201
|
Jacobsson H, Harrison H, Hughes É, Persson E, Rhost S, Fitzpatrick P, Gustafsson A, Andersson D, Gregersson P, Magnusson Y, Ståhlberg A, Landberg G. Hypoxia-induced secretion stimulates breast cancer stem cell regulatory signalling pathways. Mol Oncol 2019; 13:1693-1705. [PMID: 31066211 PMCID: PMC6670019 DOI: 10.1002/1878-0261.12500] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/09/2019] [Accepted: 05/07/2019] [Indexed: 01/08/2023] Open
Abstract
It is well known that tumour cells are dependent on communication with the tumour microenvironment. Previously, it has been shown that hypoxia (HX) induces pronounced, diverse and direct effects on cancer stem cell (CSC) qualities in different breast cancer subtypes. Here, we describe the mechanism by which HX-induced secretion influences the spreading of CSCs. Conditioned media (CM) from estrogen receptor (ER)-α-positive hypoxic breast cancer cell cultures increased the fraction of CSCs compared to normal growth conditions, as determined using sets of CSC assays and model systems. In contrast, media from ERα-negative hypoxic cell cultures instead decreased this key subpopulation of cancer cells. Further, there was a striking overrepresentation of JAK-STAT-associated cytokines in both the ERα-positive and ERα-negative linked hypoxic responses as determined by a protein screen of the CM. JAK-STAT inhibitors and knockdown experiments further supported the hypothesis that this pathway is critical for the CSC-activating and CSC-inactivating effects induced by hypoxic secretion. We also observed that the interleukin-6-JAK2-STAT3 axis was specifically central for the ERα-negative hypoxic behaviour. Our results underline the importance of considering breast cancer subtypes in treatments targeting JAK-STAT or HX-associated processes and indicate that HX is not only a confined tumour biological event, but also influences key tumour properties in widespread normoxic microenvironments.
Collapse
Affiliation(s)
- Hanna Jacobsson
- Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Sweden
| | - Hannah Harrison
- Breakthrough Breast Cancer Unit, Centre for Molecular Pathology, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, UK.,Manchester Cancer Research Centre, The University of Manchester, UK
| | - Éamon Hughes
- Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Sweden
| | - Emma Persson
- Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Sweden
| | - Sara Rhost
- Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Sweden
| | - Paul Fitzpatrick
- Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Sweden
| | - Anna Gustafsson
- Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Sweden
| | - Daniel Andersson
- Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Sweden
| | - Pernilla Gregersson
- Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Sweden
| | - Ylva Magnusson
- Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Sweden
| | - Anders Ståhlberg
- Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden.,Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Göran Landberg
- Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Sweden.,Breakthrough Breast Cancer Unit, Centre for Molecular Pathology, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, UK
| |
Collapse
|
202
|
Smigiel JM, Taylor SE, Bryson BL, Tamagno I, Polak K, Jackson MW. Cellular plasticity and metastasis in breast cancer: a pre- and post-malignant problem. JOURNAL OF CANCER METASTASIS AND TREATMENT 2019; 5:47. [PMID: 32355893 PMCID: PMC7192216 DOI: 10.20517/2394-4722.2019.26] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
As a field we have made tremendous strides in treating breast cancer, with a decline in the past 30 years of overall breast cancer mortality. However, this progress is met with little affect once the disease spreads beyond the primary site. With a 5-year survival rate of 22%, 10-year of 13%, for those patients with metastatic breast cancer (mBC), our ability to effectively treat wide spread disease is minimal. A major contributing factor to this ineffectiveness is the complex make-up, or heterogeneity, of the primary site. Within a primary tumor, secreted factors, malignant and pre-malignant epithelial cells, immune cells, stromal fibroblasts and many others all reside alongside each other creating a dynamic environment contributing to metastasis. Furthermore, heterogeneity contributes to our lack of understanding regarding the cells' remarkable ability to undergo epithelial/non-cancer stem cell (CSC) to mesenchymal/CSC (E-M/CSC) plasticity. The enhanced invasion & motility, tumor-initiating potential, and acquired therapeutic resistance which accompanies E-M/CSC plasticity implicates a significant role in metastasis. While most work trying to understand E-M/CSC plasticity has been done on malignant cells, recent evidence is emerging concerning the ability for pre-malignant cells to undergo E-M/CSC plasticity and contribute to the metastatic process. Here we will discuss the importance of E-M/CSC plasticity within malignant and pre-malignant populations of the tumor. Moreover, we will discuss how one may potentially target these populations, ultimately disrupting the metastatic cascade and increasing patient survival for those with mBC.
Collapse
Affiliation(s)
- Jacob M. Smigiel
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Sarah E. Taylor
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Benjamin L. Bryson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ilaria Tamagno
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Kelsey Polak
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Mark W. Jackson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
203
|
Gaspar D, Peixoto R, De Pieri A, Striegl B, Zeugolis DI, Raghunath M. Local pharmacological induction of angiogenesis: Drugs for cells and cells as drugs. Adv Drug Deliv Rev 2019; 146:126-154. [PMID: 31226398 DOI: 10.1016/j.addr.2019.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 05/12/2019] [Accepted: 06/16/2019] [Indexed: 12/12/2022]
Abstract
The past decades have seen significant advances in pro-angiogenic strategies based on delivery of molecules and cells for conditions such as coronary artery disease, critical limb ischemia and stroke. Currently, three major strategies are evolving. Firstly, various pharmacological agents (growth factors, interleukins, small molecules, DNA/RNA) are locally applied at the ischemic region. Secondly, preparations of living cells with considerable bandwidth of tissue origin, differentiation state and preconditioning are delivered locally, rarely systemically. Thirdly, based on the notion, that cellular effects can be attributed mostly to factors secreted in situ, the cellular secretome (conditioned media, exosomes) has come into the spotlight. We review these three strategies to achieve (neo)angiogenesis in ischemic tissue with focus on the angiogenic mechanisms they tackle, such as transcription cascades, specific signalling steps and cellular gases. We also include cancer-therapy relevant lymphangiogenesis, and shall seek to explain why there are often conflicting data between in vitro and in vivo. The lion's share of data encompassing all three approaches comes from experimental animal work and we shall highlight common technical obstacles in the delivery of therapeutic molecules, cells, and secretome. This plethora of preclinical data contrasts with a dearth of clinical studies. A lack of adequate delivery vehicles and standardised assessment of clinical outcomes might play a role here, as well as regulatory, IP, and manufacturing constraints of candidate compounds; in addition, completed clinical trials have yet to reveal a successful and efficacious strategy. As the biology of angiogenesis is understood well enough for clinical purposes, it will be a matter of time to achieve success for well-stratified patients, and most probably with a combination of compounds.
Collapse
Affiliation(s)
- Diana Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Rita Peixoto
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Andrea De Pieri
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Proxy Biomedical Ltd., Coilleach, Spiddal, Galway, Ireland
| | - Britta Striegl
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Michael Raghunath
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland.
| |
Collapse
|
204
|
Tiwari P, Blank A, Cui C, Schoenfelt KQ, Zhou G, Xu Y, Khramtsova G, Olopade F, Shah AM, Khan SA, Rosner MR, Becker L. Metabolically activated adipose tissue macrophages link obesity to triple-negative breast cancer. J Exp Med 2019; 216:1345-1358. [PMID: 31053611 PMCID: PMC6547867 DOI: 10.1084/jem.20181616] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/27/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022] Open
Abstract
Tiwari et al. identify metabolically activated macrophages in obese mammary adipose tissue as an important source of IL-6, which fuels triple-negative breast cancer stemness and tumorigenesis through GP130 signaling. These mechanistic insights provide potential targets for treating obesity-associated triple-negative breast cancer. Obesity is associated with increased incidence and severity of triple-negative breast cancer (TNBC); however, mechanisms underlying this relationship are incompletely understood. Here, we show that obesity reprograms mammary adipose tissue macrophages to a pro-inflammatory metabolically activated phenotype (MMe) that alters the niche to support tumor formation. Unlike pro-inflammatory M1 macrophages that antagonize tumorigenesis, MMe macrophages are pro-tumorigenic and represent the dominant macrophage phenotype in mammary adipose tissue of obese humans and mice. MMe macrophages release IL-6 in an NADPH oxidase 2 (NOX2)–dependent manner, which signals through glycoprotein 130 (GP130) on TNBC cells to promote stem-like properties including tumor formation. Deleting Nox2 in myeloid cells or depleting GP130 in TNBC cells attenuates obesity-augmented TNBC stemness. Moreover, weight loss reverses the effects of obesity on MMe macrophage inflammation and TNBC tumor formation. Our studies implicate MMe macrophage accumulation in mammary adipose tissue as a mechanism for promoting TNBC stemness and tumorigenesis during obesity.
Collapse
Affiliation(s)
- Payal Tiwari
- Committee on Cancer Biology, The University of Chicago, Chicago, IL.,Ben May Department for Cancer Research, The University of Chicago, Chicago, IL
| | - Ariane Blank
- Committee on Cancer Biology, The University of Chicago, Chicago, IL.,Ben May Department for Cancer Research, The University of Chicago, Chicago, IL
| | - Chang Cui
- Committee on Cancer Biology, The University of Chicago, Chicago, IL.,Ben May Department for Cancer Research, The University of Chicago, Chicago, IL
| | - Kelly Q Schoenfelt
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL
| | - Guolin Zhou
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL
| | - Yanfei Xu
- Department of Surgery and Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine of Northwestern University, Northwestern University, Chicago, IL
| | - Galina Khramtsova
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, IL
| | - Funmi Olopade
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, IL
| | - Ajay M Shah
- School of Cardiovascular Medicine and Sciences, King's College, London British Hearth Foundation Centre, London, UK
| | - Seema A Khan
- Department of Surgery and Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine of Northwestern University, Northwestern University, Chicago, IL
| | - Marsha Rich Rosner
- Committee on Cancer Biology, The University of Chicago, Chicago, IL .,Ben May Department for Cancer Research, The University of Chicago, Chicago, IL
| | - Lev Becker
- Committee on Cancer Biology, The University of Chicago, Chicago, IL .,Ben May Department for Cancer Research, The University of Chicago, Chicago, IL.,Committee on Molecular Metabolism and Nutrition, The University of Chicago, Chicago, IL
| |
Collapse
|
205
|
Donnenberg AD, Luketich JD, Dhupar R, Donnenberg VS. Treatment of malignant pleural effusions: the case for localized immunotherapy. J Immunother Cancer 2019; 7:110. [PMID: 30999958 PMCID: PMC6472034 DOI: 10.1186/s40425-019-0590-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/02/2019] [Indexed: 12/31/2022] Open
Abstract
Malignant pleural effusions (MPE) are a common terminal pathway for many cancers, with an estimated United States incidence of more than 150,000 cases per year. MPE is an aggressive disease with a uniformly fatal prognosis and a life expectancy of only 3 to 12 months. The development of an effective targeted therapy represents a pressing unmet need. This commentary focuses on how cellular and humoral components condition the pleural space as a tumor-promoting, wound-healing environment. Despite an abundance of potential antigen presenting and effector cells in the pleura, their physical isolation by the mesothelial barrier, the concentration of cytokines and chemokines driving the epithelial to mesenchymal transition (EMT) and M2 /Th-2 polarization, suppress tumor-specific immune effector responses. We argue that local immune repolarization must precede either immune checkpoint or cellular therapy to successfully eradicate pleural tumor. We further hypothesize that, because of its cellular content, a repolarized pleural space will provide an effective immune environment for generation of systemic anti-tumor response.
Collapse
Affiliation(s)
- Albert D Donnenberg
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Departments of Medicine and Infectious Disease and Microbiology, University of Pittsburgh, School of Medicine and Graduate School of Public Health, Hillman Cancer Center, Research Pavilion, 5117 Centre Ave, Pittsburgh, PA, 15213, USA.
| | - James D Luketich
- Department of Cardiothoracic Surgery, Division of Thoracic and Foregut Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rajeev Dhupar
- Department of Cardiothoracic Surgery, Division of Thoracic and Foregut Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Vera S Donnenberg
- Department of Cardiothoracic Surgery, Division of Thoracic and Foregut Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA.
- Departments of Cardiothoracic Surgery and Pharmaceutical Sciences, University of Pittsburgh, Schools of Medicine and Pharmacy, Hillman Cancer Center, Research Pavilion, 5117 Centre Ave, Suite 2.42, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
206
|
Ruiu R, Tarone L, Rolih V, Barutello G, Bolli E, Riccardo F, Cavallo F, Conti L. Cancer stem cell immunology and immunotherapy: Harnessing the immune system against cancer's source. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 164:119-188. [PMID: 31383404 DOI: 10.1016/bs.pmbts.2019.03.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite recent advances in diagnosis and therapy having improved cancer outcome, many patients still do not respond to treatments, resulting in the progression or relapse of the disease, eventually impairing survival expectations. The limited efficacy of therapy is often attributable to its inability to affect cancer stem cells (CSCs), a small population of cells resistant to current radio- and chemo-therapies. CSCs are characterized by self-renewal and tumor-initiating capabilities, and function as a reservoir for the local and distant recurrence of the disease. Therefore, new therapeutic approaches able to effectively target and deplete CSCs are urgently needed. Immunotherapy is facing a renewed interest for its potential in cancer treatment, and the possibility of harnessing the immune system to target CSCs is being addressed by a new exciting research field. In this chapter, we discuss the cancer stem cell model and illustrate CSC biological and molecular properties, critically addressing theoretical and practical issues linked with their definition and study. We then review the existing literature regarding the immunological properties of CSCs and the complex interplay occurring between CSCs and immune cells. Finally, we present up-to-date studies on CSC immunotargeting and its potential future perspective. In conclusion, understanding the interplay between CSC biology and tumor immunology will provide a deeper understanding of the mechanisms that regulate CSC immunological properties. This will contribute to the design of new CSC-directed immunotherapeutic strategies with the potential of strongly improving cancer outcomes.
Collapse
Affiliation(s)
- Roberto Ruiu
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Valeria Rolih
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| |
Collapse
|
207
|
Umansky V, Adema GJ, Baran J, Brandau S, Van Ginderachter JA, Hu X, Jablonska J, Mojsilovic S, Papadaki HA, Pico de Coaña Y, Santegoets KCM, Santibanez JF, Serre K, Si Y, Sieminska I, Velegraki M, Fridlender ZG. Interactions among myeloid regulatory cells in cancer. Cancer Immunol Immunother 2019; 68:645-660. [PMID: 30003321 PMCID: PMC11028297 DOI: 10.1007/s00262-018-2200-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022]
Abstract
Mounting evidence has accumulated on the critical role of the different myeloid cells in the regulation of the cancerous process, and in particular in the modulation of the immune reaction to cancer. Myeloid cells are a major component of host cells infiltrating tumors, interacting with each other, with tumor cells and other stromal cells, and demonstrating a prominent plasticity. We describe here various myeloid regulatory cells (MRCs) in mice and human as well as their relevant therapeutic targets. We first address the role of the monocytes and macrophages that can contribute to angiogenesis, immunosuppression and metastatic dissemination. Next, we discuss the differential role of neutrophil subsets in tumor development, enhancing the dual and sometimes contradicting role of these cells. A heterogeneous population of immature myeloid cells, MDSCs, was shown to be generated and accumulated during tumor progression as well as to be an important player in cancer-related immune suppression. Lastly, we discuss the role of myeloid DCs, which can either contribute to effective anti-tumor responses or play a more regulatory role. We believe that MRCs play a critical role in cancer-related immune regulation and suggest that future anti-cancer therapies will focus on these abundant cells.
Collapse
Affiliation(s)
- Viktor Umansky
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht Karl University of Heidelberg, Mannheim, Germany.
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Jaroslaw Baran
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Kraków, Poland
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jo A Van Ginderachter
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Xiaoying Hu
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht Karl University of Heidelberg, Mannheim, Germany
| | - Jadwiga Jablonska
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Slavko Mojsilovic
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Helen A Papadaki
- Department of Hematology, School of Medicine, University of Crete, Heraklion, Greece
| | - Yago Pico de Coaña
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kim C M Santegoets
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Juan F Santibanez
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Karine Serre
- Faculty of Medicine, Institute of Molecular Medicine (IMM)-João Lobo Antunes, University of Lisbon, Lisbon, Portugal
| | - Yu Si
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Isabela Sieminska
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Kraków, Poland
| | - Maria Velegraki
- Department of Hematology, School of Medicine, University of Crete, Heraklion, Greece
| | - Zvi G Fridlender
- Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
208
|
Wei C, Yang C, Wang S, Shi D, Zhang C, Lin X, Liu Q, Dou R, Xiong B. Crosstalk between cancer cells and tumor associated macrophages is required for mesenchymal circulating tumor cell-mediated colorectal cancer metastasis. Mol Cancer 2019; 18:64. [PMID: 30927925 PMCID: PMC6441214 DOI: 10.1186/s12943-019-0976-4] [Citation(s) in RCA: 490] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/22/2019] [Indexed: 02/08/2023] Open
Abstract
Background Tumor-associated macrophages (TAMs) are major components of tumor microenvironment that frequently associated with tumor metastasis in human cancers. Circulating tumor cell (CTC), originating from primary tumor sites, is considered to be the precursors of tumor metastasis. However, the regulatory mechanism of TAMs in CTC-mediated tumor metastasis still remains unclear. Methods Immunohistochemical staining was used to detect the macrophages infiltration (CD68 and CD163), epithelial–mesenchymal transition (EMT) markers (E-cadherin and Vimentin) expression in serial sections of human colorectal cancer (CRC) specimens. Then, the correlations between macrophages infiltration and clinicopathologic features, mesenchymal CTC ratio, and patients’ prognosis were analyzed. A co-culture assay in vitro was used to evaluate the role of TAMs on CRC EMT, migration and invasion, and ELISA, luciferase reporter assay and CHIP were performed to uncover the underlying mechanism. Furthermore, an in vivo model was carried out to confirm the effect of TAMs on mesenchymal CTC-mediated metastasis. Results Clinically, CD163+ TAMs infiltrated in invasive front was associated with EMT, mesenchymal CTC ratio, and poor prognosis in patients with CRC. CRC–conditioned macrophages regulated EMT program to enhance CRC cells migration and invasion by secreting IL6. TAMs-derived IL6 activated the JAK2/STAT3 pathway, and activated STAT3 transcriptionally inhibited the tumor suppressor miR-506-3p in CRC cells. miR-506-3p, a key miRNA regulating FoxQ1, was downregulated in CRC cells, resulting in increased FoxQ1 expression, which in turn led to the production of CCL2 that promoted macrophage recruitment. Inhibition of CCL2 or IL6 broke this loop and reduced macrophage migration and mesenchymal CTC-mediated metastasis, respectively. Conclusions Our data indicates that TAMs induce EMT program to enhance CRC migration, invasion, and CTC-mediated metastasis by regulating the JAK2/STAT3/miR-506-3p/FoxQ1 axis, which in turn leads to the production of CCL2 that promote macrophage recruitment, revealing a new cross-talk between immune cells and tumor cells in CRC microenvironment. Electronic supplementary material The online version of this article (10.1186/s12943-019-0976-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chen Wei
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Chaogang Yang
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Shuyi Wang
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Dongdong Shi
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Chunxiao Zhang
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Xiaobin Lin
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Qing Liu
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Rongzhang Dou
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Bin Xiong
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China. .,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China. .,Hubei Cancer Clinical Study Center, Wuhan, China.
| |
Collapse
|
209
|
Guo L, Cheng X, Chen H, Chen C, Xie S, Zhao M, Liu D, Deng Q, Liu Y, Wang X, Chen X, Wang J, Yin Z, Qi S, Gao J, Ma Y, Guo N, Shi M. Induction of breast cancer stem cells by M1 macrophages through Lin-28B-let-7-HMGA2 axis. Cancer Lett 2019; 452:213-225. [PMID: 30917918 DOI: 10.1016/j.canlet.2019.03.032] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 03/12/2019] [Accepted: 03/20/2019] [Indexed: 12/24/2022]
Abstract
Proinflammatory macrophage (M1) is now being suggested as a potential therapeutic strategy for cancer because of its tumoricidal capacity. However, few studies have been focused directly on the effects of M1 macrophages on cancer cells. Here, we found that M1 induced a subpopulation of CD44high/CD24-/low or ALDH1+ cells with CSC-like phenotypes from different types of breast cancer cells (BCCs) in a paracrine manner. Stat3/NF-κB pathways in BCCs were activated by proinflammatory cytokines, igniting Lin-28B-let-7-HMGA2 axis to induce CSC through epithelial-mesenchymal transition (EMT). Previously, we reported that Stat3-coordinated Lin-28B-let-7-HMGA2 axis initiated EMT in BCCs. Here, inhibition of Stat3/NF-κB pathways or Lin-28B-let-7-HMGA2 axis suppressed EMT/CSCs program. Notably, HMGA2 knockdown directly repressed M1-induced CSC formation and expression of Klf-4 and Nanog. Meanwhile, prolonged coculture with BCCs endowed M1 with M2 properties. M1 supernatant induced CSC from non-stem cancer cells, while M2 supernatant sustained a higher proportion of ALDH1+ cells. Our data suggest that macrophages might modulate CSC formation and maintenance by transferring between M1/M2 phenotype. Given that M1 are being considered as a promising immunotherapy tool, it is important to inhibit their CSC-inducing potential by targeting key molecules and pathways.
Collapse
Affiliation(s)
- Liang Guo
- Institute of Basic Medical Sciences, Beijing, 100850, PR China.
| | - Xiang Cheng
- Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Hongyu Chen
- Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Changguo Chen
- Department of Clinical Laboratory, The Navy General Hospital, Beijing, 100048, PR China
| | - Shuai Xie
- Laboratory of Cellular and Molecular Immunology, Medical School of Henan University, Kaifeng, 475004, PR China
| | - Min Zhao
- Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Dan Liu
- Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Que Deng
- Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Yanjun Liu
- Laboratory of Cellular and Molecular Immunology, Medical School of Henan University, Kaifeng, 475004, PR China
| | - Xiaomeng Wang
- Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Xintian Chen
- Department of Cancer Biotherapy, Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, PR China
| | - Jiangong Wang
- Department of Cancer Biotherapy, Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, PR China
| | - Zhaoyang Yin
- Department of Urology, The First Affiliated Hospital, General Hospital of PLA, Beijing, 100048, PR China
| | - Siyong Qi
- Department of Urology, The First Affiliated Hospital, General Hospital of PLA, Beijing, 100048, PR China
| | - Jiangping Gao
- Department of Urology, The First Affiliated Hospital, General Hospital of PLA, Beijing, 100048, PR China
| | - Yuanfang Ma
- Laboratory of Cellular and Molecular Immunology, Medical School of Henan University, Kaifeng, 475004, PR China
| | - Ning Guo
- Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Ming Shi
- Institute of Basic Medical Sciences, Beijing, 100850, PR China.
| |
Collapse
|
210
|
Wang Y, Chen J, Yang L, Li J, Wu W, Huang M, Lin L, Su S. Tumor-Contacted Neutrophils Promote Metastasis by a CD90-TIMP-1 Juxtacrine-Paracrine Loop. Clin Cancer Res 2019; 25:1957-1969. [PMID: 30482778 DOI: 10.1158/1078-0432.ccr-18-2544] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/08/2018] [Accepted: 11/21/2018] [Indexed: 02/05/2023]
Abstract
PURPOSE The different prognostic values of tumor-infiltrating neutrophils (TIN) in different tissue compartments are unknown. In this study, we investigated their different prognostic roles and the underlying mechanism.Experimental Design: We evaluated CD66b+ neutrophils in primary tumors from 341 patients with breast cancer from Sun Yat-sen Memorial Hospital by IHC. The association between stromal and parenchymal neutrophil counts and clinical outcomes was assessed in a training set (170 samples), validated in an internal validation set (171 samples), and further confirmed in an external validation set (105 samples). In addition, we isolated TINs from clinical samples and screened the cytokine profile by antibody microarray. The interaction between neutrophils and tumor cells was investigated in transwell and 3D Matrigel coculture systems. The therapeutic potential of indicated cytokines was evaluated in tumor-bearing immunocompetent mice. RESULTS We observed that the neutrophils in tumor parenchyma, rather than those in stroma, were an independent poor prognostic factor in the training [HR = 5.00, 95% confidence interval (CI): 2.88-8.68, P < 0.001], internal validation (HR = 3.56, 95% CI: 2.07-6.14, P < 0.001), and external validation set (HR = 5.07, 95% CI: 2.27-11.33, P < 0.001). The mechanistic study revealed that neutrophils induced breast cancer epithelial-mesenchymal transition (EMT) via tissue inhibitor of matrix metalloprotease (TIMP-1). Reciprocally, breast cancer cells undergoing EMT enhanced neutrophils' TIMP-1 secretion by CD90 in a cell-contact manner. In vivo, TIMP-1 neutralization or CD90 blockade significantly reduced metastasis. More importantly, TIMP-1 and CD90 were positively correlated in breast cancer (r 2 = 0.6079; P < 0.001) and associated with poor prognosis of patients. CONCLUSIONS Our findings unravel a location-dictated interaction between tumor cells and neutrophils and provide a rationale for new antimetastasis treatments.
Collapse
Affiliation(s)
- Ying Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Linbin Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiaqian Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ling Lin
- Department of Internal Medicine, The First Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
211
|
LSECtin on tumor-associated macrophages enhances breast cancer stemness via interaction with its receptor BTN3A3. Cell Res 2019; 29:365-378. [PMID: 30858559 DOI: 10.1038/s41422-019-0155-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 02/21/2019] [Indexed: 12/14/2022] Open
Abstract
Macrophages have been suggested to contribute to constructing a cancer stem cell (CSC) niche. However, whether and how macrophages regulate the activity of CSCs through juxtacrine signaling are poorly understood. Here we report LSECtin, a transmembrane protein highly expressed on tumor-associated macrophages (TAMs), enhances stemness of breast cancer cells (BCCs). We identified BTN3A3, a B7 family member with previously unknown functions as the receptor for LSECtin on BCCs responsible for stemness-promoting effect of LSECtin. In mice bearing human tumor xenografts, either macrophage-specific ablation of LSECtin or silencing of BTN3A3 in BCCs decreased CSC frequency and tumor growth. Admixture of LSECtin-positive macrophages increased the tumorigenic activity of BCCs dependent on BTN3A3. Disruption of the LSECtin-BTN3A3 axis with BTN3A3-Fc or anti-BTN3A3 mAb has a therapeutic effect on breast cancer. These findings define a juxtacrine signaling mechanism by which TAMs promote cancer stemness. Targeting this axis in the CSC niche may provide potential therapies to breast cancer.
Collapse
|
212
|
CXCL12/CXCR4 pathway orchestrates CSC-like properties by CAF recruited tumor associated macrophage in OSCC. Exp Cell Res 2019; 378:131-138. [PMID: 30857971 DOI: 10.1016/j.yexcr.2019.03.013] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 02/17/2019] [Accepted: 03/07/2019] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophage (TAM), a crucial component of immune cell infiltrated in tumor microenvironment, is associated with progression of oral squamous cell carcinoma (OSCC). However, it is still unclear how TAM is induced/accumulated and activated around/in OSCC. In the study herein, we tried to understand how TAM accumulates and activates in the OSCC and how TAM promotes OSCC to convert cancer stem cell (CSC). In this study, first important finding was that the M2 macrophages significantly increased in all twenty human OSCC samples in vivo. Cancer-associated fibroblast (CAF)-derived CXCL12 effectively attracted monocytes, which displayed M2 macrophage phenotype. Blocking CXCL12 receptor (CXCR4) significantly reduced chemotaxis of M2 macrophage. Polarized M2 macrophage promoted CSC-like transition in OSCC cell line, Cal27 cells. These CSC-like cells significantly expressed higher Sox2, Oct4, and Nanog genes, were stronger positive for CD44 and CD105, increased cell proliferation with less apoptosis, enhanced cell migration, and were resistant to chemotherapy drug, vineristine. These results indicate that CAF effectively attracts monocytes via the CXCL12/CXCR4 pathway and induces their differentiation to M2 macrophages. Interestingly, these polarized M2 macrophages promote formation of CSC-like cells from the OSCC lead to enhance OSCC proliferation with less apoptosis. Therefore, our findings have potential to lead to novel therapy for the OSCC to target CXCL12-mediated TAM recruitment.
Collapse
|
213
|
Primitive Cancer Cell States: A Target for Drug Screening? Trends Pharmacol Sci 2019; 40:161-171. [DOI: 10.1016/j.tips.2019.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/05/2018] [Accepted: 01/07/2019] [Indexed: 12/26/2022]
|
214
|
Sanchez LR, Borriello L, Entenberg D, Condeelis JS, Oktay MH, Karagiannis GS. The emerging roles of macrophages in cancer metastasis and response to chemotherapy. J Leukoc Biol 2019; 106:259-274. [PMID: 30720887 DOI: 10.1002/jlb.mr0218-056rr] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/17/2022] Open
Abstract
Macrophages represent a heterogeneous group of cells, capable of carrying out distinct functions in a variety of organs and tissues. Even within individual tissues, their functions can vary with location. Tumor-associated macrophages (TAMs) specialize into three major subtypes that carry out multiple tasks simultaneously. This is especially true in the context of metastasis, where TAMs establish most of the cellular and molecular prerequisites for successful cancer cell dissemination and seeding to the secondary site. Perivascular TAMs operate in the perivascular niche, where they promote tumor angiogenesis and aid in the assembly of intravasation sites called tumor microenvironment of metastasis (TMEM). Streaming TAMs co-migrate with tumor cells (irrespective of the perivascular niche) and promote matrix remodeling, tumor cell invasiveness, and an immunosuppressive local microenvironment. Premetastatic TAMs are recruited to the premetastatic niche, where they can assist in tumor cell extravasation, seeding, and metastatic colonization. The dynamic interplay between TAMs and tumor cells can also modify the ability of the latter to resist cytotoxic chemotherapy (a phenotype known as environment-mediated drug resistance) and induce chemotherapy-mediated pro-metastatic microenvironmental changes. These observations suggest that future therapeutics should be designed to target TAMs with the aim of suppressing the metastatic potential of tumors and rendering chemotherapy more efficient.
Collapse
Affiliation(s)
- Luis Rivera Sanchez
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Surgery, Montefiore Medical Center, Bronx, New York, USA
| | - Lucia Borriello
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - David Entenberg
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Surgery, Montefiore Medical Center, Bronx, New York, USA.,Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Pathology, Montefiore Medical Center, Bronx, New York, USA
| | - George S Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
215
|
Hypoxia-inducible factors promote breast cancer stem cell specification and maintenance in response to hypoxia or cytotoxic chemotherapy. Adv Cancer Res 2019; 141:175-212. [PMID: 30691683 DOI: 10.1016/bs.acr.2018.11.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Clinical studies have revealed that breast cancers contain regions of intratumoral hypoxia (reduced oxygen availability), which activates hypoxia-inducible factors (HIFs). The relationship between intratumoral hypoxia, distant metastasis and cancer mortality has been well established. A major mechanism by which intratumoral hypoxia contributes to disease progression is through induction of the breast cancer stem cell (BCSC) phenotype. BCSCs are a small subpopulation of cells with the capability for self-renewal. BCSCs have been implicated in resistance to chemotherapy, disease recurrence, and metastasis. In this review, we will discuss our current understanding of the molecular mechanisms underlying HIF-dependent induction of the BCSC phenotype in response to hypoxia or chemotherapy.
Collapse
|
216
|
Hossain F, Sorrentino C, Ucar DA, Peng Y, Matossian M, Wyczechowska D, Crabtree J, Zabaleta J, Morello S, Del Valle L, Burow M, Collins-Burow B, Pannuti A, Minter LM, Golde TE, Osborne BA, Miele L. Notch Signaling Regulates Mitochondrial Metabolism and NF-κB Activity in Triple-Negative Breast Cancer Cells via IKKα-Dependent Non-canonical Pathways. Front Oncol 2018; 8:575. [PMID: 30564555 PMCID: PMC6289043 DOI: 10.3389/fonc.2018.00575] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 11/15/2018] [Indexed: 12/21/2022] Open
Abstract
Triple negative breast cancer (TNBC) patients have high risk of recurrence and metastasis, and current treatment options remain limited. Cancer stem-like cells (CSCs) have been linked to cancer initiation, progression and chemotherapy resistance. Notch signaling is a key pathway regulating TNBC CSC survival. Treatment of TNBC with PI3K or mTORC1/2 inhibitors results in drug-resistant, Notch-dependent CSC. However, downstream mechanisms and potentially druggable Notch effectors in TNBC CSCs are largely unknown. We studied the role of the AKT pathway and mitochondrial metabolism downstream of Notch signaling in TNBC CSC from cell lines representative of different TNBC molecular subtypes as well as a novel patient-derived model. We demonstrate that exposure of TNBC cells to recombinant Notch ligand Jagged1 leads to rapid AKT phosphorylation in a Notch1-dependent but RBP-Jκ independent fashion. This requires mTOR and IKKα. Jagged1 also stimulates mitochondrial respiration and fermentation in an AKT- and IKK-dependent fashion. Notch1 co-localizes with mitochondria in TNBC cells. Pharmacological inhibition of Notch cleavage by gamma secretase inhibitor PF-03084014 in combination with AKT inhibitor MK-2206 or IKK-targeted NF-κB inhibitor Bay11-7082 blocks secondary mammosphere formation from sorted CD90hi or CD44+CD24low (CSCs) cells. A TNBC patient-derived model gave comparable results. Besides mitochondrial oxidative metabolism, Jagged1 also triggers nuclear, NF-κB-dependent transcription of anti-apoptotic gene cIAP-2. This requires recruitment of Notch1, IKKα and NF-κB to the cIAP-2 promoter. Our observations support a model where Jagged1 triggers IKKα-dependent, mitochondrial and nuclear Notch1 signals that stimulate AKT phosphorylation, oxidative metabolism and transcription of survival genes in PTEN wild-type TNBC cells. These data suggest that combination treatments targeting the intersection of the Notch, AKT and NF-κB pathways have potential therapeutic applications against CSCs in TNBC cases with Notch1 and wild-type PTEN expression.
Collapse
Affiliation(s)
- Fokhrul Hossain
- Louisiana State University Health Sciences Center, Stanley S. Scott Cancer Center, New Orleans, LA, United States.,Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Claudia Sorrentino
- Louisiana State University Health Sciences Center, Stanley S. Scott Cancer Center, New Orleans, LA, United States.,Department of Pharmacy, University of Salerno, Salerno, Italy
| | - Deniz A Ucar
- Louisiana State University Health Sciences Center, Stanley S. Scott Cancer Center, New Orleans, LA, United States.,Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Yin Peng
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, China
| | - Margarite Matossian
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Dorota Wyczechowska
- Louisiana State University Health Sciences Center, Stanley S. Scott Cancer Center, New Orleans, LA, United States
| | - Judy Crabtree
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Jovanny Zabaleta
- Louisiana State University Health Sciences Center, Stanley S. Scott Cancer Center, New Orleans, LA, United States
| | - Silvana Morello
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - Luis Del Valle
- Louisiana State University Health Sciences Center, Stanley S. Scott Cancer Center, New Orleans, LA, United States
| | - Matthew Burow
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Bridgette Collins-Burow
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Antonio Pannuti
- Louisiana State University Health Sciences Center, Stanley S. Scott Cancer Center, New Orleans, LA, United States
| | - Lisa M Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts at Amherst, Amherst, MA, United States
| | - Todd E Golde
- Department of Neuroscience, McKnight Brain Institute, University of Florida at Gainesville, Gainesville, FL, United States
| | - Barbara A Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts at Amherst, Amherst, MA, United States
| | - Lucio Miele
- Louisiana State University Health Sciences Center, Stanley S. Scott Cancer Center, New Orleans, LA, United States.,Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
217
|
Tan T, Wang H, Cao H, Zeng L, Wang Y, Wang Z, Wang J, Li J, Wang S, Zhang Z, Li Y. Deep Tumor-Penetrated Nanocages Improve Accessibility to Cancer Stem Cells for Photothermal-Chemotherapy of Breast Cancer Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1801012. [PMID: 30581704 PMCID: PMC6299727 DOI: 10.1002/advs.201801012] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/29/2018] [Indexed: 05/26/2023]
Abstract
Cancer stem cells (CSCs) are proposed to account for the initiation of cancer metastasis, but their accessibility remains a great challenge. This study reports deep tumor-penetrated biomimetic nanocages to augment the accessibility to CSCs fractions in tumor for anti-metastasis therapy. The nanocages can load photothermal agent of 1,1-dioctadecyl-3,3,3,3-tetramethylindotricarbocyanine iodide (DBN) and chemotherapeutic epirubicin (EBN) to eradicate CSCs for photothermal-chemotherapy of breast cancer metastasis. In metastatic 4T1-indcued tumor model, both DBN and EBN can efficiently accumulate in tumor sites and feasibly permeate throughout the tumor mass. These biomimetic nanosystems can be preferentially internalized by cancer cells and effectively accessed to CSCs fractions in tumor. The DBN+laser/EBN treatment produces considerable depression of primary tumor growth, drastically eradicates around 80% of CSCs fractions in primary tumor, and results in 95.2% inhibition of lung metastasis. Thus, the biomimetic nanocages can be a promising delivery nanovehicle with preferential CSCs-accessibility for effective anti-metastasis therapy.
Collapse
Affiliation(s)
- Tao Tan
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of PharmacyShenyang Pharmaceutical UniversityShenyang110016LiaoningChina
| | - Hong Wang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Haiqiang Cao
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Lijuan Zeng
- School of PharmacyShenyang Pharmaceutical UniversityShenyang110016LiaoningChina
| | - Yuqi Wang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of PharmacyShenyang Pharmaceutical UniversityShenyang110016LiaoningChina
| | - Zhiwan Wang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Jing Wang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Siling Wang
- School of PharmacyShenyang Pharmaceutical UniversityShenyang110016LiaoningChina
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| |
Collapse
|
218
|
Mollen EWJ, Ient J, Tjan-Heijnen VCG, Boersma LJ, Miele L, Smidt ML, Vooijs MAGG. Moving Breast Cancer Therapy up a Notch. Front Oncol 2018; 8:518. [PMID: 30515368 PMCID: PMC6256059 DOI: 10.3389/fonc.2018.00518] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/22/2018] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is the second most common malignancy, worldwide. Treatment decisions are based on tumor stage, histological subtype, and receptor expression and include combinations of surgery, radiotherapy, and systemic treatment. These, together with earlier diagnosis, have resulted in increased survival. However, initial treatment efficacy cannot be guaranteed upfront, and these treatments may come with (long-term) serious adverse effects, negatively affecting a patient's quality of life. Gene expression-based tests can accurately estimate the risk of recurrence in early stage breast cancers. Disease recurrence correlates with treatment resistance, creating a major need to resensitize tumors to treatment. Notch signaling is frequently deregulated in cancer and is involved in treatment resistance. Preclinical research has already identified many combinatory therapeutic options where Notch involvement enhances the effectiveness of radiotherapy, chemotherapy or targeted therapies for breast cancer. However, the benefit of targeting Notch has remained clinically inconclusive. In this review, we summarize the current knowledge on targeting the Notch pathway to enhance current treatments for breast cancer and to combat treatment resistance. Furthermore, we propose mechanisms to further exploit Notch-based therapeutics in the treatment of breast cancer.
Collapse
Affiliation(s)
- Erik W J Mollen
- Department of Radiotherapy, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands.,Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre+, Maastricht, Netherlands.,Division of Medical Oncology, Department of Surgery, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Jonathan Ient
- Department of Radiotherapy, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Vivianne C G Tjan-Heijnen
- Department of Radiotherapy, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands.,Division of Medical Oncology, Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Liesbeth J Boersma
- Department of Radiotherapy, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands.,Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Marjolein L Smidt
- Department of Radiotherapy, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands.,Division of Medical Oncology, Department of Surgery, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Marc A G G Vooijs
- Department of Radiotherapy, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands.,Department of Radiation Oncology (MAASTRO), Maastricht University Medical Centre+, Maastricht, Netherlands
| |
Collapse
|
219
|
Gross ETE, Peinado CD, Jung Y, Han S, Liu B, Santosa EK, Bui JD. Identification and editing of stem-like cells in methylcholanthrene-induced sarcomas. Oncoimmunology 2018; 8:e1404212. [PMID: 30546937 DOI: 10.1080/2162402x.2017.1404212] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/24/2017] [Accepted: 10/27/2017] [Indexed: 12/18/2022] Open
Abstract
The cancer stem cell (CSC) paradigm posits that specific cells within a tumor, so-called CSC-like cells, have differing levels of tumorigenicity and chemoresistance. Original studies of CSCs identified them in human cancers and utilized mouse xenograft models to define the cancer initiating properties of these cells, thereby hampering the understanding of how immunity could affect CSCs. Indeed, few studies have characterized CSCs in the context of cancer immunoediting, and it is currently not clear how immunity could impact on the levels or stem-like behavior of CSCs. Using the well-studied 3'methylcholanthrene (MCA) model of primary sarcoma formation, we have defined a CSC-like population within MCA-induced sarcomas as expressing high levels of stem cell antigen-1 (Sca-1) and low levels of CD90. These Sca-1+CD90- CSC-like cells had higher tumor initiating ability, could spontaneously give rise to Sca-1-negative cells, and formed more sarcospheres than corresponding non-CSC-like cells. Moreover, when examining MCA-induced sarcomas that were in the equilibrium phase of cancer growth, higher levels of CSC-like cells were found compared to MCA-induced sarcomas in the escape phase of cancer progression. Notably, CSC-like cells also emerged during escape from anti-PD-1 or anti-CTLA4 therapy, thus suggesting that CSC-like cells could evade immune therapy. Finally, we demonstrate that paradoxically, interferon (IFN)-γ produced in vivo by immune cells could promote the emergence of CSC-like cells. Our findings define the existence of a Sca1+CD90- CSC-like population in the MCA-sarcoma model capable of differentiation, tumorsphere formation, and increased tumor initiation in vivo. These cells may also act as mediators of immune resistance during cancer immunoediting and immune therapy.
Collapse
Affiliation(s)
- Emilie T E Gross
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Carlos D Peinado
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Yujin Jung
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Semi Han
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Beichen Liu
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Endi K Santosa
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Jack D Bui
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
220
|
Ponert JM, Gockel LM, Henze S, Schlesinger M. Unfractionated and Low Molecular Weight Heparin Reduce Platelet Induced Epithelial-Mesenchymal Transition in Pancreatic and Prostate Cancer Cells. Molecules 2018; 23:molecules23102690. [PMID: 30347648 PMCID: PMC6222876 DOI: 10.3390/molecules23102690] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/12/2018] [Accepted: 10/18/2018] [Indexed: 12/17/2022] Open
Abstract
The interaction with platelets is of crucial importance for tumor cells passing through hematogenous metastasis. Platelets protect cancer cells from immune surveillance and exhibit many other prometastatic effects. Notably, platelets can change the epithelial tumor phenotype, a process termed epithelial-mesenchymal transition (EMT), which confers stem cell-like properties onto tumor cells associated with an increased motility and drug resistance. The aim of the study is to investigate the impact of heparin on the platelet induced EMT program in pancreatic and prostate tumor cells. Platelet activation and interaction with cancer cells were determined by static adhesion assays. Applying ELISAs, the platelet release of EMT inducing mediators was quantified. EMT marker protein expression by tumor cells was explored by western blot and qPCR. Our data show that different tumor cell entities have different platelet binding capacities and also that a weak interaction is sufficient to change tumor cell phenotype. Additionally, unfractionated heparin (UFH) as well as low molecular weight heparin (LMWH) reduced tumor cell platelet interaction. Subsequently, attenuated platelet-derived mediator release resulted in reduced EMT marker protein and transcription factor expression by the cancer cells and decreased cell migration. These data suggest that heparin reduces platelet induced EMT program and prevents the formation of cancer cells with stem cell-like properties. This additional mechanism argues for the use of heparin in oncological applications.
Collapse
Affiliation(s)
- Jan Moritz Ponert
- Department of Pharmacy, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany.
| | - Lukas Maria Gockel
- Department of Pharmacy, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany.
| | - Svenja Henze
- Department of Pharmacy, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany.
| | - Martin Schlesinger
- Department of Pharmacy, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany.
| |
Collapse
|
221
|
Dianat-Moghadam H, Heidarifard M, Jahanban-Esfahlan R, Panahi Y, Hamishehkar H, Pouremamali F, Rahbarghazi R, Nouri M. Cancer stem cells-emanated therapy resistance: Implications for liposomal drug delivery systems. J Control Release 2018; 288:62-83. [DOI: 10.1016/j.jconrel.2018.08.043] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/29/2018] [Accepted: 08/31/2018] [Indexed: 12/17/2022]
|
222
|
Epigenetic Silencing of THY1 Tracks the Acquisition of the Notch1–EGFR Signaling in a Xenograft Model of CD44+/CD24low/CD90+ Myoepithelial Cells. Mol Cancer Res 2018; 17:628-641. [DOI: 10.1158/1541-7786.mcr-17-0324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/13/2017] [Accepted: 09/13/2018] [Indexed: 11/16/2022]
|
223
|
Castaño Z, San Juan BP, Spiegel A, Pant A, DeCristo MJ, Laszewski T, Ubellacker JM, Janssen SR, Dongre A, Reinhardt F, Henderson A, Del Rio AG, Gifford AM, Herbert ZT, Hutchinson JN, Weinberg RA, Chaffer CL, McAllister SS. IL-1β inflammatory response driven by primary breast cancer prevents metastasis-initiating cell colonization. Nat Cell Biol 2018; 20:1084-1097. [PMID: 30154549 PMCID: PMC6511979 DOI: 10.1038/s41556-018-0173-5] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/19/2018] [Indexed: 02/07/2023]
Abstract
Lack of insight into mechanisms governing breast cancer metastasis has precluded the development of curative therapies. Metastasis-initiating cancer cells (MICs) are uniquely equipped to establish metastases, causing recurrence and therapeutic resistance. Using various metastasis models, we discovered that certain primary tumours elicit a systemic inflammatory response involving interleukin-1β (IL-1β)-expressing innate immune cells that infiltrate distant MIC microenvironments. At the metastatic site, IL-1β maintains MICs in a ZEB1-positive differentiation state, preventing MICs from generating highly proliferative E-cadherin-positive progeny. Thus, when the inherent plasticity of MICs is impeded, overt metastases cannot be established. Ablation of the pro-inflammatory response or inhibition of the IL-1 receptor relieves the differentiation block and results in metastatic colonization. Among patients with lymph node-positive breast cancer, high primary tumour IL-1β expression is associated with better overall survival and distant metastasis-free survival. Our data reveal complex interactions that occur between primary tumours and disseminated MICs that could be exploited to improve patient survival.
Collapse
Affiliation(s)
- Zafira Castaño
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Beatriz P San Juan
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Asaf Spiegel
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Ayush Pant
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Molly J DeCristo
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Tyler Laszewski
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jessalyn M Ubellacker
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | - Anushka Dongre
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | | | - Ayana Henderson
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Ana Garcia Del Rio
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Ann M Gifford
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Zachary T Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, MA, USA
| | - John N Hutchinson
- Department of Biostatistics, Harvard T.H. Chan, School of Public Health, Boston, MA, USA
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- MIT Department of Biology and Ludwig/MIT Center for Molecular Oncology, Cambridge, MA, USA
| | - Christine L Chaffer
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
| | - Sandra S McAllister
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
224
|
Bailey PC, Lee RM, Vitolo MI, Pratt SJP, Ory E, Chakrabarti K, Lee CJ, Thompson KN, Martin SS. Single-Cell Tracking of Breast Cancer Cells Enables Prediction of Sphere Formation from Early Cell Divisions. iScience 2018; 8:29-39. [PMID: 30268511 PMCID: PMC6170521 DOI: 10.1016/j.isci.2018.08.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/31/2018] [Accepted: 08/16/2018] [Indexed: 12/31/2022] Open
Abstract
The mammosphere assay has become widely employed to quantify stem-like cells in a population. However, the problem is there is no standard protocol employed by the field. Cell seeding densities of 1,000 to 100,000 cells/mL have been reported. These high densities lead to cellular aggregation. To address this, we have individually tracked 1,127 single MCF-7 and 696 single T47D human breast tumor cells by eye over the course of 14 days. This tracking has given us detailed information for the commonly used endpoints of 5, 7, and 14 days that is unclouded by cellular aggregation. This includes mean sphere sizes, sphere-forming efficiencies, and a well-defined minimum size for both lines. Importantly, we have correlated early cell division with eventual sphere formation. At 24 hr post seeding, we can predict the total spheres on day 14 with 98% accuracy in both lines. This approach removes cell aggregation and potentially shortens a 5- to 14-day assay to a 24 hours. Single-cell tracking removes confounding aggregation from the mammosphere assay Tracking reveals sphere-forming efficiencies much higher than commonly reported True clonal spheres are smaller than commonly reported At 24 hours, tracking can predict total day 14 spheres with 98% accuracy
Collapse
Affiliation(s)
- Patrick C Bailey
- Graduate Program in Biochemistry, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD 21201, USA
| | - Rachel M Lee
- University of Maryland School of Medicine, Bressler Research Building Rm 10-29, 655 W. Baltimore St., Baltimore, MD 21201, USA; University of Maryland College Park, College Park, MD 20742, USA
| | - Michele I Vitolo
- University of Maryland School of Medicine, Bressler Research Building Rm 10-29, 655 W. Baltimore St., Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene St., Baltimore, MD 21201, USA; Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA
| | - Stephen J P Pratt
- Graduate Program in Biochemistry, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD 21201, USA
| | - Eleanor Ory
- University of Maryland School of Medicine, Bressler Research Building Rm 10-29, 655 W. Baltimore St., Baltimore, MD 21201, USA
| | - Kristi Chakrabarti
- University of Maryland School of Medicine, Bressler Research Building Rm 10-29, 655 W. Baltimore St., Baltimore, MD 21201, USA
| | - Cornell J Lee
- University of Maryland School of Medicine, Bressler Research Building Rm 10-29, 655 W. Baltimore St., Baltimore, MD 21201, USA
| | - Keyata N Thompson
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene St., Baltimore, MD 21201, USA
| | - Stuart S Martin
- Graduate Program in Biochemistry, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD 21201, USA; University of Maryland School of Medicine, Bressler Research Building Rm 10-29, 655 W. Baltimore St., Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene St., Baltimore, MD 21201, USA; Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA.
| |
Collapse
|
225
|
Gao L, Zhang W, Zhong WQ, Liu ZJ, Li HM, Yu ZL, Zhao YF. Tumor associated macrophages induce epithelial to mesenchymal transition via the EGFR/ERK1/2 pathway in head and neck squamous cell carcinoma. Oncol Rep 2018; 40:2558-2572. [PMID: 30132555 PMCID: PMC6151899 DOI: 10.3892/or.2018.6657] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 08/03/2018] [Indexed: 01/06/2023] Open
Abstract
The development of head and neck squamous cell carcinoma (HNSCC) is closely associated with inflammation. Tumor associated macrophages (TAMs), the largest population of inflammatory cells in the tumor stroma, serve an important role in accelerating cancer progression. The present study aimed to investigate the role of TAMs in the metastasis of HNSCC. TAM biomarkers and epithelial to mesenchymal transition (EMT)-associated proteins were detected using immunohistochemical and immunofluorescence staining in HNSCC. Then, direct and indirect co-culture systems of TAMs and HNSCC cells were established. The EMT-associated proteins and associated signaling pathways in HNSCC cells of the co-culture system were measured by reverse transcription-quantitative polymerase chain reaction and western blotting. Finally, hierarchical clustering was performed to analyze associations among TAM biomarkers, epidermal growth factor receptor (EGFR), activated extracellular signal-regulated protein kinase 1/2 (ERK1/2) and EMT-associated proteins in HNSCC tissues. The results indicated that the expression of EMT-associated proteins was positively associated with M2 macrophage biomarkers in HNSCC tissues. Cal27 cells were isolated from the co-culture system by fluorescence-activated cell sorting, and it was identified that E-cadherin was downregulated in Cal27 cells, while Vimentin and Slug were upregulated. Furthermore, the results indicated that EGF released by M2 macrophages in the co-culture served an important role by activating ERK1/2. The correlation and cluster analyses indicated that activated ERK1/2 was positively correlated with cluster of differentiation-163, EGFR, Vimentin and Slug. This suggested that TAMs may induce the EMT of cancer cells by activating the EGFR/ERK1/2 signaling pathway in HNSCC, which may be a promising approach to suppressing cancer metastasis.
Collapse
Affiliation(s)
- Lu Gao
- Department of Oral Anatomy, College of Stomotology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Wei Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‑MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Wen-Qun Zhong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‑MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Zhuo-Jue Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‑MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Hui-Min Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‑MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Zi-Li Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‑MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Yi-Fang Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‑MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| |
Collapse
|
226
|
Donnenberg VS, Corselli M, Normolle DP, Meyer EM, Donnenberg AD. Flow cytometric detection of most proteins in the cell surface proteome is unaffected by trypsin treatment. Cytometry A 2018; 93:803-810. [DOI: 10.1002/cyto.a.23525] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/14/2018] [Accepted: 06/18/2018] [Indexed: 10/28/2022]
Affiliation(s)
- Vera S. Donnenberg
- Department of Cardiothoracic Surgery University of Pittsburgh School of Medicine Pittsburgh Pennsylvania
- UPMC Hillman Cancer Center Pittsburgh Pennsylvania
- McGowan Institute of Regenerative Medicine Pittsburgh Pennsylvania
| | | | - Daniel P. Normolle
- UPMC Hillman Cancer Center Pittsburgh Pennsylvania
- Department of Biostatistics University of Pittsburgh Graduate School of Public Health Pittsburgh Pennsylvania
| | | | - Albert D. Donnenberg
- UPMC Hillman Cancer Center Pittsburgh Pennsylvania
- McGowan Institute of Regenerative Medicine Pittsburgh Pennsylvania
- Department of Medicine University of Pittsburgh School of Medicine Pittsburgh Pennsylvania
| |
Collapse
|
227
|
Viewing the Eph receptors with a focus on breast cancer heterogeneity. Cancer Lett 2018; 434:160-171. [PMID: 30055288 DOI: 10.1016/j.canlet.2018.07.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 02/07/2023]
Abstract
Aberrant expression of different family members of the Eph/ephrin system, which comprises the Eph receptors (Ephs) and their ligands (ephrins), has been implicated in various malignancies including breast cancer. The latter presents as a heterogeneous disease with diverse molecular, morphologic and clinical behavior signatures. This review reflects the existing Eph/ephrin literature while focusing on breast cancer heterogeneity. Hormone positive, HER2 positive and triple negative breast cancer (TNBC) cell lines, xenografts/mutant animal models and patient samples are examined separately as, in humans, they represent entities with differences in prognosis and treatment. EphA2, EphB4 and EphB6 are the members most extensively studied in breast cancer. Existing research points to the potential use of various Eph/ephrin members as biomarkers for assessing prognosis and selecting the most suitable therapeutic strategies in variable clinical scenarios, also for overcoming drug resistance, in the era of breast cancer heterogeneity.
Collapse
|
228
|
Zhang Y, Weinberg RA. Epithelial-to-mesenchymal transition in cancer: complexity and opportunities. Front Med 2018; 12:361-373. [PMID: 30043221 PMCID: PMC6186394 DOI: 10.1007/s11684-018-0656-6] [Citation(s) in RCA: 431] [Impact Index Per Article: 71.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 07/02/2018] [Indexed: 02/07/2023]
Abstract
The cell-biological program termed the epithelial-to-mesenchymal transition (EMT) plays an important role in both development and cancer progression. Depending on the contextual signals and intracellular gene circuits of a particular cell, this program can drive fully epithelial cells to enter into a series of phenotypic states arrayed along the epithelial-mesenchymal phenotypic axis. These cell states display distinctive cellular characteristics, including stemness, invasiveness, drug-resistance and the ability to form metastases at distant organs, and thereby contribute to cancer metastasis and relapse. Currently we still lack a coherent overview of the molecular and biochemical mechanisms inducing cells to enter various states along the epithelial-mesenchymal phenotypic spectrum. An improved understanding of the dynamic and plastic nature of the EMT program has the potential to yield novel therapies targeting this cellular program that may aid in the management of high-grade malignancies.
Collapse
Affiliation(s)
- Yun Zhang
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA.
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA. .,MIT Department of Biology, Cambridge, MA, 02142, USA. .,Ludwig/MIT Center for Molecular Oncology, Cambridge, MA, 02142, USA.
| |
Collapse
|
229
|
Abstract
The concept that progression of cancer is regulated by interactions of cancer cells with their microenvironment was postulated by Stephen Paget over a century ago. Contemporary tumour microenvironment (TME) research focuses on the identification of tumour-interacting microenvironmental constituents, such as resident or infiltrating non-tumour cells, soluble factors and extracellular matrix components, and the large variety of mechanisms by which these constituents regulate and shape the malignant phenotype of tumour cells. In this Timeline article, we review the developmental phases of the TME paradigm since its initial description. While illuminating controversies, we discuss the importance of interactions between various microenvironmental components and tumour cells and provide an overview and assessment of therapeutic opportunities and modalities by which the TME can be targeted.
Collapse
Affiliation(s)
- Shelly Maman
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Isaac P Witz
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
230
|
Yang WH, Cha JH, Xia W, Lee HH, Chan LC, Wang YN, Hsu JL, Ren G, Hung MC. Juxtacrine Signaling Inhibits Antitumor Immunity by Upregulating PD-L1 Expression. Cancer Res 2018; 78:3761-3768. [PMID: 29789418 DOI: 10.1158/0008-5472.can-18-0040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/09/2018] [Accepted: 05/17/2018] [Indexed: 01/21/2023]
Abstract
Programmed death-ligand 1 (PD-L1) is a well-known immune checkpoint protein that helps cancer cells evade immune response. Anti-PD-L1 immune therapy has been approved for the treatment of several advanced human cancers. Therefore, further understanding of the regulatory mechanisms of PD-L1 is critical to improve PD-L1-targeting immunotherapy. Recent studies indicated that contact-dependent pathways may regulate anticancer immunity, highlighting the importance of cell contact-induced signaling in cancer immunity. Here, we show that tumor cell contact upregulates PD-L1 expression and reduces T-cell-mediated cell killing through the membrane receptor tyrosine kinase ephrin receptor A10 (EphA10), which is not expressed in normal tissues except testis and is known to mediate cell contact-dependent juxtacrine signaling. Knockout of EphA10 in tumor cells increased T-cell-mediated antitumor immunity in syngeneic mouse models. EphA10 expression also correlated positively with PD-L1 in human breast tumor tissues. Together, our data reveal that in addition to paracrine/autocrine signaling, cell contact-mediated juxtacrine signaling also promotes PD-L1 expression, implying that tumor cells may escape immune surveillance via this mechanism and that targeting EphA10 to boost antitumor immunity may be a new immune checkpoint blockade strategy for female patients with breast cancer.Significance: Regulation of PD-L1 expression by cell contact-mediated signaling promotes immune escape in breast cancer and may lead to the development of an immunotherapy with less adverse effects in female patients. Cancer Res; 78(14); 3761-8. ©2018 AACR.
Collapse
Affiliation(s)
- Wen-Hao Yang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jong-Ho Cha
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Weiya Xia
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heng-Huan Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Li-Chuan Chan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, Texas
| | - Ying-Nai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer L Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Guoxin Ren
- Department of Oral and Maxillofacial Head and Neck Oncology, Affiliated 9th People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, Texas
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
231
|
Zhang D, Tang DG, Rycaj K. Cancer stem cells: Regulation programs, immunological properties and immunotherapy. Semin Cancer Biol 2018; 52:94-106. [PMID: 29752993 DOI: 10.1016/j.semcancer.2018.05.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023]
Abstract
It is becoming increasingly clear that virtually all types of human cancers harbor a small population of stem-like cancer cells (i.e., cancer stem cells, CSCs). These CSCs preexist in primary tumors, can self-renew and are more tolerant of standard treatments, such as antimitotic and molecularly targeted agents, most of which preferentially eliminate differentiated and proliferating cancer cells. CSCs are therefore postulated as the root of therapy resistance, relapse and metastasis. Aside from surgery, radiation, and chemotherapy, immunotherapy is now established as the fourth pillar in the therapeutic armamentarium for patients with cancer, especially late-stage and advanced cancers. A better understanding of CSC immunological properties should lead to development of novel immunologic approaches targeting CSCs, which, in turn, may help prevent tumor recurrence and eliminate residual diseases. Here, with a focus on CSCs in solid tumors, we review CSC regulation programs and recent transcriptomics-based immunological profiling data specific to CSCs. By highlighting CSC antigens that could potentially be immunogenic, we further discuss how CSCs can be targeted immunologically.
Collapse
Affiliation(s)
- Dingxiao Zhang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA; Key Lab of Agricultural Animal Genetics, Breeding & Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA; Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Kiera Rycaj
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
232
|
Mammary Stem Cells and Breast Cancer Stem Cells: Molecular Connections and Clinical Implications. Biomedicines 2018; 6:biomedicines6020050. [PMID: 29734696 PMCID: PMC6026898 DOI: 10.3390/biomedicines6020050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/29/2018] [Accepted: 03/31/2018] [Indexed: 12/27/2022] Open
Abstract
Cancer arises from subpopulations of transformed cells with high tumor initiation and repopulation ability, known as cancer stem cells (CSCs), which share many similarities with their normal counterparts. In the mammary gland, several studies have shown common molecular regulators between adult mammary stem cells (MaSCs) and breast cancer stem cells (bCSCs). Cell plasticity and self-renewal are essential abilities for MaSCs to maintain tissue homeostasis and regenerate the gland after pregnancy. Intriguingly, these properties are similarly executed in breast cancer stem cells to drive tumor initiation, tumor heterogeneity and recurrence after chemotherapy. In addition, both stem cell phenotypes are strongly influenced by external signals from the microenvironment, immune cells and supportive specific niches. This review focuses on the intrinsic and extrinsic connections of MaSC and bCSCs with clinical implications for breast cancer progression and their possible therapeutic applications.
Collapse
|
233
|
Petersen CP, Meyer AR, DeSalvo C, Choi E, Schlegel C, Petersen A, Engevik AC, Prasad N, Levy SE, Peebles RS, Pizarro TT, Goldenring JR. A signalling cascade of IL-33 to IL-13 regulates metaplasia in the mouse stomach. Gut 2018; 67:805-817. [PMID: 28196875 PMCID: PMC5681443 DOI: 10.1136/gutjnl-2016-312779] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 01/13/2017] [Accepted: 01/15/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Alternatively activated macrophages (M2) are associated with the progression of spasmolytic polypeptide-expressing metaplasia (SPEM) in the stomach. However, the precise mechanism(s) and critical mediators that induce SPEM are unknown. DESIGN To determine candidate genes important in these processes, macrophages from the stomach corpus of mice with SPEM (DMP-777-treated) or advanced SPEM (L635-treated) were isolated and RNA sequenced. Effects on metaplasia development after acute parietal cell loss induced by L635 were evaluated in interleukin (IL)-33, IL-33 receptor (ST2) and IL-13 knockout (KO) mice. RESULTS Profiling of metaplasia-associated macrophages in the stomach identified an M2a-polarised macrophage population. Expression of IL-33 was significantly upregulated in macrophages associated with advanced SPEM. L635 induced metaplasia in the stomachs of wild-type mice, but not in the stomachs of IL-33 and ST2 KO mice. While IL-5 and IL-9 were not required for metaplasia induction, IL-13 KO mice did not develop metaplasia in response to L635. Administration of IL-13 to ST2 KO mice re-established the induction of metaplasia following acute parietal cell loss. CONCLUSIONS Metaplasia induction and macrophage polarisation after parietal cell loss is coordinated through a cytokine signalling network of IL-33 and IL-13, linking a combined response to injury by both intrinsic mucosal mechanisms and infiltrating M2 macrophages.
Collapse
Affiliation(s)
- Christine P. Petersen
- Departments of Cell and Developmental Biology, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Anne R. Meyer
- Departments of Cell and Developmental Biology, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Carlo DeSalvo
- Department of Pathology, Case Western Reserve School of Medicine, Cleveland, OH
| | - Eunyoung Choi
- Department of Nashville VA Medical Center, Vanderbilt University, Nashville, TN,Department of Surgery, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Cameron Schlegel
- Department of Surgery, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Alec Petersen
- Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Amy C. Engevik
- Department of Surgery, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Nripesh Prasad
- Department of HudsonAlpha Institute for Biotechnology, Huntsville, AL
| | - Shawn E. Levy
- Department of HudsonAlpha Institute for Biotechnology, Huntsville, AL
| | | | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve School of Medicine, Cleveland, OH
| | - James R. Goldenring
- Department of Nashville VA Medical Center, Vanderbilt University, Nashville, TN,Departments of Cell and Developmental Biology, Vanderbilt University, Nashville, TN,Department of Surgery, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| |
Collapse
|
234
|
Huang H, Wang C, Liu F, Li HZ, Peng G, Gao X, Dong KQ, Wang HR, Kong DP, Qu M, Dai LH, Wang KJ, Zhou Z, Yang J, Yang ZY, Cheng YQ, Tian QQ, Liu D, Xu CL, Xu DF, Cui XG, Sun YH. Reciprocal Network between Cancer Stem-Like Cells and Macrophages Facilitates the Progression and Androgen Deprivation Therapy Resistance of Prostate Cancer. Clin Cancer Res 2018; 24:4612-4626. [PMID: 29691294 DOI: 10.1158/1078-0432.ccr-18-0461] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/03/2018] [Accepted: 04/20/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Cancer stem-like cells (CSC) contribute to the progression and androgen deprivation therapy (ADT) resistance of prostate cancer. As CSCs depend on their specific niche, including tumor-associated macrophages (TAM), elucidating the network between CSCs and TAMs may help to effectively inhibit the progression and ADT resistance of prostate cancer.Experimental Design: The underlying intracellular mechanism that sustains the stem-like characteristics of CSCs in prostate cancer was assessed via RNA sequencing, co-immunoprecipitation, chromatin immunoprecipitation, and other assays. A coculture system and cytokine antibody arrays were used to examine the interaction network between CSCs and TAMs. In addition, an orthotopic prostate cancer model was established to evaluate the in vivo effects of the combined targeting of CSCs and their interaction with TAMs on ADT resistance.Results: Autophagy-related gene 7 (ATG7) facilitated the transcription of OCT4 via β-catenin, which binds to the OCT4 promoter, promoting CSC characteristics in prostate cancer, including self-renewal, tumor initiation, and drug resistance. In addition, CSCs remodeled their specific niche by educating monocytes/macrophages toward TAMs, and the CSC-educated TAMs reciprocally promoted the stem-like properties of CSCs, progression and ADT resistance of prostate cancer via IL6/STAT3. Furthermore, the combined targeting of CSCs and their interaction with TAMs by inhibiting ATG7/OCT4 and IL6 receptor effectively ameliorated ADT resistance in an orthotopic prostate cancer model.Conclusions: Targeting CSCs and their niche may prove to be a more powerful strategy than targeting CSCs alone, providing a rational approach to ameliorating ADT resistance in prostate cancer. Clin Cancer Res; 24(18); 4612-26. ©2018 AACR.
Collapse
Affiliation(s)
- Hai Huang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chao Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Fei Liu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hui-Zhen Li
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Guang Peng
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xu Gao
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ke-Qin Dong
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hong-Ru Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - De-Pei Kong
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Min Qu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Li-He Dai
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Kai-Jian Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhe Zhou
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jun Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ze-Yu Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yan-Qiong Cheng
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qin-Qin Tian
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Dan Liu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chuan-Liang Xu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Dan-Feng Xu
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xin-Gang Cui
- Department of Urinary Surgery, The Third Affiliated Hospital of Second Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China.
- Department of Urinary Surgery, Gongli Hospital, Second Military Medical University, Shanghai, China
| | - Ying-Hao Sun
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
235
|
Shahin SA, Wang R, Simargi SI, Contreras A, Parra Echavarria L, Qu L, Wen W, Dellinger T, Unternaehrer J, Tamanoi F, Zink JI, Glackin CA. Hyaluronic acid conjugated nanoparticle delivery of siRNA against TWIST reduces tumor burden and enhances sensitivity to cisplatin in ovarian cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1381-1394. [PMID: 29665439 DOI: 10.1016/j.nano.2018.04.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/19/2018] [Accepted: 04/08/2018] [Indexed: 12/29/2022]
Abstract
TWIST protein is critical to development and is activated in many cancers. TWIST regulates epithelial-mesenchymal transition, and is linked to angiogenesis, metastasis, cancer stem cell phenotype, and drug resistance. The majority of epithelial ovarian cancer (EOC) patients with metastatic disease respond well to first-line chemotherapy but most relapse with disease that is both metastatic and drug resistant, leading to a five-year survival rate under 20%. We are investigating the role of TWIST in mediating these relapses. We demonstrate TWIST-siRNA (siTWIST) and a novel nanoparticle delivery platform to reverse chemoresistance in an EOC model. Hyaluronic-acid conjugated mesoporous silica nanoparticles (MSN-HAs) carried siTWIST into target cells and led to sustained TWIST knockdown in vitro. Mice treated with siTWIST-MSN-HA and cisplatin exhibited specific tumor targeting and reduction of tumor burden. This platform has potential application for overcoming clinical challenges of tumor cell targeting, metastasis and chemoresistance in ovarian and other TWIST overexpressing cancers.
Collapse
Affiliation(s)
- Sophia A Shahin
- Irell & Manella Graduate School of Biological Sciences, City of Hope - Beckman Research Institute, Duarte, California, USA; Department of Stem Cell and Developmental Biology, City of Hope - Beckman Research Institute, Duarte, California, USA
| | - Ruining Wang
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California Los Angeles, Los Angeles, California, USA
| | - Shirleen I Simargi
- Department of Stem Cell and Developmental Biology, City of Hope - Beckman Research Institute, Duarte, California, USA; Department of Biological Sciences, California State University, Pomona, CA
| | - Altagracia Contreras
- Department of Stem Cell and Developmental Biology, City of Hope - Beckman Research Institute, Duarte, California, USA; Department of Biological Sciences, California State University, Long Beach, CA
| | - Liliana Parra Echavarria
- Department of Stem Cell and Developmental Biology, City of Hope - Beckman Research Institute, Duarte, California, USA
| | - Louise Qu
- Irell & Manella Graduate School of Biological Sciences, City of Hope - Beckman Research Institute, Duarte, California, USA; Department of Stem Cell and Developmental Biology, City of Hope - Beckman Research Institute, Duarte, California, USA
| | - Wei Wen
- Department of Surgery, City of Hope - Beckman Research Institute, Duarte, California, USA
| | - Thanh Dellinger
- Department of Surgery, City of Hope - Beckman Research Institute, Duarte, California, USA
| | - Juli Unternaehrer
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA
| | - Fuyuhiko Tamanoi
- Department of Microbiology, Immunology, and Molecular Genetics, Jonsson Comprehensive Cancer Center, California NanoSystems Institute, University of California Los Angeles, Los Angeles, California, USA
| | - Jeffrey I Zink
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California Los Angeles, Los Angeles, California, USA
| | - Carlotta A Glackin
- Irell & Manella Graduate School of Biological Sciences, City of Hope - Beckman Research Institute, Duarte, California, USA; Department of Stem Cell and Developmental Biology, City of Hope - Beckman Research Institute, Duarte, California, USA.
| |
Collapse
|
236
|
Monterisi S, Lo Riso P, Russo K, Bertalot G, Vecchi M, Testa G, Di Fiore PP, Bianchi F. HOXB7 overexpression in lung cancer is a hallmark of acquired stem-like phenotype. Oncogene 2018; 37:3575-3588. [PMID: 29576613 DOI: 10.1038/s41388-018-0229-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/31/2018] [Accepted: 02/28/2018] [Indexed: 12/24/2022]
Abstract
HOXB7 is a homeodomain (HOX) transcription factor involved in regional body patterning of invertebrates and vertebrates. We previously identified HOXB7 within a ten-gene prognostic signature for lung adenocarcinoma, where increased expression of HOXB7 was associated with poor prognosis. This raises the question of how HOXB7 overexpression can influence the metastatic behavior of lung adenocarcinoma. Here, we analyzed publicly available microarray and RNA-seq lung cancer expression datasets and found that HOXB7-overexpressing tumors are enriched in gene signatures characterizing adult and embryonic stem cells (SC), and induced pluripotent stem cells (iPSC). Experimentally, we found that HOXB7 upregulates several canonical SC/iPSC markers and sustains the expansion of a subpopulation of cells with SC characteristics, through modulation of LIN28B, an emerging cancer gene and pluripotency factor, which we discovered to be a direct target of HOXB7. We validated this new circuit by showing that HOXB7 enhances reprogramming to iPSC with comparable efficiency to LIN28B or its target c-MYC, which is a canonical reprogramming factor.
Collapse
Affiliation(s)
- Simona Monterisi
- Molecular Medicine Program, European Institute of Oncology, 20141, Milan, Italy.,IFOM, The FIRC Institute for Molecular Oncology Foundation, 20139, Milan, Italy.,Humanitas Clinical and Research Center, 20089 Rozzano (MI), Italy
| | - Pietro Lo Riso
- Department of Experimental Oncology, European Institute of Oncology, 20141, Milan, Italy
| | - Karin Russo
- IFOM, The FIRC Institute for Molecular Oncology Foundation, 20139, Milan, Italy
| | - Giovanni Bertalot
- Molecular Medicine Program, European Institute of Oncology, 20141, Milan, Italy
| | - Manuela Vecchi
- IFOM, The FIRC Institute for Molecular Oncology Foundation, 20139, Milan, Italy
| | - Giuseppe Testa
- Department of Experimental Oncology, European Institute of Oncology, 20141, Milan, Italy.,DIPO, Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
| | - Pier Paolo Di Fiore
- Molecular Medicine Program, European Institute of Oncology, 20141, Milan, Italy.,IFOM, The FIRC Institute for Molecular Oncology Foundation, 20139, Milan, Italy.,DIPO, Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
| | - Fabrizio Bianchi
- Molecular Medicine Program, European Institute of Oncology, 20141, Milan, Italy. .,ISBREMIT, Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo (FG), Italy.
| |
Collapse
|
237
|
TRIM28 multi-domain protein regulates cancer stem cell population in breast tumor development. Oncotarget 2018; 8:863-882. [PMID: 27845900 PMCID: PMC5352203 DOI: 10.18632/oncotarget.13273] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 10/19/2016] [Indexed: 12/21/2022] Open
Abstract
The expression of Tripartite motif-containing protein 28 (TRIM28)/Krüppel-associated box (KRAB)-associated protein 1 (KAP1), is elevated in at least 14 tumor types, including solid and hematopoietic tumors. High level of TRIM28 is associated with triple-negative subtype of breast cancer (TNBC), which shows higher aggressiveness and lower survival rates. Interestingly, TRIM28 is essential for maintaining the pluripotent phenotype in embryonic stem cells. Following on that finding, we evaluated the role of TRIM28 protein in the regulation of breast cancer stem cells (CSC) populations and tumorigenesis in vitro and in vivo. Downregulation of TRIM28 expression in xenografts led to deceased expression of pluripotency and mesenchymal markers, as well as inhibition of signaling pathways involved in the complex mechanism of CSC maintenance. Moreover, TRIM28 depletion reduced the ability of cancer cells to induce tumor growth when subcutaneously injected in limiting dilutions. Our data demonstrate that the downregulation of TRIM28 gene expression reduced the ability of CSCs to self-renew that resulted in significant reduction of tumor growth. Loss of function of TRIM28 leads to dysregulation of cell cycle, cellular response to stress, cancer cell metabolism, and inhibition of oxidative phosphorylation. All these mechanisms directly regulate maintenance of CSC population. Our original results revealed the role of the TRIM28 in regulating the CSC population in breast cancer. These findings may pave the way to novel and more effective therapies targeting cancer stem cells in breast tumors.
Collapse
|
238
|
Liu L, Yang L, Yan W, Zhai J, Pizzo DP, Chu P, Chin AR, Shen M, Dong C, Ruan X, Ren X, Somlo G, Wang SE. Chemotherapy Induces Breast Cancer Stemness in Association with Dysregulated Monocytosis. Clin Cancer Res 2018; 24:2370-2382. [PMID: 29500278 DOI: 10.1158/1078-0432.ccr-17-2545] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/20/2017] [Accepted: 03/01/2018] [Indexed: 12/17/2022]
Abstract
Purpose: Preoperative or neoadjuvant therapy (NT) is increasingly used in patients with locally advanced or inflammatory breast cancer to allow optimal surgery and aim for pathologic response. However, many breast cancers are resistant or relapse after treatment. Here, we investigated conjunctive chemotherapy-triggered events occurring systemically and locally, potentially promoting a cancer stem-like cell (CSC) phenotype and contributing to tumor relapse.Experimental Design: We started by comparing the effect of paired pre- and post-NT patient sera on the CSC properties of breast cancer cells. Using cell lines, patient-derived xenograft models, and primary tumors, we investigated the regulation of CSCs and tumor progression by chemotherapy-induced factors.Results: In human patients and mice, we detected a therapy-induced CSC-stimulatory activity in serum, which was attributed to therapy-associated monocytosis leading to systemic elevation of monocyte chemoattractant proteins (MCP). The post-NT hematopoietic regeneration in the bone marrow highlighted both altered monocyte-macrophage differentiation and biased commitment of stimulated hematopoietic stem cells toward monocytosis. Chemotherapeutic agents also induce monocyte expression of MCPs through a JNK-dependent mechanism. Genetic and pharmacologic inhibitions of the MCP-CCR2 pathway blocked chemotherapy's adverse effect on CSCs. Levels of nuclear Notch and ALDH1 were significantly elevated in primary breast cancers following NT, whereas higher levels of CCR2 in pre-NT tumors were associated with a poor response to NT.Conclusions: Our data establish a mechanism of chemotherapy-induced cancer stemness by linking the cellular events in the bone marrow and tumors, and suggest pharmacologic inhibition of CCR2 as a potential cotreatment during conventional chemotherapy in neoadjuvant and adjuvant settings. Clin Cancer Res; 24(10); 2370-82. ©2018 AACR.
Collapse
Affiliation(s)
- Liang Liu
- Department of Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lin Yang
- Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wei Yan
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - Jing Zhai
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Donald P Pizzo
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - Peiguo Chu
- Department of Pathology, City of Hope National Medical Center and Comprehensive Cancer Center, Duarte, California
| | - Andrew R Chin
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - Meng Shen
- Department of Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chuan Dong
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - Xianhui Ruan
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiubao Ren
- Department of Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - George Somlo
- Department of Medical Oncology, City of Hope National Medical Center and Comprehensive Cancer Center, Duarte, California
| | - Shizhen Emily Wang
- Department of Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
- Department of Pathology, University of California, San Diego, La Jolla, California
| |
Collapse
|
239
|
Donnenberg VS, Zhang JJ, Moravcikova E, Meyer EM, Lu H, Carson CT, Donnenberg AD. Antibody-based cell-surface proteome profiling of metastatic breast cancer primary explants and cell lines. Cytometry A 2018; 93:448-457. [PMID: 29498809 DOI: 10.1002/cyto.a.23300] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/09/2016] [Accepted: 12/28/2016] [Indexed: 01/06/2023]
Abstract
Flow cytometric cell surface proteomics provides a new and powerful tool to determine changes accompanying neoplastic transformation and invasion, providing clues to essential interactions with the microenvironment as well as leads for potential therapeutic targets. One of the most important advantages of flow cytometric cell surface proteomics is that it can be performed on living cells that can be sorted for further characterization and functional studies. Here, we document the surface proteome of clonogenic metastatic breast cancer (MBrCa) explants, which was strikingly similar to that of normal mesenchymal stromal cells (P = 0.017, associated with Pearson correlation coefficient) and transformed mammary epithelial cells (P = 0.022). Markers specifically upregulated on MBrCa included CD200 (Ox2), CD51/CD61 (Integrin α5/β3), CD26 (dipeptidyl peptidase-4), CD165 (c-Cbl), and CD54 (ICAM-1). Proteins progressively upregulated in a model of neoplastic transformation and invasion included CD26, CD63 (LAMP3), CD105 (Endoglin), CD107a (LAMP1), CD108 (Semaphorin 7A), CD109 (Integrin β4), CD151 (Raph blood group), and disialoganglioside G2. The proteome of the commonly used cell lines MDA-MB-231, MCF7, and BT-474 were uncorrelated with that of MBrCa (P = 1.0, 1.0, 0.9, respectively). The comparison has demonstrated the mesenchymal nature of clonogenic cells isolated by short-term culture of metastatic breast cancer, provided several leads for biomarkers and potential targets for anti-invasive therapy, including CD200, and highlighted the limitations of breast cancer cell lines for representing the cell surface biology of breast cancer. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Vera S Donnenberg
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA.,University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.,McGowan Institute of Regenerative Medicine, Pittsburgh, Pennsylvania
| | - Jayce Jieming Zhang
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.,Xiangya School of Medicine, Central South University, Changsha City, China
| | - Erika Moravcikova
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Haihui Lu
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | | | - Albert D Donnenberg
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.,McGowan Institute of Regenerative Medicine, Pittsburgh, Pennsylvania.,Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
240
|
Proteolysis-a characteristic of tumor-initiating cells in murine metastatic breast cancer. Oncotarget 2018; 7:58244-58260. [PMID: 27542270 PMCID: PMC5295428 DOI: 10.18632/oncotarget.11309] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/27/2016] [Indexed: 11/25/2022] Open
Abstract
Tumor initiating cells (TICs) have been identified and functionally characterized in hematological malignancies as well as in solid tumors such as breast cancer. In addition to their high tumor-initiating potential, TICs are founder cells for metastasis formation and are involved in chemotherapy resistance. In this study we explored molecular pathways which enable this tumor initiating potential for a cancer cell subset of the transgenic MMTV-PyMT mouse model for metastasizing breast cancer. The cell population, characterized by the marker profile CD24+CD90+CD45-, showed a high tumorigenicity compared to non-CD24+CD90+CD45- cancer cells in colony formation assays, as well as upon orthotopic transplantation into the mammary fat pad of mice. In addition, these orthotopically grown CD24+CD90+CD45- TICs metastasized to the lungs. The transcriptome of TICs freshly isolated from primary tumors by cell sorting was compared with that of sorted non-CD24+CD90+CD45- cancer cells by RNA-seq. In addition to more established TIC signatures, such as epithelial-to-mesenchymal transition or mitogen signaling, an upregulated gene set comprising several classes of proteolytic enzymes was uncovered in the TICs. Accordingly, TICs showed high intra- and extracellular proteolytic activity. Application of a broad range of protease inhibitors to TICs in a colony formation assay reduced anchorage independent growth and had an impact on colony morphology in 3D cell culture assays. We conclude that CD24+CD90+CD45- cells of the MMTV- PyMT mouse model possess an upregulated proteolytic signature which could very well represent a functional hallmark of metastatic TICs from mammary carcinomas.
Collapse
|
241
|
Yan R, Chen X, Wang W, Liang L, Zhou C, Wei W, Yi H, Wu X, Liu G, Zhong M, Yu Y. [Association between HLA-A and HLA-DRB1 allele polymorphisms and susceptibility to tuberculosis in southern Chinese population]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:101-105. [PMID: 33177033 DOI: 10.3969/j.issn.1673-4254.2018.01.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the correlation between tumor-associated macrophages (TAMs) and the development of high risk human papilloma virus (hr-HPV)-related cervical cancer. METHODS A total of 112 cases of cervical tissue were collected, including 16 normal cervical tissues, 55 cervical intraepithelial neoplasia (CIN) tissues and 41 squamous cervical cancer (SCC) tissues. The expression of CD163+ macrophages in the cervical tissues was detected by immunohistochemical method, and the results were analyzed in relation with the clinical data of the patients. RESULTS Immunohistochemical analysis showed that the cell density of CD163+ macrophages increased progressively with the increase in the tissue malignancy, in the order of normal cervical tissue, CIN Ⅰ, CIN Ⅱ-Ⅲ, and SCC. Correlation analysis revealed a positive relationship between CD163+ macrophage density and tissue malignancy (P=0.000). The density of CD163+ macrophages was significantly upregulated in HR-HPV-positive SCC tissue (P < 0.05). CD163+ macrophages were positively correlated with cervical lymph node metastasis (P=0.005) and FIGO stage (P=0.004) of SCC. CONCLUSIONS The expression of CD163+ macrophages is positively correlated with malignant transformation of cervical tissues, and hr-HPV infection is significantly correlated with CD163 expression level in the macrophages. CD163+ macrophages can be used as predictors of the occurrence and progression of cervical cancer caused by hr-HPV infection.
Collapse
Affiliation(s)
- Ruiming Yan
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Xiaojing Chen
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Wei Wang
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Li Liang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Chenfei Zhou
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Wenfei Wei
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Hongyan Yi
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Xiangguang Wu
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Guobing Liu
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Mei Zhong
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Yanhong Yu
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| |
Collapse
|
242
|
Sultan M, Vidovic D, Paine AS, Huynh TT, Coyle KM, Thomas ML, Cruickshank BM, Dean CA, Clements DR, Kim Y, Lee K, Gujar SA, Weaver IC, Marcato P. Epigenetic Silencing of TAP1 in Aldefluor+Breast Cancer Stem Cells Contributes to Their Enhanced Immune Evasion. Stem Cells 2018; 36:641-654. [DOI: 10.1002/stem.2780] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 12/20/2017] [Accepted: 01/10/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Mohammad Sultan
- Departments of Pathology, Dalhousie University; Halifax Nova Scotia Canada
| | - Dejan Vidovic
- Departments of Pathology, Dalhousie University; Halifax Nova Scotia Canada
| | - Arianne S. Paine
- Departments of Pathology, Dalhousie University; Halifax Nova Scotia Canada
| | - Thomas T. Huynh
- Departments of Pathology, Dalhousie University; Halifax Nova Scotia Canada
| | - Krysta M. Coyle
- Departments of Pathology, Dalhousie University; Halifax Nova Scotia Canada
| | - Margaret L. Thomas
- Departments of Pathology, Dalhousie University; Halifax Nova Scotia Canada
| | | | - Cheryl A. Dean
- Departments of Pathology, Dalhousie University; Halifax Nova Scotia Canada
| | - Derek R. Clements
- Departments of Pathology, Dalhousie University; Halifax Nova Scotia Canada
| | - Youra Kim
- Departments of Pathology, Dalhousie University; Halifax Nova Scotia Canada
| | - Kristen Lee
- Psychology and Neuroscience, Dalhousie University; Halifax Nova Scotia Canada
| | - Shashi A. Gujar
- Departments of Pathology, Dalhousie University; Halifax Nova Scotia Canada
- Microbiology and Immunology, Dalhousie University; Halifax Nova Scotia Canada
| | - Ian C.G. Weaver
- Psychology and Neuroscience, Dalhousie University; Halifax Nova Scotia Canada
- Psychiatry and Brain Repair Centre; Dalhousie University; Halifax Nova Scotia Canada
| | - Paola Marcato
- Departments of Pathology, Dalhousie University; Halifax Nova Scotia Canada
- Microbiology and Immunology, Dalhousie University; Halifax Nova Scotia Canada
| |
Collapse
|
243
|
Ponert JM, Schwarz S, Haschemi R, Müller J, Pötzsch B, Bendas G, Schlesinger M. The mechanisms how heparin affects the tumor cell induced VEGF and chemokine release from platelets to attenuate the early metastatic niche formation. PLoS One 2018; 13:e0191303. [PMID: 29346400 PMCID: PMC5773218 DOI: 10.1371/journal.pone.0191303] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/01/2018] [Indexed: 11/18/2022] Open
Abstract
Metastasis is responsible for the majority of cancer associated fatalities. Tumor cells leaving the primary tumor and entering the blood flow immediately interact with platelets. Activated platelets contribute in different ways to cancer cell survival and proliferation, e.g. in formation of the early metastatic niche by release of different growth factors and chemokines. Here we show that a direct interaction between platelets and MV3 melanoma or MCF7 breast cancer cells induces platelet activation and a VEGF release in citrated plasma that cannot be further elevated by the coagulation cascade and generated thrombin. In contrast, the release of platelet-derived chemokines CXCL5 and CXCL7 depends on both, a thrombin-mediated platelet activation and a direct interaction between tumor cells and platelets. Preincubation of platelets with therapeutic concentrations of unfractionated heparin reduces the tumor cell initiated VEGF release from platelets. In contrast, tumor cell induced CXCL5 and CXCL7 release from platelets was not impacted by heparin pretreatment in citrated plasma. In defibrinated, recalcified plasma, on the contrary, heparin is able to reduce CXCL5 and CXCL7 release from platelets by thrombin inhibition. Our data indicate that different chemokines and growth factors in diverse platelet granules are released in tightly regulated processes by various trigger mechanisms. We show for the first time that heparin is able to reduce the mediator release induced by different tumor cells both in a contact and coagulation dependent manner.
Collapse
Affiliation(s)
- Jan Moritz Ponert
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
| | - Svenja Schwarz
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
| | - Reza Haschemi
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
| | - Jens Müller
- Institute for Experimental Hematology and Transfusion Medicine, University of Bonn Medical Centre, Bonn, Germany
| | - Bernd Pötzsch
- Institute for Experimental Hematology and Transfusion Medicine, University of Bonn Medical Centre, Bonn, Germany
| | - Gerd Bendas
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
| | - Martin Schlesinger
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
- * E-mail:
| |
Collapse
|
244
|
Functional and prognostic significance of long non-coding RNA MALAT1 as a metastasis driver in ER negative lymph node negative breast cancer. Oncotarget 2018; 7:40418-40436. [PMID: 27250026 PMCID: PMC5130017 DOI: 10.18632/oncotarget.9622] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/09/2016] [Indexed: 12/15/2022] Open
Abstract
MALAT1 (metastasis associated lung adenocarcinoma transcript1) is a conserved long non-coding RNA, known to regulate gene expression by modulating transcription and post-transcriptional pre-mRNA processing of a large number of genes. MALAT1 expression is deregulated in various tumors, including breast cancer. However, the significance of such abnormal expression is yet to be fully understood. In this study, we demonstrate that regulation of aggressive breast cancer cell traits by MALAT1 is not predicted solely based on an elevated expression level but is context specific. By performing loss- and gain-of-function studies, both under in vitro and in vivo conditions, we demonstrate that MALAT1 facilitates cell proliferation, tumor progression and metastasis of triple-negative breast cancer (TNBC) cells despite having a comparatively lower expression level than ER or HER2-positive breast cancer cells. Furthermore, MALAT1 regulates the expression of several cancer metastasis-related genes, but displays molecular subtype specific correlations with such genes. Assessment of the prognostic significance of MALAT1 in human breast cancer (n=1992) revealed elevated MALAT1 expression was associated with decreased disease-specific survival in ER negative, lymph node negative patients of the HER2 and TNBC molecular subtypes. Multivariable analysis confirmed MALAT1 to have independent prognostic significance in the TNBC lymph node negative patient subset (HR=2.64, 95%CI 1.35 − 5.16, p=0.005). We propose that the functional significance of MALAT1 as a metastasis driver and its potential use as a prognostic marker is most promising for those patients diagnosed with ER negative, lymph node negative breast cancer who might otherwise mistakenly be stratified to have low recurrence risk.
Collapse
|
245
|
Chemotherapy-induced metastasis: mechanisms and translational opportunities. Clin Exp Metastasis 2018; 35:269-284. [PMID: 29307118 DOI: 10.1007/s10585-017-9870-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
Tumors often overcome the cytotoxic effects of chemotherapy through either acquired or environment-mediated drug resistance. In addition, signals from the microenvironment obfuscate the beneficial effects of chemotherapy and may facilitate progression and metastatic dissemination. Seminal mediators in chemotherapy-induced metastasis appear to be a wide range of hematopoietic, mesenchymal and immune progenitor cells, originating from the bone marrow. The actual purpose of these cells is to orchestrate the repair response to the cytotoxic damage of chemotherapy. However, these repair responses are exploited by tumor cells at every step of the metastatic cascade, ranging from tumor cell invasion, intravasation and hematogenous dissemination to extravasation and effective colonization at the metastatic site. A better understanding of the mechanistic underpinnings of chemotherapy-induced metastasis will allow us to better predict which patients are more likely to exhibit pro-metastatic responses to chemotherapy and will help develop new therapeutic strategies to neutralize chemotherapy-driven prometastatic changes.
Collapse
|
246
|
Sehl ME, Wicha MS. Modeling of Interactions between Cancer Stem Cells and their Microenvironment: Predicting Clinical Response. Methods Mol Biol 2018; 1711:333-349. [PMID: 29344897 PMCID: PMC6322404 DOI: 10.1007/978-1-4939-7493-1_16] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Mathematical models of cancer stem cells are useful in translational cancer research for facilitating the understanding of tumor growth dynamics and for predicting treatment response and resistance to combined targeted therapies. In this chapter, we describe appealing aspects of different methods used in mathematical oncology and discuss compelling questions in oncology that can be addressed with these modeling techniques. We describe a simplified version of a model of the breast cancer stem cell niche, illustrate the visualization of the model, and apply stochastic simulation to generate full distributions and average trajectories of cell type populations over time. We further discuss the advent of single-cell data in studying cancer stem cell heterogeneity and how these data can be integrated with modeling to advance understanding of the dynamics of invasive and proliferative populations during cancer progression and response to therapy.
Collapse
Affiliation(s)
- Mary E Sehl
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Department of Biomathematics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Max S Wicha
- Department of Internal Medicine, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
247
|
Socorro M, Criscimanna A, Riva P, Tandon M, Prasadan K, Guo P, Humar A, Husain SZ, Leach SD, Gittes GK, Esni F. Identification of Newly Committed Pancreatic Cells in the Adult Mouse Pancreas. Sci Rep 2017; 7:17539. [PMID: 29235528 PMCID: PMC5727523 DOI: 10.1038/s41598-017-17884-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/01/2017] [Indexed: 12/21/2022] Open
Abstract
Multipotent epithelial cells with high Aldehyde dehydrogenase activity have been previously reported to exist in the adult pancreas. However, whether they represent true progenitor cells remains controversial. In this study, we isolated and characterized cells with ALDH activity in the adult mouse or human pancreas during physiological conditions or injury. We found that cells with ALDH activity are abundant in the mouse pancreas during early postnatal growth, pregnancy, and in mouse models of pancreatitis and type 1 diabetes (T1D). Importantly, a similar population of cells is found abundantly in healthy children, or in patients with pancreatitis or T1D. We further demonstrate that cells with ALDH activity can commit to either endocrine or acinar lineages, and can be divided into four sub-populations based on CD90 and Ecadherin expression. Finally, our in vitro and in vivo studies show that the progeny of ALDH1+/CD90−/Ecad− cells residing in the adult mouse pancreas have the ability to initiate Pancreatic and duodenal homeobox (Pdx1) expression for the first time. In summary, we provide evidence for the existence of a sortable population of multipotent non-epithelial cells in the adult pancreas that can commit to the pancreatic lineage following proliferation and mesenchymal to epithelial transition (MET).
Collapse
Affiliation(s)
- Mairobys Socorro
- Department of Surgery, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA.,Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA
| | - Angela Criscimanna
- Department of Surgery, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA.,Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA
| | - Patricia Riva
- Department of Surgery, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA.,Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA
| | - Manuj Tandon
- Department of Surgery, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA.,Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA
| | - Krishna Prasadan
- Department of Surgery, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA.,Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA
| | - Ping Guo
- Department of Surgery, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA.,Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA.,Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, 1881 East Road, 3SCR6.4621, Houston, Texas, 77054, USA
| | - Abhinav Humar
- Department of Surgery, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA
| | - Sohail Z Husain
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, 15244, USA
| | - Steven D Leach
- Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - George K Gittes
- Department of Surgery, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA.,Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA
| | - Farzad Esni
- Department of Surgery, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA. .,Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, One Children's Drive, 4401 Penn Avenue, Rangos Research Center, Pittsburgh, PA, 15244, USA. .,Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, 15244, USA. .,Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, 15244, USA. .,University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15123, USA.
| |
Collapse
|
248
|
Jiang X, Wang J, Chen X, Hong Y, Wu T, Chen X, Xia J, Cheng B. Elevated autocrine chemokine ligand 18 expression promotes oral cancer cell growth and invasion via Akt activation. Oncotarget 2017; 7:16262-72. [PMID: 26919103 PMCID: PMC4941312 DOI: 10.18632/oncotarget.7585] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/05/2016] [Indexed: 11/29/2022] Open
Abstract
Chemokine (C-C motif) ligand 18 (CCL18) has been implicated in the pathogenesis and progression of various cancers; however, in oral squamous cell carcinoma (OSCC), the role of CCL18 is unknown. In this study, we found that CCL18 was overexpressed in primary OSCC tissues and was associated with an advanced clinical stage. CCL18 was found in both the cytoplasm and cell membrane of OSCC cells and was predominantly produced by cancer epithelial cells, as opposed to tumor-infiltrating macrophages. In vitro studies indicated that the effects of endogenous CCL18 on OSCC cell growth, migration, and invasion could be blocked by treatment with a neutralizing anti-CCL18 antibody or CCL18 knockdown, while exogenous recombinant CCL18 (rCCL18) rescued those effects. Akt was activated in rCCL18-treated OSCC cells, while LY294002, a pan-PI3K inhibitor, abolished both endogenous and exogenous CCL18-induced OSCC cell invasion. In vivo, LY294002 treatment attenuated rCCL18-induced OSCC cell growth. Our results indicate that CCL18 acts in an autocrine manner via Akt activation to stimulate OSCC cell growth and invasion during OSCC progression. They also provide a potential therapeutic target for the treatment of oral cancer.
Collapse
Affiliation(s)
- Xiao Jiang
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China.,Guangdong Provincial Stomatological Hospital, Guangzhou, Guangdong 510280, China
| | - Juan Wang
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Xijuan Chen
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Yun Hong
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Tong Wu
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Xiaobing Chen
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Juan Xia
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Bin Cheng
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| |
Collapse
|
249
|
Prieto-Vila M, Takahashi RU, Usuba W, Kohama I, Ochiya T. Drug Resistance Driven by Cancer Stem Cells and Their Niche. Int J Mol Sci 2017; 18:ijms18122574. [PMID: 29194401 PMCID: PMC5751177 DOI: 10.3390/ijms18122574] [Citation(s) in RCA: 336] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/11/2022] Open
Abstract
Drug resistance represents one of the greatest challenges in cancer treatment. Cancer stem cells (CSCs), a subset of cells within the tumor with the potential for self-renewal, differentiation and tumorigenicity, are thought to be the major cause of cancer therapy failure due to their considerable chemo- and radioresistance, resulting in tumor recurrence and eventually metastasis. CSCs are situated in a specialized microenvironment termed the niche, mainly composed of fibroblasts and endothelial, mesenchymal and immune cells, which also play pivotal roles in drug resistance. These neighboring cells promote the molecular signaling pathways required for CSC maintenance and survival and also trigger endogenous drug resistance in CSCs. In addition, tumor niche components such as the extracellular matrix also physically shelter CSCs from therapeutic agents. Interestingly, CSCs contribute directly to the niche in a bilateral feedback loop manner. Here, we review the recent advances in the study of CSCs, the niche and especially their collective contribution to resistance, since increasingly studies suggest that this interaction should be considered as a target for therapeutic strategies.
Collapse
Affiliation(s)
- Marta Prieto-Vila
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045, Japan.
| | - Ryou-U Takahashi
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045, Japan.
| | - Wataru Usuba
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045, Japan.
| | - Isaku Kohama
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045, Japan.
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045, Japan.
| |
Collapse
|
250
|
Cortés M, Sanchez-Moral L, de Barrios O, Fernández-Aceñero MJ, Martínez-Campanario MC, Esteve-Codina A, Darling DS, Győrffy B, Lawrence T, Dean DC, Postigo A. Tumor-associated macrophages (TAMs) depend on ZEB1 for their cancer-promoting roles. EMBO J 2017; 36:3336-3355. [PMID: 29038174 PMCID: PMC5686549 DOI: 10.15252/embj.201797345] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/18/2017] [Accepted: 09/20/2017] [Indexed: 01/06/2023] Open
Abstract
Accumulation of tumor-associated macrophages (TAMs) associates with malignant progression in cancer. However, the mechanisms that drive the pro-tumor functions of TAMs are not fully understood. ZEB1 is best known for driving an epithelial-to-mesenchymal transition (EMT) in cancer cells to promote tumor progression. However, a role for ZEB1 in macrophages and TAMs has not been studied. Here we describe that TAMs require ZEB1 for their tumor-promoting and chemotherapy resistance functions in a mouse model of ovarian cancer. Only TAMs that expressed full levels of Zeb1 accelerated tumor growth. Mechanistically, ZEB1 expression in TAMs induced their polarization toward an F4/80low pro-tumor phenotype, including direct activation of Ccr2 In turn, expression of ZEB1 by TAMs induced Ccl2, Cd74, and a mesenchymal/stem-like phenotype in cancer cells. In human ovarian carcinomas, TAM infiltration and CCR2 expression correlated with ZEB1 in tumor cells, where along with CCL2 and CD74 determined poorer prognosis. Importantly, ZEB1 in TAMs was a factor of poorer survival in human ovarian carcinomas. These data establish ZEB1 as a key factor in the tumor microenvironment and for maintaining TAMs' tumor-promoting functions.
Collapse
Affiliation(s)
- Marlies Cortés
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, Barcelona, Spain
| | - Lidia Sanchez-Moral
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, Barcelona, Spain
| | - Oriol de Barrios
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, Barcelona, Spain
| | | | - M C Martínez-Campanario
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science & Technology, and Universitat Pompeu Fabra, Barcelona, Spain
| | - Douglas S Darling
- Department of Oral Immunology, and Center for Genetics and Molecular Medicine, University of Louisville, Louisville, KY, USA
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, and Semmelweis University 2 Department of Pediatrics, Budapest, Hungary
| | - Toby Lawrence
- Centre d'Immunologie de Marseille-Luminy, INSERM U1104 and CNRS MR7280, Marseille, France
| | - Douglas C Dean
- Department of Ophthalmology and Visual Sciences and Birth Defects Center, University of Louisville, Louisville, KY, USA
- Molecular Targets Program, James G. Brown Cancer Center, Louisville, KY, USA
| | - Antonio Postigo
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, Barcelona, Spain
- Molecular Targets Program, James G. Brown Cancer Center, Louisville, KY, USA
- ICREA, Barcelona, Spain
| |
Collapse
|