to conventional therapies.
- Published
- 2015
- Full Text
- View/download PDF
47. Origin and interpretation of cancer transcriptome profiling: the essential role of the stroma in determining prognosis and drug resistance.
- Author
-
Goossens N, Hoshida Y, and Aguirre-Ghiso JA
- Subjects
- Animals, Humans, Prognosis, Drug Resistance, Neoplasm, Gene Expression Profiling, Neoplasms diagnosis, Neoplasms pathology, Tumor Microenvironment
- Published
- 2015
- Full Text
- View/download PDF
48. The In Ovo Chick Chorioallantoic Membrane (CAM) Assay as an Efficient Xenograft Model of Hepatocellular Carcinoma.
- Author
-
Li M, Pathak RR, Lopez-Rivera E, Friedman SL, Aguirre-Ghiso JA, and Sikora AG
- Subjects
- Animals, Carcinoma, Hepatocellular blood supply, Cell Line, Tumor, Chick Embryo, Chorioallantoic Membrane metabolism, Extracellular Matrix Proteins metabolism, Humans, Liver Neoplasms blood supply, Neovascularization, Pathologic pathology, Carcinoma, Hepatocellular pathology, Chorioallantoic Membrane growth & development, Liver Neoplasms pathology, Neoplasm Transplantation methods, Xenograft Model Antitumor Assays methods
- Abstract
The chick chorioallantoic membrane (CAM) begins to develop by day 7 after fertilization and matures by day 12. The CAM is naturally immunodeficient and highly vascularized, making it an ideal system for tumor implantation. Furthermore, the CAM contains extracellular matrix proteins such as fibronectin, laminin, collagen, integrin alpha(v)beta3, and MMP-2, making it an attractive model to study tumor invasion and metastasis. Scientists have long taken advantage of the physiology of the CAM by using it as a model of angiogenesis. More recently, the CAM assay has been modified to work as an in vivo xenograft model system for various cancers that bridges the gap between basic in vitro work and more complex animal cancer models. The CAM assay allows for the study of tumor growth, anti-tumor therapies, and pro-tumor molecular pathways in a biologically relevant system that is both cost- and time-effective. Here, we describe the development of CAM xenograft model of hepatocellular carcinoma (HCC) with embryonic survival rates of up to 93% and reliable tumor take leading to growth of three-dimensional, vascularized tumors.
- Published
- 2015
- Full Text
- View/download PDF
49. Identification of markers that functionally define a quiescent multiple myeloma cell sub-population surviving bortezomib treatment.
- Author
-
Adomako A, Calvo V, Biran N, Osman K, Chari A, Paton JC, Paton AW, Moore K, Schewe DM, and Aguirre-Ghiso JA
- Subjects
- Adult, Aged, Animals, Apoptosis drug effects, Azacitidine administration & dosage, Cell Survival drug effects, Cyclin-Dependent Kinase 6 genetics, Endoplasmic Reticulum Chaperone BiP, Female, Gene Expression Regulation, Neoplastic drug effects, Heat-Shock Proteins genetics, Humans, Male, Mice, Middle Aged, Multiple Myeloma genetics, Multiple Myeloma pathology, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, Xenograft Model Antitumor Assays, p21-Activated Kinases genetics, Bortezomib administration & dosage, Cyclin-Dependent Kinase 6 biosynthesis, Heat-Shock Proteins biosynthesis, Multiple Myeloma drug therapy, p21-Activated Kinases biosynthesis
- Abstract
Background: The mechanisms allowing residual multiple myeloma (MM) cells to persist after bortezomib (Bz) treatment remain unclear. We hypothesized that studying the biology of bortezomib-surviving cells may reveal markers to identify these cells and survival signals to target and kill residual MM cells., Methods: We used H2B-GFP label retention, biochemical tools and in vitro and in vivo experiments to characterize growth arrest and the unfolded protein responses in quiescent Bz-surviving cells. We also tested the effect of a demethylating agent, 5-Azacytidine, on Bz-induced quiescence and whether inhibiting the chaperone GRP78/BiP (henceforth GRP78) with a specific toxin induced apoptosis in Bz-surviving cells. Finally, we used MM patient samples to test whether GRP78 levels might associate with disease progression. Statistical analysis employed t-test and Mann-Whitney tests at a 95% confidence., Results: We report that Bz-surviving MM cells in vitro and in vivo enter quiescence characterized by p21(CIP1) upregulation. Bz-surviving MM cells also downregulated CDK6, Ki67 and P-Rb. H2B-GFP label retention showed that Bz-surviving MM cells are either slow-cycling or deeply quiescent. The Bz-induced quiescence was stabilized by low dose (500nM) of 5-azacytidine (Aza) pre-treatment, which also potentiated the initial Bz-induced apoptosis. We also found that expression of GRP78, an unfolded protein response (UPR) survival factor, persisted in MM quiescent cells. Importantly, GRP78 downregulation using a specific SubAB bacterial toxin killed Bz-surviving MM cells. Finally, quantification of Grp78(high)/CD138+ MM cells from patients suggested that high levels correlated with progressive disease., Conclusions: We conclude that Bz-surviving MM cells display a GRP78(HIGH)/p21(HIGH)/CDK6(LOW)/P-Rb(LOW) profile, and these markers may identify quiescent MM cells capable of fueling recurrences. We further conclude that Aza + Bz treatment of MM may represent a novel strategy to delay recurrences by enhancing Bz-induced apoptosis and quiescence stability.
- Published
- 2015
- Full Text
- View/download PDF
50. Epithelial Xbp1 is required for cellular proliferation and differentiation during mammary gland development.
- Author
-
Hasegawa D, Calvo V, Avivar-Valderas A, Lade A, Chou HI, Lee YA, Farias EF, Aguirre-Ghiso JA, and Friedman SL
- Subjects
- Animals, Cell Differentiation, Cells, Cultured, DNA-Binding Proteins analysis, DNA-Binding Proteins genetics, Epithelial Cells cytology, Epithelial Cells metabolism, Female, Gene Deletion, Mammary Glands, Animal ultrastructure, Mice, Mice, Inbred C57BL, Prolactin analysis, Prolactin metabolism, Receptor, ErbB-4 analysis, Receptor, ErbB-4 metabolism, Regulatory Factor X Transcription Factors, STAT5 Transcription Factor analysis, STAT5 Transcription Factor metabolism, Transcription Factors analysis, Transcription Factors genetics, Unfolded Protein Response, X-Box Binding Protein 1, Cell Proliferation, DNA-Binding Proteins metabolism, Lactation, Mammary Glands, Animal cytology, Mammary Glands, Animal growth & development, Transcription Factors metabolism
- Abstract
Xbp1, a key mediator of the unfolded protein response (UPR), is activated by IRE1α-mediated splicing, which results in a frameshift to encode a protein with transcriptional activity. However, the direct function of Xbp1 in epithelial cells during mammary gland development is unknown. Here we report that the loss of Xbp1 in the mammary epithelium through targeted deletion leads to poor branching morphogenesis, impaired terminal end bud formation, and spontaneous stromal fibrosis during the adult virgin period. Additionally, epithelial Xbp1 deletion induces endoplasmic reticulum (ER) stress in the epithelium and dramatically inhibits epithelial proliferation and differentiation during lactation. The synthesis of milk and its major components, α/β-casein and whey acidic protein (WAP), is significantly reduced due to decreased prolactin receptor (Prlr) and ErbB4 expression in Xbp1-deficient mammary epithelium. Reduction of Prlr and ErbB4 expression and their diminished availability at the cell surface lead to reduced phosphorylated Stat5, an essential regulator of cell proliferation and differentiation during lactation. As a result, lactating mammary glands in these mice produce less milk protein, leading to poor pup growth and postnatal death. These findings suggest that the loss of Xbp1 induces a terminal UPR which blocks proliferation and differentiation during mammary gland development., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.