1
|
McMinn PH, Ahmed A, Huttenlocher A, Beebe DJ, Kerr SC. The lymphatic endothelium-derived follistatin: activin A axis regulates neutrophil motility in response to Pseudomonas aeruginosa. Integr Biol (Camb) 2023; 15:zyad003. [PMID: 36781971 PMCID: PMC10101905 DOI: 10.1093/intbio/zyad003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/02/2022] [Accepted: 01/23/2023] [Indexed: 02/15/2023]
Abstract
The lymphatic system plays an active role during infection, however the role of lymphatic-neutrophil interactions in host-defense responses is not well understood. During infection with pathogens such as Pseudomonas aeruginosa, Staphylococcus aureus and Yersinia pestis, neutrophils traffic from sites of infection through the lymphatic vasculature, to draining lymph nodes to interact with resident lymphocytes. This process is poorly understood, in part, due to the lack of in vitro models of the lymphatic system. Here we use a 3D microscale lymphatic vessel model to examine neutrophil-lymphatic cell interactions during host defense responses to pathogens. In previous work, we have shown that follistatin is secreted at high concentrations by lymphatic endothelial cells during inflammation. Follistatin inhibits activin A, a member of the TGF-β superfamily, and, together, these molecules form a signaling pathway that plays a role in regulating both innate and adaptive immune responses. Although follistatin and activin A are constitutively produced in the pituitary, gonads and skin, their major source in the serum and their effects on neutrophils are poorly understood. Here we report a microfluidic model that includes both blood and lymphatic endothelial vessels, and neutrophils to investigate neutrophil-lymphatic trafficking during infection with P. aeruginosa. We found that lymphatic endothelial cells produce secreted factors that increase neutrophil migration toward P. aeruginosa, and are a significant source of both follistatin and activin A during Pseudomonas infection. We determined that follistatin produced by lymphatic endothelial cells inhibits activin A, resulting in increased neutrophil migration. These data suggest that the follistatin:activin A ratio influences neutrophil trafficking during infection with higher ratios increasing neutrophil migration.
Collapse
Affiliation(s)
- Patrick H McMinn
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Adeel Ahmed
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Beebe
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Sheena C Kerr
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
2
|
Inactivating the Uninhibited: The Tale of Activins and Inhibins in Pulmonary Arterial Hypertension. Int J Mol Sci 2023; 24:ijms24043332. [PMID: 36834742 PMCID: PMC9963072 DOI: 10.3390/ijms24043332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Advances in technology and biomedical knowledge have led to the effective diagnosis and treatment of an increasing number of rare diseases. Pulmonary arterial hypertension (PAH) is a rare disorder of the pulmonary vasculature that is associated with high mortality and morbidity rates. Although significant progress has been made in understanding PAH and its diagnosis and treatment, numerous unanswered questions remain regarding pulmonary vascular remodeling, a major factor contributing to the increase in pulmonary arterial pressure. Here, we discuss the role of activins and inhibins, both of which belong to the TGF-β superfamily, in PAH development. We examine how these relate to signaling pathways implicated in PAH pathogenesis. Furthermore, we discuss how activin/inhibin-targeting drugs, particularly sotatercep, affect pathophysiology, as these target the afore-mentioned specific pathway. We highlight activin/inhibin signaling as a critical mediator of PAH development that is to be targeted for therapeutic gain, potentially improving patient outcomes in the future.
Collapse
|
3
|
Ganjoo S, Puebla-Osorio N, Nanez S, Hsu E, Voss T, Barsoumian H, Duong LK, Welsh JW, Cortez MA. Bone morphogenetic proteins, activins, and growth and differentiation factors in tumor immunology and immunotherapy resistance. Front Immunol 2022; 13:1033642. [PMID: 36353620 PMCID: PMC9638036 DOI: 10.3389/fimmu.2022.1033642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2024] Open
Abstract
The TGF-β superfamily is a group of secreted polypeptides with key roles in exerting and regulating a variety of physiologic effects, especially those related to cell signaling, growth, development, and differentiation. Although its central member, TGF-β, has been extensively reviewed, other members of the family-namely bone morphogenetic proteins (BMPs), activins, and growth and differentiation factors (GDFs)-have not been as thoroughly investigated. Moreover, although the specific roles of TGF-β signaling in cancer immunology and immunotherapy resistance have been extensively reported, little is known of the roles of BMPs, activins, and GDFs in these domains. This review focuses on how these superfamily members influence key immune cells in cancer progression and resistance to treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
4
|
Li F, Long Y, Yu X, Tong Y, Gong L. Different Immunoregulation Roles of Activin A Compared With TGF-β. Front Immunol 2022; 13:921366. [PMID: 35774793 PMCID: PMC9237220 DOI: 10.3389/fimmu.2022.921366] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Activin A, a critical member of the transforming growth factor-β (TGF-β) superfamily, is a pluripotent factor involved in allergies, autoimmune diseases, cancers and other diseases with immune disorder. Similar to its family member, TGF-β, activin A also transmits signals through SMAD2/SMAD3, however, they bind to distinct receptors. Recent studies have uncovered that activin A plays a pivotal role in both innate and adaptive immune systems. Here we mainly focus its effects on activation, differentiation, proliferation and function of cells which are indispensable in the immune system and meanwhile make some comparisons with those of TGF-β.
Collapse
Affiliation(s)
- Fanglin Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiru Long
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaolu Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yongliang Tong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Likun Gong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- *Correspondence: Likun Gong,
| |
Collapse
|
5
|
Qian H, Shan Y, Gong R, Lin D, Zhang M, Wang C, Wang L. Fibroblasts in Scar Formation: Biology and Clinical Translation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4586569. [PMID: 35602101 PMCID: PMC9119755 DOI: 10.1155/2022/4586569] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 11/17/2022]
Abstract
Scarring, which develops due to fibroblast activation and excessive extracellular matrix deposition, can cause physical, psychological, and cosmetic problems. Fibroblasts are the main type of connective tissue cells and play important roles in wound healing. However, the underlying mechanisms of fibroblast in reaching scarless wound healing require more exploration. Herein, we systematically reviewed how fibroblasts behave in response to skin injuries, as well as their functions in regeneration and scar formation. Several biocompatible materials, including hydrogels and nanoparticles, were also suggested. Moreover, factors that concern transformation from fibroblasts into cancer-associated fibroblasts are mentioned due to a tight association between scar formation and primary skin cancers. These findings will help us better understand skin fibrotic pathogenesis, as well as provide potential targets for scarless wound healing therapies.
Collapse
Affiliation(s)
- Huan Qian
- Department of Plastic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yihan Shan
- Wenzhou Medical University, Wenzhou, China
| | | | - Danfeng Lin
- Department of Breast Surgery, The First Affifiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengwen Zhang
- Department of Plastic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chen Wang
- Department of Plastic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lu Wang
- Starbody plastic surgery Clinic, Hangzhou, China
| |
Collapse
|
6
|
Gutiérrez-Seijo A, García-Martínez E, Barrio-Alonso C, Parra-Blanco V, Avilés-Izquierdo JA, Sánchez-Mateos P, Samaniego R. Activin A Sustains the Metastatic Phenotype of Tumor-Associated Macrophages and Is a Prognostic Marker in Human Cutaneous Melanoma. J Invest Dermatol 2022; 142:653-661.e2. [PMID: 34499901 DOI: 10.1016/j.jid.2021.07.179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/10/2021] [Accepted: 07/26/2021] [Indexed: 12/29/2022]
Abstract
Tumor cells attract and dynamically interact with monocytes/macrophages to subvert their differentiation into tumor-associated macrophages (TAMs), which mainly promote immune suppression and neoplastic progression, but the pathways and microenvironmental cues governing their protumoral deviation are not completely understood. To identify the molecular pathways responsible for TAM differentiation, we screened the biomarkers secreted during melanoma‒macrophage interactions using Quantibody microarrays and RNA sequencing of macrophages. We found that activin A, a member of the transforming GF family, plays an instrumental role in the cross-talk between melanoma cells and monocytes/macrophages, which results in the upregulation of distinct tumor-sustaining genes and the achievement of proinvasive and immunosuppressive functions of TAMs. Blockade of activin reduces the upregulation of part of these genes and prevents the acquisition of protumoral functions, facilitating human melanoma rejection by transferred human lymphocytes in a xenograft mouse model. Remarkably, screening of two independent cutaneous primary melanoma collections showed that activin A is enriched in TAMs and melanoma cells from patients with worse outcomes and constitutes a new and independent prognostic marker. Thus, we identify activin A as a key intermediary in the protumoral and immunosuppressive functions of TAMs, with significant potential as a disease biomarker as well as an immunotherapeutic target.
Collapse
Affiliation(s)
- Alba Gutiérrez-Seijo
- Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Laboratorio de Inmuno-oncología, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Elena García-Martínez
- Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Laboratorio de Inmuno-oncología, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Celia Barrio-Alonso
- Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Laboratorio de Inmuno-oncología, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Verónica Parra-Blanco
- Servicio de Anatomía Patológica, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | - Paloma Sánchez-Mateos
- Laboratorio de Inmuno-oncología, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Rafael Samaniego
- Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.
| |
Collapse
|
7
|
Barany N, Rozsas A, Megyesfalvi Z, Grusch M, Hegedus B, Lang C, Boettiger K, Schwendenwein A, Tisza A, Renyi-Vamos F, Schelch K, Hoetzenecker K, Hoda MA, Paku S, Laszlo V, Dome B. Clinical relevance of circulating activin A and follistatin in small cell lung cancer. Lung Cancer 2021; 161:128-135. [PMID: 34583221 DOI: 10.1016/j.lungcan.2021.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Circulating levels of activin A (ActA) and follistatin (FST) have been investigated in various disorders including malignancies. However, to date, their diagnostic and prognostic relevance is largely unknown in small cell lung cancer (SCLC). Our aim was to evaluate circulating ActA and FST levels as potential biomarkers in this devastating disease. METHODS Seventy-nine Caucasian SCLC patients and 67 age- and sex-matched healthy volunteers were included in this study. Circulating ActA and FST concentrations were measured by ELISA and correlated with clinicopathological parameters and long-term outcomes. RESULTS Plasma ActA and FST concentrations were significantly elevated in SCLC patients when compared to healthy volunteers (p < 0.0001). Furthermore, extensive-stage SCLC patients had significantly higher circulating ActA levels than those with limited-stage disease (p = 0.0179). Circulating FST concentration was not associated with disease stage (p = 0.6859). Notably, patients with high (≥548.8 pg/ml) plasma ActA concentration exhibited significantly worse median overall survival (OS) compared to those with low (<548.8 pg/ml) ActA levels (p = 0.0009). Moreover, Cox regression analysis adjusted for clinicopathological parameters revealed that high ActA concentration is an independent predictor of shorter OS (HR: 1.932; p = 0.023). No significant differences in OS have been observed with regards to plasma FST levels (p = 0.1218). CONCLUSION Blood ActA levels are elevated and correlate with disease stage in SCLC patients. Measurement of circulating ActA levels might help in the estimation of prognosis in patients with SCLC.
Collapse
Affiliation(s)
- Nandor Barany
- National Koranyi Institute of Pulmonology, Budapest, Hungary; 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Anita Rozsas
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Zsolt Megyesfalvi
- National Koranyi Institute of Pulmonology, Budapest, Hungary; Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria; Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
| | - Michael Grusch
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Balazs Hegedus
- Department of Thoracic Surgery, Ruhrlandklinik, University Clinic Essen, Essen, Germany
| | - Christian Lang
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Kristiina Boettiger
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Anna Schwendenwein
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Anna Tisza
- National Koranyi Institute of Pulmonology, Budapest, Hungary; 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ferenc Renyi-Vamos
- National Koranyi Institute of Pulmonology, Budapest, Hungary; Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
| | - Karin Schelch
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria; Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Sandor Paku
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Viktoria Laszlo
- National Koranyi Institute of Pulmonology, Budapest, Hungary; Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.
| | - Balazs Dome
- National Koranyi Institute of Pulmonology, Budapest, Hungary; Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria; Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary.
| |
Collapse
|
8
|
Rezzola S, Sigmund EC, Halin C, Ronca R. The lymphatic vasculature: An active and dynamic player in cancer progression. Med Res Rev 2021; 42:576-614. [PMID: 34486138 PMCID: PMC9291933 DOI: 10.1002/med.21855] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/29/2021] [Accepted: 08/26/2021] [Indexed: 12/16/2022]
Abstract
The lymphatic vasculature has been widely described and explored for its key functions in fluid homeostasis and in the organization and modulation of the immune response. Besides transporting immune cells, lymphatic vessels play relevant roles in tumor growth and tumor cell dissemination. Cancer cells that have invaded into afferent lymphatics are propagated to tumor‐draining lymph nodes (LNs), which represent an important hub for metastatic cell arrest and growth, immune modulation, and secondary dissemination to distant sites. In recent years many studies have reported new mechanisms by which the lymphatic vasculature affects cancer progression, ranging from induction of lymphangiogenesis to metastatic niche preconditioning or immune modulation. In this review, we provide an up‐to‐date description of lymphatic organization and function in peripheral tissues and in LNs and the changes induced to this system by tumor growth and progression. We will specifically focus on the reported interactions that occur between tumor cells and lymphatic endothelial cells (LECs), as well as on interactions between immune cells and LECs, both in the tumor microenvironment and in tumor‐draining LNs. Moreover, the most recent prognostic and therapeutic implications of lymphatics in cancer will be reported and discussed in light of the new immune‐modulatory roles that have been ascribed to LECs.
Collapse
Affiliation(s)
- Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Elena C Sigmund
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
9
|
Wound Repair, Scar Formation, and Cancer: Converging on Activin. Trends Mol Med 2020; 26:1107-1117. [PMID: 32878730 DOI: 10.1016/j.molmed.2020.07.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
Wound repair is a highly regulated process that requires the interaction of various cell types. It has been shown that cancers use the mechanisms of wound healing to promote their own growth. Therefore, it is of importance to identify common regulators of wound repair and tumor formation and to unravel their functions and mechanisms of action. An exciting example is activin, which acts on multiple cell types in wounds and tumors, thereby promoting healing, but also scar formation and tumorigenesis. Here, we summarize current knowledge on the role of activin in these processes and highlight the therapeutic potential of activin or activin antagonists for the treatment of impaired healing or excessive scarring and cancer, respectively.
Collapse
|
10
|
Ries A, Schelch K, Falch D, Pany L, Hoda MA, Grusch M. Activin A: an emerging target for improving cancer treatment? Expert Opin Ther Targets 2020; 24:985-996. [PMID: 32700590 DOI: 10.1080/14728222.2020.1799350] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Activin A is involved in the regulation of a surprisingly broad number of processes that are relevant for cancer development and treatment; it is implicated in cell autonomous functions and multiple regulatory functions in the tumor microenvironment. AREAS COVERED This article summarizes the current knowledge about activin A in cell growth and death, migration and metastasis, angiogenesis, stemness and drug resistance, regulation of antitumor immunity, and cancer cachexia. We explore the role of activin A as a biomarker and discuss strategies for using it as target for cancer therapy. Literature retrieved from Medline until 25 June 2020 was considered. EXPERT OPINION While many functions of activin A were investigated in preclinical models, there is currently limited experience from clinical trials. Activin A has growth- and migration-promoting effects, contributes to immune evasion and cachexia and is associated with shorter survival in several cancer types. Targeting activin A could offer the chance to simultaneously limit tumor growth and spreading, improve drug response, boost antitumor immune responses and improve cancer-associated or treatment-associated cachexia, bone loss, and anemia. Nevertheless, defining which patients have the highest likelihood of benefiting from these effects is challenging and will require further work.
Collapse
Affiliation(s)
- Alexander Ries
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna , Vienna, Austria
| | - Karin Schelch
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna , Vienna, Austria
| | - David Falch
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna , Vienna, Austria
| | - Laura Pany
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna , Vienna, Austria
| | - Mir Alireza Hoda
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna , Vienna, Austria
| | - Michael Grusch
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna , Vienna, Austria
| |
Collapse
|
11
|
Cangkrama M, Wietecha M, Mathis N, Okumura R, Ferrarese L, Al‐Nuaimi D, Antsiferova M, Dummer R, Innocenti M, Werner S. A paracrine activin A-mDia2 axis promotes squamous carcinogenesis via fibroblast reprogramming. EMBO Mol Med 2020; 12:e11466. [PMID: 32150356 PMCID: PMC7136968 DOI: 10.15252/emmm.201911466] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 12/18/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are key regulators of tumorigenesis and promising targets for next-generation therapies. We discovered that cancer cell-derived activin A reprograms fibroblasts into pro-tumorigenic CAFs. Mechanistically, this occurs via Smad2-mediated transcriptional regulation of the formin mDia2, which directly promotes filopodia formation and cell migration. mDia2 also induces expression of CAF marker genes through prevention of p53 nuclear accumulation, resulting in the production of a pro-tumorigenic matrisome and secretome. The translational relevance of this finding is reflected by activin A overexpression in tumor cells and of mDia2 in the stroma of skin cancer and other malignancies and the correlation of high activin A/mDia2 levels with poor patient survival. Blockade of this signaling axis using inhibitors of activin, activin receptors, or mDia2 suppressed cancer cell malignancy and squamous carcinogenesis in 3D organotypic cultures, ex vivo, and in vivo, providing a rationale for pharmacological inhibition of activin A-mDia2 signaling in stratified cancer patients.
Collapse
Affiliation(s)
- Michael Cangkrama
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Mateusz Wietecha
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Nicolas Mathis
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Rin Okumura
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Luca Ferrarese
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Dunja Al‐Nuaimi
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| | - Maria Antsiferova
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
- Present address:
Roche Glycart AGSchlierenSwitzerland
| | - Reinhard Dummer
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
| | - Metello Innocenti
- Heidelberg University Biochemistry Center (BZH)Heidelberg UniversityHeidelbergGermany
| | - Sabine Werner
- Department of BiologyInstitute of Molecular Health SciencesETH ZurichZurichSwitzerland
| |
Collapse
|
12
|
Neuditschko B, Janker L, Niederstaetter L, Brunmair J, Krivanek K, Izraely S, Sagi-Assif O, Meshel T, Keppler BK, Del Favero G, Witz IP, Gerner C. The Challenge of Classifying Metastatic Cell Properties by Molecular Profiling Exemplified with Cutaneous Melanoma Cells and Their Cerebral Metastasis from Patient Derived Mouse Xenografts. Mol Cell Proteomics 2020; 19:478-489. [PMID: 31892524 PMCID: PMC7050108 DOI: 10.1074/mcp.ra119.001886] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Indexed: 12/20/2022] Open
Abstract
The prediction of metastatic properties from molecular analyses still poses a major challenge. Here we aimed at the classification of metastasis-related cell properties by proteome profiling making use of cutaneous and brain-metastasizing variants from single melanomas sharing the same genetic ancestry. Previous experiments demonstrated that cultured cells derived from these xenografted variants maintain a stable phenotype associated with a differential metastatic behavior: The brain metastasizing variants produce more spontaneous micro-metastases than the corresponding cutaneous variants. Four corresponding pairs of cutaneous and metastatic cells were obtained from four individual patients, resulting in eight cell-lines presently investigated. Label free proteome profiling revealed significant differences between corresponding pairs of cutaneous and cerebellar metastases from the same patient. Indeed, each brain metastasizing variant expressed several apparently metastasis-associated proteomic alterations as compared with the corresponding cutaneous variant. Among the differentially expressed proteins we identified cell adhesion molecules, immune regulators, epithelial to mesenchymal transition markers, stem cell markers, redox regulators and cytokines. Similar results were observed regarding eicosanoids, considered relevant for metastasis, such as PGE2 and 12-HETE. Multiparametric morphological analysis of cells also revealed no characteristic alterations associated with the cutaneous and brain metastasis variants. However, no correct classification regarding metastatic potential was yet possible with the present data. We thus concluded that molecular profiling is able to classify cells according to known functional categories but is not yet able to predict relevant cell properties emerging from networks consisting of many interconnected molecules. The presently observed broad diversity of molecular patterns, irrespective of restricting to one tumor type and two main classes of metastasis, highlights the important need to develop meta-analysis strategies to predict cell properties from molecular profiling data. Such base knowledge will greatly support future individualized precision medicine approaches.
Collapse
Affiliation(s)
- Benjamin Neuditschko
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna; Department of Inorganic Chemistry, Faculty of Chemistry, University of Vienna
| | - Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna
| | | | - Julia Brunmair
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna
| | - Katharina Krivanek
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna; Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna
| | - Sivan Izraely
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University
| | - Orit Sagi-Assif
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University
| | - Tsipi Meshel
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University
| | - Bernhard K Keppler
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Vienna
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna; Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna
| | - Isaac P Witz
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna; Joint Metabolome Facility, Faculty of Chemistry, University of Vienna.
| |
Collapse
|
13
|
Gong MM, Lugo-Cintron KM, White BR, Kerr SC, Harari PM, Beebe DJ. Human organotypic lymphatic vessel model elucidates microenvironment-dependent signaling and barrier function. Biomaterials 2019; 214:119225. [PMID: 31154151 DOI: 10.1016/j.biomaterials.2019.119225] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 12/12/2022]
Abstract
The lymphatic system is an active player in the pathogenesis of several human diseases, including lymphedema and cancer. Relevant models are needed to advance our understanding of lymphatic biology in disease progression to improve therapy and patient outcomes. Currently, there are few 3D in vitro lymphatic models that can recapitulate the physiological structure, function, and interactions of lymphatic vessels in normal and diseased microenvironments. Here, we developed a 3D microscale lymphatic vessel (μLYMPH) system for generating human lymphatic vessels with physiological tubular structure and function. Consistent with characteristics of lymphatic vessels in vivo, the endothelium of cultured vessels was leaky with an average permeability of 1.38 × 10-5 ± 0.29 × 10-5 cm/s as compared to 0.68 × 10-5 ± 0.13 × 10-5 cm/s for blood vessels. This leakiness also resulted in higher uptake of solute by the lymphatic vessels under interstitial flow, demonstrating recapitulation of their natural draining function. The vessels secreted appropriate growth factors and inflammatory mediators. Our system identified the follistatin/activin axis as a novel pathway in lymphatic vessel maintenance and inflammation. Moreover, the μLYMPH system provided a platform for examining crosstalk between lymphatic vessels and tumor microenvironmental components, such as breast cancer-associated fibroblasts (CAFs). In co-culture with CAFs, vessel barrier function was significantly impaired by CAF-secreted IL-6, a possible pro-metastatic mechanism of lymphatic metastasis. Targeted blocking of the IL-6/IL-6R signaling pathway with an IL-6 neutralizing antibody fully rescued the vessels, demonstrating the potential of our system for screening therapeutic targets. These results collectively demonstrate the μLYMPH system as a powerful model for advancing lymphatic biology in health and disease.
Collapse
Affiliation(s)
- Max M Gong
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1451 Engineering Dr., Madison, WI, 53706, USA
| | - Karina M Lugo-Cintron
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1451 Engineering Dr., Madison, WI, 53706, USA
| | - Bridget R White
- Department of Engineering Physics, University of Wisconsin-Platteville, 1 University Plaza, Platteville, WI, 53818, USA
| | - Sheena C Kerr
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1451 Engineering Dr., Madison, WI, 53706, USA
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin-Madison, 600 Highland Ave., Madison, WI, 53792, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1451 Engineering Dr., Madison, WI, 53706, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 1685 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
14
|
Böhm I, Gehrke S, Kleb B, Hungerbühler M, Müller R, Klose KJ, Alfke H. Monitoring of tumor burden in vivo by optical imaging in a xenograft SCID mouse model: evaluation of two fluorescent proteins of the GFP-superfamily. Acta Radiol 2019; 60:315-326. [PMID: 29890843 DOI: 10.1177/0284185118780896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Mouse models of human-malignant-melanoma (MM) are important tools to study tumor dynamics. The enhanced green fluorescent protein (EGFP) is widely used in molecular imaging approaches, together with optical scanners, and fluorescence imaging. PURPOSE Currently, there are no data available as to whether other fluorescent proteins are more suitable. The goal of this preclinical study was to analyze two fluorescent proteins of the GFP superfamily under real-time in vivo conditions using fluorescence reflectance imaging (FRI). MATERIAL AND METHODS The human melanoma cell line MeWo was stable transfected with one plasmid: pEGFP-C1 or pDsRed1-N1. We investigated two severe combined immunodeficiency (SCID)-mice groups: A (solid xenografts) and B (xenografts as metastases). After three weeks, the animals were weekly imaged by FRI. Afterwards the mice were euthanized and metastases were imaged in situ: to quantify the cutis-dependent reduction of emitted light, we compared signal intensities obtained by metastases in vivo with signal intensities obtained by in situ liver parenchyma preparations. RESULTS More than 90% of cells were stable transfected. EGFP-/DsRed-xenograft tumors had identical growth kinetics. In vivo the emitted light by DsRed tumors/metastases was much brighter than by EGFP. DsRed metastases were earlier (3 vs. 5 weeks) and much more sensitive detectable than EGFP metastases. Cutis-dependent reduction of emitted light was greater in EGFP than in DsRed mice (tenfold). Autofluorescence of DsRed was lower than of EGFP. CONCLUSION We established an in vivo xenograft mouse model (DsRed-MeWo) that is reliable, reproducible, and superior to the EGFP model as a preclinical tool to study innovative therapies by FRI under real-time in vivo conditions.
Collapse
Affiliation(s)
- Ingrid Böhm
- Department of Diagnostic, Interventional, and Pediatric Radiology, Inselspital, University of Bern, Bern, Switzerland
- Radiology Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Stephan Gehrke
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Beate Kleb
- Department of Experimental Ophthalmology, Philipps University of Marburg, Marburg, Germany
| | - Martin Hungerbühler
- Department of Diagnostic, Interventional, and Pediatric Radiology, Inselspital, University of Bern, Bern, Switzerland
- Radiology Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Rolf Müller
- Institute of Molecular Tumor Biology and Cancer Gene Therapy (IMT), Philipps University of Marburg, Marburg, Germany
| | - Klaus J Klose
- Deans Office, Faculty of Medicine, Philipps University of Marburg, Marburg, Germany
| | - Heiko Alfke
- Department of Diagnostic Radiology and Interventional Radiology, Klinikum Lüdenscheid, Lüdenscheid, Germany
| |
Collapse
|
15
|
Hoda MA, Rozsas A, Lang E, Klikovits T, Lohinai Z, Torok S, Berta J, Bendek M, Berger W, Hegedus B, Klepetko W, Renyi-Vamos F, Grusch M, Dome B, Laszlo V. High circulating activin A level is associated with tumor progression and predicts poor prognosis in lung adenocarcinoma. Oncotarget 2017; 7:13388-99. [PMID: 26950277 PMCID: PMC4924649 DOI: 10.18632/oncotarget.7796] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/09/2016] [Indexed: 12/22/2022] Open
Abstract
Activin A (ActA)/follistatin (FST) signaling has been shown to be deregulated in different tumor types including lung adenocarcinoma (LADC). Here, we report that serum ActA protein levels are significantly elevated in LADC patients (n=64) as compared to controls (n=46, p=0.015). ActA levels also correlated with more advanced disease stage (p<0.0001) and T (p=0.0035) and N (p=0.0002) factors. M1 patients had significantly higher ActA levels than M0 patients (p<0.001). High serum ActA level was associated with poor overall survival (p<0.0001) and was confirmed as an independent prognostic factor (p=0.004). Serum FST levels were increased only in female LADC patients (vs. female controls, p=0.031). Two out of five LADC cell lines secreted biologically active ActA, while FST was produced in all of them. Transcripts of both type I and II ActA receptors were detected in all five LADC cell lines. In conclusion, our study does not only suggest that measuring blood ActA levels in LADC patients might improve the prediction of prognosis, but also indicates that this parameter might be a novel non-invasive biomarker for identifying LADC patients with organ metastases.
Collapse
Affiliation(s)
- Mir Alireza Hoda
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria.,Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Anita Rozsas
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria.,National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Elisabeth Lang
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Thomas Klikovits
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Zoltan Lohinai
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Szilvia Torok
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Judit Berta
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Matyas Bendek
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Walter Berger
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Balazs Hegedus
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria.,MTA-SE Molecular Oncology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Walter Klepetko
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Ferenc Renyi-Vamos
- Department of Thoracic Surgery, National Institute of Oncology and Semmelweis University, Budapest, Hungary
| | - Michael Grusch
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Balazs Dome
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria.,National Koranyi Institute of Pulmonology, Budapest, Hungary.,Department of Thoracic Surgery, National Institute of Oncology and Semmelweis University, Budapest, Hungary.,Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Viktoria Laszlo
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Donovan P, Dubey OA, Kallioinen S, Rogers KW, Muehlethaler K, Müller P, Rimoldi D, Constam DB. Paracrine Activin-A Signaling Promotes Melanoma Growth and Metastasis through Immune Evasion. J Invest Dermatol 2017; 137:2578-2587. [DOI: 10.1016/j.jid.2017.07.845] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 06/26/2017] [Accepted: 07/25/2017] [Indexed: 01/19/2023]
|
17
|
Jiang L, Si T, Yu M, Zeng X, Morse HC, Lu Y, Ouyang G, Zhou JX. The tumor suppressive role of inhibin βA in diffuse large B-cell lymphoma. Leuk Lymphoma 2017; 59:1202-1212. [PMID: 28877610 DOI: 10.1080/10428194.2017.1372574] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INHBA (inhibin βA), a subunit of a ligand of the transforming growth factor-β superfamily, is known to play diverse roles in various solid tumors. However, its role in hematologic malignancies remains unexplored. Here, we investigated the function of INHBA in diffuse large B-cell lymphoma (DLBCL). Both mRNA and protein levels of INHBA were significantly downregulated in primary DLBCL tissues, irrespective of germinal center B-cell-like (GCB) or non-GCB subtype, compared to those in benign tonsils. The low level of INHBA in patients with de novo DLBCL was correlated with reduced overall and progression-free survival. Ectopic expression of INHBA in DLBCL cell lines (OCI-Ly01 and SUDHL-10) resulted in reduced cell proliferation, increased spontaneous apoptosis and arrested cell cycle in vitro and suppressed xenograft tumor growth in vivo. Moreover, INHBA enhanced the chemosensitivity of DLBCL cells. Thus, our results provide novel evidence that INHBA functions as a tumor suppressor in DLBCL.
Collapse
Affiliation(s)
- Lei Jiang
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China
| | - Ting Si
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China.,b Department of Hematology , Affiliated Yinzhou Hospital, Ningbo University School of Medicine , Ningbo , China
| | - Mei Yu
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China
| | - Xinli Zeng
- c Department of ENT , Ningbo Second People's Hospital , Ningbo , China
| | - Herbert C Morse
- d Laboratory of Immunopathology , National Institute of Allergy and Infectious Diseases, National Institutes of Health , Rockville , MD , USA
| | - Ying Lu
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China.,b Department of Hematology , Affiliated Yinzhou Hospital, Ningbo University School of Medicine , Ningbo , China
| | - Guifang Ouyang
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China.,e Department of Hematology , Ningbo First People's Hospital , Ningbo , China
| | - Jeff X Zhou
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China
| |
Collapse
|
18
|
Arcidiacono P, Ragonese F, Stabile A, Pistilli A, Kuligina E, Rende M, Bottoni U, Calvieri S, Crisanti A, Spaccapelo R. Antitumor activity and expression profiles of genes induced by sulforaphane in human melanoma cells. Eur J Nutr 2017; 57:2547-2569. [PMID: 28864908 PMCID: PMC6182666 DOI: 10.1007/s00394-017-1527-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/11/2017] [Indexed: 01/02/2023]
Abstract
Purpose Human melanoma is a highly aggressive incurable cancer due to intrinsic cellular resistance to apoptosis, reprogramming, proliferation and survival during tumour progression. Sulforaphane (SFN), an isothiocyanate found in cruciferous vegetables, plays a role in carcinogenesis in many cancer types. However, the cytotoxic molecular mechanisms and gene expression profiles promoted by SFN in human melanoma remain unknown. Methods Three different cell lines were used: two human melanoma A375 and 501MEL and human epidermal melanocytes (HEMa). Cell viability and proliferation, cell cycle analysis, cell migration and invasion and protein expression and phosphorylation status of Akt and p53 upon SFN treatment were determined. RNA-seq of A375 was performed at different time points after SFN treatment. Results We demonstrated that SFN strongly decreased cell viability and proliferation, induced G2/M cell cycle arrest, promoted apoptosis through the activation of caspases 3, 8, 9 and hampered migration and invasion abilities in the melanoma cell lines. Remarkably, HEMa cells were not affected by SFN treatment. Transcriptomic analysis revealed regulation of genes involved in response to stress, apoptosis/cell death and metabolic processes. SFN upregulated the expression of pro-apoptotic genes, such as p53, BAX, PUMA, FAS and MDM2; promoted cell cycle inhibition and growth arrest by upregulating EGR1, GADD45B, ATF3 and CDKN1A; and simultaneously acted as a potent inhibitor of genotoxicity by launching the stress-inducible protein network (HMOX1, HSPA1A, HSPA6, SOD1). Conclusion Overall, the data show that SFN cytotoxicity in melanoma derives from complex and concurrent mechanisms during carcinogenesis, which makes it a promising cancer prevention agent. Electronic supplementary material The online version of this article (doi:10.1007/s00394-017-1527-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paola Arcidiacono
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United KingdomDepartment of Experimental Medicine, University of Perugia, Piazza Lucio Severi, 06132, Perugia, Italy.,Dermatology Clinic, Department of Internal Medicine and Medical Specialties, University of Rome, Rome, Italy
| | - Francesco Ragonese
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United KingdomDepartment of Experimental Medicine, University of Perugia, Piazza Lucio Severi, 06132, Perugia, Italy
| | - Anna Stabile
- Department of Surgery and Biomedical Sciences, University of Perugia, 06132, Perugia, Italy
| | - Alessandra Pistilli
- Department of Surgery and Biomedical Sciences, University of Perugia, 06132, Perugia, Italy
| | - Ekaterina Kuligina
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United KingdomDepartment of Experimental Medicine, University of Perugia, Piazza Lucio Severi, 06132, Perugia, Italy.,N.N. Petrov Institute of Oncology, Saint Petersburg, 197758, Russia
| | - Mario Rende
- Department of Surgery and Biomedical Sciences, University of Perugia, 06132, Perugia, Italy
| | - Ugo Bottoni
- Dermatology Clinic, Department of Internal Medicine and Medical Specialties, University of Rome, Rome, Italy.,University Magna Graecia, Catanzaro, Italy
| | - Stefano Calvieri
- Dermatology Clinic, Department of Internal Medicine and Medical Specialties, University of Rome, Rome, Italy
| | - Andrea Crisanti
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Roberta Spaccapelo
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United KingdomDepartment of Experimental Medicine, University of Perugia, Piazza Lucio Severi, 06132, Perugia, Italy.
| |
Collapse
|
19
|
Tanaka M, Iwakiri Y. The Hepatic Lymphatic Vascular System: Structure, Function, Markers, and Lymphangiogenesis. Cell Mol Gastroenterol Hepatol 2016; 2:733-749. [PMID: 28105461 PMCID: PMC5240041 DOI: 10.1016/j.jcmgh.2016.09.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/02/2016] [Indexed: 02/06/2023]
Abstract
The lymphatic vascular system has been minimally explored in the liver despite its essential functions including maintenance of tissue fluid homeostasis. The discovery of specific markers for lymphatic endothelial cells has advanced the study of lymphatics by methods including imaging, cell isolation, and transgenic animal models and has resulted in rapid progress in lymphatic vascular research during the last decade. These studies have yielded concrete evidence that lymphatic vessel dysfunction plays an important role in the pathogenesis of many diseases. This article reviews the current knowledge of the structure, function, and markers of the hepatic lymphatic vascular system as well as factors associated with hepatic lymphangiogenesis and compares liver lymphatics with those in other tissues.
Collapse
Key Words
- CCl4, carbon tetrachloride
- Cirrhosis
- EHE, epithelioid hemangioendothelioma
- HA, hyaluronan
- HBx Ag, hepatitis B x antigen
- HCC, hepatocellular carcinoma
- IFN, interferon
- IL, interleukin
- Inflammation
- LSEC, liver sinusoidal endothelial cell
- LYVE-1, lymphatic vessel endothelial hyaluronan receptor 1
- LyEC, lymphatic endothelial cell
- NO, nitric oxide
- Portal Hypertension
- Prox1, prospero homeobox protein 1
- VEGF
- VEGF, vascular endothelial growth factor
- VEGFR, vascular endothelial growth factor receptor
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
| | - Yasuko Iwakiri
- Reprint requests Address requests for reprints to: Yasuko Iwakiri, PhD, Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, TAC S223B, 333 Cedar Street, New Haven, Connecticut 06520. fax: (203) 785-7273.Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineTAC S223B, 333 Cedar StreetNew HavenConnecticut 06520
| |
Collapse
|
20
|
Shurin MR, Ma Y, Keskinov AA, Zhao R, Lokshin A, Agassandian M, Shurin GV. BAFF and APRIL from Activin A-Treated Dendritic Cells Upregulate the Antitumor Efficacy of Dendritic Cells In Vivo. Cancer Res 2016; 76:4959-69. [PMID: 27364554 DOI: 10.1158/0008-5472.can-15-2668] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 06/14/2016] [Indexed: 12/12/2022]
Abstract
The members of the TGFβ superfamily play a key role in regulating developmental and homeostasis programs by controlling differentiation, proliferation, polarization, and survival of different cell types. Although the role of TGFβ1 in inflammation and immunity is well evident, the contribution of other TGFβ family cytokines in the modulation of the antitumor immune response remains less documented. Here we show that activin A triggers SMAD2 and ERK1/2 pathways in dendritic cells (DC) expressing type I and II activin receptors, and upregulates production of the TNFα family cytokines BAFF (TALL-1, TNFSF13B) and APRIL (TALL-2, TNFSF13A), which is blocked by SMAD2 and ERK1/2 inhibitors, respectively. BAFF and APRIL derived from activin A-treated DCs upregulate proliferation and survival of T cells expressing the corresponding receptors, BAFF-R and TACI. In vivo, activin A-stimulated DCs demonstrate a significantly increased ability to induce tumor-specific CTLs and inhibit the growth of melanoma and lung carcinoma, which relies on DC-derived BAFF and APRIL, as knockdown of the BAFF and APRIL gene expression in activin A-treated DCs blocks augmentation of their antitumor potential. Although systemic administration of activin A, BAFF, or APRIL for the therapeutic purposes is not likely due to the pluripotent effects on malignant and nonmalignant cells, our data open a novel opportunity for improving the efficacy of DC vaccines. In fact, a significant augmentation of the antitumor activity of DC pretreated with activin A and the proven role of DC-derived BAFF and APRIL in the induction of antitumor immunity in vivo support this direction. Cancer Res; 76(17); 4959-69. ©2016 AACR.
Collapse
Affiliation(s)
- Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Yang Ma
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Anton A Keskinov
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ruijing Zhao
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Anna Lokshin
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Marianna Agassandian
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Galina V Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| |
Collapse
|
21
|
Puujalka E, Heinz M, Hoesel B, Friedl P, Schweighofer B, Wenzina J, Pirker C, Schmid JA, Loewe R, Wagner EF, Berger W, Petzelbauer P. Opposing Roles of JNK and p38 in Lymphangiogenesis in Melanoma. J Invest Dermatol 2016; 136:967-977. [PMID: 26829032 DOI: 10.1016/j.jid.2016.01.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/11/2015] [Accepted: 01/04/2016] [Indexed: 01/14/2023]
Abstract
In primary melanoma, the amount of vascular endothelial growth factor C (VEGF-C) expression and lymphangiogenesis predicts the probability of metastasis to sentinel nodes, but conditions boosting VEGF-C expression in melanoma are poorly characterized. By comparative mRNA expression analysis of a set of 22 human melanoma cell lines, we found a striking negative correlation between VEGF-C and microphthalmia-associated transcription factor (MITF) expression, which was confirmed by data mining in GEO databases of human melanoma Affymetrix arrays. Moreover, in human patients, high VEGF-C and low MITF levels in primary melanoma significantly correlated with the chance of metastasis. Pathway analysis disclosed the respective c-Jun N-terminal kinase and p38/mitogen-activated protein kinase activities as being responsible for the inverse regulation of VEGF-C and MITF. Predominant c-Jun N-terminal kinase signaling results in a VEGF-C(low)/MITF(high) phenotype; these melanoma cells are highly proliferative, show low mobility, and are poorly lymphangiogenic. Predominant p38 signaling results in a VEGF-C(high)/MITF(low) phenotype, corresponding to a slowly cycling, highly mobile, lymphangiogenic, and metastatic melanoma. In conclusion, the relative c-Jun N-terminal kinase and p38 activities determine the biological behavior of melanoma. VEGF-C and MITF levels serve as surrogate markers for the respective c-Jun N-terminal kinase and p38 activities and may be used to predict the risk of metastasis in primary melanoma.
Collapse
Affiliation(s)
- Emmi Puujalka
- Department of Dermatology, Skin and Endothelium Research Division (SERD), Medical University of Vienna, Austria
| | - Magdalena Heinz
- Department of Dermatology, Skin and Endothelium Research Division (SERD), Medical University of Vienna, Austria
| | - Bastian Hoesel
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Peter Friedl
- Department of Dermatology, Skin and Endothelium Research Division (SERD), Medical University of Vienna, Austria
| | - Bernhard Schweighofer
- Department of Dermatology, Skin and Endothelium Research Division (SERD), Medical University of Vienna, Austria
| | - Judith Wenzina
- Department of Dermatology, Skin and Endothelium Research Division (SERD), Medical University of Vienna, Austria
| | - Christine Pirker
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Austria
| | - Johannes A Schmid
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Robert Loewe
- Department of Dermatology, Skin and Endothelium Research Division (SERD), Medical University of Vienna, Austria
| | - Erwin F Wagner
- BBVA Foundation-CNIO Cancer Cell Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Austria
| | - Peter Petzelbauer
- Department of Dermatology, Skin and Endothelium Research Division (SERD), Medical University of Vienna, Austria.
| |
Collapse
|