1
|
Guo H, Li W, Guo Y, Chen N, Cui J. Molecular classification of small cell lung cancer subtypes: Characteristics, prognostic factors, and clinical translation. Chin Med J (Engl) 2024; 137:130-139. [PMID: 37660289 PMCID: PMC10798698 DOI: 10.1097/cm9.0000000000002693] [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: 02/15/2023] [Indexed: 09/04/2023] Open
Abstract
ABSTRACT Small cell lung cancer (SCLC) is a highly malignant tumor with a very poor prognosis; therefore, more effective treatments are urgently needed for patients afflicted with the disease. In recent years, emerging molecular classifications based on key transcription factors of SCLC have provided more information on the tumor pathophysiology, metastasis, immune microenvironment, and acquired therapeutic resistance and reflected the intertumoral heterogeneity of the various SCLC phenotypes. Additionally, advances in genomics and single-cell sequencing analysis have further revealed the high intratumoral heterogeneity and plasticity of the disease. Herein, we review and summarize these recent lines of evidence and discuss the possible pathogenesis of SCLC.
Collapse
Affiliation(s)
| | | | | | | | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
2
|
Yuan M, Zhao Y, Arkenau HT, Lao T, Chu L, Xu Q. Signal pathways and precision therapy of small-cell lung cancer. Signal Transduct Target Ther 2022; 7:187. [PMID: 35705538 PMCID: PMC9200817 DOI: 10.1038/s41392-022-01013-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 03/05/2022] [Accepted: 04/29/2022] [Indexed: 12/24/2022] Open
Abstract
Small-cell lung cancer (SCLC) encounters up 15% of all lung cancers, and is characterized by a high rate of proliferation, a tendency for early metastasis and generally poor prognosis. Most of the patients present with distant metastatic disease at the time of clinical diagnosis, and only one-third are eligible for potentially curative treatment. Recently, investigations into the genomic make-up of SCLC show extensive chromosomal rearrangements, high mutational burden and loss-of-function mutations of several tumor suppressor genes. Although the clinical development of new treatments for SCLC has been limited in recent years, a better understanding of oncogenic driver alterations has found potential novel targets that might be suitable for therapeutic approaches. Currently, there are six types of potential treatable signaling pathways in SCLC, including signaling pathways targeting the cell cycle and DNA repair, tumor development, cell metabolism, epigenetic regulation, tumor immunity and angiogenesis. At this point, however, there is still a lack of understanding of their role in SCLC tumor biology and the promotion of cancer growth. Importantly optimizing drug targets, improving drug pharmacology, and identifying potential biomarkers are the main focus and further efforts are required to recognize patients who benefit most from novel therapies in development. This review will focus on the current learning on the signaling pathways, the status of immunotherapy, and targeted therapy in SCLC.
Collapse
Affiliation(s)
- Min Yuan
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China
| | - Yu Zhao
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China
| | | | - Tongnei Lao
- Department of Oncology, Centro Medico BO CHI, Macao, SAR, China
| | - Li Chu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China.
| | - Qing Xu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, 200072, Shanghai, China.
| |
Collapse
|
3
|
Nisar S, Yousuf P, Masoodi T, Wani NA, Hashem S, Singh M, Sageena G, Mishra D, Kumar R, Haris M, Bhat AA, Macha MA. Chemokine-Cytokine Networks in the Head and Neck Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22094584. [PMID: 33925575 PMCID: PMC8123862 DOI: 10.3390/ijms22094584] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/03/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are aggressive diseases with a dismal patient prognosis. Despite significant advances in treatment modalities, the five-year survival rate in patients with HNSCC has improved marginally and therefore warrants a comprehensive understanding of the HNSCC biology. Alterations in the cellular and non-cellular components of the HNSCC tumor micro-environment (TME) play a critical role in regulating many hallmarks of cancer development including evasion of apoptosis, activation of invasion, metastasis, angiogenesis, response to therapy, immune escape mechanisms, deregulation of energetics, and therefore the development of an overall aggressive HNSCC phenotype. Cytokines and chemokines are small secretory proteins produced by neoplastic or stromal cells, controlling complex and dynamic cell-cell interactions in the TME to regulate many cancer hallmarks. This review summarizes the current understanding of the complex cytokine/chemokine networks in the HNSCC TME, their role in activating diverse signaling pathways and promoting tumor progression, metastasis, and therapeutic resistance development.
Collapse
Affiliation(s)
- Sabah Nisar
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
| | - Parvaiz Yousuf
- Department of Zoology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India;
| | - Tariq Masoodi
- Department of Genomic Medicine, Genetikode 400102, India;
| | - Nissar A. Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India;
| | - Sheema Hashem
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
| | - Mayank Singh
- Departmental of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India;
| | | | - Deepika Mishra
- Centre for Dental Education and Research, Department of Oral Pathology and Microbiology, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Rakesh Kumar
- Centre for Advanced Research, School of Biotechnology and Indian Council of Medical Research, Shri Mata Vaishno Devi University, Katra 182320, India;
| | - Mohammad Haris
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
- Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar
| | - Ajaz A. Bhat
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
- Correspondence: (A.A.B.); or (M.A.M.); Tel.: +974-40037703 (A.A.B.); +91-8082326900 (M.A.M.)
| | - Muzafar A. Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora 192122, India
- Correspondence: (A.A.B.); or (M.A.M.); Tel.: +974-40037703 (A.A.B.); +91-8082326900 (M.A.M.)
| |
Collapse
|
4
|
Abstract
Small-cell lung cancer (SCLC) represents about 15% of all lung cancers and is marked by an exceptionally high proliferative rate, strong predilection for early metastasis and poor prognosis. SCLC is strongly associated with exposure to tobacco carcinogens. Most patients have metastatic disease at diagnosis, with only one-third having earlier-stage disease that is amenable to potentially curative multimodality therapy. Genomic profiling of SCLC reveals extensive chromosomal rearrangements and a high mutation burden, almost always including functional inactivation of the tumour suppressor genes TP53 and RB1. Analyses of both human SCLC and murine models have defined subtypes of disease based on the relative expression of dominant transcriptional regulators and have also revealed substantial intratumoural heterogeneity. Aspects of this heterogeneity have been implicated in tumour evolution, metastasis and acquired therapeutic resistance. Although clinical progress in SCLC treatment has been notoriously slow, a better understanding of the biology of disease has uncovered novel vulnerabilities that might be amenable to targeted therapeutic approaches. The recent introduction of immune checkpoint blockade into the treatment of patients with SCLC is offering new hope, with a small subset of patients deriving prolonged benefit. Strategies to direct targeted therapies to those patients who are most likely to respond and to extend the durable benefit of effective antitumour immunity to a greater fraction of patients are urgently needed and are now being actively explored.
Collapse
Affiliation(s)
- Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Elisabeth Brambilla
- Institute for Advanced Biosciences, Université Grenoble Alpes, Grenoble, France
| | - Corinne Faivre-Finn
- Department of Clinical Oncology, The Christie Hospital NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Julien Sage
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| |
Collapse
|
5
|
Popper H. Primary tumor and metastasis-sectioning the different steps of the metastatic cascade. Transl Lung Cancer Res 2020; 9:2277-2300. [PMID: 33209649 PMCID: PMC7653118 DOI: 10.21037/tlcr-20-175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Patients with lung cancer in the majority die of metastases. Treatment options include surgery, chemo- and radiotherapy, targeted therapy by tyrosine kinase inhibitors (TKIs), and immuno-oncologic treatment. Despite the success with these treatment options, cure of lung cancer is achieved in only a very small proportion of patients. In most patients’ recurrence and metastasis will occur, and finally kill the patient. Metastasis is a multistep procedure. It requires a change in adhesion of tumor cells for detachment from their neighboring cells. The next step is migration either as single cells [epithelial-mesenchymal transition (EMT)], or as cell clusters (hybrid-EMT or bulk migration). A combination of genetic changes is required to facilitate migration. Then tumor cells have to orient themselves along matrix proteins, detect oxygen concentrations, prevent attacks by immune cells, and induce a tumor-friendly switch of stroma cells (macrophages, myofibroblasts, etc.). Having entered the blood stream tumor cells need to adapt to shear stress, avoid being trapped by coagulation, but also use coagulation in small veins for adherence to endothelia, and express homing molecules for extravasation. Within a metastatic site, tumor cells need a well-prepared niche to establish a metastatic focus. Tumor cells again have to establish a vascular net for maintaining nutrition and oxygen supply, communicate with stroma cells, grow out and set further metastases. In this review the different steps will be discussed with a focus on pulmonary carcinomas. The vast amount of research manuscripts published so far are not easy to analyze: in most reports’ single steps of the metastatic cascade are interpreted as evidence for the whole process; for example, migration is interpreted as evidence for metastasis. In lung cancer most often latency periods are shorter, in between 1–5 years. In other cases, despite widespread migration occurs, tumor cells die within the circulation and do not reach a metastatic site. Therefore, migration is a requisite, but does not necessarily predict metastasis. The intention of this review is to point to these different aspects and hopefully provoke research directed into a more functional analysis of the metastatic process.
Collapse
Affiliation(s)
- Helmut Popper
- Institute of Pathology, Medical University of Graz, Graz, Austria
| |
Collapse
|
6
|
Sung GH, Chang H, Lee JY, Song SY, Kim HS. Pancreatic-cancer-cell-derived trefoil factor 2 impairs maturation and migration of human monocyte-derived dendritic cells in vitro. Anim Cells Syst (Seoul) 2018; 22:368-381. [PMID: 30533259 PMCID: PMC6282439 DOI: 10.1080/19768354.2018.1527721] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/07/2018] [Accepted: 09/05/2018] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is a challenging disease with a high mortality rate. While the importance of crosstalk between cancer and immune cells has been well documented, the understanding of this complex molecular network is incomplete. Thus, identification of the secreted proteins contributing to the immunosuppressive microenvironment in pancreatic cancer is crucial for effective diagnosis and/or therapy. We utilized a public microarray dataset (GSE16515) from the Gene Expression Omnibus database to identify genes for secreted proteins in pancreatic cancer. RT-PCR and ELISA of the pancreatic cancer cell lines validated the cellular origin of the selected genes. For functional assay of the selected proteins, we utilized human-monocyte-derived dendritic cells (DCs). From the list of the secreted proteins, trefoil factor 2 (TFF2) was further examined as a potential chemokine/cytokine. While TFF2 did not significantly affect the phenotypic maturation and the allostimulatory capacity of DCs, TFF2 preferentially attracted immature (but not mature) DCs and inhibited their endocytic activity. Our data suggest that TFF2 from pancreatic cancer cells may attract immature DCs and affect the initial stage of DC maturation, thereby contributing to the induction of immune tolerance against pancreatic cancer.
Collapse
Affiliation(s)
- Gi-Ho Sung
- Institute for Healthcare and Life Science and Institute for Translational and Clinical Research, Catholic Kwandong University International St. Mary’s Hospital, Incheon, Republic of Korea
- Department of Microbiology, Catholic Kwandong University College of Medicine, Gangneung-si, Gangwon-do, Republic of Korea
| | - Hyun Chang
- Hematology and Medical Oncology, International St Mary’s Hospital, Catholic Kwandong University College of Medicine, Incheon, Republic of Korea
| | - Ji-Yong Lee
- Department of Anatomy, Yonsei University Wonju College of Medicine, Wonju-si, Gangwon-do, Republic of Korea
| | - Si Young Song
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Han-Soo Kim
- Institute for Healthcare and Life Science and Institute for Translational and Clinical Research, Catholic Kwandong University International St. Mary’s Hospital, Incheon, Republic of Korea
- Department of Biomedical Sciences, College of Medical Convergence, Catholic Kwandong University, Gangneung-si, Gangwon-do, Republic of Korea
| |
Collapse
|
7
|
Abstract
Metastasis in lung cancer is a multifaceted process. In this review, we will dissect the process in several isolated steps such as angiogenesis, hypoxia, circulation, and establishment of a metastatic focus. In reality, several of these processes overlap and occur even simultaneously, but such a presentation would be unreadable. Metastasis requires cell migration toward higher oxygen tension, which is based on changing the structure of the cell (epithelial-mesenchymal transition), orientation within the stroma and stroma interaction, and communication with the immune system to avoid attack. Once in the blood stream, cells have to survive trapping by the coagulation system, to survive shear stress in small blood vessels, and to find the right location for extravasation. Once outside in the metastatic locus, tumor cells have to learn the communication with the “foreign” stroma cells to establish vascular supply and again express molecules, which induce immune tolerance.
Collapse
|
8
|
Zong J, Keskinov AA, Shurin GV, Shurin MR. Tumor-derived factors modulating dendritic cell function. Cancer Immunol Immunother 2016; 65:821-33. [PMID: 26984847 PMCID: PMC11028482 DOI: 10.1007/s00262-016-1820-y] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/26/2016] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DC) play unique and diverse roles in the tumor occurrence, development, progression and response to therapy. First of all, DC can actively uptake tumor-associated antigens, process them and present antigenic peptides to T cells inducing and maintaining tumor-specific T cell responses. DC interaction with different immune effector cells may also support innate antitumor immunity, as well as humoral responses also known to inhibit tumor development in certain cases. On the other hand, DC are recruited to the tumor site by specific tumor-derived and stroma-derived factors, which may also impair DC maturation, differentiation and function, thus resulting in the deficient formation of antitumor immune response or development of DC-mediated tolerance and immune suppression. Identification of DC-stimulating and DC-suppressing/polarizing factors in the tumor environment and the mechanism of DC modulation are important for designing effective DC-based vaccines and for recovery of immunodeficient resident DC responsible for maintenance of clinically relevant antitumor immunity in patients with cancer. DC-targeting tumor-derived factors and their effects on resident and administered DC in the tumor milieu are described and discussed in this review.
Collapse
Affiliation(s)
- Jinbao Zong
- Department of Pathology, University of Pittsburgh Medical Center, Scaife Hall S735, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao City, China
| | - Anton A Keskinov
- Department of Pathology, University of Pittsburgh Medical Center, Scaife Hall S735, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Galina V Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Scaife Hall S735, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Scaife Hall S735, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
9
|
Elakoum R, Gauchotte G, Oussalah A, Wissler MP, Clément-Duchêne C, Vignaud JM, Guéant JL, Namour F. CARM1 and PRMT1 are dysregulated in lung cancer without hierarchical features. Biochimie 2013; 97:210-8. [PMID: 24211191 DOI: 10.1016/j.biochi.2013.10.021] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/23/2013] [Indexed: 01/13/2023]
Abstract
CARM1 and PRMT1 are 2 Protein Arginine Methyl Transferases (PRMT) dysregulated in cancer. CARM1 function is contradictory and depicted as facilitating proliferation or differentiation. PRMT1 is required for cell proliferation. CARM1 and PRMT1 cooperate for gene regulation. We report that CARM1 and PRMT1 are significantly overexpressed in 60 patients with Non-Small Cell Lung Carcinomas (NSCLC). CARM1 and PRMT1 correlated in healthy but not tumor tissue. Their levels of expression in tumor tissue were proportional to their levels of expression in the counterpart healthy tissue. Only CARM1 expression was found to be correlated with tumor differentiation and neither CARM1 nor PRMT1 expression was correlated with survival. Accordingly, CARM1 and PRMT1 are overexpressed in 2 NSCLC cell lines, A549 and H1299. Targeting PRMT1 with siRNA reduced proliferation, by decreasing cell growth and inhibiting soft agar colony formation, and promoted differentiation, by increasing the epithelial markers cytokeratin 7 and 8 and decreasing Neuromedin B receptor, which binds a mitogenic factor. siCARM1 yielded similar consequences but the conditions with siCARM1 reflected inhibition of both CARM1 and PRMT1. Together these results suggest that CARM1 and PRMT1 are involved in proliferation in lung cancer with no hierarchy of one protein over the other. The fact that CARM1 targeting suppresses PRMT1 in addition to CARM1 reinforces the functional importance of CARM1/PRMT1 interaction.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Farès Namour
- INSERM NGERE UMR 954, Vandoeuvre F-54500, France.
| |
Collapse
|
10
|
Immunosuppressive mechanisms of regulatory dendritic cells in cancer. CANCER MICROENVIRONMENT 2013; 6:159-67. [PMID: 23749739 DOI: 10.1007/s12307-013-0133-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 05/07/2013] [Indexed: 12/20/2022]
Abstract
Three major functional subsets of dendritic cells (DCs) have been described in the tumor microenvironment in patients with cancer and tumor-bearing animals: (i) conventional DCs with intact antigen-presenting capabilities, (ii) functionally defective DCs with decreased motility and low ability to uptake, process and present antigens or produce cytokines and (iii) regulatory DCs with high capacity to suppress T cell proliferation, induce differentiation of regulatory T cells or support immune tolerance. Phenotypic characteristics of regulatory DCs (regDCs), as well as the mechanisms of T cell inhibition, vary in different experimental conditions and environments, suggesting high level of their plasticity and probably different origin. Although new data demonstrate that regDCs may play an important role at early stages of tumor development, functional differences and clinical significance of emergence of different myeloid regulatory cells (MDSCs, regDCs, M2 macrophages, N2 neutrophils, mast cells) in cancer remain to be determined.
Collapse
|
11
|
Ma Y, Shurin GV, Peiyuan Z, Shurin MR. Dendritic cells in the cancer microenvironment. J Cancer 2012; 4:36-44. [PMID: 23386903 PMCID: PMC3564245 DOI: 10.7150/jca.5046] [Citation(s) in RCA: 258] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 12/01/2012] [Indexed: 01/01/2023] Open
Abstract
The complexity of the tumor immunoenvironment is underscored by the emergence and discovery of different subsets of immune effectors and regulatory cells. Tumor-induced polarization of immune cell differentiation and function makes this unique environment even more intricate and variable. Dendritic cells (DCs) represent a special group of cells that display different phenotype and activity at the tumor site and exhibit differential pro-tumorigenic and anti-tumorigenic functions. DCs play a key role in inducing and maintaining the antitumor immunity, but in the tumor environment their antigen-presenting function may be lost or inefficient. DCs might be also polarized into immunosuppressive/tolerogenic regulatory DCs, which limit activity of effector T cells and support tumor growth and progression. Although various factors and signaling pathways have been described to be responsible for abnormal functioning of DCs in cancer, there are still no feasible therapeutic modalities available for preventing or reversing DC malfunction in tumor-bearing hosts. Thus, better understanding of DC immunobiology in cancer is pivotal for designing novel or improved therapeutic approaches that will allow proper functioning of DCs in patients with cancer.
Collapse
Affiliation(s)
- Yang Ma
- 1. Departments of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
12
|
Li X, Lv Y, Yuan A, Li Z. Gastrin-releasing peptide links stressor to cancer progression. J Cancer Res Clin Oncol 2010; 136:483-91. [PMID: 20140628 DOI: 10.1007/s00432-010-0766-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 01/13/2010] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Gastrin-releasing peptide (GRP) plays an important role in cancer growth and metastasis; however, the mechanisms of how GRP affects cancer progression are not well understood. Recent studies revealed that chronic stress is a major risk factor for cancer progression, and this effect may be mediated by GRP. In this review, we will discuss the mechanisms and implications of GRP linking stressor to cancer progression. MATERIALS AND METHODS We retrieved the studies of the relationship between GRP, stress and cancers through PubMed using systematic methods to search, select, and evaluate the findings. RESULTS The results suggested that GRP can mediate the effects of stress on cancers at systemic, tissue and cellular levels: Stress elicits the secretion of GRP in the brain and GRP in turn activates the stress response pathways resulting in an elevation of stress hormones and GRP in the plasma and tissues. GRP in synergy with stress hormones stimulates the growth and invasion of cancer cells by suppressing the anti-tumor immune function and directly activating the pro-proliferative and pro-migratory signaling pathways in cancer cells. CONCLUSION GRP is a multi-functional peptide, which acts as a stress mediator as well as a growth factor linking stressor to cancer progression. GRP and its high-affinity receptor are useful targets for the diagnosis and treatment of cancers.
Collapse
Affiliation(s)
- Xinqiu Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277, West Yanta Road, 710061, Xi'an, People's Republic of China
| | | | | | | |
Collapse
|
13
|
Zeng Z, Yao W, Xu X, Xu G, Long J, Wang X, Wen Z, Chien S. Hepatocellular Carcinoma Cells Deteriorate the Biophysical Properties of Dendritic Cells. Cell Biochem Biophys 2009; 55:33-43. [DOI: 10.1007/s12013-009-9055-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2009] [Accepted: 06/18/2009] [Indexed: 10/20/2022]
|
14
|
Degan S, Lopez GY, Kevill K, Sunday ME. Gastrin-releasing peptide, immune responses, and lung disease. Ann N Y Acad Sci 2009; 1144:136-47. [PMID: 19076373 DOI: 10.1196/annals.1418.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Gastrin-releasing peptide (GRP) is produced by pulmonary neuroendocrine cells (PNECs), with highest numbers of GRP-positive cells present in fetal lung. Normally GRP-positive PNECs are relatively infrequent after birth, but PNEC hyperplasia is frequently associated with chronic lung diseases. To address the hypothesis that GRP mediates chronic lung injury, we present the cumulative evidence implicating GRP in bronchopulmonary dysplasia (BPD), the chronic lung disease of premature infants who survive acute respiratory distress syndrome. The availability of well-characterized animal models of BPD was a critical tool for demonstrating that GRP plays a direct role in the early pathogenesis of this disease. Potential mechanisms by which GRP contributes to injury are analyzed, with the main focus on innate immunity. Autoreactive T cells may contribute to lung injury late in the course of disease. A working model is proposed with GRP triggering multiple cell types in both the innate and adaptive immune systems, promoting cascades culminating in chronic lung disease. These observations represent a paradigm shift in the understanding of the early pathogenesis of BPD, and suggest that GRP blockade could be a novel treatment to prevent this lung disease in premature infants.
Collapse
Affiliation(s)
- Simone Degan
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
15
|
Jensen RT, Battey JF, Spindel ER, Benya RV. International Union of Pharmacology. LXVIII. Mammalian bombesin receptors: nomenclature, distribution, pharmacology, signaling, and functions in normal and disease states. Pharmacol Rev 2008; 60:1-42. [PMID: 18055507 PMCID: PMC2517428 DOI: 10.1124/pr.107.07108] [Citation(s) in RCA: 395] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The mammalian bombesin receptor family comprises three G protein-coupled heptahelical receptors: the neuromedin B (NMB) receptor (BB(1)), the gastrin-releasing peptide (GRP) receptor (BB(2)), and the orphan receptor bombesin receptor subtype 3 (BRS-3) (BB(3)). Each receptor is widely distributed, especially in the gastrointestinal (GI) tract and central nervous system (CNS), and the receptors have a large range of effects in both normal physiology and pathophysiological conditions. The mammalian bombesin peptides, GRP and NMB, demonstrate a broad spectrum of pharmacological/biological responses. GRP stimulates smooth muscle contraction and GI motility, release of numerous GI hormones/neurotransmitters, and secretion and/or hormone release from the pancreas, stomach, colon, and numerous endocrine organs and has potent effects on immune cells, potent growth effects on both normal tissues and tumors, potent CNS effects, including regulation of circadian rhythm, thermoregulation; anxiety/fear responses, food intake, and numerous CNS effects on the GI tract as well as the spinal transmission of chronic pruritus. NMB causes contraction of smooth muscle, has growth effects in various tissues, has CNS effects, including effects on feeding and thermoregulation, regulates thyroid-stimulating hormone release, stimulates various CNS neurons, has behavioral effects, and has effects on spinal sensory transmission. GRP, and to a lesser extent NMB, affects growth and/or differentiation of various human tumors, including colon, prostate, lung, and some gynecologic cancers. Knockout studies show that BB(3) has important effects in energy balance, glucose homeostasis, control of body weight, lung development and response to injury, tumor growth, and perhaps GI motility. This review summarizes advances in our understanding of the biology/pharmacology of these receptors, including their classification, structure, pharmacology, physiology, and role in pathophysiological conditions.
Collapse
Affiliation(s)
- R T Jensen
- Digestive Diseases Branch, National Institute of Diabetes Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | |
Collapse
|
16
|
Ross JW, Ashworth MD, White FJ, Johnson GA, Ayoubi PJ, DeSilva U, Whitworth KM, Prather RS, Geisert RD. Premature estrogen exposure alters endometrial gene expression to disrupt pregnancy in the pig. Endocrinology 2007; 148:4761-73. [PMID: 17640989 DOI: 10.1210/en.2007-0599] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Establishment and maintenance of pregnancy in the pig involve intricate communication between the developing conceptuses and maternal endometrium. Conceptus synthesis and release of estrogen during trophoblastic elongation are essential factors involved with establishing conceptus-uterine communication. The present study identified endometrial changes in gene expression associated with implantation failure and complete pregnancy loss after premature exposure of pregnant gilts to exogenous estrogen. Gilts were treated with either 5 mg estradiol cypionate (EC) or corn oil on d-9 and -10 gestation, which was associated with complete conceptus degeneration by d-17 gestation. Microarray analysis of gene expression revealed that a total of eight, 32, and five genes were up-regulated in the EC endometrium, whereas one, 39, and 16 genes were down-regulated, on d 10, 13, and 15, respectively. Four endometrial genes altered by EC, aldose reductase (AKR1B1), secreted phosphoprotein 1 (SPP1), CD24 antigen (CD24), and neuromedin B (NMB), were evaluated using quantitative RT-PCR and in situ hybridization. In situ hybridization localized gene expression for NMB, CD24, AKR1B1, and SPP1 in the luminal epithelium, and confirmed the expression patterns from RT-PCR analysis. The aberrant expression patterns of endometrial AKR1B1, SPP1, CD24, and NMB 3-4 d after premature estrogen exposure to pregnant gilts may be involved with conceptus attachment failure to the uterine surface epithelium and induction of endometrial responses that disrupt the establishment of a viable pregnancy.
Collapse
Affiliation(s)
- Jason W Ross
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74078, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Segain JP, Rolli-Derkinderen M, Gervois N, Raingeard de la Blétière D, Loirand G, Pacaud P. Urotensin II is a new chemotactic factor for UT receptor-expressing monocytes. THE JOURNAL OF IMMUNOLOGY 2007; 179:901-9. [PMID: 17617581 DOI: 10.4049/jimmunol.179.2.901] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Urotensin II (U-II), a vasoactive cyclic neuropeptide which activates the G protein-coupled receptor UT receptor, exerts various cardiovascular effects and may play a role in the pathophysiology of atherosclerosis. In this study, we report that the UT receptor is expressed and functional on human PBMC and rat splenocytes. PBMC surface expression of the UT receptor was mainly found in monocytes and NK cells, also in a minority of B cells, but not in T cells. Stimulation of monocytes with LPS increased UT receptor mRNA and protein expression. Cloning and functional characterization of the human UT receptor gene promoter revealed the presence of NF-kappaB-binding sites involved in the stimulation of UT receptor gene expression by LPS. Activation of the UT receptor by U-II induced chemotaxis with maximal activity at 10 and 100 nM. This U-II effect was restricted to monocytes. Analysis of the signaling pathway involved indicated that U-II-mediated chemotaxis was related to RhoA and Rho kinase activation and actin cytoskeleton reorganization. The present results thus identify U-II as a chemoattractant for UT receptor-expressing monocytes and indicate a pivotal role of the RhoA-Rho kinase signaling cascade in the chemotaxis induced by U-II.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Base Sequence
- Cells, Cultured
- Chemotaxis, Leukocyte/immunology
- Cloning, Molecular
- Flow Cytometry
- Gene Expression
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Molecular Sequence Data
- Monocytes/immunology
- Monocytes/metabolism
- Mutagenesis, Site-Directed
- NF-kappa B/metabolism
- Promoter Regions, Genetic
- RNA, Messenger/analysis
- Rats
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/immunology
- Receptors, G-Protein-Coupled/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/immunology
- Transfection
- Urotensins/immunology
- Urotensins/metabolism
- rhoA GTP-Binding Protein/biosynthesis
Collapse
Affiliation(s)
- Jean-Pierre Segain
- Institut National de la Recherche Agronomique, Unité Mixte de Recherche 1280-Physiologie des Adaptations Nutritionnelles, Nantes, France
| | | | | | | | | | | |
Collapse
|
18
|
Shurin MR, Shurin GV, Lokshin A, Yurkovetsky ZR, Gutkin DW, Chatta G, Zhong H, Han B, Ferris RL. Intratumoral cytokines/chemokines/growth factors and tumor infiltrating dendritic cells: friends or enemies? Cancer Metastasis Rev 2007; 25:333-56. [PMID: 17029028 DOI: 10.1007/s10555-006-9010-6] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The tumor microenvironment consists of a variable combination of tumor cells, stromal fibroblasts, endothelial cells and infiltrating leukocytes, such as macrophages, T lymphocytes, and dendritic cells. A variety of cytokines, chemokines and growth factors are produced in the local tumor environment by different cells accounting for a complex cell interaction and regulation of differentiation, activation, function and survival of multiple cell types. The interaction between cytokines, chemokines, growth factors and their receptors forms a comprehensive network at the tumor site, which is primary responsible for overall tumor progression and spreading or induction of antitumor immune responses and tumor rejection. Although the general thought is that dendritic cells are among the first cells migrating to the tumor site and recognizing tumor cells for the induction of specific antitumor immunity, the clinical relevance of dendritic cells at the site of the tumor remains a matter of debate regarding their role in the generation of successful antitumor immune responses in human cancers. While several lines of evidence suggest that intratumoral dendritic cells play an important role in antitumor immune responses, understanding the mechanisms of dendritic cell/tumor cell interaction and modulation of activity and function of different dendritic cell subtypes at the tumor site is incomplete. This review is limited to discussing the role of intratumoral cytokine network in the understanding immunobiology of tumor-associated dendritic cells, which seems to possess different regulatory functions at the tumor site.
Collapse
Affiliation(s)
- Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zeng Z, Xu X, Zhang Y, Xing J, Long J, Gu L, Wang X, Sun D, Ka W, Yao W, Wen Z, Chien S. Tumor-derived factors impaired motility and immune functions of dendritic cells through derangement of biophysical characteristics and reorganization of cytoskeleton. ACTA ACUST UNITED AC 2007; 64:186-98. [PMID: 17183544 DOI: 10.1002/cm.20175] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The generation and progress of tumors are accompanied with a marked suppression of human immune system. To explore the mechanisms by which tumors escape from immune recognition, we studied the influences of tumor microenvironment on differentiation of dendritic cells (DCs), which play an important role in tumor immunology, by biophysical and immunological methods. It was found that the cytokines derived from tumors caused an increase in osmotic fragility and a decrease in membrane fluidity of DCs, disordering and elevated expression levels of cytoskeleton, and changes of the gene transcriptional levels and energy status of the cells. Moreover, IL-12 production and the expression levels of some surface-marker molecules were also suppressed. These changes led to impaired capabilities of antigen uptake, cell motility and naïve T cell activation; the abnormal biophysical characteristics of DCs may be one aspect of the immune escape mechanism of tumor. These results provide insights into the importance of the reconstruction of tumor microenvironment for immunotherapy based on the anti-cancer activities of DCs.
Collapse
Affiliation(s)
- Zhu Zeng
- Department of Biophysics, Health Science Center, Peking University, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Watson SA, Grabowska AM, El-Zaatari M, Takhar A. Gastrin - active participant or bystander in gastric carcinogenesis? Nat Rev Cancer 2006; 6:936-46. [PMID: 17128210 DOI: 10.1038/nrc2014] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gastrin is a pro-proliferative, anti-apoptotic hormone with a central role in acid secretion in the gastric mucosa and a long-standing association with malignant progression in transgenic mouse models. However, its exact role in human gastric malignancy requires further validation. Gastrin expression is tightly regulated by two closely associated hormones, somatostatin and gastrin-releasing peptide, and aspects of their interaction may be deregulated during progression to gastric adenocarcinoma. Furthermore, agonists and antagonists of the receptors for all three hormones have shown modest clinical efficacy against gastric adenocarcinoma, which might provide useful information on the future combined use of these agents.
Collapse
Affiliation(s)
- Susan A Watson
- Academic Unit of Cancer Studies, University of Nottingham, Nottingham, NG7 2UH, UK.
| | | | | | | |
Collapse
|
21
|
Tan YR, Qi MM, Qin XQ, Xiang Y, Li X, Wang Y, Qu F, Liu HJ, Zhang JS. Wound repair and proliferation of bronchial epithelial cells enhanced by bombesin receptor subtype 3 activation. Peptides 2006; 27:1852-8. [PMID: 16426703 DOI: 10.1016/j.peptides.2005.12.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Revised: 12/19/2005] [Accepted: 12/19/2005] [Indexed: 11/26/2022]
Abstract
The present study was designed to investigate the role of bombesin receptor subtype 3 (BRS-3) in airway wound repair. The results showed that: (1) There was few expression of BRS-3 mRNA in the control group. In contrast, the expression of BRS-3 mRNA was gradually increased in the early 2 days, and peaked on the fourth day, and then decreased in the ozone-stressed AHR animal. BRS-3 mRNA was distributed in the ciliated columnar epithelium, monolayer columnar epithelium cells, scattered mesenchymal cells and Type II alveolar cells; (2) The wound repair and proliferation of bronchial epithelial cells (BECs) were accelerated in a concentration-dependent manner by BRS-3 activation with P3513, which could be inhibited by PKA inhibitor H89. The study demostrated that activation of BRS-3 may play an important role in wound repair of AHR.
Collapse
Affiliation(s)
- Yu-Rong Tan
- Xiangya School of Medicine, Central South University, Changsha 410078, Hunan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lang K, Entschladen F, Weidt C, Zaenker KS. Tumor immune escape mechanisms: impact of the neuroendocrine system. Cancer Immunol Immunother 2006; 55:749-60. [PMID: 16435128 PMCID: PMC11030197 DOI: 10.1007/s00262-006-0126-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Accepted: 01/04/2006] [Indexed: 12/24/2022]
Abstract
Tumor cells act upon, and react to both their proximate and more distant environment, the mechanisms by which this is achieved being both autocrine and paracrine in nature. This interaction, however, takes place not only between adjacent malignant cells, but also non-malignant cells such as those of the immune system, the latter also partaking in the modeling of the tumor environment. Although tumor cells descend from normal tissue cells and thus bear in classical immunological terms 'self signals', it is evident that the immune system is able to recognize tumor cells as a harassment for the body and in consequence tries to eliminate these cells. On the counterpart, tumor cells acquire various characteristics which allow them to evade this immunological surveillance, and have been collectively coined with the term "tumor escape mechanisms". This review will describe and summarize current understanding of tumor escape strategies, and also more closely elaborate on the modulatory role of the neuroendocrine system in the immune system-tumor cell interaction.
Collapse
Affiliation(s)
- Kerstin Lang
- Institute of Immunology, Witten/Herdecke University, Stockumer Str. 10, 58448 Witten, Germany.
| | | | | | | |
Collapse
|
23
|
Tsoumakidou M, Tzanakis N, Papadaki HA, Koutala H, Siafakas NM. Isolation of myeloid and plasmacytoid dendritic cells from human bronchoalveolar lavage fluid. Immunol Cell Biol 2006; 84:267-73. [PMID: 16509829 DOI: 10.1111/j.1440-1711.2006.01428.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Studies of bronchoalveolar lavage fluid (BALF) dendritic cells (DC) have been hampered by the scarcity of DC and the lack of DC-specific surface markers. Four surface Ag have been recently described as specific markers for distinct subsets of DC and have been used for the isolation and characterization of fresh noncultured DC from lung resection specimens: BDCA-1 (CD1c) and BDCA-3 for myeloid DC type 1 and type 2, respectively, and BDCA-2 and BDCA-4 for plasmacytoid DC. The aim of this study was to develop a new method for the isolation of BALF DC, using immunomagnetic separation of BDCA+ cells. Mononuclear cells were obtained from BALF after Ficoll-Paque density gradient centrifugation. Monocytes, T cells and B cells were magnetically labelled and depleted. The unlabelled cell fraction was incubated with BDCA-1, BDCA-3 and BDCA-4 beads and the total BDCA+ DC were retained. The ability of isolated DC to induce T-cell responses was evaluated by coculturing the isolated DC with immunomagnetically sorted naive T cells. The above procedure resulted in a population of viable DC that showed a strong capacity in induce T-cell responses. Functionally intact human BALF myeloid DC types 1 and 2 as well as plasmacytoid DC can be easily obtained by immunomagnetic isolation. Considering that bronchoalveolar lavage is a minimally invasive procedure, these cells are optimal candidates with which to elucidate the properties and capabilities of pulmonary DC.
Collapse
Affiliation(s)
- Maria Tsoumakidou
- Department of Thoracic Medicine, Medical School, University of Crete, Heraklion, Crete, Greece
| | | | | | | | | |
Collapse
|
24
|
Àvila-Moreno F, López-González JS, Galindo-Rodríguez G, Prado-García H, Bajaña S, Sánchez-Torres C. Lung squamous cell carcinoma and adenocarcinoma cell lines use different mediators to induce comparable phenotypic and functional changes in human monocyte-derived dendritic cells. Cancer Immunol Immunother 2006; 55:598-611. [PMID: 16133109 PMCID: PMC11029896 DOI: 10.1007/s00262-005-0060-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2005] [Accepted: 07/18/2005] [Indexed: 01/03/2023]
Abstract
Tumor-derived immunosuppressive factors contribute to the evasion of malignant cells from the immune response, partially by hampering dendritic cell (DC) differentiation. Here, we analyze whether soluble mediators released by the most frequent histological types of non-small cell lung carcinoma, squamous cell carcinoma (SCC), and adenocarcinoma (AD) cells, affect the development and functionality of DC. Monocytes from healthy donors were differentiated in vitro into DC with granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)-4, in the absence or presence of soluble factors (SF) from SCC or AD cell lines. Monocytes were differentiated in parallel into macrophages (MPhi s) with macrophage colony-stimulating factor (M-CSF). SF-treated DC were phenotypically and functionally more similar to MPhi s than to untreated DC [control DC (Ctrl-DC)]. Both tumors increased myelomonocytic markers (CD14, CD16, CD32, and CD163) and impaired CD1a expression on DC. SF-treated DC increased their endocytic capacity, and released higher levels of IL-6, IL-10, and lower levels of IL-12, compared to Ctrl-DC. SF-treated DC were poor stimulators in mixed lymphocyte reactions, and naïve CD4(+) T lymphocytes stimulated by SF-treated DC secreted lower levels of interferon (IFN)-gamma and higher amounts of IL-10 than controls. In contrast to AD, the effects caused by SCC were mostly abolished by IL-6 neutralization during monocyte differentiation. However, tumor-derived prostanoid blockade recovered the IFN-gamma levels secreted by lymphocytes stimulated with SF-treated DC, whereas prostanoid/IL-6 or prostanoid/IL-10 blockade decreased IL-10 production only by SCC-DC-stimulated lymphocytes. Thus, we provide evidence that lung SCC and AD cause comparable deficiencies on DC in vitro, skewing monocyte differentiation from DC to MPhi -like cells, but most of these changes occurred via different mediators.
Collapse
Affiliation(s)
- Federico Àvila-Moreno
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados-IPN (CINVESTAV-IPN), Av. IPN 2508, CP 07360 Mexico City, Mexico
- Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias (INER), Mexico City, Mexico
| | - José Sullivan López-González
- Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias (INER), Mexico City, Mexico
| | - Griselda Galindo-Rodríguez
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados-IPN (CINVESTAV-IPN), Av. IPN 2508, CP 07360 Mexico City, Mexico
- Rheumatic Disease Unit, Hospital de Especialidades Centro Medico Nacional La Raza, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Heriberto Prado-García
- Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias (INER), Mexico City, Mexico
| | - Sandra Bajaña
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del IPN, Mexico City, Mexico
| | - Carmen Sánchez-Torres
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados-IPN (CINVESTAV-IPN), Av. IPN 2508, CP 07360 Mexico City, Mexico
| |
Collapse
|
25
|
Rosen D, Lee JH, Cuttitta F, Rafiqi F, Degan S, Sunday ME. Accelerated thymic maturation and autoreactive T cells in bronchopulmonary dysplasia. Am J Respir Crit Care Med 2006; 174:75-83. [PMID: 16574933 PMCID: PMC2662921 DOI: 10.1164/rccm.200511-1784oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Bronchopulmonary dysplasia (BPD), a chronic lung disease of newborns triggered by oxygen and barotrauma, is characterized by arrested alveolarization. Increased levels of bombesin-like peptides shortly after birth mediate lung injury: anti-bombesin antibody 2A11 protects against BPD in two baboon models. The role of adaptive immunity in BPD has not been explored previously. OBJECTIVES Our goal was to test the hypothesis that thymic architecture and/or T-cell function is altered with BPD, leading to autoimmunity and immunodeficiency. METHODS Thymic structure was analyzed by histopathology of thymic architecture and immunohistochemistry for thymic maturation markers (terminal deoxynucleotidyl transferase, proliferating cell nuclear antigen, CD4, and CD8). Thymic cortical epithelial cells (nurse cells) were studied using HLA-DR and protein gene product 9.5 as markers. Functional analysis was performed with "mixed lymphocyte reaction" of thymocyte or splenocyte responder cells with autologous lung cells as the stimulators. MEASUREMENTS AND MAIN RESULTS 2A11 treatment attenuates thymic cortical involution in BPD animals, sustaining terminal deoxynucleotidyl transferase-positive prothymocytes and thymocyte proliferation. BPD animals have increased CD4(+) cells in thymic cortex and lung interstitium, which are reduced by 2A11. Conversely, cortical protein gene product 9.5/HLA-DR-positive thymic nurse cells are depleted in BPD animals, but are preserved by 2A11-treatment. Whereas fetal thymocytes and splenocytes respond to phythemagglutinin/ionomycin and to a lesser extent, to autologous lung, BPD thymocytes and splenocytes are phythemagglutinin/ionomycin-unresponsive, and yet react strongly to autologous lung. The 2A11 normalizes these responses. CONCLUSIONS These observations suggest that bombesin-like peptides mediate premature thymic maturation and thymic nurse-cell depletion, leading to autoreactive T cells that could contribute to lung injury.
Collapse
Affiliation(s)
- Dennis Rosen
- Division of Pulmonary Medicine, Department of Medicine, Children's and Brigham and Women's Hospitals, and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
26
|
Medina S, Del Rio M, Hernanz A, Guaza C, De la Fuente M. Nitric oxide released by accessory cells mediates the gastrin-releasing peptide effect on murine lymphocyte chemotaxis. ACTA ACUST UNITED AC 2005; 131:46-53. [PMID: 15996770 DOI: 10.1016/j.regpep.2005.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2005] [Revised: 06/01/2005] [Accepted: 06/07/2005] [Indexed: 11/28/2022]
Abstract
Several neuropeptides, including gastrin-releasing peptide (GRP), modulate the immune response, specifically lymphocyte chemotaxis. In the present work the effect of GRP on the chemotaxis of murine lymphocytes from different immune locations in both, total leukocyte populations and populations depleted of adherent cells have been studied. Specificity of the GRP effect on chemotaxis using an antagonist of the GRP receptor, as well as the implication of nitric oxide (NO), using inhibitors of NO synthase and donors of NO, were investigated. The effects of GRP stimulating the chemotaxis of lymphocytes from peritoneum, axillary nodes and spleen and decreasing the chemotaxis from thymus were receptor-specific and disappeared in lymphocytes from populations depleted of adherent cells. NO synthase inhibitors blocked the GRP effect on lymphocyte chemotaxis, and this action was reversed in the presence of l-arginine. Thus, the effect of GRP on murine lymphocyte chemotaxis appears to be mediated by NO secreted by adherent cells.
Collapse
Affiliation(s)
- Sonia Medina
- Department of Animal Physiology, Faculty of Biology, Complutense University, Spain
| | | | | | | | | |
Collapse
|
27
|
Abstract
Dendritic cells (DCs) are leukocytes that are emerging as chief orchestrators of immune responses. The crucial task of DCs is the continuous surveillance of antigen-exposed sites throughout the body, and their unique responsibility is to decide whether to present sampled antigen in an immunogenic or tolerogenic way. Any misstep can either lead to a flawed immune defense or to allergy, even autoimmunity. It comes as no surprise that the lungs become increasingly the subject of DC-related investigations, as they represent a vast interface between the body and the outer world. This constitutes an enormous challenge for the immune system: "firing up" immune responses inappropriately could have devastating results for the fragile gas exchange structures. Evidence accumulates that DCs play a pivotal role in maintaining the delicate balance between tolerance and active immune response in our respiratory system. The exponentially growing body of DC-related publications is a big challenge. This article aims to provide researchers and clinicians with an up-to-date view on DC biology and its relevance to pulmonary medicine. A developing trend in the field of DCs is the shift from fundamental immunologic research toward exciting clinical insights and applications. For the pulmonary clinician, this heralds the dawn of promising therapies in various domains such as infections, allergy, and cancer.
Collapse
Affiliation(s)
- Karim Vermaelen
- Department of Respiratory Diseases, Ghent University Hospital, 7K12ie, De Pintelaan 185, Ghent B-9000, Belgium.
| | | |
Collapse
|