1
|
Zhang Y, Dong X, Zhang Y, Chen Z, Zhou G, Chen N, Shen W, Yang K, Pei P. Biomaterials to regulate tumor extracellular matrix in immunotherapy. J Control Release 2024; 376:149-166. [PMID: 39389365 DOI: 10.1016/j.jconrel.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
The tumor extracellular matrix (ECM) provides physical support and influences tumor development, metastasis, and the tumor microenvironment, creating barriers to immune drug delivery and cell infiltration. Therefore, modulating or degrading the ECM is of significant importance to enhance the efficacy of tumor immunotherapy. This manuscript initially summarizes the main strategies and mechanisms of biomaterials in modulating various components of the ECM, including collagen, fibronectin, hyaluronic acid, and in remodeling the ECM. Subsequently, it discusses the benefits of biomaterials for immunotherapy following ECM modulation, such as promoting the infiltration of drugs and immune cells, regulating immune cell function, and alleviating the immunosuppressive microenvironment. The manuscript also briefly introduces the application of biomaterials that utilize and mimic the ECM for tumor immunotherapy. Finally, it addresses the current challenges and future directions in this field, providing a comprehensive overview of the potential and innovation in leveraging biomaterials to enhance cancer treatment outcomes. Our work will offer a comprehensive overview of ECM modulation strategies and their application in biomaterials to enhance tumor immunotherapy.
Collapse
Affiliation(s)
- Yujie Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xuexue Dong
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Yanxiang Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zetong Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Ni Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| | - Wenhao Shen
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Jiangsu, China.
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pei Pei
- Department of Nuclear Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province 230022, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| |
Collapse
|
2
|
Nishimura J, Morita Y, Tobe-Nishimoto A, Kitahira Y, Takayama S, Kishimoto S, Matsumiya-Matsumoto Y, Takeshita A, Matsunaga K, Imai T, Uzawa N. CDDP-induced desmoplasia-like changes in oral cancer tissues are related to SASP-related factors induced by the senescence of cancer cells. Int Immunopharmacol 2024; 136:112377. [PMID: 38838554 DOI: 10.1016/j.intimp.2024.112377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/10/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
The tumor microenvironment (TME) concept has been proposed and is currently being actively studied. The development of extracellular matrix (ECM) in the TME is known as desmoplasia and is observed in many solid tumors. It has also been strongly associated with poor prognosis and resistance to drug therapy. Recently, cellular senescence has gained attention as an effect of drug therapy on cancer cells. Cellular senescence is a phenomenon wherein proliferating cells become resistant to growth-promoting stimuli, secrete the SASP (senescence-associated phenotypic) factors, and stably arrest the cell cycle. These proteins are rich in pro-inflammatory factors, such as interleukin (IL)-6, IL-8, C-X-C motif chemokine ligand 1, C-C motif chemokine ligand (CCL)2, CCL5, and matrix metalloproteinase 3. This study aimed to investigate the desmoplasia-like changes in the TME before and after cancer drug therapy in oral squamous cell carcinomas, evaluate the effect of anticancer drugs on the TME, and the potential involvement of cancer cell senescence. Using a syngeneic oral cancer transplant mouse model, we confirmed that cis-diamminedichloroplatinum (II) (CDDP) administration caused desmoplasia-like changes in cancer tissues. Furthermore, CDDP treatment-induced senescence in tumor-bearing mouse tumor tissues and cultured cancer cells. These results suggest CDDP administration-induced desmoplasia-like structural changes in the TME are related to cellular senescence. Our findings suggest that the administration of anticancer drugs alters the TME of oral cancer cells. Additionally, oral cancer cells undergo senescence, which may influence the TME through the production of SASP factors.
Collapse
Affiliation(s)
- Junya Nishimura
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Suita-shi, Osaka 565-0871, Japan
| | - Yoshihiro Morita
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Suita-shi, Osaka 565-0871, Japan.
| | - Ayano Tobe-Nishimoto
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Suita-shi, Osaka 565-0871, Japan
| | - Yukiko Kitahira
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Suita-shi, Osaka 565-0871, Japan
| | - Shun Takayama
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Suita-shi, Osaka 565-0871, Japan
| | - Satoko Kishimoto
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Suita-shi, Osaka 565-0871, Japan
| | - Yuka Matsumiya-Matsumoto
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Suita-shi, Osaka 565-0871, Japan
| | - Akinori Takeshita
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Suita-shi, Osaka 565-0871, Japan
| | - Kazuhide Matsunaga
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Suita-shi, Osaka 565-0871, Japan
| | - Tomoaki Imai
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Suita-shi, Osaka 565-0871, Japan
| | - Narikazu Uzawa
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Suita-shi, Osaka 565-0871, Japan
| |
Collapse
|
3
|
Boutas I, Kontogeorgi A, Kalantaridou SN, Dimitrakakis C, Patsios P, Kalantzi M, Xanthos T. Reverse Onco-Cardiology: What Is the Evidence for Breast Cancer? A Systematic Review of the Literature. Int J Mol Sci 2023; 24:16500. [PMID: 38003690 PMCID: PMC10671526 DOI: 10.3390/ijms242216500] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Breast cancer and cardiovascular diseases (CVD) represent significant global health challenges, with CVD being the leading cause of mortality and breast cancer, showing a complex pattern of incidence and mortality. We explore the intricate interplay between these two seemingly distinct medical conditions, shedding light on their shared risk factors and potential pathophysiological connections. A specific connection between hypertension (HTN), atrial fibrillation (AF), myocardial infarction (MI), and breast cancer was evaluated. HTN is explored in detail, emphasizing the role of aging, menopause, insulin resistance, and obesity as common factors linking HTN and breast cancer. Moreover, an attempt is made to identify the potential impact of antihypertensive medications and highlight the increased risk of breast cancer among those women, with a focus on potential mechanisms. A summary of key findings underscores the need for a multisystem approach to understanding the relationship between CVD and breast cancer is also explored with a highlight for all the gaps in current research, such as the lack of clinical observational data on MI and breast cancer in humans and the need for studies specifically designed for breast cancer. This paper concludes that there should be a focus on potential clinical applications of further investigation in this field, including personalized prevention and screening strategies for women at risk. Overall, the authors attempt to provide a comprehensive overview of the intricate connections between breast cancer and cardiovascular diseases, emphasizing the importance of further research in this evolving field of cardio-oncology.
Collapse
Affiliation(s)
- Ioannis Boutas
- Breast Unit, Rea Maternity Hospital, 383 Andrea Siggrou Ave., Paleo Faliro, 175 64 Athens, Greece
| | - Adamantia Kontogeorgi
- Medical School, University of Crete, 13 Andrea Kalokairinoy Ave., 715 00 Giofirakia, Greece
| | - Sophia N. Kalantaridou
- Third Department of Obstetrics and Gynecology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 1 Rimini Str., 124 62 Chaidari, Greece;
| | - Constantine Dimitrakakis
- First Department of Obstetrics and Gynecology, Alexandra University Hospital, Medical School, National and Kapodistrian University of Athens, 4-2 Lourou Str., 115 28 Athens, Greece;
| | - Panagiotis Patsios
- Cardiology Department, Elpis General Hospital, 7 Dimitsana Str., 115 22 Athens, Greece;
| | - Maria Kalantzi
- Post Graduate Study Program “Cardiopulmonary Resuscitation”, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 115 27 Athens, Greece;
| | - Theodoros Xanthos
- School of Health Sciences, University of West Attica, 28 Aghiou Spyridonos Str., 122 43 Aigaleo, Greece;
| |
Collapse
|
4
|
Shakiba D, Genin GM, Zustiak SP. Mechanobiology of cancer cell responsiveness to chemotherapy and immunotherapy: Mechanistic insights and biomaterial platforms. Adv Drug Deliv Rev 2023; 196:114771. [PMID: 36889646 PMCID: PMC10133187 DOI: 10.1016/j.addr.2023.114771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/17/2022] [Accepted: 03/03/2023] [Indexed: 03/08/2023]
Abstract
Mechanical forces are central to how cancer treatments such as chemotherapeutics and immunotherapies interact with cells and tissues. At the simplest level, electrostatic forces underlie the binding events that are critical to therapeutic function. However, a growing body of literature points to mechanical factors that also affect whether a drug or an immune cell can reach a target, and to interactions between a cell and its environment affecting therapeutic efficacy. These factors affect cell processes ranging from cytoskeletal and extracellular matrix remodeling to transduction of signals by the nucleus to metastasis of cells. This review presents and critiques the state of the art of our understanding of how mechanobiology impacts drug and immunotherapy resistance and responsiveness, and of the in vitro systems that have been of value in the discovery of these effects.
Collapse
Affiliation(s)
- Delaram Shakiba
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO, USA; Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, USA
| | - Guy M Genin
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO, USA; Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, USA.
| | - Silviya P Zustiak
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO, USA; Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, St. Louis, MO, USA.
| |
Collapse
|
5
|
Faisal SM, Comba A, Varela ML, Argento AE, Brumley E, Abel C, Castro MG, Lowenstein PR. The complex interactions between the cellular and non-cellular components of the brain tumor microenvironmental landscape and their therapeutic implications. Front Oncol 2022; 12:1005069. [PMID: 36276147 PMCID: PMC9583158 DOI: 10.3389/fonc.2022.1005069] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022] Open
Abstract
Glioblastoma (GBM), an aggressive high-grade glial tumor, is resistant to therapy and has a poor prognosis due to its universal recurrence rate. GBM cells interact with the non-cellular components in the tumor microenvironment (TME), facilitating their rapid growth, evolution, and invasion into the normal brain. Herein we discuss the complexity of the interactions between the cellular and non-cellular components of the TME and advances in the field as a whole. While the stroma of non-central nervous system (CNS) tissues is abundant in fibrillary collagens, laminins, and fibronectin, the normal brain extracellular matrix (ECM) predominantly includes proteoglycans, glycoproteins, and glycosaminoglycans, with fibrillary components typically found only in association with the vasculature. However, recent studies have found that in GBMs, the microenvironment evolves into a more complex array of components, with upregulated collagen gene expression and aligned fibrillary ECM networks. The interactions of glioma cells with the ECM and the degradation of matrix barriers are crucial for both single-cell and collective invasion into neighboring brain tissue. ECM-regulated mechanisms also contribute to immune exclusion, resulting in a major challenge to immunotherapy delivery and efficacy. Glioma cells chemically and physically control the function of their environment, co-opting complex signaling networks for their own benefit, resulting in radio- and chemo-resistance, tumor recurrence, and cancer progression. Targeting these interactions is an attractive strategy for overcoming therapy resistance, and we will discuss recent advances in preclinical studies, current clinical trials, and potential future clinical applications. In this review, we also provide a comprehensive discussion of the complexities of the interconnected cellular and non-cellular components of the microenvironmental landscape of brain tumors to guide the development of safe and effective therapeutic strategies against brain cancer.
Collapse
Affiliation(s)
- Syed M. Faisal
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrea Comba
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria L. Varela
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anna E. Argento
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Emily Brumley
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Clifford Abel
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria G. Castro
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pedro R. Lowenstein
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Pedro R. Lowenstein,
| |
Collapse
|
6
|
Tajaldini M, Saeedi M, Amiriani T, Amiriani AH, Sedighi S, Mohammad Zadeh F, Dehghan M, Jahanshahi M, Zanjan Ghandian M, Khalili P, Poorkhani AH, Alizadeh AM, Khori V. Cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs); where do they stand in tumorigenesis and how they can change the face of cancer therapy? Eur J Pharmacol 2022; 928:175087. [PMID: 35679891 DOI: 10.1016/j.ejphar.2022.175087] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/18/2022] [Accepted: 06/03/2022] [Indexed: 11/03/2022]
Abstract
The tumor microenvironment (TME) and its components have recently attracted tremendous attention in cancer treatment strategies, as alongside the genetic and epigenetic alterations in tumor cells, TME could also provide a fertile background for malignant cells to survive and proliferate. Interestingly, TME plays a vital role in the mediation of cancer metastasis and drug resistance even against immunotherapeutic agents. Among different cells that are presenting in TME, tumor-associated macrophages (TAMs) and cancer-associated fibroblasts (CAFs) have shown to have significant value in the regulation of angiogenesis, tumor metastasis, and drug-resistance through manipulating the composition as well as the organization of extracellular matrix (ECM). Evidence has shown that the presence of both TAMs and CAFs in TME is associated with poor prognosis and failure of chemotherapeutic agents. It seems that these cells together with ECM form a shield around tumor cells to protect them from the toxic agents and even the adaptive arm of the immune system, which is responsible for tumor surveillance. Given this, targeting TAMs and CAFs seems to be an essential approach to potentiate the cytotoxic effects of anti-cancer agents, either conventional chemotherapeutic drugs or immunotherapies. In the present review, we aimed to take a deep look at the mechanobiology of CAFs and TAMs in tumor progression and to discuss the available therapeutic approaches for harnessing these cells in TME.
Collapse
Affiliation(s)
- Mahboubeh Tajaldini
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohsen Saeedi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Taghi Amiriani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Amir Hossein Amiriani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sima Sedighi
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Fatemeh Mohammad Zadeh
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohammad Dehghan
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Jahanshahi
- Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maziar Zanjan Ghandian
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Pedram Khalili
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Ali Mohammad Alizadeh
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Khori
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
7
|
Brokhman I, Watkin AMT, Bacher JC, Glazer SA, Galea AM. A Novel Method for the Production of an Autologous Anti-Inflammatory and Anti-Catabolic Product (Cytorich) from Human Blood: A Prospective Treatment for the COVID-19-Induced Cytokine Storm. Med Sci Monit 2021; 27:e934365. [PMID: 34795200 PMCID: PMC8609770 DOI: 10.12659/msm.934365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Autologous blood-derived products can target specific inflammatory molecular pathways and have potentially beneficial therapeutic effects on inflammatory pathologies. The purpose of this study was to assess in vitro the anti-inflammatory and anti-catabolic potential of an autologous blood product as a possible treatment for COVID-19-induced cytokine storm. Material/Methods Blood samples from healthy donors and donors who had recovered from COVID-19 were incubated using different techniques and analyzed for the presence of anti-inflammatory, anti-catabolic, regenerative, pro-inflammatory, and procatabolic molecules. Results The highest concentrations of therapeutic molecules for targeting inflammatory pathways were found in the blood that had been incubated for 24 h at 37°C, whereas a significant increase was observed after 6 h of incubation in blood from COVID-19-recovered donors. Beneficially, the 6-h incubation process did not downregulate anti-COVID-19 immunoglobulin G concentrations. Unfortunately, increases in matrix metalloproteinase 9, tumor necrosis factor α, and interleukin-1 were detected in the product after incubation; however, these increases could be blocked by adding citric acid, with no effect on the concentration of the target therapeutic molecules. Our data allow for safer and more effective future treatments. Conclusions An autologous blood-derived product containing anti-inflammatory and anti-catabolic molecules, which we term Cytorich, has a promising therapeutic role in the treatment of a virus-induced cytokine storm, including that associated with COVID-19.
Collapse
Affiliation(s)
- Irina Brokhman
- Department of Research and Development, The Institute of Human Mechanics, Toronto, ON, Canada
| | - Alyssia M T Watkin
- Department of Research and Development, The Institute of Human Mechanics, Toronto, ON, Canada
| | - Jeffrey C Bacher
- Department of Research and Development, The Institute of Human Mechanics, Toronto, ON, Canada
| | - Stephen A Glazer
- Toronto Critical Care Medicine, Humber River Hospital, Toronto, ON, Canada
| | - Anthony M Galea
- Department of Research and Development, The Institute of Human Mechanics, Toronto, ON, Canada
| |
Collapse
|
8
|
Hot or cold: Bioengineering immune contextures into in vitro patient-derived tumor models. Adv Drug Deliv Rev 2021; 175:113791. [PMID: 33965462 DOI: 10.1016/j.addr.2021.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, immune checkpoint inhibitors (ICI) have proven to be tremendously effective for a subset of cancer patients. However, it is difficult to predict the response of individual patients and efforts are now directed at understanding the mechanisms of ICI resistance. Current models of patient tumors poorly recapitulate the immune contexture, which describe immune parameters that are associated with patient survival. In this Review, we discuss parameters that influence the induction of different immune contextures found within tumors and how engineering strategies may be leveraged to recapitulate these contextures to develop the next generation of immune-competent patient-derived in vitro models.
Collapse
|
9
|
Huang J, Zhang L, Wan D, Zhou L, Zheng S, Lin S, Qiao Y. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct Target Ther 2021; 6:153. [PMID: 33888679 PMCID: PMC8062524 DOI: 10.1038/s41392-021-00544-0] [Citation(s) in RCA: 312] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 02/17/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) is one of the major components of tumors that plays multiple crucial roles, including mechanical support, modulation of the microenvironment, and a source of signaling molecules. The quantity and cross-linking status of ECM components are major factors determining tissue stiffness. During tumorigenesis, the interplay between cancer cells and the tumor microenvironment (TME) often results in the stiffness of the ECM, leading to aberrant mechanotransduction and further malignant transformation. Therefore, a comprehensive understanding of ECM dysregulation in the TME would contribute to the discovery of promising therapeutic targets for cancer treatment. Herein, we summarized the knowledge concerning the following: (1) major ECM constituents and their functions in both normal and malignant conditions; (2) the interplay between cancer cells and the ECM in the TME; (3) key receptors for mechanotransduction and their alteration during carcinogenesis; and (4) the current therapeutic strategies targeting aberrant ECM for cancer treatment.
Collapse
Affiliation(s)
- Jiacheng Huang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Lele Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Dalong Wan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Shengzhang Lin
- School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310000, China.
| | - Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China.
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China.
| |
Collapse
|
10
|
Pilkington SM, Bulfone-Paus S, Griffiths CE, Watson RE. Inflammaging and the Skin. J Invest Dermatol 2021; 141:1087-1095. [DOI: 10.1016/j.jid.2020.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/09/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022]
|
11
|
Deligne C, Midwood KS. Macrophages and Extracellular Matrix in Breast Cancer: Partners in Crime or Protective Allies? Front Oncol 2021; 11:620773. [PMID: 33718177 PMCID: PMC7943718 DOI: 10.3389/fonc.2021.620773] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/15/2021] [Indexed: 12/11/2022] Open
Abstract
Solid cancers such as breast tumors comprise a collection of tumor, stromal and immune cells, embedded within a network of tumor-specific extracellular matrix. This matrix is associated with tumor aggression, treatment failure, chemo- and radio-resistance, poor survival and metastasis. Recent data report an immunomodulatory role for the matrix in cancer, via the creation of niches that control the migration, localization, phenotype and function of tumor-infiltrating immune cells, ultimately contributing to escape of immune surveillance. Macrophages are crucial components of the immune infiltrate in tumors; they are associated with a poor prognosis in breast cancer and contribute to shaping the anti-tumor immune response. We and others have described how matrix molecules commonly upregulated within the tumor stroma, such as tenascin-C, fibronectin and collagen, exert a complex influence over macrophage behavior, for example restricting or enhancing their infiltration into the tumor, and driving their polarization towards or away from a pro-tumoral phenotype, and how in turn macrophages can modify matrix production in the tumor to favor tumor growth and metastasis. Targeting specific domains of matrix molecules to reinstate an efficient anti-tumor immune response, and effectively control tumor growth and spread, is emerging as a promising field offering a new angle for cancer therapy. Here, we review current knowledge on the interactions between tumor-associated macrophages and matrix molecules that occur within the tumor microenvironment of breast cancer, and discuss how these pathways can be targeted for new immunotherapies for hard to treat, desmoplastic tumors.
Collapse
Affiliation(s)
- Claire Deligne
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Kim S Midwood
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
12
|
Wei Z, Chen L, Meng L, Han W, Huang L, Xu A. LncRNA HOTAIR promotes the growth and metastasis of gastric cancer by sponging miR-1277-5p and upregulating COL5A1. Gastric Cancer 2020; 23:1018-1032. [PMID: 32583079 DOI: 10.1007/s10120-020-01091-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Emerging studies have shown that HOTAIR acts as an oncogene in gastric cancer (GC). However, its role in the extracellular matrix and in tumor immune infiltration remains unknown. METHODS HOTAIR and COL5A1 levels were analyzed by bioinformatics analysis and validated by qRT-PCR, western blotting and immunohistochemistry assays. The regulatory relationships between components of the HOTAIR/miR-1277-5p/COL5A1 axis and the role of this axis in GC were predicted by bioinformatics analysis, and validated by in vitro and in vivo experiments. The correlation between COL5A1 and GC immune infiltration was assessed by bioinformatics analysis and a COL5A1-based predictive nomogram was established using the Stomach Adenocarcinoma dataset from The Cancer Genome Atlas. RESULTS We found that HOTAIR and COL5A1 were overexpressed in GC compared to normal controls, which predicted poor prognosis. The regulatory relationship of the HOTAIR/miR-1277-5p/COL5A1 axis in GC was demonstrated, and HOTAIR and COL5A1 were found to promote GC growth while miR-1277-5p exerted the reverse effects. In addition, COL5A1 was negatively associated with tumor purity but positively associated with immune infiltration, which suggested that COL5A1-mediated GC growth may be partially mediated by the regulation of immune infiltration. Additionally, the established COL5A1-based nomogram showed that COL5A1 can serve as a prognostic biomarker in GC. CONCLUSIONS HOTAIR regulates GC growth by sponging miR-1277-5p and upregulating COL5A1, and COL5A1-mediated GC cell proliferation may be mediated by effects on the tumor microenvironment, which provides novel targets for GC treatment.
Collapse
Affiliation(s)
- Zhijian Wei
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, PR China
| | - Lei Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, People's Republic of China.,Institute of Urology, Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, People's Republic of China
| | - Lei Meng
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, 100 HuaiHe Avenue, Hefei, 230002, Anhui, People's Republic of China
| | - Wenxiu Han
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, PR China.
| | - Lei Huang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, PR China.
| | - Aman Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, PR China.
| |
Collapse
|
13
|
Klepfish M, Gross T, Vugman M, Afratis NA, Havusha-Laufer S, Brazowski E, Solomonov I, Varol C, Sagi I. LOXL2 Inhibition Paves the Way for Macrophage-Mediated Collagen Degradation in Liver Fibrosis. Front Immunol 2020; 11:480. [PMID: 32296422 PMCID: PMC7136575 DOI: 10.3389/fimmu.2020.00480] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/02/2020] [Indexed: 01/06/2023] Open
Abstract
Liver fibrosis is characterized by the excessive accumulation of extracellular matrix (ECM) proteins and enzymes, especially fibrillary collagens, and represents a major cause of morbidity and mortality worldwide. Lysyl oxidases (LOXs) drive covalent crosslinking of collagen fibers, thereby promoting stabilization and accumulation of liver fibrosis while limiting its resolution. Here we show in a carbon tetrachloride (CCl4)-induced liver fibrosis murine model that treatment with a novel anti-lysyl oxidase like 2 (LOXL2) neutralizing antibody, which targets extracellular LOXL2, significantly improves fibrosis resolution. LOXL2 inhibition following the onset of fibrosis accelerated and augmented collagen degradation. This was accompanied by increased localization of reparative monocyte-derived macrophages (MoMFs) in the proximity of fibrotic fibers and their representation in the liver. These cells secreted collagenolytic matrix metalloproteinases (MMPs) and, in particular, the membrane-bound MT1-MMP (MMP-14) collagenase. Inducible and selective ablation of infiltrating MoMFs negated the increased "on-fiber" accumulation of MMP-14-expressing MoMFs and the accelerated collagenolytic activity observed in the anti-LOXL2-treated mice. Many studies of liver fibrosis focus on preventing the progression of the fibrotic process. In contrast, the therapeutic mechanism of LOXL2 inhibition presented herein aims at reversing existing fibrosis and facilitating endogenous liver regeneration by paving the way for collagenolytic macrophages.
Collapse
Affiliation(s)
- Mordehay Klepfish
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Gross
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Milena Vugman
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv-Yafo, Israel
| | - Nikolaos A Afratis
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Sapir Havusha-Laufer
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Eli Brazowski
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv-Yafo, Israel
| | - Inna Solomonov
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Chen Varol
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv-Yafo, Israel.,Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
14
|
Henke E, Nandigama R, Ergün S. Extracellular Matrix in the Tumor Microenvironment and Its Impact on Cancer Therapy. Front Mol Biosci 2020; 6:160. [PMID: 32118030 PMCID: PMC7025524 DOI: 10.3389/fmolb.2019.00160] [Citation(s) in RCA: 567] [Impact Index Per Article: 141.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
Solid tumors are complex organ-like structures that consist not only of tumor cells but also of vasculature, extracellular matrix (ECM), stromal, and immune cells. Often, this tumor microenvironment (TME) comprises the larger part of the overall tumor mass. Like the other components of the TME, the ECM in solid tumors differs significantly from that in normal organs. Intratumoral signaling, transport mechanisms, metabolisms, oxygenation, and immunogenicity are strongly affected if not controlled by the ECM. Exerting this regulatory control, the ECM does not only influence malignancy and growth of the tumor but also its response toward therapy. Understanding the particularities of the ECM in solid tumor is necessary to develop approaches to interfere with its negative effect. In this review, we will also highlight the current understanding of the physical, cellular, and molecular mechanisms by which the pathological tumor ECM affects the efficiency of radio-, chemo-, and immunotherapy. Finally, we will discuss the various strategies to target and modify the tumor ECM and how they could be utilized to improve response to therapy.
Collapse
Affiliation(s)
- Erik Henke
- Department of Medicine, Institute of Anatomy and Cell Biology, Universität Würzburg, Würzburg, Germany
| | - Rajender Nandigama
- Department of Medicine, Institute of Anatomy and Cell Biology, Universität Würzburg, Würzburg, Germany
| | - Süleyman Ergün
- Department of Medicine, Institute of Anatomy and Cell Biology, Universität Würzburg, Würzburg, Germany
| |
Collapse
|
15
|
Varol C. Tumorigenic Interplay Between Macrophages and Collagenous Matrix in the Tumor Microenvironment. Methods Mol Biol 2019; 1944:203-220. [PMID: 30840245 DOI: 10.1007/978-1-4939-9095-5_15] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment is a heterogeneous tissue that in addition to tumor cells, contain tumor-associated cell types such as immune cells, fibroblasts, and endothelial cells. Considerably important in the tumor microenvironment is its noncellular component, namely, the extracellular matrix (ECM). In particular, the collagenous matrix is subjected to significant alterations in its composition and structure that create a permissive environment for tumor growth, invasion, and dissemination. Among tumor-infiltrating immune cells, tumor-associated macrophages (TAMs) are numerous in the tumor stroma and are locally educated to mediate important biological functions that profoundly affect tumor initiation, growth, and dissemination. While the influence of TAMs and mechanical properties of the collagenous matrix on tumor invasion and progression have been comprehensively investigated individually, their interaction within the complex tumor microenvironment was overlooked. This review summarizes accumulating evidence that indicate the existence of an intricate tumorigenic crosstalk between TAMs and collagenous matrix. A better mechanistic comprehension of this reciprocal interplay may open a novel arena for cancer therapeutics.
Collapse
Affiliation(s)
- Chen Varol
- The Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
16
|
3D type I collagen environment leads up to a reassessment of the classification of human macrophage polarizations. Biomaterials 2019; 208:98-109. [PMID: 31005702 DOI: 10.1016/j.biomaterials.2019.04.018] [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] [Received: 01/14/2019] [Revised: 03/21/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
Abstract
Macrophages have multiple roles in development, tissue homeostasis and repair and present a high degree of phenotypic plasticity embodied in the concept of polarization. One goal of macrophage biology field is to characterize these polarizations at the molecular level. To achieve this task, it is necessary to integrate how physical environment signals are interpreted by macrophages under immune stimulation. In this work, we study how a 3D scaffold obtained from polymerized fibrillar rat type I collagen modulates the polarizations of human macrophages and reveal that some traditionally used markers should be reassessed. We demonstrate that integrin β2 is a regulator of STAT1 phosphorylation in response to IFNγ/LPS as well as responsible for the inhibition of ALOX15 expression in response to IL-4/IL-13 in 3D. Meanwhile, we also find that the CCL19/CCL20 ratio is reverted in 3D under IFNγ/LPS stimulation. 3D also induces the priming of the NLRP3 inflammasome resulting in an increased IL-1β and IL-6 secretion. These results give the molecular basis for assessing collagen induced immunomodulation of human macrophages in various physiological and pathological contexts such as cancer.
Collapse
|
17
|
Long KB, Collier AI, Beatty GL. Macrophages: Key orchestrators of a tumor microenvironment defined by therapeutic resistance. Mol Immunol 2017; 110:3-12. [PMID: 29273393 DOI: 10.1016/j.molimm.2017.12.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/22/2017] [Accepted: 12/02/2017] [Indexed: 12/12/2022]
Abstract
Macrophages have emerged as promising therapeutic targets in cancer. Within tumor tissue, macrophages foster tumor development, invasion, and metastasis. As the phenotype of macrophages is inherently pliable and dependent on cues received from the surrounding microenvironment, macrophages co-evolve with malignant and other non-malignant cells during cancer progression. In doing so, they establish a microenvironment that is therapeutically resistant and thwarts the productivity of T cell immunosuveillance. Strategies designed to deplete, inhibit, or redirect macrophages with anti-tumor activity are being explored to reverse the pro-tumor properties of macrophages that are commonly observed in cancer. In this review, we discuss our current understanding of the mechanisms that regulate macrophage recruitment to tumors, their impact on the tumor microenvironment, and their promise as therapeutic targets for improving the efficacy of cytotoxic- and immune-based therapies.
Collapse
Affiliation(s)
- Kristen B Long
- Department of Biology, Mansfield University, Mansfield, PA 16933, USA
| | - Arthur I Collier
- Department of Biology, Mansfield University, Mansfield, PA 16933, USA
| | - Gregory L Beatty
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Varol C, Sagi I. Phagocyte-extracellular matrix crosstalk empowers tumor development and dissemination. FEBS J 2017; 285:734-751. [DOI: 10.1111/febs.14317] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/01/2017] [Accepted: 10/31/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Chen Varol
- The Research Center for Digestive Tract and Liver Diseases; Tel-Aviv Sourasky Medical Center; Sackler Faculty of Medicine; Tel-Aviv University; Israel
- Department of Clinical Microbiology and Immunology; Sackler Faculty of Medicine; Tel Aviv University; Israel
| | - Irit Sagi
- Department of Biological Regulation; Weizmann Institute of Science; Rehovot Israel
| |
Collapse
|
19
|
Comunanza V, Corà D, Orso F, Consonni FM, Middonti E, Di Nicolantonio F, Buzdin A, Sica A, Medico E, Sangiolo D, Taverna D, Bussolino F. VEGF blockade enhances the antitumor effect of BRAFV600E inhibition. EMBO Mol Med 2017; 9:219-237. [PMID: 27974353 PMCID: PMC5286370 DOI: 10.15252/emmm.201505774] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The development of resistance remains a major obstacle to long‐term disease control in cancer patients treated with targeted therapies. In BRAF‐mutant mouse models, we demonstrate that although targeted inhibition of either BRAF or VEGF initially suppresses the growth of BRAF‐mutant tumors, combined inhibition of both pathways results in apoptosis, long‐lasting tumor responses, reduction in lung colonization, and delayed onset of acquired resistance to the BRAF inhibitor PLX4720. As well as inducing tumor vascular normalization and ameliorating hypoxia, this approach induces remodeling of the extracellular matrix, infiltration of macrophages with an M1‐like phenotype, and reduction in cancer‐associated fibroblasts. At the molecular level, this therapeutic regimen results in a de novo transcriptional signature, which sustains and explains the observed efficacy with regard to cancer progression. Collectively, our findings offer new biological rationales for the management of clinical resistance to BRAF inhibitors based on the combination between BRAFV600E inhibitors with anti‐angiogenic regimens.
Collapse
Affiliation(s)
- Valentina Comunanza
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute IRCCS, Candiolo, Italy
| | - Davide Corà
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute IRCCS, Candiolo, Italy.,Center for Molecular Systems Biology, University of Torino, Orbassano, Italy
| | - Francesca Orso
- Center for Molecular Systems Biology, University of Torino, Orbassano, Italy.,Molecular Biotechnology Center (MBC), Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | | | - Emanuele Middonti
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute IRCCS, Candiolo, Italy
| | - Federica Di Nicolantonio
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute IRCCS, Candiolo, Italy
| | - Anton Buzdin
- Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.,National Research Centre "Kurchatov Institute", Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, Moscow, Russia
| | - Antonio Sica
- Humanitas Clinical and Research Center, Rozzano, Italy.,Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Enzo Medico
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute IRCCS, Candiolo, Italy
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute IRCCS, Candiolo, Italy
| | - Daniela Taverna
- Center for Molecular Systems Biology, University of Torino, Orbassano, Italy.,Molecular Biotechnology Center (MBC), Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, Candiolo, Italy .,Candiolo Cancer Institute IRCCS, Candiolo, Italy.,Center for Molecular Systems Biology, University of Torino, Orbassano, Italy
| |
Collapse
|
20
|
Sangaletti S, Chiodoni C, Tripodo C, Colombo MP. Common extracellular matrix regulation of myeloid cell activity in the bone marrow and tumor microenvironments. Cancer Immunol Immunother 2017; 66:1059-1067. [PMID: 28501940 PMCID: PMC11029001 DOI: 10.1007/s00262-017-2014-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 05/06/2017] [Indexed: 01/09/2023]
Abstract
The complex interaction between cells undergoing transformation and the various stromal and immunological cell components of the tumor microenvironment (TME) crucially influences cancer progression and diversification, as well as endowing clinical and prognostic significance. The immunosuppression characterizing the TME depends on the recruitment and activation of different cell types including regulatory T cells, myeloid-derived suppressor cells, and tumor-associated macrophages. Less considered is the non-cellular component of the TME. Here, we focus on the extracellular matrix (ECM) regulatory activities that, within the TME, actively contribute to many aspects of tumor progression, acting on both tumor and immune cells. Particularly, ECM-mediated regulation of tumor-associated immunosuppression occurs through the modulation of myeloid cell expansion, localization, and functional activities. Such regulation is not limited to the TME but occurs also within the bone marrow, wherein matricellular proteins contribute to the maintenance of specialized hematopoietic stem cell niches thereby regulating their homeostasis as well as the generation and expansion of myeloid cells under both physiological and pathological conditions. Highlighting the commonalities among ECM-myeloid cell interactions in bone marrow and TME, in this review we present a picture in which myeloid cells might sense and respond to ECM modifications, providing different ECM-myeloid cell interfaces that may be useful to define prognostic groups and to tailor therapeutic interventions.
Collapse
Affiliation(s)
- Sabina Sangaletti
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo, 42, 20133, Milan, Italy
| | - Claudia Chiodoni
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo, 42, 20133, Milan, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, University of Palermo, Palermo, Italy
| | - Mario P Colombo
- Molecular Immunology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo, 42, 20133, Milan, Italy.
| |
Collapse
|
21
|
Fornetti J, Martinson HA, Betts CB, Lyons TR, Jindal S, Guo Q, Coussens LM, Borges VF, Schedin P. Mammary gland involution as an immunotherapeutic target for postpartum breast cancer. J Mammary Gland Biol Neoplasia 2014; 19:213-28. [PMID: 24952477 PMCID: PMC4363120 DOI: 10.1007/s10911-014-9322-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 06/09/2014] [Indexed: 12/24/2022] Open
Abstract
Postpartum mammary gland involution has been identified as tumor-promotional and is proposed to contribute to the increased rates of metastasis and poor survival observed in postpartum breast cancer patients. In rodent models, the involuting mammary gland microenvironment is sufficient to induce enhanced tumor cell growth, local invasion, and metastasis. Postpartum involution shares many attributes with wound healing, including upregulation of genes involved in immune responsiveness and infiltration of tissue by immune cells. In rodent models, treatment with non-steroidal anti-inflammatory drugs (NSAIDs) ameliorates the tumor-promotional effects of involution, consistent with the immune milieu of the involuting gland contributing to tumor promotion. Currently, immunotherapy is being investigated as a means of breast cancer treatment with the purpose of identifying ways to enhance anti-tumor immune responses. Here we review evidence for postpartum mammary gland involution being a uniquely defined 'hot-spot' of pro-tumorigenic immune cell infiltration, and propose that immunotherapy should be explored for prevention and treatment of breast cancers that arise in this environment.
Collapse
Affiliation(s)
- Jaime Fornetti
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
- Program in Reproductive Sciences, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Holly A. Martinson
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USA
| | - Courtney B. Betts
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
- Cell Biology, Stem cells, and Development, 12801 E 17th Ave, Aurora, CO 80045, USA
| | - Traci R. Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
| | - Sonali Jindal
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
| | - Qiuchen Guo
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USA
| | - Lisa M. Coussens
- Department of Cell & Developmental Biology, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Virginia F. Borges
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
| | - Pepper Schedin
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, 1665 Aurora Court, Aurora, CO 80045, USA
- Program in Reproductive Sciences, University of Colorado Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USA
- Cell Biology, Stem cells, and Development, 12801 E 17th Ave, Aurora, CO 80045, USA
- Department of Cell & Developmental Biology, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
22
|
Collagen as a double-edged sword in tumor progression. Tumour Biol 2013; 35:2871-82. [PMID: 24338768 PMCID: PMC3980040 DOI: 10.1007/s13277-013-1511-7] [Citation(s) in RCA: 363] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 12/03/2013] [Indexed: 12/11/2022] Open
Abstract
It has been recognized that cancer is not merely a disease of tumor cells, but a disease of imbalance, in which stromal cells and tumor microenvironment play crucial roles. Extracellular matrix (ECM) as the most abundant component in tumor microenvironment can regulate tumor cell behaviors and tissue tension homeostasis. Collagen constitutes the scaffold of tumor microenvironment and affects tumor microenvironment such that it regulates ECM remodeling by collagen degradation and re-deposition, and promotes tumor infiltration, angiogenesis, invasion and migration. While collagen was traditionally regarded as a passive barrier to resist tumor cells, it is now evident that collagen is also actively involved in promoting tumor progression. Collagen changes in tumor microenvironment release biomechanical signals, which are sensed by both tumor cells and stromal cells, trigger a cascade of biological events. In this work, we discuss how collagen can be a double-edged sword in tumor progression, both inhibiting and promoting tumor progression at different stages of cancer development.
Collapse
|
23
|
Abstract
The local microenvironment, or niche, of a cancer cell plays important roles in cancer development. A major component of the niche is the extracellular matrix (ECM), a complex network of macromolecules with distinctive physical, biochemical, and biomechanical properties. Although tightly controlled during embryonic development and organ homeostasis, the ECM is commonly deregulated and becomes disorganized in diseases such as cancer. Abnormal ECM affects cancer progression by directly promoting cellular transformation and metastasis. Importantly, however, ECM anomalies also deregulate behavior of stromal cells, facilitate tumor-associated angiogenesis and inflammation, and thus lead to generation of a tumorigenic microenvironment. Understanding how ECM composition and topography are maintained and how their deregulation influences cancer progression may help develop new therapeutic interventions by targeting the tumor niche.
Collapse
Affiliation(s)
- Pengfei Lu
- Breakthrough Breast Cancer Research Unit, University of Manchester, Manchester M20 4BX, England, UK
| | | | | |
Collapse
|
24
|
Kees T, Egeblad M. Innate immune cells in breast cancer--from villains to heroes? J Mammary Gland Biol Neoplasia 2011; 16:189-203. [PMID: 21789554 DOI: 10.1007/s10911-011-9224-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 07/18/2011] [Indexed: 12/13/2022] Open
Abstract
The innate immune system ensures effective protection against foreign pathogens and plays important roles in tissue remodeling. There are many types of innate immune cells, including monocytes, macrophages, dendritic cells, and granulocytes. Interestingly, these cells accumulate in most solid tumors, including those of the breast. There, they play a tumor-promoting role through secretion of growth and angiogenic factors, as well as immunosuppressive molecules. This is in strong contrast to the tumor-suppressing effects that innate immune cells exert in vitro upon proper activation. Therapeutic approaches have been developed with the aim of achieving similar suppressive activities in vivo. However, multiple factors in the tumor microenvironment, many of which are immunosuppressive, represent a major obstacle to effective treatment. Here, we discuss the potential of combating breast cancer through activation of the innate immune system, including possible strategies to enhance the success of immunotherapy.
Collapse
Affiliation(s)
- Tim Kees
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | | |
Collapse
|
25
|
Dynamic interplay between the collagen scaffold and tumor evolution. Curr Opin Cell Biol 2010; 22:697-706. [PMID: 20822891 DOI: 10.1016/j.ceb.2010.08.015] [Citation(s) in RCA: 657] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 08/10/2010] [Accepted: 08/11/2010] [Indexed: 12/11/2022]
Abstract
The extracellular matrix (ECM) is a key regulator of cell and tissue function. Traditionally, the ECM has been thought of primarily as a physical scaffold that binds cells and tissues together. However, the ECM also elicits biochemical and biophysical signaling. Controlled proteolysis and remodeling of the ECM network regulate tissue tension, generate pathways for migration, and release ECM protein fragments to direct normal developmental processes such as branching morphogenesis. Collagens are major components of the ECM of which basement membrane type IV and interstitial matrix type I are the most prevalent. Here we discuss how abnormal expression, proteolysis and structure of these collagens influence cellular functions to elicit multiple effects on tumors, including proliferation, initiation, invasion, metastasis, and therapy response.
Collapse
|
26
|
de Fougerolles AR, Koteliansky VE. Regulation of monocyte gene expression by the extracellular matrix and its functional implications. Immunol Rev 2002; 186:208-20. [PMID: 12234373 DOI: 10.1034/j.1600-065x.2002.18617.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
By binding to extracellular matrix (ECM) proteins, integrins integrate signals from outside the cell and transmit them inwards, thereby providing cells with information about location and allowing them to respond to stimuli in a manner appropriate to their environment. This is particularly important for monocytes and macrophages, given their wide distribution throughout the body and the vital role they play in immune and inflammatory responses. Integrin-mediated interaction of monocytes with ECM is a potent regulator of gene expression and is strongly synergized by the presence of growth factors. This synergy between growth factors and integrins is also apparent in the overlap seen in their signaling pathways. Integrin-mediated interaction with ECM results in increased expression of numerous inflammatory and immune response genes, revealing an important role for ECM-integrin interaction in affecting monocyte function and thus impacting on the development of pathologies. This is of particular relevance in the context of immune and inflammatory responses, where integrin-mediated adhesive interactions with the ECM-rich peripheral tissues are central to the localization of both resident and infiltrating monocytes at inflammatory sites. Here, we will review the functional effects of integrin-ECM interactions on monocytes, with particular attention to the regulation of gene expression by ECM and its functional implications.
Collapse
|
27
|
Düesberg U, Schneiders AM, Flieger D, Inchauspé G, Sauerbruch T, Spengler U. Natural cytotoxicity and antibody-dependent cellular cytotoxicity (ADCC) is not impaired in patients suffering from chronic hepatitis C. J Hepatol 2001; 35:650-7. [PMID: 11690712 DOI: 10.1016/s0168-8278(01)00194-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND/AIMS Recent observations suggest that natural killer (NK) cell activity might be impaired in chronic hepatitis C. However, to date antibody-dependent cellular cytotoxicity (ADCC) has not been studied in chronic hepatitis C in detail. METHODS Therefore, we investigated spontaneous and cytokine-induced (interleukin-2 and interferon-gamma) natural cytotoxicity and ADCC in 29 patients suffering from chronic hepatitis C and 19 healthy controls. Cytotoxicity was determined with a flow-cytometric assay, which can also assess monocyte cytotoxicity. As target cells we used the colorectal tumor cell line HT29 and the lymphoma cell line Raji. RESULTS We found no significant differences with respect to spontaneous cytotoxicity (HCV versus healthy controls (32 vs. 46%) and 17-1A specific ADCC (59 vs. 48%), even if isolated monocytes or NK cells were studied. Preincubation and stimulation of effector cells with cytokines increased both natural cytotoxicity and ADCC by 20-30%. However, natural cytotoxicity and ADCC after stimulation did not differ between the two groups. CONCLUSIONS Our data obtained with a long-term cytotoxicity assay do not reveal impaired cytolytic capacity of the innate immune system in chronic hepatitis C, even when isolated monocytes and NK cells were studied as effector cells.
Collapse
Affiliation(s)
- U Düesberg
- Department of Internal Medicine I, University of Bonn, Sigmund Freud-Strasse d25, D-53105 Bonn, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Yoneda Y, Yoshida R. The Role of T Cells in Allografted Tumor Rejection: IFN-γ Released from T Cells Is Essential for Induction of Effector Macrophages in the Rejection Site. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.12.6012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Allografted Meth A tumor rejection is T cell dependent, but T cells are inactive toward the allograft; rather, the main effector cells are allograft-induced macrophages (AIM) with MHC haplotype specificity. Here, we examined the role of T cells in the induction of AIM in the rejection site. On day 4.5 after i.p. transplantation of Meth A fibrosarcoma cells to C57BL/6 (B6) mice, we obtained a kind of precursor of AIM (pro-AIM) from the transplantation site by an enrichment technique involving adherence to serum-coated dishes. The noncytotoxic pro-AIM-rich population put into a diffusion chamber became cytotoxic against Meth A cells after 2 days in the peritoneal cavity of an untreated B6 mouse. Similar activation of the chambered B6 pro-AIM-rich population occurred in IFN-γ −/− B6 mice, whereas there was no activation when chambers containing an IFN-γ −/− mouse-derived pro-AIM-rich population were placed in normal or IFN-γ −/− mice, suggesting that IFN-γ is involved in the activation. RT-PCR experiments demonstrated that among bulk infiltrates T cells were the major producer of IFN-γ; and most of the cells in a T cell-eliminated pro-AIM population in a diffusion chamber kept for 2 days in a B6 mouse did not become AIM. Furthermore, IFN-γ −/− B6 mice could not reject allografted Meth A tumor cells, whereas the grafts were rejected by i.p. injections of IFN-γ into the mutant mice. These results indicate that IFN-γ released from allograft-induced T cells is essential for both the activation of a kind of pro-AIM to AIM in the transplantation site and the rejection of an allografted tumor.
Collapse
Affiliation(s)
- Yukio Yoneda
- Department of Cell Biology, Osaka Bioscience Institute, Suita, Osaka, Japan
| | - Ryotaro Yoshida
- Department of Cell Biology, Osaka Bioscience Institute, Suita, Osaka, Japan
| |
Collapse
|
29
|
Delpech B, Girard N, Olivier A, Maingonnat C, van Driessche G, van Beeumen J, Bertrand P, Duval C, Delpech A, Bourguignon J. The origin of hyaluronectin in human tumors. Int J Cancer 1997; 72:942-8. [PMID: 9378555 DOI: 10.1002/(sici)1097-0215(19970917)72:6<942::aid-ijc4>3.0.co;2-p] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The origin of tumor stroma hyaluronectin (HN), a glycoprotein that binds to hyaluronan (HA), has long remained unknown. Histological observations of human tumors suggest that tumor HN could originate from stroma fibroblasts, and in some cases from inflammatory cells. The fibroblast origin was confirmed by the discovery of HN-like antigen along with hyaluronan in culture medium of tumor-derived fibroblasts. An HA-binding protein was characterized in the culture medium of peripheral blood mononuclear cells (PBMC) in both normal subjects and tumor-bearing patients and was found to be human HN. Cultivated monocytes did not produce HA. HN was not related to the HA-binding site CD44. Sequencing of brain HN-derived peptides demonstrated that each determined peptide sequence was similar to a sequence of the proteoglycan PG-M/versican, suggesting that HN is the HA-binding moiety of the proteoglycan. One probe was synthesized from human PBMC by polymerase chain reaction with primers derived from HN sequences also found in versican. Northern blots were positive only with HN-producing cells. The main RNAs were in the 6-8 kb range, and there was a limited proportion of smaller RNA, which was compatible with the size expected from the HN molecular mass. Southern blotting of monocytes and tumor cells demonstrated that the gene was limited to a unique band. We conclude that HN, an extracellular component of brain, connective embryonic, inflammatory and tumoral tissues, is a PG-M/versican-derived molecule. Our results suggest that tumor HN, which originates from fibroblasts and monocytes of tumor stroma, is a molecular component of the host-tumor relationship and could play a role in the regulation of HA activity in oncogenesis.
Collapse
Affiliation(s)
- B Delpech
- Laboratoire d'Oncologie Moléculaire, Centre Henri-Becquerel, Rouen, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Cobb RR, Molony JL. Interleukin-1beta expression is induced by adherence and is enhanced by Fc-receptor binding to immune complex in THP-1 cells. FEBS Lett 1996; 394:241-6. [PMID: 8830651 DOI: 10.1016/0014-5793(96)00960-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Adherence of monocytes to endothelial cells and subsequently to basement membrane represents the initial steps in monocyte migration from the vasculature to the interstitium. We investigated the role of adhesion to endothelial cells and basement membrane in the induction of the cytokine IL-1beta. We demonstrated that mRNA for IL-1beta is induced in adherent THP-1 cells, but not in a matrix-specific manner. Adherence to fibrinogen, however, causes an increase in mRNA for IL-1beta. A background level of IL-1beta mRNA induction was observed in cells adherent to all matrices, including the non-specific human serum albumin substrate, as compared to non-adherent cells cultured in teflon troughs. In addition, antibodies to CD11a, CD11b, beta1 integrin, VLA4, (alpha)v(beta)3 (VNR), and ICAM-1 did not induce significant IL-1beta mRNA when THP-1 cells were adherent to those immunoglobulins. THP-1 cells adherent to immune complexes of anti-CD11a, anti-CD11b, anti-VLA4, anti-VNR, and anti-ICAM-1 showed greater mRNA induction than cells adherent to primary antibodies alone. THP-1 cells adherent to non-specific immune complexes gave the highest level of mRNA induction. Secretion of IL-1beta protein, measured by ELISA at 24 h, was greatest when cells were adherent to immobilized immune complexes or to fibrinogen. Our results demonstrate that a general adherence-induced increase in IL-1beta gene expression is greatly enhanced by the presence of immune complex.
Collapse
Affiliation(s)
- R R Cobb
- Department of Biology, Tanabe Research Laboratories, San Diego, CA 92121, USA
| | | |
Collapse
|
31
|
Barker K, Fan H, Carroll C, Kaplan G, Barker J, Hellmann W, Cohn ZA. Nonadherent cultures of human monocytes kill Mycobacterium smegmatis, but adherent cultures do not. Infect Immun 1996; 64:428-33. [PMID: 8550187 PMCID: PMC173781 DOI: 10.1128/iai.64.2.428-433.1996] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Human peripheral blood monocytes are permissive for the growth of Mycobacterium tuberculosis, but the fate of nonpathogenic Mycobacterium smegmatis in these cells is not known. Since M. smegmatis may be used as a host with which to express and screen for M. tuberculosis genes needed for survival in monocytes, we determined whether human peripheral blood monocytes could restrict the growth of Mycobacterium smegmatis. Adherent human peripheral blood monocytes were permissive for the growth of M. smegmatis, as measured by ex vivo [3H]uracil uptake. However, human peripheral blood monocytes which were cultured nonadherently in Teflon wells were able to restrict the growth of M. smegmatis while remaining permissive for the growth of M. tuberculosis H37Ra. The loss of viability of M. smegmatis in nonadherent cells was correlated with an increase in nonspacious phagocytic vacuoles. The killing of M. smegmatis was not blocked by NG-monomethyl-L-arginine, suggesting that it was not due to the production of reactive nitrogen intermediates. Incubation of the monocytes for 1 to 7 days before infection had no effect on the fate of M. smegmatis, suggesting that adherence versus nonadherence, and not differentiation, was the key determinant for the difference in functional ability. Nonadherent human peripheral blood monocytes may be a more appropriate model than adherent cells for the study of factors employed by bacterial to survive within monocytes and for selection screening of bacterial genes needed for intracellular survival.
Collapse
Affiliation(s)
- K Barker
- Rockefeller University, New York, New York 10021, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Thomson AW, Lu L, Murase N, Demetris AJ, Rao AS, Starzl TE. Microchimerism, dendritic cell progenitors and transplantation tolerance. Stem Cells 1995; 13:622-39. [PMID: 8590864 PMCID: PMC2963943 DOI: 10.1002/stem.5530130607] [Citation(s) in RCA: 145] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The recent discovery of multilineage donor leukocyte microchimerism in allograft recipients up to three decades after organ transplantation implies the migration and survival of donor stem cells within the host. It has been postulated that in chimeric graft recipients, reciprocal modulation of immune responsiveness between donor and recipient leukocytes may lead, eventually, to the induction of mutual immunologic nonreactivity (tolerance). A prominent donor leukocyte, both in human organ transplant recipients and in animals, has invariably been the bone marrow-derived dendritic cell (DC). These cells have been classically perceived as the most potent antigen-presenting cells but evidence also exists for their tolerogenicity. The liver, despite its comparatively heavy leukocyte content, is the whole organ that is most capable of inducing tolerance. We have observed that DC progenitors propagated from normal mouse liver in response to GM-CSF express only low levels of major histocompatibility complex (MHC) class II antigen and little or no cell surface B7 family T cell costimulatory molecules. They fail to activate resting naive allogeneic T cells. When injected into normal allogeneic recipients, these DC progenitors migrate to T-dependent areas of host lymphoid tissue, where some at least upregulate cell surface MHC class II. These donor-derived cells persist indefinitely, recapitulating the behavior pattern of donor leukocytes after the successful transplantation of all whole organs, but most dramatically after the orthotopic (replacement) engraftment of the liver. A key finding is that in mice, progeny of these donor-derived DC progenitors can be propagated ex vivo from the bone marrow and other lymphoid tissues of nonimmunosuppressed spontaneously tolerant liver allograft recipients. In humans, donor DC can also be grown from the blood of organ allograft recipients whose organ-source chimerism is augmented with donor bone marrow infusion. DC progenitors cannot, however, be propagated from the lymphoid tissue of nonimmunosuppressed cardiac-allografted mice that reject their grafts. These findings are congruent with the possibility that bidirectional leukocyte migration and donor cell chimerism play key roles in acquired transplantation tolerance. Although the cell interactions are undoubtedly complex, a discrete role can be identified for DC under well-defined experimental conditions. Bone marrow-derived DC progenitors (MHC class II+, B7-1dim, B7-2-) induce alloantigen-specific hyporesponsiveness (anergy) in naive T cells in vitro. Moreover, costimulatory molecule-deficient DC progenitors administered systemically prolong the survival of mouse heart or pancreatic islet allografts. How the regulation of donor DC phenotype and function relates to the balance between the immunogenicity and tolerogenicity of organ allografts remains to be determined.
Collapse
Affiliation(s)
- A W Thomson
- Pittsburgh Transplantation Institute, University of Pittsburgh Medical Center, Pennsylvania 15213, USA
| | | | | | | | | | | |
Collapse
|
33
|
Flieger D, Gruber R, Schlimok G, Reiter C, Pantel K, Riethmüller G. A novel non-radioactive cellular cytotoxicity test based on the differential assessment of living and killed target and effector cells. J Immunol Methods 1995; 180:1-13. [PMID: 7897241 DOI: 10.1016/0022-1759(94)00293-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Monocyte/macrophage-mediated cytotoxicity and antibody-dependent cellular cytotoxicity (ADCC) are slow processes, requiring cocultivation of effector and target cells for up to several days. Because of the high spontaneous release and possible reutilization of isotopic labels, the conventional radioactive release assays are unsuited for measuring long term cytotoxicity. We developed a non-radioactive flow cytometric assay for the quantitative analysis of cell-mediated cytotoxicity. Because dead cells can dissolve and disappear during the incubation period (lysis, phagocytosis), we determined the absolute numbers of living cells in the well. Prior to incubation the effector cells are stained with the red lipophilic fluorescent dye PKH26 and the target cells with the green fluorescent dye PKH2. At the end of the incubation (1-6 days) a defined number of bright fluorescent cell standards and propidium iodide for staining of dead cells was added to each well. Using flow cytometric analysis, we determined the ratio of targets to standards and calculated the absolute target cell number by multiplication with the known number of standards added. The main advantages of the assay are the possibility of extended incubation periods, the avoidance of radioactivity and its potential applicability to autologous culture systems, where effector and tumor cells are derived from the same patient. The assay opens new avenues for preclinical testing of tumor therapeutics such as monoclonal antibodies and/or cytokines.
Collapse
Affiliation(s)
- D Flieger
- Institute for Immunology, University of Munich, Germany
| | | | | | | | | | | |
Collapse
|
34
|
Audran R, Dazord L, Toujas L. Interactions between human macrophages and tumor cells in three-dimensional cultures. Cancer Immunol Immunother 1994; 39:299-304. [PMID: 7987861 PMCID: PMC11038061 DOI: 10.1007/bf01519982] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/1994] [Accepted: 07/04/1994] [Indexed: 01/28/2023]
Abstract
Human blood mononuclear cells were cultured for 7 days in hydrophobic plastic bags. Macrophages differentiated from monocytes and purified by elutriation were then cocultured with round-shaped aggregates of epithelial cells (spheroids). Spheroids prepared from the SK-MES-1 carcinoma cell line were cultured individually, under constant stirring, in multiwell plates coated with agarose. Macrophage/spheroid interactions were investigated under various experimental conditions. Macrophages activated with interferon gamma aggregated to each other and to spheroids, in contrast to control unactivated macrophages. Histological examination, after staining with a macrophage-specific monoclonal antibody, showed that both control and interferon-gamma-activated macrophages migrated between epithelial tumor cells and infiltrated the spheroids. The addition of anti-ICAM-1 monoclonal antibody inhibited macrophage homotypic aggregation as well as aggregation to and penetration into spheroids. The macrophages did not exert cytolytic effects, as judged by a chromium-51 release assay, but provoked a diminution of tritiated thymidine incorporation by tumor cells. Cytostatic activity was observed with effector: target ratios as low as 1:16, and was maximal (99% at a 1:1 E:T ratio) with macrophages differentiated in the presence of granulocyte/macrophage-colony-stimulating factor. The cytostatic effect was not related to tumor necrosis factor alpha secretion.
Collapse
Affiliation(s)
- R Audran
- Centre régional de lutte contre la cancer, Rennes, France
| | | | | |
Collapse
|
35
|
Armstrong JW, Chapes SK. Effects of extracellular matrix proteins on macrophage differentiation, growth, and function: comparison of liquid and agar culture systems. THE JOURNAL OF EXPERIMENTAL ZOOLOGY 1994; 269:178-87. [PMID: 8014614 DOI: 10.1002/jez.1402690303] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Both spaceflight and skeletal unloading suppress the haematopoietic differentiation of macrophages (Sonnenfeld et al., Aviat. Space Environ. Med., 61:648-653, 1990; Armstrong et al., J. Appl. Physiol., 75:2734-2739, 1993). The mechanism behind this reduction in haematopoiesis has yet to be elucidated. However, changes in bone marrow extracellular matrix (ECM) may be involved. To further understand the role of ECM products in macrophage differentiation, we have performed experiments evaluating the effects of fibronectin, laminin, collagen type I, and collagen type IV on macrophage development and function. Bone marrow-derived macrophages cultured on four different ECM substrates in liquid culture medium showed less growth than those cultured on plastic. Significant morphological differences were seen on each of the substrates used. Phenotypically and functionally, as measured by class II major histocompatibility molecule (MHCII) expression, MAC-2 expression, and the secretion of interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha), these macrophages were similar. In contrast, bone marrow-derived macrophages cultured in suspension, using agar, showed no difference in growth when exposed to ECM proteins. However, IL-6 and TNF-alpha secretion was affected by fibronectin, laminin, collagen type I, and collagen type IV in a concentration-dependent manner. We conclude that the ECM products fibronectin, laminin, collagen type I, and collagen type IV have profound effects on macrophage development and function. Additionally, we suggest that an ECM-supplemented agar culture system provides an environment more analogous to in vivo bone marrow than does a traditional liquid culture system.
Collapse
Affiliation(s)
- J W Armstrong
- Division of Biology, NASA Specialized Center of Research and Training, Kansas State University, Manhattan 66506
| | | |
Collapse
|
36
|
Armbrust T, Schwögler S, Zöhrens G, Ramadori G. C1 esterase inhibitor gene expression in rat Kupffer cells, peritoneal macrophages and blood monocytes: modulation by interferon gamma. J Exp Med 1993; 178:373-80. [PMID: 8340749 PMCID: PMC2191141 DOI: 10.1084/jem.178.2.373] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Kupffer cells (KC) represent the main part of the tissue macrophages. Beside phagocytosis of particulate material, involvement of KC in immunological and inflammatory reactions has been supposed. As C1 esterase inhibitor (C1-INH) is a serine protease inhibitor involved in such processes, the aim of this work was to study C1-INH synthesis in KC and, by comparison, in peritoneal macrophages (PM) and blood monocytes (MC) of the rat. C1-INH synthesis was studied on the protein level by biosynthetic labeling, immunoprecipitation, and sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis, and on the RNA level by Northern blotting of total RNA or by in situ hybridization. KC were found to express C1-INH gene spontaneously. C1-INH synthesis represents 1.3 +/- 0.2% of total protein synthesis at day 1 of culture and the absolute amount each cell synthesis remains constant during the whole time in culture. Transcripts of C1-INH were detected both in freshly isolated and in cultured KC. In contrast, spontaneous C1-INH gene expression was not detectable in freshly isolated PM, but only in cultured PM. In MC, C1-INH was not detectable at any time, whatever. Treatment of the cells with interferon gamma increased C1-INH synthesis in KC and in PM and caused an induction of C1-INH synthesis in MC. The results suggest that constitutive C1-INH synthesis is a functional marker for mature tissue macrophages.
Collapse
Affiliation(s)
- T Armbrust
- Abteilung für Gastroenterologie und Endokrinologie, Georg-August-Universität Göttingen, Germany
| | | | | | | |
Collapse
|
37
|
Fisker S, Kudahl K, Sonne O. In vivo inflammatory stimulation induces a transient change in the binding of thrombin to rat peritoneal macrophages. Exp Cell Res 1992; 201:145-53. [PMID: 1319345 DOI: 10.1016/0014-4827(92)90358-f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The binding of 125I-labeled thrombin to rat peritoneal macrophages isolated 20 h after the ip injection of thioglycollate broth or lipopolysaccharide decreased to 20% of the value found in resident macrophages due to a decrease in the number of receptors. The binding returned to normal values within a week after the injection. The decline parallelled more or less the Vmax for the 5'-nucleotidase activity. This decrease in the binding of thrombin could not be explained by an immigration of monocytes into the peritoneal cavity, since the binding of 125I-labeled alpha 2-macroglobulin-trypsin complex increased 4.5-fold in the same cell population due to an increase in the number of receptors, and blood monocytes do not bind alpha 2-macroglobulin-trypsin complex. The increase in the binding of alpha 2-macroglobulin-protease complex parallelled an increase in the incorporation of glucosamine, although the latter did not increase to the same extent. Engulfment of plasma membrane after phagocytosis did not result in a decreased binding of thrombin, but preincubation at 37 degrees C with concanavalin A caused a minor reduction in the binding. There was a positive correlation between the binding of alpha 2-macroglobulin-trypsin complex and the fraction of polymorphonuclear leukocytes in the peritoneal exudate and a negative correlation between the binding of thrombin and the fraction of polymorphonuclear leukocytes in the exudate, when the inflammation was induced by a milder stimulus, sterile NaCl, indicating a common signal for the polymorphonuclear leukocyte chemotaxis and the macrophage differentiation.
Collapse
Affiliation(s)
- S Fisker
- Institute of Physiology, University of Aarhus, Denmark
| | | | | |
Collapse
|
38
|
Pacifici R, Basilico C, Roman J, Zutter MM, Santoro SA, McCracken R. Collagen-induced release of interleukin 1 from human blood mononuclear cells. Potentiation by fibronectin binding to the alpha 5 beta 1 integrin. J Clin Invest 1992; 89:61-7. [PMID: 1729281 PMCID: PMC442819 DOI: 10.1172/jci115586] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PBMC express cell surface receptors for extracellular matrix components known as integrins. We have recently shown that ligand binding to one PBMC integrin, the collagen receptor alpha 2 beta 1, stimulates the secretion of interleukin 1 (IL-1). We have now investigated the role of fibronectin (Fn), an adherence protein that has binding sites for both PBMC and collagen, in the generation of the IL-1 response to collagen. In contrast to collagen, Fn did not stimulate IL-1 release but Fn-depleted serum decreased the release of IL-1 induced by collagen. A polyclonal antiserum directed against Fn also decreased the collagen-induced IL-1 secretion. The IL-1 response to collagen from cells incubated in Fn-depleted serum was restored by the addition of either purified Fn or the 120-kD cell-binding fragment of Fn, which contains the cell-binding site but not the collagen-binding domain. Smaller Arg-Gly-Asp (RGD) peptides failed to enhance the PBMC response to collagen but inhibited in a concentration-dependent fashion the potentiating effect Fn. As expected, a MAb against the alpha 2 beta 1 collagen receptor decreased collagen-induced IL-1 release. However collagen-induced IL-1 release was also inhibited by a MAb against the alpha 5 beta 1 Fn receptor. The effect of the two MAbs was not additive, suggesting that the occupancy of both receptors by ligands is required in order for collagen to induce an maximal response from PBMC. The mechanism by which Fn exerts its effect remains unknown. However, flow-cytometric analysis revealed that Fn does not alter expression of the alpha2beta1 receptor on PBMC. These data demonstrate a potentiating effect of Fn on the collagen-induced secretion of IL-1 from human PBMC and suggest that this effect is mediated via the integrin alpha5beta1. These findings indicate a complex interactive role for specific integrin receptors in the regulation of the mononuclear cell immune response.
Collapse
Affiliation(s)
- R Pacifici
- Division of Endocrinology and Bone Metabolism, Jewish Hospital of St. Louis, Washington University Medical Center, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
39
|
Martin A, Audran R, Collet B, Lancien G, Toujas L. Monoclonal antibody AMH152 reacts with human monocytes in culture and with inflammatory macrophages. RESEARCH IN IMMUNOLOGY 1992; 143:39-47. [PMID: 1565845 DOI: 10.1016/0923-2494(92)80078-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Monoclonal antibodies (mAb) raised against human peritoneal macrophages were selected for their non-reactivity with freshly sampled blood cells. One of these mAb, AMH152, initially non-reactive, bound to monocytes after 18 h of culture, a property which was not shared by an unrelated antibody of the same isotype (IgG1). The induction of the expression of the antigen detected by AMH152 on monocytes in culture was not influenced by the addition of serum or by the substrate used, plastic that favoured adhesion or teflon bags. Overnight incubation at 4 degrees C in adhesion conditions did not enable antigen expression. A 1-h treatment with phorbol myristate acetate or formyl-methionyl-leucyl-phenylalanine did not increase AMH152 binding. Culturing monocytes with cycloheximide tended to inhibit antigen expression. These observations suggested that antigen expression represents an active phenomenon, requiring protein synthesis. The antigen recognized by mAb AMH152 could be visualized on sections of formalin-fixed and paraffin-embedded tissues. Macrophages of healthy lymphoid organs and tissues that expressed CD68 antigen failed to bind AMH152. In contrast, chronic inflammatory lesions, like those of sarcoidosis, tuberculosis and cat scratch disease, contained epithelioid and multinucleated giant cells that reacted with AMH152. In serous exudates of cancer metastases, 10-40% of macrophages were also stained. The antigenic material was essentially present at the cell periphery. Thus, mAb AMH152 recognized a surface antigen, detectable on paraffin-embedded tissue sections, and which accompanied differentiation of monocytes into inflammatory cells. The expression of this antigen on monocytes in culture suggests that these cells underwent an activation process, even when maintained for some hours in teflon bags and in a serum-free medium.
Collapse
Affiliation(s)
- A Martin
- Laboratoire d'Immunologie-Immunothérapie, Centre Régional de Lutte contre le Cancer, Rennes, France
| | | | | | | | | |
Collapse
|
40
|
Gudewicz PW, Frewin MB. Surface contact modulation of inflammatory macrophage antibody dependent cytotoxicity and prostanoid release. J Cell Physiol 1991; 149:195-201. [PMID: 1660899 DOI: 10.1002/jcp.1041490204] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Adherence to extracellular matrix proteins modulates the functional and secretory activities of mononuclear phagocytes, although the mechanisms regulating these adherence-dependent changes are poorly understood. In this study, the ability of rat inflammatory peritoneal macrophages (PM) to adhere to an endothelial cell-derived extracellular matrix or a denatured collagen/fibronectin-coated surface and perform antibody dependent cell cytotoxicity (ADCC) and secrete reactive oxygen intermediates was compared with PM adherent to tissue culture plastic. Prostaglandin E2 (PGE2) and thromboxane B2 (TxB2), two major cyclooxygenase products released by inflammatory macrophages, were also measured by PM adherent to the protein coated surfaces. Rat exudate PM were equally adherent to tissue culture plastic or wells coated with either endothelial cell derived matrix or denatured collagen (gelatin)/fibronectin. PM adherent to a denatured collagen/fibronectin-coated wells demonstrated significantly less cytolytic activity (15 +/- 2% lysis) when compared with either tissue culture plastic adherent PM (43 +/- 7% lysis) or PM adherent to extracellular matrix (59 +/- 11% lysis). PM adherent to extracellular matrix released twofold more TxB2 than plastic adherent PM, while PM adherent to denatured collagen/fibronectin released 40% more PGE2 than cells adherent to tissue culture plastic or 80% more PGE2 than PM adherent to the extracellular matrix. PM adherent to denatured collagen/fibronectin release less superoxide anion (27 +/- .9 nmoles/10(6) PM) than PM adherent to either tissue culture plastic (43 +/- 1 nmoles/10(6) PM) or the extracellular matrix (60 +/- 0.5 nmoles/10(6) PM). Furthermore, incubation of plastic adherent PM with exogenous PGE2 reduced superoxide production in a dose-dependent manner. These results demonstrate that the inhibition of ADCC and secretion of reactive oxygen intermediates by PM adherent to a denatured collagen/fibronectin surface correlated with an increased release of the immunosuppressive prostanoid PGE2. Furthermore, the addition of exogenous PGE2 to plastic adherent PM reproduced the depression in ADCC and superoxide anion production observed by PM adherent to a denatured collagen/fibronectin surface. These studies suggest that the increased production and release of PGE2 by inflammatory macrophages adherent to a denatured collagen surface may act to suppress cytotoxic mechanisms and thereby constitutes part of an autocrine feedback mechanism regulating macrophage function during wound injury.
Collapse
Affiliation(s)
- P W Gudewicz
- Department of Physiology and Cell Biology, Albany Medical College, New York 12208
| | | |
Collapse
|
41
|
Liesveld JL, Frediani KE, Winslow JM, Duerst RE, Abboud CN. Cytokine effects and role of adhesive proteins and Fc receptors in human macrophage-mediated antibody dependent cellular cytotoxicity. J Cell Biochem 1991; 45:381-90. [PMID: 1675219 DOI: 10.1002/jcb.240450412] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Mononuclear phagocytes participate in host immunological defense against tumors. We have investigated the role of selected recombinant cytokines on human macrophage-mediated tumor cytotoxicity in vitro utilizing a human colon cancer cell line target, SW1116, and murine monoclonal antibody 17-1A. Blood monocytes were kept in continuous culture to allow differentiation into macrophages. Maximum antibody dependent cellular cytotoxicity (ADCC) as measured in a 3H-thymidine release assay occurred after culturing the monocytes for 5-7 days. Human recombinant macrophage colony stimulating factor (CSF) (1,000 U/ml) did not increase ADCC above control levels whereas recombinant human granulocyte-macrophage colony stimulating factor, interleukin 4, and interleukin 3 were all capable of increasing ADCC. Antibodies to the CD11/CD18 integrin receptors did not significantly inhibit ADCC. When the ADCC incubation occurred in the presence of antibodies to the human Fc receptors, ADCC was inhibited significantly only by anti-FcRIII (3G8). A role for tumor necrosis factor alpha or other soluble mediators of cytotoxicity was not demonstrable in this system. These studies suggest avenues for manipulation and augmentation of macrophage-mediated antitumor ADCC.
Collapse
Affiliation(s)
- J L Liesveld
- Department of Medicine, University of Rochester School of Medicine, New York
| | | | | | | | | |
Collapse
|
42
|
Abstract
Proteoglycans are produced by all types of haemopoietic cells including mature cells and the undifferentiated stem cells. The proteinase-resistant secretory granule proteoglycan (serglycin; Ref. 14), is the most prevalent and best characterised of these proteoglycans. Although its complete pattern of distribution in the haemopoietic system is unknown, serglycin has been identified in the mast cells, basophils and NK cells, in which secretion is regulated, and in HL-60 cells and a monocytoid cell line (Kolset, S.O., unpublished data) in which secretion is constitutive. Proteinase-resistant proteoglycans have been detected in human T-lymphocytes and murine stem cells (FDCP-mix) and the core proteins may be closely related to serglycin. A variety of glycosaminoglycan chains are assembled on the serglycin protein and it is likely that this class of proteoglycan can carry out a wide variety of functions in haemopoietic cells including the regulation of immune responses, inflammatory reactions and blood coagulation. There is strong evidence that in mast cells, NK cells and platelets, the proteoglycans are complexed to basic proteins (including enzymes and cytolytic agents) and amines in secretory granules and such complexes may dissociate following secretion from the cell. The stability of the complexes may be regulated by the ambient pH which may be acidic in the granules and neutral or above in the external medium. However, proteinase-proteoglycan complexes in mast cell granules seem to remain stable after secretion and it has been proposed that the proteoglycan regulates activity of proteinases released into the pericellular domain. The functions of proteoglycans which are constitutively secreted from cells are less clear. If cells have no requirement for storage of basic proteins why do they utilise the same design of proteoglycan as cells which accumulate secretory material prior to regulated release? We should stress that the so-called constitutive secretory pathway has been identified in haemopoietic cells in culture, which are usually maintained and grown in the presence of mitogenic factors (e.g., IL-2, IL-3). the cells are therefore activated and it has not been established that continuous proteoglycan secretion occurs in quiescent cells circulating in the peripheral blood. It is possible that lymphocytes, monocytes and macrophages, in which the constitutive secretion pathway operates in vitro, may store proteoglycan in vivo unless stimulated by mitogens or other activating agents.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- S O Kolset
- Institute of Medical Biology, University of Tromsö, Norway
| | | |
Collapse
|
43
|
Newman SL, Tucci MA. Regulation of human monocyte/macrophage function by extracellular matrix. Adherence of monocytes to collagen matrices enhances phagocytosis of opsonized bacteria by activation of complement receptors and enhancement of Fc receptor function. J Clin Invest 1990; 86:703-14. [PMID: 2168442 PMCID: PMC296784 DOI: 10.1172/jci114766] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In inflammation monocytes emigrate from the peripheral circulation into an extravascular area rich in extracellular matrix proteins. In this milieu, phagocytes ingest and kill invading pathogens. In the present studies, we found that monocytes adhered to type I collagen gels phagocytized 2.5-12-fold more opsonized Escherichia coli, Staphylococcus aureus, Streptococcus pyogenes, and Streptococcus pneumoniae than plastic-adherent monocytes. The rate of phagocytosis and the number of bacteria ingested by collagen-adherent monocytes was equal to, or greater than, the number of bacteria ingested by 7-d cultured macrophages (M phi). Although both collagen- and plastic-adherent monocytes were bactericidal for E. coli and S. aureus, more bacteria were killed by collagen-adherent monocytes by virtue of their enhanced phagocytic capacity. Cultured M phi only were bacteriostatic. Adherence of monocytes to collagen gels activated C receptors (CR) types 1 and 3 for phagocytosis, and enhanced Fc receptor (FcR)-mediated phagocytosis. Collagen- and plastic-adherent monocytes produced equivalent amounts of superoxide anion in response to phorbol myristate acetate and opsonized zymosan. Thus, the enhanced phagocytosis and killing of opsonized bacteria by collagen-adherent monocytes appear to be by regulation of the function of membrane CR and FcR, without apparent enhancement of the respiratory burst. These data suggest that adherence of monocytes to the extracellular matrix during inflammation may rapidly activate these cells for enhanced phagocytic bactericidal activity.
Collapse
Affiliation(s)
- S L Newman
- Department of Internal Medicine, University of Cincinnati Medical Center, Ohio 45267
| | | |
Collapse
|
44
|
Ganter U, Bauer J, Majello B, Gerok W, Ciliberto G. Characterization of mononuclear-phagocyte terminal maturation by mRNA phenotyping using a set of cloned cDNA probes. EUROPEAN JOURNAL OF BIOCHEMISTRY 1989; 185:291-6. [PMID: 2583184 DOI: 10.1111/j.1432-1033.1989.tb15114.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The terminal step in the maturation of mononuclear cells from circulating monocytes to resident macrophages is accompanied by dramatic changes in cell morphology and physiology. Applying a cultivation system which allows peripheral monocytes to undergo terminal maturation in vitro under absolutely endotoxin-free conditions, we have determined the pattern of expression of a set of eight genes by mRNA phenotyping. The results can be summarized as follows: the two protease inhibitors alpha 1-antitrypsin and alpha 2-macroglobulin show a inverse pattern of expression. alpha 1-Antitrypsin mRNA is repressed, alpha 2-macroglobulin mRNA is strongly induced during maturation to macrophages. Therefore, these two genes are excellent markers of the terminal maturation. In addition, ferritin-light-chain mRNA progressively increases during the course of differentiation, providing a further marker for maturation. Gene expression as a function of activation was studied in mononuclear cells stimulated with bacterial endotoxin (lipopolysaccharide). In monocytes, complement-factor-B, interleukin-1 and interleukin-6 mRNAs are drastically induced upon lipopolysaccharide activation whereas lysozyme RNA is strongly repressed. However, the ability of all four genes to respond to endotoxin was markedly diminished or abolished in mature macrophages, indicating that susceptibility to a certain type of activation may be restricted to a specific stage of maturation. Our data show that mRNA phenotyping is excellently suited for the characterization of the differentiation and activation state of mononuclear phagocytes.
Collapse
Affiliation(s)
- U Ganter
- Medizinische Universitätsklinik, Freiburg
| | | | | | | | | |
Collapse
|
45
|
Uhlin-Hansen L, Eskeland T, Kolset SO. Modulation of the Expression of Chondroitin Sulfate Proteoglycan in Stimulated Human Monocytes. J Biol Chem 1989. [DOI: 10.1016/s0021-9258(18)63789-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
46
|
Affiliation(s)
- D L Thiele
- Department of Internal Medicine, University of Texas, Southwestern Medical Center, Dallas 75235
| |
Collapse
|
47
|
Griffith RL, Virella GT, Stevenson HC, Lopes-Virella MF. Low density lipoprotein metabolism by human macrophages activated with low density lipoprotein immune complexes. A possible mechanism of foam cell formation. J Exp Med 1988; 168:1041-59. [PMID: 3171477 PMCID: PMC2189038 DOI: 10.1084/jem.168.3.1041] [Citation(s) in RCA: 135] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Human macrophages play a key role in atherogenesis and are believed to be the progenitors of the cholesteryl ester (CE)-laden foam cells present in early atherosclerotic lesions. Several mechanisms by which macrophages accumulate CE have been recently described. One involves a perturbation in LDL metabolism subsequent to macrophage activation. Thus, we decided to study the effect of macrophage activation by immune complexes on N-LDL metabolism. Initially, LDL-containing immune complexes (LDL-IC) were chosen, since increased plasma levels of these IC have been reported in patients with coronary heart disease. Human macrophages stimulated for 22 h with LDL-IC (250 micrograms/ml) and incubated afterwards for 20 h with 10 micrograms/ml 125I-N-LDL showed a six- and fourfold increase in the accumulation and degradation, respectively, of 125I-N-LDL over the values observed in nonstimulated cells. Scatchard analysis of 125I-N-LDL-specific binding suggests an increase (20-fold) in the number of LDL receptors in macrophages stimulated with LDL-IC. We studied other immune complexes varying in size and antigen composition. Some of the IC were able to stimulate, although to a lesser degree, the uptake of N-LDL by macrophages. Lipoprotein IC are more efficient and have the greatest capacity to increase N-LDL uptake and CE accumulation. We conclude that human macrophage activation by LDL-IC leads to an increase in LDL receptor activity and promotes in vitro foam cell formation.
Collapse
Affiliation(s)
- R L Griffith
- Department of Basic and Clinical Immunology and Microbiology, Medical University of South Carolina, Charleston 29425
| | | | | | | |
Collapse
|
48
|
Andreesen R, Gadd S, Costabel U, Leser HG, Speth V, Cesnik B, Atkins RC. Human macrophage maturation and heterogeneity: restricted expression of late differentiation antigens in situ. Cell Tissue Res 1988; 253:271-9. [PMID: 3044599 DOI: 10.1007/bf00222281] [Citation(s) in RCA: 32] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Terminal maturation of human macrophages is an important step for creation of cell diversity amongst site-specific subpopulations and their functional competence in situ. As monocytes undergo differentiation in vitro, they start to express lineage-restricted antigens specific for differentiation stages beyond the blood monocyte level as detected by monoclonal antibodies of the MAX series. We have analyzed the expression of MAX.1, MAX.2, MAX.3 and MAX.11 on exudate-type macrophages from pleural and peritoneal cavity and the alveolar space, as well as on resident and activated tissue macrophages in cryostat sections of spleen, lymph node, tonsil, liver, gut mucosa, skin, placenta, kidney and bone. It was found that "free" macrophages in serous cavities expressed MAX antigens in a heterogenous pattern, whereas none of the organ-specific tissue macrophages subsets did so (with the exception being the weak label of MAX.2 on Kupffer cells). Only during allograft rejection were infiltrating macrophages found to express MAX antigens but not at sites of "non-specific" inflammation or granuloma formation. However, Cyclosporin A treatment seems to suppress the induction of MAX antigen expression on intragraft macrophages. In addition, freshly harvested MAX-negative exudate macrophages converted to the complete Max+ phenotype on further cultivation. Isolated Kupffer cells were able only to express the MAX.2 antigen in culture but still did not react with the MAX.1 and MAX.3 monoclonal antibodies. Some MAX antigens are co-expressed on glomerular mesangial cells, dendritic reticulum cells and placental cells (MAX.1/.11) as well as on capillary endothelium within tissues of active immune response (MAX.2).(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- R Andreesen
- Department of Nephrology, Monash University, Prince Henry's Hospital, Melbourne, Australia
| | | | | | | | | | | | | |
Collapse
|
49
|
Kolset SO. Interactions between extracellular matrix components and proteoglycans released from monocytesin vitro. Glycoconj J 1988. [DOI: 10.1007/bf01061207] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
50
|
Richter M, Kazaniwsky N. Surface receptors and immune activity of purified human circulating mononuclear cells. III. The mechanisms of lysis, by lymphocytes and monocytes, of target cells in the lymphocyte-dependent and monocyte-dependent ADCC, NK, NOCC, and MICC cytotoxic reactions. Cell Immunol 1987; 108:297-312. [PMID: 3621351 DOI: 10.1016/0008-8749(87)90214-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The abilities of unfractionated mononuclear cells (MNC), monocytes (98-99% pure), and lymphocytes (98-99% pure) to carry out the lysis of target cells in the ADCC, NK, NOCC, and MICC assays were compared. Lymphocytes by themselves were able to lyse the CRBC (ADCC), K-562 (NK), and RRBC (MICC) target cells. The monocytes were very effective in the lysis of the CRBC (MICC) target cells. However, the lysis of two other target cells--RRBC (NOCC) and HRBC (ADCC)--required the simultaneous presence of both lymphocytes and monocytes in order to effect optimal lysis. Soluble factor(s) secreted by the cytotoxic cells capable of lysing the target cells were detected only in the NK assay. The activity of the soluble cytotoxic factor (NKCF) was only 25-40% of that exhibited by the cytotoxic NK cells and it was secreted by the cytotoxic cells after 48 hr of culture and not 24 hr of culture which is the usual assay condition. The NKCF was cytotoxic only to the NK target cells and not to the target cells used in the ADCC, NOCC, and MICC cytotoxic assays. Different classes of lymphocytes were cytotoxic in the monocyte-independent assays [ADCC (CRBC), NK (K-562), and MICC (RRBC)]. The null lymphocytes and the T lymphocytes were the primary cytotoxic cells in the ADCC and MICC assays, respectively, whereas the T, B, and null cells were almost equally cytotoxic in the NK assay. With respect to the monocyte-dependent assays [ADCC (HRBC), NOCC (RRBC), and MICC (CRBC)], the cytotoxic activity of any one class of lymphocytes failed to approach that of the unfractionated MNC. The T cells were the most cytotoxic; the B cells exhibited limited cytotoxic activity in only the ADCC assay and the null cells showed no cytotoxic activity. However, the combination of T and non-T cells and, to a lesser extent, T and B cells, exhibited much greater cytotoxic activity than the individual cells and together were as cytotoxic as the unfractionated MNC. It is concluded that, depending upon the selection of the target cells, lysis in the ADCC, NK, NOCC, and MICC assays may be effected by lymphocytes only, by monocytes only, by both monocytes and lymphocytes, or as a result of lymphocyte-monocyte collaboration. In the latter instance more than one class of lymphocytes must be present in order for maximum cytotoxic activity to be expressed.
Collapse
|