1
|
Ezennia SC, Beane Freeman LE, Chang VC, Xie S, Sandler DP, Andreotti G, Parks CG, Friesen MC, Hofmann JN. Estimated exposure to endotoxin and circulating immunological markers among male farmers in the Biomarkers of Exposure and Effect in Agriculture study. Occup Environ Med 2025; 81:635-638. [PMID: 39746796 DOI: 10.1136/oemed-2024-109646] [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: 05/30/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Occupational exposure to endotoxin has been associated with reduced lung cancer risk. The mechanisms underlying this association are unclear, though immunological alterations likely play a role. Farmers who perform certain tasks (eg, raising hogs) can be highly exposed to endotoxin. We, therefore, leveraged measurements of circulating immune markers from a prior investigation among male farmers in the Biomarkers of Exposure and Effect in Agriculture study to evaluate associations with newly developed estimates of endotoxin exposure. METHODS Our investigation included 122 non-smoking farmers from Iowa, oversampling those raising hogs. Serum levels of 60 markers were measured using multiplex bead-based assays and ELISA. Based on an algorithm linking measurement-based task intensity estimates with self-reported task frequency, we estimated cumulative endotoxin exposure in the 30 days up to sample collection. We used multivariable linear regression to estimate geometric mean ratios of immune markers across exposure quartiles. RESULTS Higher endotoxin exposure in the last 30 days was associated with increased levels of fibroblast growth factor-2, macrophage inflammatory protein-3 alpha/CCL20 and sIL-4R (Ptrend≤0.02) and decreased levels of macrophage-derived chemokine/CCL22 (Ptrend=0.02). We also identified novel associations with several additional markers; those with the highest (vs lowest) exposure to endotoxin had decreased levels of TARC/CCL17, sCD27 and IL-1B. CONCLUSIONS Several circulating immune markers were associated with endotoxin levels in an exposure-response manner. Our findings are consistent with prior work on hog farming and suggest possible biological mechanisms through which endotoxin may confer a reduced risk of lung cancer.
Collapse
Affiliation(s)
- Somayina C Ezennia
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Laura E Beane Freeman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Vicky C Chang
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Shuai Xie
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Gabriella Andreotti
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Christine G Parks
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Melissa C Friesen
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Jonathan N Hofmann
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, Maryland, USA
| |
Collapse
|
2
|
Yan S, Gan Y, Xu H, Piao H. Bacterial carrier-mediated drug delivery systems: a promising strategy in cancer therapy. Front Bioeng Biotechnol 2025; 12:1526612. [PMID: 39845371 PMCID: PMC11750792 DOI: 10.3389/fbioe.2024.1526612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Cancer is a major killer threatening modern human health and a leading cause of death worldwide. Due to the heterogeneity and complexity of cancer, traditional treatments have limited effectiveness. To address this problem, an increasing number of researchers and medical professionals are working to develop new ways to treat cancer. Bacteria have chemotaxis that can target and colonize tumor tissue, as well as activate anti-tumor immune responses, which makes them ideal for biomedical applications. With the rapid development of nanomedicine and synthetic biology technologies, bacteria are extensively used as carriers for drug delivery to treat tumors, which holds the promise of overcoming the limitations of conventional cancer treatment regimens. This paper summarizes examples of anti-cancer drugs delivered by bacterial carriers, and their strengths and weaknesses. Further, we emphasize the promise of bacterial carrier delivery systems in clinical translation.
Collapse
Affiliation(s)
- Sizuo Yan
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Yu Gan
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Huizhe Xu
- Institute of Cancer Medicine, Dalian University of Technology, Dalian, China
- Central Laboratory, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Haozhe Piao
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
- Institute of Cancer Medicine, Dalian University of Technology, Dalian, China
| |
Collapse
|
3
|
Metsäniitty M, Hasnat S, Öhman C, Salo T, Eklund KK, Oscarsson J, Salem A. Zebrafish larvae as a model for studying the impact of oral bacterial vesicles on tumor cell growth and metastasis. Hum Cell 2024; 37:1696-1705. [PMID: 39138804 PMCID: PMC11481661 DOI: 10.1007/s13577-024-01114-6] [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: 03/13/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024]
Abstract
Oral bacteria naturally secrete extracellular vesicles (EVs), which have attracted attention for their promising biomedical applications including cancer therapeutics. However, our understanding of EV impact on tumor progression is hampered by limited in vivo models. In this study, we propose a facile in vivo platform for assessing the effect of EVs isolated from different bacterial strains on oral cancer growth and dissemination using the larval zebrafish model. EVs were isolated from: wild-type Aggregatibacter actinomycetemcomitans and its mutant strains lacking the cytolethal distending toxin (CDT) or lipopolysaccharide (LPS) O-antigen; and wild-type Porphyromonas gingivalis. Cancer cells pretreated with EVs were xenotransplanted into zebrafish larvae, wherein tumor growth and metastasis were screened. We further assessed the preferential sites for the metastatic foci development. Interestingly, EVs from the CDT-lacking A. actinomycetemcomitans resulted in an increased tumor growth, whereas EVs lacking the lipopolysaccharide O-antigen reduced the metastasis rate. P. gingivalis-derived EVs showed no significant effects. Cancer cells pretreated with EVs from the mutant A. actinomycetemcomitans strains tended to metastasize less often to the head and tail compared to the controls. In sum, the proposed approach provided cost- and labor-effective yet efficient model for studying bacterial EVs in oral carcinogenesis, which can be easily extended for other cancer types. Furthermore, our results support the notion that these nanosized particles may represent promising targets in cancer therapeutics.
Collapse
Affiliation(s)
- Marjut Metsäniitty
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, 00014, Helsinki, Finland
| | - Saika Hasnat
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, 00014, Helsinki, Finland
| | - Carina Öhman
- Oral Microbiology, Department of Odontology, Umeå University, 90187, Umeå, Sweden
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, 00014, Helsinki, Finland
| | - Kari K Eklund
- Department of Rheumatology, University of Helsinki and Helsinki University Hospital, 00014, Helsinki, Finland
- Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, 00014, Helsinki, Finland
| | - Jan Oscarsson
- Oral Microbiology, Department of Odontology, Umeå University, 90187, Umeå, Sweden
| | - Abdelhakim Salem
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, 00014, Helsinki, Finland.
- Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, 00014, Helsinki, Finland.
| |
Collapse
|
4
|
Liu B, Zhou H, Tan L, Siu KTH, Guan XY. Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduct Target Ther 2024; 9:175. [PMID: 39013849 PMCID: PMC11252281 DOI: 10.1038/s41392-024-01856-7] [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: 01/23/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 07/18/2024] Open
Abstract
Traditional therapeutic approaches such as chemotherapy and radiation therapy have burdened cancer patients with onerous physical and psychological challenges. Encouragingly, the landscape of tumor treatment has undergone a comprehensive and remarkable transformation. Emerging as fervently pursued modalities are small molecule targeted agents, antibody-drug conjugates (ADCs), cell-based therapies, and gene therapy. These cutting-edge treatment modalities not only afford personalized and precise tumor targeting, but also provide patients with enhanced therapeutic comfort and the potential to impede disease progression. Nonetheless, it is acknowledged that these therapeutic strategies still harbour untapped potential for further advancement. Gaining a comprehensive understanding of the merits and limitations of these treatment modalities holds the promise of offering novel perspectives for clinical practice and foundational research endeavours. In this review, we discussed the different treatment modalities, including small molecule targeted drugs, peptide drugs, antibody drugs, cell therapy, and gene therapy. It will provide a detailed explanation of each method, addressing their status of development, clinical challenges, and potential solutions. The aim is to assist clinicians and researchers in gaining a deeper understanding of these diverse treatment options, enabling them to carry out effective treatment and advance their research more efficiently.
Collapse
Affiliation(s)
- Beilei Liu
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Hongyu Zhou
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Licheng Tan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Kin To Hugo Siu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China.
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, China.
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China.
| |
Collapse
|
5
|
Xuanzhuang LU, Qiuxia QIU, Chunyu YANG, Caichen LI, Jianfu LI, Shan XIONG, Bo CHENG, Chujing ZHOU, Xiaoqin DU, Yi ZHANG, Jianxing HE, Wenhua LIANG, Nanshan ZHONG. [Results of Lung Cancer Screening with Low-dose Computed Tomography
and Exploration of Risk Factors in Guangzhou]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:345-358. [PMID: 38880922 PMCID: PMC11183313 DOI: 10.3779/j.issn.1009-3419.2024.101.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Both of lung cancer incidence and mortality rank first among all cancers in China. Previous lung cancer screening trials were mostly selective screening for high-risk groups such as smokers. Non-smoking women accounted for a considerable proportion of lung cancer cases in Asia. This study aimed to evaluate the outcome of community-based mass screening in Guangzhou and identify the high-risk factors for lung cancer. METHODS Residents aged 40-74 years in Guangzhou were screened with low-dose computed tomography (LDCT) for lung cancer and the pulmonary nodules were classified and managed according to China National Lung Cancer Screening Guideline with Low-dose Computed Tomography (2018 version). The detection rate of positive nodules was calculated. Before the LDCT examination, residents were required to complete a "lung cancer risk factors questionnaire". The risk factors of the questionnaire were analyzed by least absolute shrinkage and selection operator (LASSO) penalized Logistic regression analysis. RESULTS A total of 6256 residents were included in this study. 1228 positive nodules (19.63%) and 117 lung cancers were confirmed, including 6 cases of Tis, 103 cases of stage I (accounting for 88.03% of lung cancer). The results of LASSO penalized Logistic regression analysis indicated that age ≥50 yr (OR=1.07, 95%CI: 1.06-1.07), history of cancer (OR=3.29, 95%CI: 3.22-3.37), textile industry (OR=1.10, 95%CI: 1.08-1.13), use coal for cooking in childhood (OR=1.14, 95%CI: 1.13-1.16) and food allergy (OR=1.10, 95%CI: 1.07-1.13) were risk factors of lung cancer for female in this district. CONCLUSIONS This study highlighted that numerous early stages of lung cancer cases were detected by LDCT, which could be applied to screening of lung cancer in women. Besides, age ≥50 yr, personal history of cancer, textile industry and use coal for cooking in childhood are risk factors for women in this district, which suggested that it's high time to raise the awareness of early lung cancer screening in this group.
Collapse
|
6
|
Láng L, McArthur S, Lazar AS, Pourtau L, Gaudout D, Pontifex MG, Müller M, Vauzour D. Dietary (Poly)phenols and the Gut-Brain Axis in Ageing. Nutrients 2024; 16:1500. [PMID: 38794738 PMCID: PMC11124177 DOI: 10.3390/nu16101500] [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: 04/03/2024] [Revised: 05/07/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
As the population ages, the incidence of age-related neurodegenerative diseases is rapidly increasing, and novel approaches to mitigate this soaring prevalence are sorely needed. Recent studies have highlighted the importance of gut microbial homeostasis and its impact on brain functions, commonly referred to as the gut-brain axis, in maintaining overall health and wellbeing. Nonetheless, the mechanisms by which this system acts remains poorly defined. In this review, we will explore how (poly)phenols, a class of natural compounds found in many plant-based foods and beverages, can modulate the gut-brain axis, and thereby promote neural health. While evidence indicates a beneficial role of (poly)phenol consumption as part of a balanced diet, human studies are scarce and mechanistic insight is still lacking. In this regard, we make the case that dietary (poly)phenols should be further explored to establish their therapeutic efficacy on brain health through modulation of the gut-brain axis, with much greater emphasis on carefully designed human interventions.
Collapse
Affiliation(s)
- Léonie Láng
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich NR4 7TJ, UK; (L.L.); (M.M.)
| | - Simon McArthur
- Faculty of Medicine & Dentistry, Queen Mary, University of London, Blizard Institute, London E1 2AT, UK;
| | - Alpar S. Lazar
- Faculty of Medicine and Health Sciences, The Queen’s Building, University of East Anglia, Norwich NR4 7TJ, UK; (A.S.L.); (M.G.P.)
| | - Line Pourtau
- Activ’Inside, 33750 Beychac et Caillau, France; (L.P.); (D.G.)
| | - David Gaudout
- Activ’Inside, 33750 Beychac et Caillau, France; (L.P.); (D.G.)
| | - Matthew G. Pontifex
- Faculty of Medicine and Health Sciences, The Queen’s Building, University of East Anglia, Norwich NR4 7TJ, UK; (A.S.L.); (M.G.P.)
| | - Michael Müller
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich NR4 7TJ, UK; (L.L.); (M.M.)
| | - David Vauzour
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich NR4 7TJ, UK; (L.L.); (M.M.)
| |
Collapse
|
7
|
Metsäniitty M, Hasnat S, Öhman C, Salo T, Eklund KK, Oscarsson J, Salem A. Extracellular vesicles from Aggregatibacter actinomycetemcomitans exhibit potential antitumorigenic effects in oral cancer: a comparative in vitro study. Arch Microbiol 2024; 206:244. [PMID: 38702412 PMCID: PMC11068833 DOI: 10.1007/s00203-024-03976-8] [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: 03/11/2024] [Accepted: 04/24/2024] [Indexed: 05/06/2024]
Abstract
Aggregatibacter actinomycetemcomitans is an opportunistic Gram-negative periodontopathogen strongly associated with periodontitis and infective endocarditis. Recent evidence suggests that periodontopathogens can influence the initiation and progression of oral squamous cell carcinoma (OSCC). Herein we aimed to investigate the effect of A. actinomycetemcomitans-derived extracellular vesicles (EVs) on OSCC cell behavior compared with EVs from periodontopathogens known to associate with carcinogenesis. EVs were isolated from: A. actinomycetemcomitans and its mutant strains lacking the cytolethal distending toxin (CDT) or lipopolysaccharide (LPS) O-antigen; Porphyromonas gingivalis; Fusobacterium nucleatum; and Parvimonas micra. The effect of EVs on primary and metastatic OSCC cells was assessed using cell proliferation, apoptosis, migration, invasion, and tubulogenesis assays. A. actinomycetemcomitans-derived EVs reduced the metastatic cancer cell proliferation, invasion, tubulogenesis, and increased apoptosis, mostly in CDT- and LPS O-antigen-dependent manner. EVs from F. nucleatum impaired the metastatic cancer cell proliferation and induced the apoptosis rates in all OSCC cell lines. EVs enhanced cancer cell migration regardless of bacterial species. In sum, this is the first study demonstrating the influence of A. actinomycetemcomitans-derived EVs on oral cancer in comparison with other periodontopathogens. Our findings revealed a potential antitumorigenic effect of these EVs on metastatic OSCC cells, which warrants further in vivo investigations.
Collapse
Affiliation(s)
- Marjut Metsäniitty
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Shrabon Hasnat
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Carina Öhman
- Oral Microbiology, Department of Odontology, Umeå University, Umeå, 90187, Sweden
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Kari K Eklund
- Department of Rheumatology, University of Helsinki and Helsinki University Hospital, Helsinki, 00014, Finland
- Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Jan Oscarsson
- Oral Microbiology, Department of Odontology, Umeå University, Umeå, 90187, Sweden
| | - Abdelhakim Salem
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland.
- Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland.
| |
Collapse
|
8
|
Mourino N, Varela-Lema L, Ruano-Ravina A, Peiteado C, Candal-Pedreira C, Rey-Brandariz J, Torres-Cadavid E, García G, Pérez-Ríos M. Occupational exposure to endotoxins and small cell lung cancer: a systematic review with meta-analysis. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2024; 27:91-105. [PMID: 38369511 DOI: 10.1080/10937404.2024.2316151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
The relationship of occupational exposure to endotoxins with different histologic subtypes of lung cancer has not been established. Our objective was to conduct a systematic review with meta-analysis to assess the effect of exposure to endotoxins on the development of small cell lung cancer (SCLC). A bibliographic search was conducted using MEDLINE, Embase, CENTRAL, and Web of Science databases until December 2022, including all cohort and/or case-control studies that examined occupational exposure to endotoxins and SCLC. Risk of bias was assessed using the U.S. Office of Health Assessment and Translation tool. A random effects model was applied, publication bias were assessed, and a sensitivity analysis was conducted. Four papers were selected for meta-analysis purposes. A total of 144 incident cases of SCLC and 897 population or hospital controls were included. Occupational exposure to endotoxins was considered for textile/leather industry and agricultural sector workers exposed to endotoxins originating from wool, cotton, or leather dust. Except for one study, all investigations were classified as having a low probability of risk of biases. The results of the meta-analysis were not statistically significant (pooled OR: 0.86; 95% CI:0.69-1.08). In addition, neither between-study heterogeneity (I2=0%;p=0.92) nor publication bias was observed (p=0.49). The results of the sensitivity analysis, after including five studies that assessed the risk of SCLC among textile industry and crop/livestock farm workers (not specifically exposed to endotoxins), showed a negative statistically non-significant association and low between-study heterogeneity (pooled OR: 0.90; 95% CI:0.79-1.02; I2=22%;p=0.23). Subjects exposed to occupational exposure to endotoxins seem to exhibit a negative association with the development of SCLC, although the results are not conclusive.
Collapse
Affiliation(s)
- Nerea Mourino
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Leonor Varela-Lema
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Alberto Ruano-Ravina
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Cristina Peiteado
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Cristina Candal-Pedreira
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Julia Rey-Brandariz
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Eliana Torres-Cadavid
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Guadalupe García
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Mónica Pérez-Ríos
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
9
|
Remigio RV, Andreotti G, Sandler DP, Erickson PA, Koutros S, Albert PS, Hurwitz LM, Parks CG, Lubin JH, Hofmann JN, Beane Freeman LE. An Updated Evaluation of Atrazine-Cancer Incidence Associations among Pesticide Applicators in the Agricultural Health Study Cohort. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:27010. [PMID: 38381478 PMCID: PMC10880817 DOI: 10.1289/ehp13684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Atrazine is a common agricultural herbicide in the United States. Few epidemiologic studies have evaluated cancer risks. Previous analyses within the Agricultural Health Study (AHS) have found some evidence of associations with cancer at some sites. OBJECTIVE We updated exposure information, incident cases, and follow-up time to assess the associations between atrazine use and cancer at specific sites in the AHS. METHODS Information about lifetime pesticide use was reported at enrollment (1993-1997) and follow-up (1999-2005). Among 53,562 pesticide applicators in North Carolina and Iowa, we identified 8,915 incident cases through cancer registry linkages through 2014 (North Carolina)/2017 (Iowa). We used Poisson regression to evaluate the association between ever/never and intensity-weighted lifetime days of atrazine use and incident cancer risk controlling for several confounders. We also evaluated lagged exposures and age-stratified risk. RESULTS Approximately 71.2% of applicators reported ever using atrazine, which was associated with lung cancer [rate ratios ( RR ) = 1.24 ; 95% confidence interval (CI): 1.04, 1.46]. Aggressive prostate cancer risk was increased in the highest quartile (RR Q 4 = 1.20 ; 95% CI: 0.95, 1.52; p -trend = 0.19 ), particularly among those < 60 years old (RR Q 4 = 3.04 ; 95% CI: 1.61, 5.75; p -trend < 0.001 ; p -interaction = 0.04 ). Among applicators < 50 years of age, ever-atrazine use was associated with non-Hodgkin lymphoma (NHL) (RR = 2.43 ; 95% CI: 1.10, 5.38; p -interaction = 0.60 ). For soft tissue sarcoma, there was an elevated risk in the highest tertile of exposure (RR T 3 : 2.54; 95% CI: 0.97, 6.62; p -trend = 0.31 ). In analyses with exposure lagged by 25 years, there was an elevated risk of pharyngeal (RR T 3 = 3.04 ; 95% CI: 1.45, 6.36; p -trend = 0.07 ) and kidney (RR Q 4 = 1.62 ; 95% CI: 1.15, 2.29; p -trend < 0.005 ) cancers. DISCUSSION We observed suggestive associations with some malignancies in overall, age-specific, and lagged analyses. Associations with aggressive prostate cancer and NHL were apparent among those diagnosed at younger ages and with cancers of the pharynx and kidney, and soft tissue sarcomas were observed in lagged analyses. Further work is needed to confirm these observed associations and elucidate potential underlying mechanisms. https://doi.org/10.1289/EHP13684.
Collapse
Affiliation(s)
- Richard V. Remigio
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Gabriella Andreotti
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Dale P. Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Patricia A. Erickson
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Stella Koutros
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Paul S. Albert
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Lauren M. Hurwitz
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Christine G. Parks
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Jay H. Lubin
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Jonathan N. Hofmann
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Laura E. Beane Freeman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Diniz-Lima I, da Fonseca LM, Dos Reis JS, Decote-Ricardo D, Morrot A, Previato JO, Previato LM, Freire-de-Lima CG, Freire-de-Lima L. Non-self glycan structures as possible modulators of cancer progression: would polysaccharides from Cryptococcus spp. impact this phenomenon? Braz J Microbiol 2023; 54:907-919. [PMID: 36840821 PMCID: PMC10235250 DOI: 10.1007/s42770-023-00936-0] [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: 11/26/2022] [Accepted: 02/15/2023] [Indexed: 02/26/2023] Open
Abstract
Invasive fungal infections (IFI) are responsible for a large number of annual deaths. Most cases are closely related to patients in a state of immunosuppression, as is the case of patients undergoing chemotherapy. Cancer patients are severely affected by the worrisome proportions that an IFI can take during cancer progression, especially in an already immunologically and metabolically impaired patient. There is scarce knowledge about strategies to mitigate cancer progression in these cases, beyond conventional treatment with antifungal drugs with a narrow therapeutic range. However, in recent years, ample evidence has surfaced describing the possible interferences that IFI may have both on the progression of pre-existing cancers and in the induction of newly transformed cells. The leading gambit for modulation of tumor progression comes from the ability of fungal virulence factors to modulate the host's immune system, since they are found in considerable concentrations in the tumor microenvironment during infection. In this context, cryptococcosis is of particular concern, since the main virulence factor of the pathogenic yeast is its polysaccharide capsule, which carries constituents with high immunomodulatory properties and cytotoxic potential. Therefore, we open a discussion on what has already been described regarding the progression of cryptococcosis in the context of cancer progression, and the possible implications that fungal glycan structures may take in both cancer development and progression.
Collapse
Affiliation(s)
- Israel Diniz-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Leonardo Marques da Fonseca
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Jhenifer Santos Dos Reis
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Debora Decote-Ricardo
- Departamento de Microbiologia E Imunologia Veterinária, Instituto de Veterinária, Universidade Federal Rural Do Rio de Janeiro, Rio de Janeiro, 23890-000, Brazil
| | - Alexandre Morrot
- Faculdade de Medicina, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
- Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, 21040-360, Brazil
| | - Jose Osvaldo Previato
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Lucia Mendonça Previato
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Celio Geraldo Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Leonardo Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| |
Collapse
|
11
|
Hsieh CC, Wu CH, Peng SH, Chang CH. Seed-derived peptide lunasin suppressed breast cancer cell growth by regulating inflammatory mediators, aromatase, and estrogen receptors. Food Nutr Res 2023; 67:8991. [PMID: 36794014 PMCID: PMC9899045 DOI: 10.29219/fnr.v67.8991] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/27/2022] [Accepted: 11/19/2022] [Indexed: 01/26/2023] Open
Abstract
Background Breast cancer is one of the most prevalent cancers in women. Its pathology comprises tumor cells and nearby stromal cells, accompanied by cytokines and stimulated molecules, resulting in a favorable microenvironment for tumor progression. Lunasin is a seed peptide with multiple bioactivities derived from seeds. However, the chemopreventive effect of lunasin on different characteristics of breast cancer has not been fully explored. Objective This study aims to explore the chemopreventive mechanisms of lunasin through inflammatory mediators and estrogen-related molecules in breast cancer cells. Design Estrogen-dependent MCF-7 and independent MDA-MB-231 breast cancer cells were used. The β-estradiol was used to mimic physiological estrogen. The gene expression, mediator secretion, cell vitality, and apoptosis impacting breast malignancy were explored. Results Lunasin did not affect normal MCF-10A cell growth but inhibited breast cancer cell growth, increased interleukin (IL)-6 gene expression and protein production at 24 h, and decreased its secretion at 48 h. In both breast cancer cells, aromatase gene and activity and estrogen receptor (ER)α gene expression were decreased by lunasin treatment, while ERβ gene levels were significantly increased in MDA-MB-231 cells. Moreover, lunasin decreased vascular endothelial growth factor (VEGF) secretion and cell vitality and induced cell apoptosis in both breast cancer cell lines. However, lunasin only decreased leptin receptor (Ob-R) mRNA expression in MCF-7 cells. Additionally, β-estradiol increased MCF-7-cell proliferation but not the proliferation of other cells; in particular, lunasin still inhibited MCF-7-cell growth and cell vitality in the presence of β-estradiol. Conclusion Seed peptide lunasin inhibited breast cancer cell growth by regulating inflammatory, angiogenic, and estrogen-related molecules, suggesting that lunasin is a promising chemopreventive agent.
Collapse
Affiliation(s)
- Chia-Chien Hsieh
- Department of Biochemical Science & Technology, National Taiwan University, Taipei, Taiwan,Chia-Chien Hsieh Department of Biochemical Science & Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan.
| | - Chi-Hao Wu
- School of Life Science, Undergraduate and Graduate Programs of Nutrition Science, National Taiwan Normal University, Taipei, Taiwan
| | - Shih-Han Peng
- School of Life Science, Undergraduate and Graduate Programs of Nutrition Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chia-Hsin Chang
- School of Life Science, Undergraduate and Graduate Programs of Nutrition Science, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
12
|
Chen J, Wang R, Liu C, Xiong B, Miao Y, Rao C, Sun H, Gao Q, Xu B. Velvet antler water extract protects porcine oocytes from lipopolysaccharide-induced meiotic defects. Cell Prolif 2023:e13392. [PMID: 36596647 DOI: 10.1111/cpr.13392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 01/05/2023] Open
Abstract
Previous studies have demonstrated that lipopolysaccharide (LPS), as a central toxic factor of gram-negative bacteria, can induce oxidative stress and cellular inflammation to result in the impairment of female fertility in different organisms. Particularly, it has harmful effects on the oocyte quality and subsequent embryonic development. However, the approach concerning how to prevent oocytes from LPS-induced deterioration still remains largely unexplored. We assessed the effective influences of velvet antler water extract (VAWE) by immunostaining and fluorescence intensity quantification on the meiotic maturation, mitochondrial function and sperm binding ability of oocytes under oxidative stress. Here, we report that VAWE treatment restores the quality of porcine oocytes exposed to LPS. Specifically, LPS exposure contributed to the failed oocyte maturation, reduced sperm binding ability and fertilization capability by disturbing the dynamics and arrangement of meiotic apparatuses and organelles, including spindle assembly, chromosome alignment, actin polymerization, mitochondrial dynamics and cortical granule distribution, the indicators of oocyte nuclear and cytoplasmic maturation. Notably, VAWE treatment recovered these meiotic defects by removing the LPS-induced excessive ROS and thus inhibiting the apoptosis. Collectively, our study illustrates that VAWE treatment is a feasible strategy to improve the oocyte quality deteriorated by the LPS-induced oxidative stress.
Collapse
Affiliation(s)
- Jingyue Chen
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Rui Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Chunxiao Liu
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Bo Xiong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yilong Miao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Cong Rao
- College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Huimin Sun
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Qian Gao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Baozeng Xu
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
13
|
Ma L, Jiang Y, Lu F, Wang S, Liu M, Liu F, Huang L, Li Y, Jiao N, Jiang S, Yuan X, Yang W. Quantitative Proteomic Analysis of Zearalenone-Induced Intestinal Damage in Weaned Piglets. Toxins (Basel) 2022; 14:toxins14100702. [PMID: 36287972 PMCID: PMC9609629 DOI: 10.3390/toxins14100702] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/09/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
Zearalenone (ZEN), also known as the F-2 toxin, is a common contaminant in cereal crops and livestock products. This experiment aimed to reveal the changes in the proteomics of ZEN-induced intestinal damage in weaned piglets by tandem mass spectrometry tags. Sixteen weaned piglets either received a basal diet or a basal diet supplemented with 3.0 mg/kg ZEN in a 32 d study. The results showed that the serum levels of ZEN, α-zearalenol, and β-zearalenol were increased in weaned piglets exposed to ZEN (p < 0.05). Zearalenone exposure reduced apparent nutrient digestibility, increased intestinal permeability, and caused intestinal damage in weaned piglets. Meanwhile, a total of 174 differential proteins (DEPs) were identified between control and ZEN groups, with 60 up-regulated DEPs and 114 down-regulated DEPs (FC > 1.20 or <0.83, p < 0.05). Gene ontology analysis revealed that DEPs were mainly involved in substance transport and metabolism, gene expression, inflammatory, and oxidative stress. The Kyoto Encyclopedia of Genes and Genomes analysis revealed that DEPs were significantly enriched in 25 signaling pathways (p < 0.05), most of which were related to inflammation and amino acid metabolism. Our study provides valuable clues to elucidate the possible mechanism of ZEN-induced intestinal injury.
Collapse
Affiliation(s)
- Lulu Ma
- College of Animal Sciences and Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China
| | - Yanping Jiang
- Zhongcheng Feed Technology Co., Ltd., Feicheng 271600, China
| | - Fuguang Lu
- Shandong Yucheng Animal Husbandry Development Center Co., Ltd., Yucheng 251200, China
| | - Shujing Wang
- College of Animal Sciences and Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China
| | - Mei Liu
- College of Animal Sciences and Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China
| | - Faxiao Liu
- College of Animal Sciences and Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China
| | - Libo Huang
- College of Animal Sciences and Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China
| | - Yang Li
- College of Animal Sciences and Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China
| | - Ning Jiao
- College of Animal Sciences and Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China
| | - Shuzhen Jiang
- College of Animal Sciences and Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China
| | - Xuejun Yuan
- College of Life Sciences, Shandong Agricultural University, Tai’an 271018, China
- Correspondence: (X.Y.); (W.Y.)
| | - Weiren Yang
- College of Animal Sciences and Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China
- Correspondence: (X.Y.); (W.Y.)
| |
Collapse
|
14
|
Feng Q, Ma X, Cheng K, Liu G, Li Y, Yue Y, Liang J, Zhang L, Zhang T, Wang X, Gao X, Nie G, Zhao X. Engineered Bacterial Outer Membrane Vesicles as Controllable Two-Way Adaptors to Activate Macrophage Phagocytosis for Improved Tumor Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2206200. [PMID: 35985666 DOI: 10.1002/adma.202206200] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/08/2022] [Indexed: 06/15/2023]
Abstract
The most immune cells infiltrating tumor microenvironment (TME), tumor-associated macrophages (TAMs) closely resemble immunosuppressive M2-polarized macrophages. Moreover, tumor cells exhibit high expression of CD47 "don't eat me" signal, which obstructs macrophage phagocytosis. The precise and efficient activation of TAMs is a promising approach to tumor immunotherapy; however, re-education of macrophages remains a challenge. Bacteria-derived outer membrane vesicles (OMVs) are highly immunogenic nanovesicles that can robustly stimulate macrophages. Here, an OMV-based controllable two-way adaptor is reported, in which a CD47 nanobody (CD47nb) is fused onto OMV surface (OMV-CD47nb), with the outer surface coated with a polyethylene glycol (PEG) layer containing diselenide bonds (PEG/Se) to form PEG/Se@OMV-CD47nb. The PEG/Se layer modification not only mitigates the immunogenicity of OMV-CD47nb, thereby remarkedly increasing the dose that can be administered safely through intravenous injection, but also equips the formulation with radiation-triggered controlled release of OMV-CD47nb. Application of radiation to tumors in mice injected with the nanoformulation results in remodeling of TME. As two-way adaptors, OMV-CD47nb activates TAM phagocytosis of tumor cells via multiple pathways, including induction of M1 polarization and blockade of "don't eat me" signal. Moreover, this activation of TAMs results in the stimulation of T cell-mediated antitumor immunity through effective antigen presentation.
Collapse
Affiliation(s)
- Qingqing Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Xiaotu Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Yao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Yale Yue
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Lizhuo Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Tianjiao Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Xinwei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Xiaoyu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
15
|
Tanaka M, Abe S. Augmented anti-tumour effect of lipopolysaccharide with G-CSF without enhancing body weight reduction in mice bearing MH134 hepatoma. Eur J Pharmacol 2022; 934:175206. [DOI: 10.1016/j.ejphar.2022.175206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 12/01/2022]
|
16
|
Li S, Yue H, Wang S, Li X, Wang X, Guo P, Ma G, Wei W. Advances of bacteria-based delivery systems for modulating tumor microenvironment. Adv Drug Deliv Rev 2022; 188:114444. [PMID: 35817215 DOI: 10.1016/j.addr.2022.114444] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 12/13/2022]
Abstract
The components and hospitable properties of tumor microenvironment (TME) are associated with tumor progression. Recently, TME modulating vectors and strategies have garnished significant attention in cancer therapy. Although a pilot work has reviewed TME regulation via nanoparticle-based delivery systems, there is no systematical review that summarizes the natural bacteria-based anti-tumor system to modulate TME. In this review, we conclude the strategies of bacterial carriers (including whole bacteria, bacterial skeleton and bacterial components) to regulate TME from the perspective of TME components and hospitable properties, and the clinical trials of bacteria-mediated cancer therapy. Current challenges and future prospects for the design of bacteria-based carriers are also proposed that provide critical insights into this natural delivery system and related translation from the bench to the clinic.
Collapse
Affiliation(s)
- Shuping Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Hua Yue
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Xin Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Xiaojun Wang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Peilin Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
17
|
Dominguez EC, Phandthong R, Nguyen M, Ulu A, Guardado S, Sveiven S, Talbot P, Nordgren TM. Aspirin-Triggered Resolvin D1 Reduces Chronic Dust-Induced Lung Pathology without Altering Susceptibility to Dust-Enhanced Carcinogenesis. Cancers (Basel) 2022; 14:1900. [PMID: 35454807 PMCID: PMC9032113 DOI: 10.3390/cancers14081900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/28/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, with increased risk being associated with unresolved or chronic inflammation. Agricultural and livestock workers endure significant exposure to agricultural dusts on a routine basis; however, the chronic inflammatory and carcinogenic effects of these dust exposure is unclear. We have developed a chronic dust exposure model of lung carcinogenesis in which mice were intranasally challenged three times a week for 24 weeks, using an aqueous dust extract (HDE) made from dust collected in swine confinement facilities. We also treated mice with the omega-3-fatty acid lipid mediator, aspirin-triggered resolvin D1 (AT-RvD1) to provide a novel therapeutic strategy for mitigating the inflammatory and carcinogenic effects of HDE. Exposure to HDE resulted in significant immune cell influx into the lungs, enhanced lung tumorigenesis, severe tissue pathogenesis, and a pro-inflammatory and carcinogenic gene signature, relative to saline-exposed mice. AT-RvD1 treatment mitigated the dust-induced inflammatory response but did not protect against HDE + NNK-enhanced tumorigenesis. Our data suggest that chronic HDE exposure induces a significant inflammatory and pro-carcinogenic response, whereas treatment with AT-RvD1 dampens the inflammatory responses, providing a strong argument for the therapeutic use of AT-RvD1 to mitigate chronic inflammation.
Collapse
Affiliation(s)
- Edward C. Dominguez
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, CA 92521, USA; (E.C.D.); (P.T.)
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
| | - Rattapol Phandthong
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA 92521, USA;
| | - Matthew Nguyen
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
| | - Arzu Ulu
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
| | - Stephanie Guardado
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
| | - Stefanie Sveiven
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
| | - Prue Talbot
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, CA 92521, USA; (E.C.D.); (P.T.)
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA 92521, USA;
| | - Tara M. Nordgren
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, CA 92521, USA; (E.C.D.); (P.T.)
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
18
|
Bose S, Saha P, Chatterjee B, Srivastava AK. Chemokines driven ovarian cancer progression, metastasis and chemoresistance: potential pharmacological targets for cancer therapy. Semin Cancer Biol 2022; 86:568-579. [DOI: 10.1016/j.semcancer.2022.03.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 12/18/2022]
|
19
|
Lin X, Fang Y, Jin X, Zhang M, Shi K. Modulating Repolarization of Tumor-Associated Macrophages with Targeted Therapeutic Nanoparticles as a Potential Strategy for Cancer Therapy. ACS APPLIED BIO MATERIALS 2021; 4:5871-5896. [PMID: 35006894 DOI: 10.1021/acsabm.1c00461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There are always some components in the tumor microenvironment (TME), such as tumor-associated macrophages (TAMs), that help tumor cells escape the body's immune surveillance. Therefore, this situation can lead to tumor growth, progression, and metastasis, resulting in low response rates for cancer therapy. Macrophages play an important role with strong plasticity and functional diversity. Facing different microenvironmental stimulations, macrophages undergo a dynamic change in phenotype and function into two major macrophage subpopulations, namely classical activation/inflammation (M1) and alternative activation/regeneration (M2) type. Through various signaling pathways, macrophages polarize into complex groups, which can perform different immune functions. In this review, we emphasize the use of nanopreparations for macrophage related immunotherapy based on the pathological knowledge of TAMs phenotype. These macrophages targeted nanoparticles re-edit and re-educate macrophages by attenuating M2 macrophages and reducing aggregation to the TME, thereby relieving or alleviating immunosuppression. Among them, we describe in detail the cellular mechanisms and regulators of several major signaling pathways involved in the plasticity and polarization functions of macrophages. The advantages and challenges of those nanotherapeutics for these pathways have been elucidated, providing the basis and insights for the diagnosis and treatment strategies of various diseases centered on macrophages.
Collapse
Affiliation(s)
- Xiaojie Lin
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Yan Fang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Xuechao Jin
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Mingming Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Kai Shi
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, 300350 Tianjin, China
| |
Collapse
|
20
|
Hannon G, Prina-Mello A. Endotoxin contamination of engineered nanomaterials: Overcoming the hurdles associated with endotoxin testing. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1738. [PMID: 34254460 DOI: 10.1002/wnan.1738] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 12/30/2022]
Abstract
Nanomaterials are highly susceptible to endotoxin contamination due their large surface-to-volume ratios and endotoxins propensity to associate readily to hydrophobic and cationic surfaces. Additionally, the stability of endotoxin ensures it cannot be removed efficiently through conventional sterilization techniques such as autoclaving and ionizing radiation. In recent times, the true significance of this hurdle has come to light with multiple reports from the United States Nanotechnology Characterization Laboratory, in particular, along with our own experiences of endotoxin testing from multiple Horizon 2020-funded projects which highlight the importance of this issue for the clinical translation of nanomaterials. Herein, we provide an overview on the topic of endotoxin contamination of nanomaterials intended for biomedical applications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Gary Hannon
- Nanomedicine and Molecular Imaging Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Dublin, Ireland.,Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Adriele Prina-Mello
- Nanomedicine and Molecular Imaging Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, Dublin, Ireland.,Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
21
|
Secretoglobin 3A2 eliminates human cancer cells through pyroptosis. Cell Death Discov 2021; 7:12. [PMID: 33452234 PMCID: PMC7810848 DOI: 10.1038/s41420-020-00385-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/24/2020] [Accepted: 11/28/2020] [Indexed: 11/09/2022] Open
Abstract
Non-canonical inflammasome activation that recognizes intracellular lipopolysaccharide (LPS) causes pyroptosis, the inflammatory death of innate immune cells. The role of pyroptosis in innate immune cells is to rapidly eliminate pathogen-infected cells and limit the replication niche in the host body. Whether this rapid cell elimination process of pyroptosis plays a role in elimination of cancer cells is largely unknown. Our earlier study demonstrated that a multi-functional secreted protein, secretoglobin (SCGB) 3A2, chaperones LPS to cytosol, and activates caspase-11 and the non-canonical inflammasome pathway, leading to pyroptosis. Here we show that SCGB3A2 exhibits marked anti-cancer activity against 5 out of 11 of human non-small cell lung cancer cell lines in mouse xenographs, while no effect was observed in 6 of 6 small cell lung cancer cell lines examined. All SCGB3A2-LPS-sensitive cells express syndecan 1 (SDC1), a SCGB3A2 cell surface receptor, and caspase-4 (CASP4), a critical component of the non-canonical inflammasome pathway. Two epithelial-derived colon cancer cell lines expressing SDC1 and CASP4 were also susceptible to SCGB3A2-LPS treatment. TCGA analysis revealed that lung adenocarcinoma patients with higher SCGB3A2 mRNA levels exhibited better survival. These data suggest that SCGB3A2 uses the machinery of pyroptosis for the elimination of human cancer cells via the non-canonical inflammasome pathway, and that SCGB3A2 may serve as a novel therapeutic to treat cancer, perhaps in combination with immuno and/or targeted therapies.
Collapse
|
22
|
Sauvé JF, Locke SJ, Josse PR, Stapleton EM, Metwali N, Altmaier RW, Andreotti G, Thorne PS, Hofmann JN, Beane Freeman LE, Friesen MC. Characterization of inhalable endotoxin, glucan, and dust exposures in Iowa farmers. Int J Hyg Environ Health 2020; 228:113525. [PMID: 32311660 PMCID: PMC8010939 DOI: 10.1016/j.ijheh.2020.113525] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND The observed deficit of lung cancer in farmers has been partly attributed to exposure to organic dusts and endotoxins based largely on surrogate metrics. To move beyond these surrogates for etiological studies, we characterized task-based and time-weighted average (TWA) exposure to inhalable endotoxin, (1 → 3)-β-D-glucan, and dust in Iowa farmers. METHODS We collected 320 personal inhalable dust samples from 32 farmers during 69 sample days in 2015 and 2016. Samples were collected using Button aerosol samplers and analyzed for endotoxin using a kinetic chromogenic amebocyte lysate assay, and for (1 → 3)-β-D-glucan using a Limulus endpoint assay. We assessed relationships between bioaerosol concentrations and selected tasks and farm characteristics using linear mixed-effects models. RESULTS Bedding work, hog handling, and working in barn/confinement buildings, grain bins, and grain elevators were associated with higher endotoxin exposure. We found a monotonic trend between higher endotoxin concentrations and increasing number of animals. Bedding work, cleaning, and feed/grain storage work were associated with higher (1 → 3)-β-D-glucan concentrations. The median concentrations by task spanned one order of magnitude for inhalable dust and two orders of magnitude for endotoxin and (1 → 3)-β-D-glucan. Pearson correlations between endotoxin and glucan concentrations were 0.22 for TWA exposure and 0.56 for task samples. CONCLUSIONS This characterization of exposure factors that influence bioaerosol concentrations can support the development of refined bioaerosol exposure metrics for future etiologic analyses of cancer and other health outcomes in farmers.
Collapse
Affiliation(s)
- Jean-François Sauvé
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| | - Sarah J Locke
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| | - Pabitra R Josse
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| | - Emma M Stapleton
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA, United States
| | - Nervana Metwali
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA, United States
| | - Ralph W Altmaier
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA, United States
| | - Gabriella Andreotti
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| | - Peter S Thorne
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA, United States
| | - Jonathan N Hofmann
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| | - Laura E Beane Freeman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| | - Melissa C Friesen
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States.
| |
Collapse
|
23
|
Yang J, Kim EK, Park HJ, McDowell A, Kim YK. The impact of bacteria-derived ultrafine dust particles on pulmonary diseases. Exp Mol Med 2020; 52:338-347. [PMID: 32203101 PMCID: PMC7156658 DOI: 10.1038/s12276-019-0367-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/06/2019] [Accepted: 11/14/2019] [Indexed: 01/04/2023] Open
Abstract
The relationship between ambient particulate matter exposure and health has been well established. Ultrafine particles (UFP) with a diameter of 100 nm or less are known to increase pulmonary disease risk. Biological factors in dust containing UFP can cause severe inflammatory reactions. Pulmonary diseases develop primarily as a result of chronic inflammation caused by immune dysfunction. Thus, this review focuses on the adverse pulmonary effects of biological UFP, principally lipopolysaccharide (LPS), and bacterial extracellular vesicles (EVs), in indoor dust and the pathophysiological mechanisms involved in the development of chronic pulmonary diseases. The impact of LPS-induced pulmonary inflammation is based primarily on the amount of inhaled LPS. When relatively low levels of LPS are inhaled, a cascade of immune responses leads to Th2 cell induction, and IL-5 and IL-13 released by Th2 cells contributes to asthma development. Conversely, exposure to high levels of LPS induces a Th17 cell response, leading to increased production of IL-17, which is associated with asthma, COPD, and lung cancer incidence. Responses to bacterial EV exposure can similarly be broadly divided based on whether one of two mechanisms, either intracellular or extracellular, is activated, which depends on the type of the parent cell. Extracellular bacteria-derived EVs can cause neutrophilic inflammation via Th17 cell induction, which is associated with asthma, emphysema, COPD, and lung cancer. On the other hand, intracellular bacteria-derived EVs lead to mononuclear inflammation via Th1 cell induction, which increases the risk of emphysema. In conclusion, future measures should focus on the overall reduction of LPS sources in addition to the improvement of the balance of inhaled bacterial EVs in the indoor environment to minimize pulmonary disease risk.
Collapse
Affiliation(s)
- Jinho Yang
- Institute of MD Healthcare Inc., Seoul, Republic of Korea
- Department of Health and Safety Convergence Science, Graduate School of Korea University, Seoul, Republic of Korea
| | - Eun Kyoung Kim
- Institute of MD Healthcare Inc., Seoul, Republic of Korea
| | - Hyeon Ju Park
- Institute of MD Healthcare Inc., Seoul, Republic of Korea
| | | | - Yoon-Keun Kim
- Institute of MD Healthcare Inc., Seoul, Republic of Korea.
| |
Collapse
|
24
|
Kokoulin MS, Filshtein AP, Romanenko LA, Chikalovets IV, Chernikov OV. Structure and bioactivity of sulfated α-D-mannan from marine bacterium Halomonas halocynthiae KMM 1376T. Carbohydr Polym 2020; 229:115556. [DOI: 10.1016/j.carbpol.2019.115556] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/26/2019] [Accepted: 10/29/2019] [Indexed: 11/29/2022]
|
25
|
El-Zaemey S, Schinasi LH, Ferro G, Tual S, Lebailly P, Baldi I, Nordby KC, Kjaerheim K, Schüz J, Monnereau A, Brouwer M, Koutros S, Hofmann JN, Kristensen P, Kromhout H, Leon ME, Beane Freeman LE. Animal farming and the risk of lymphohaematopoietic cancers: a meta-analysis of three cohort studies within the AGRICOH consortium. Occup Environ Med 2019; 76:827-837. [PMID: 31302607 PMCID: PMC10304413 DOI: 10.1136/oemed-2018-105655] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/10/2019] [Accepted: 06/22/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Animal farming entails a variety of potential exposures, including infectious agents, endotoxins and pesticides, which may play a role in the aetiology of lymphohaematopoietic cancers (LHCs). The aim of this study was to assess whether farming specific animal species is associated with the risk of overall LHC or its subtypes. METHODS Data from three prospective cohort studies in the USA, France and Norway which are part of the Agricultural Cohort consortium and which collected information about animal farming and cancer were used. Analyses included 316 270 farmers and farm workers. Adjusted Cox models were used to investigate the associations of 13 histological subtypes of LHC (n=3282) with self-reported livestock (cattle, pigs and sheep/goats) and poultry (ever/never and numbers raised) farming. Cohort-specific HRs were combined using random-effects meta-analysis. RESULTS Ever animal farming in general or farming specific animal species was not meta-associated with overall LHC. The risk of myeloid malignancies decreased with increasing number of livestock (p trend=0.01). Increased risk of myeloproliferative neoplasms was seen with increasing number of sheep/goats (p trend <0.01), while a decreased risk was seen with increasing number of livestock (p trend=0.02). Between cohorts, we observed heterogeneity in the association of type of animal farmed and various LHC subtypes. CONCLUSIONS This large-scale study of three prospective agricultural cohorts showed no association between animal farming and LHC risk, but few associations between specific animal species and LHC subtypes were observed. The observed differences in associations by countries warrant further investigations.
Collapse
Affiliation(s)
- Sonia El-Zaemey
- Section of Environment and Radiation, International Agency for Research on Cancer (IARC), France
- School of Public Health, Curtin University, Perth, Australia
| | - Leah H. Schinasi
- Section of Environment and Radiation, International Agency for Research on Cancer (IARC), France
- Department of Environmental and Occupational Health, School of Public Health, Drexel University, Philadelphia, USA
| | - Gilles Ferro
- Section of Environment and Radiation, International Agency for Research on Cancer (IARC), France
| | - Séverine Tual
- ANTICIPE, U1086 INSERM, Université de Caen Normandie, and Centre de Lutte Contre le Cancer François Baclesse, Caen, France
| | - Pierre Lebailly
- ANTICIPE, U1086 INSERM, Université de Caen Normandie, and Centre de Lutte Contre le Cancer François Baclesse, Caen, France
| | - Isabelle Baldi
- CHU de Bordeaux, Service de Médecine du Travail et Pathologie Professionnelle, Bordeaux, France
| | - Karl-Christian Nordby
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health (STAMI), Oslo, Norway
| | | | - Joachim Schüz
- Section of Environment and Radiation, International Agency for Research on Cancer (IARC), France
| | - Alain Monnereau
- Hematological Malignancies Registry of Gironde, Bergonie Institute, Comprehensive Cancer Centre, Bordeaux, France
- University of Bordeaux, INSERM U1219 Center - EPICENE Team, CHU de Bordeaux, Bordeaux, France
| | - Maartje Brouwer
- Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, The Netherlands
| | - Stella Koutros
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), Bethesda, MD, USA
| | - Jonathan N Hofmann
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), Bethesda, MD, USA
| | - Petter Kristensen
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health (STAMI), Oslo, Norway
| | - Hans Kromhout
- Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, The Netherlands
| | - Maria E. Leon
- Section of Environment and Radiation, International Agency for Research on Cancer (IARC), France
| | - Laura E. Beane Freeman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), Bethesda, MD, USA
| |
Collapse
|
26
|
Douglas KO, Samuels TA, Gittens-St Hilaire M. Serum LPS Associated with Hantavirus and Dengue Disease Severity in Barbados. Viruses 2019; 11:v11090838. [PMID: 31505806 PMCID: PMC6783883 DOI: 10.3390/v11090838] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/25/2022] Open
Abstract
Hantavirus and dengue virus (DENV) infections are caused by RNA viruses which infect immune systems’ cells including monocytes, macrophages and dendritic cells and occur year-round in Barbados. A retrospective serological study (2008–2015) was conducted on hantavirus and dengue patient sera confirmed by IgM and IgG ELISA, NS1 and RT-PCR using Limulus amoebocyte lysate (LAL) kinetic turbidimetric method to determine serum endotoxin levels. Hantavirus patients were categorized into two groups, namely (a) hospitalized and (b) non-hospitalized. Dengue patients were categorized into 3 groups using 2009 WHO dengue guidelines (a) severe dengue (SD), (b) hospitalized non-severe dengue (non-SD) and (c) non-hospitalized non-SD. Statistical analyses were conducted to determine the association of endotoxin levels with hantavirus disease severity based on hospitalization and dengue disease severity. Serum endotoxin levels are associated with hantavirus disease severity and hospitalization and dengue disease severity (p < 0.01). Similar studies have found an association of serum endotoxin levels with dengue disease severity but never with hantavirus infection. Co-detection of hantavirus- and DENV-specific IgM in some patients were observed with elevated serum endotoxin levels. In addition, previous studies observed hantavirus replication in the gut of patients, gastrointestinal tract as a possible entry route of infection and evidence of microbial translocation and its impact on hantavirus disease severity. A significant correlation of serum endotoxin and hantavirus disease severity and hospitalization in hantavirus infected patients is reported for the first time ever. In addition, serum endotoxin levels correlated with dengue disease severity. This study adds further support to the role of endotoxin in both hantavirus and dengue virus infection and disease severity and its role as a possible therapeutic target for viral haemorrhagic fevers (VHFs).
Collapse
Affiliation(s)
- Kirk Osmond Douglas
- Faculty of Medical Sciences, University of the West Indies, Cave Hill, BB11000 St. Michael, Barbados.
| | - Thelma Alafia Samuels
- Faculty of Medical Sciences, University of the West Indies, Cave Hill, BB11000 St. Michael, Barbados.
- George Alleyne Chronic Disease Research Centre (GA-CDRC), University of the West Indies, Cave Hill, BB11000 St. Michael, Barbados.
| | - Marquita Gittens-St Hilaire
- Faculty of Medical Sciences, University of the West Indies, Cave Hill, BB11000 St. Michael, Barbados.
- Best-Dos Santos Public Health Laboratory, University of the West Indies, Cave Hill, BB11000 St. Michael, Barbados.
| |
Collapse
|
27
|
Ignacio RMC, Lee ES, Son DS. Potential Roles of Innate Immune Chemokine and Cytokine Network on Lipopolysaccharide-Based Therapeutic Approach in Ovarian Cancer. Immune Netw 2019; 19:e22. [PMID: 31281719 PMCID: PMC6597445 DOI: 10.4110/in.2019.19.e22] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/07/2019] [Accepted: 05/22/2019] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer (OC), the deadliest gynecological cancer, results in poor overall survival, urgently requiring a novel therapeutic approach. As cumulative exposures to endotoxins decreased OC risk epidemiologically, we evaluated if LPS, a Toll-like receptor 4 agonist known as active component of endotoxins, could increase survival in the murine peritoneal dissemination model of SKOV-3 OC cells. LPS significantly increased the mean survival time of more than 116 days compared with 63 days in the control. Furthermore, no tumor burden was present in three mice among eight LPS-treated mice. SKOV-3 cells were not responsive to LPS and showed unaltered chemokine signature. Rather than direct effects to OC cells, LPS was found to increase proinflammatory chemokines and cytokines, such as CXCL1, CXCL8, TNF, and IL-1B, in innate immune system. Taken together, LPS is likely to potentiate the cytotoxic-related innate immunogenicity via proinflammatory chemokines and cytokines, which attenuates the peritoneal dissemination of OC.
Collapse
Affiliation(s)
- Rosa Mistica C Ignacio
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Eun-Sook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL 32301, USA
| | - Deok-Soo Son
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
28
|
Yokoyama S, Cai Y, Murata M, Tomita T, Yoneda M, Xu L, Pilon AL, Cachau RE, Kimura S. A novel pathway of LPS uptake through syndecan-1 leading to pyroptotic cell death. eLife 2018; 7:e37854. [PMID: 30526845 PMCID: PMC6286126 DOI: 10.7554/elife.37854] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 11/08/2018] [Indexed: 01/15/2023] Open
Abstract
Intracellular lipopolysaccharide (LPS) triggers the non-canonical inflammasome pathway, resulting in pyroptosis of innate immune cells. In addition to its well-known proinflammatory role, LPS can directly cause regression of some tumors, although the underlying mechanism has remained unknown. Here we show that secretoglobin(SCGB)3A2, a small protein predominantly secreted in airways, chaperones LPS to the cytosol through the cell surface receptor syndecan-1; this leads to pyroptotic cell death driven by caspase-11. SCGB3A2 and LPS co-treatment significantly induced pyroptosis of macrophage RAW264.7 cells and decreased cancer cell proliferation in vitro, while SCGB3A2 treatment resulted in reduced progression of xenograft tumors in mice. These data suggest a conserved function for SCGB3A2 in the innate immune system and cancer cells. These findings demonstrate a critical role for SCGB3A2 as an LPS delivery vehicle; they reveal one mechanism whereby LPS enters innate immune cells leading to pyroptosis, and they clarify the direct effect of LPS on cancer cells.
Collapse
MESH Headings
- Animals
- Biological Transport
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/mortality
- Caspases/genetics
- Caspases/immunology
- Caspases, Initiator
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic
- Humans
- Immunity, Innate
- Lipopolysaccharides/pharmacology
- Lymphatic Metastasis
- Male
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/mortality
- Mice
- Mice, Transgenic
- Protein Array Analysis
- Pyroptosis/drug effects
- Pyroptosis/genetics
- Pyroptosis/immunology
- RAW 264.7 Cells
- RNA, Small Interfering/genetics
- RNA, Small Interfering/immunology
- Secretoglobins/antagonists & inhibitors
- Secretoglobins/genetics
- Secretoglobins/immunology
- Signal Transduction
- Survival Analysis
- Syndecan-1/antagonists & inhibitors
- Syndecan-1/genetics
- Syndecan-1/immunology
- Toll-Like Receptor 4/antagonists & inhibitors
- Toll-Like Receptor 4/genetics
- Toll-Like Receptor 4/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Shigetoshi Yokoyama
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Yan Cai
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Miyuki Murata
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Takeshi Tomita
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Mitsuhiro Yoneda
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Lei Xu
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaUnited States
| | | | - Raul E Cachau
- Advanced Biomedical Computing CenterFrederick National Laboratory for Cancer Research, Leidos Biomedical Inc.FrederickUnited States
| | - Shioko Kimura
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
29
|
Lin T, Pajarinen J, Kohno Y, Huang JF, Maruyama M, Romero-Lopez M, Nathan K, Yao Z, Goodman SB. Trained murine mesenchymal stem cells have anti-inflammatory effect on macrophages, but defective regulation on T-cell proliferation. FASEB J 2018; 33:4203-4211. [PMID: 30521384 DOI: 10.1096/fj.201801845r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mesenchymal stem cell (MSC)-mediated immunomodulation affects both innate and adaptive immune systems. These responses to environmental cues, such as pathogen-associated molecular patterns, damage-associated molecular patterns, or proinflammatory cytokines, are crucial for resolution of inflammation, as well as successful tissue healing and regeneration. We observed that intermittent, repeated exposure of MSCs to LPS induced stronger NF-κB activation than singular stimulation. A similar phenomenon, named innate immune memory or trained immunity, has been reported with macrophages. However, the potential regulation of "immune memory" in nonclassic immune cells, such as MSCs, has not been reported. In the current study, we chose IFN-γ plus TNF-α restimulation-induced iNOS expression as a model of MSC activation, because IFN-γ and TNF-α play crucial roles in MSC-mediated immunomodulation. The iNOS expression was enhanced in LPS-trained MSCs, 3 d after a washout period following primary stimulation. LPS-trained MSCs enhanced the anti-inflammatory (arginase 1 and CD206) marker expression, but decreased the proinflammatory marker (TNF-α, IL-1β, iNOS, and IL-6) expression using an MSC-macrophage coculture model. In contrast, LPS-trained MSCs demonstrated a defective regulation on CD4 T-cell proliferation. Mechanistic studies suggested that histone methylation and the JNK pathway are involved in LPS-trained immunomodulation in MSCs. Our results demonstrate differential immunomodulatory effects of trained MSCs on macrophages and T cells. These immunomodulatory consequences are critical, because they will have a major impact on current MSC-based cell therapies.-Lin, T., Pajarinen, J., Kohno, Y., Huang, J.-F., Maruyama, M., Romero-Lopez, M., Nathan, K., Yao, Z., Goodman, S. B. Trained murine mesenchymal stem cells have anti-inflammatory effect on macrophages, but defective regulation on T-cell proliferation.
Collapse
Affiliation(s)
- Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA; and
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA; and
| | - Yusuke Kohno
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA; and
| | - Jhih-Fong Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA; and
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA; and
| | - Monica Romero-Lopez
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA; and
| | - Karthik Nathan
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA; and
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA; and
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA; and.,Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
30
|
Kokoulin MS, Kuzmich AS, Romanenko LA, Menchinskaya ES, Mikhailov VV, Chernikov OV. Sulfated O-polysaccharide with anticancer activity from the marine bacterium Poseidonocella sedimentorum KMM 9023T. Carbohydr Polym 2018; 202:157-163. [DOI: 10.1016/j.carbpol.2018.08.107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/23/2018] [Accepted: 08/24/2018] [Indexed: 12/22/2022]
|
31
|
Müller E, Speth M, Christopoulos PF, Lunde A, Avdagic A, Øynebråten I, Corthay A. Both Type I and Type II Interferons Can Activate Antitumor M1 Macrophages When Combined With TLR Stimulation. Front Immunol 2018; 9:2520. [PMID: 30450098 PMCID: PMC6224375 DOI: 10.3389/fimmu.2018.02520] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/12/2018] [Indexed: 12/18/2022] Open
Abstract
Triggering or enhancing antitumor activity of tumor-associated macrophages is an attractive strategy for cancer treatment. We have previously shown that the cytokine interferon-γ (IFN-γ), a type II IFN, could synergize with toll-like receptor (TLR) agonists for induction of antitumor M1 macrophages. However, the toxicity of IFN-γ limits its clinical use. Here, we investigated whether the less toxic type I IFNs, IFN-α, and IFN-β, could potentially replace IFN-γ for induction of antitumor M1 macrophages. We measured in vitro the ability of type I and II IFNs to synergize with TLR agonists for transcription of inducible nitric oxide synthase (iNOS) mRNA and secretion of nitric oxide (NO) by mouse bone marrow-derived macrophages (BMDMs). An in vitro growth inhibition assay was used to measure both cytotoxic and cytostatic activity of activated macrophages against Lewis lung carcinoma (LLC) cancer cells. We found that both type I and II IFNs could synergize with TLR agonists in inducing macrophage-mediated inhibition of cancer cell growth, which was dependent on NO. The ability of high dose lipopolysaccharide (LPS) to induce tumoricidal activity in macrophages in the absence of IFN-γ was shown to depend on induction of autocrine type I IFNs. Antitumor M1 macrophages could also be generated in the absence of IFN-γ by a combination of two TLR ligands when using the TLR3 agonist poly(I:C) which induces autocrine type I IFNs. Finally, we show that encapsulation of poly(I:C) into nanoparticles improved its potency to induce M1 macrophages up to 100-fold. This study reveals the potential of type I IFNs for activation of antitumor macrophages and indicates new avenues for cancer immunotherapy based on type I IFN signaling, including combination of TLR agonists.
Collapse
Affiliation(s)
- Elisabeth Müller
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Martin Speth
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Panagiotis F Christopoulos
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Anna Lunde
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Ajna Avdagic
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Inger Øynebråten
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Alexandre Corthay
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| |
Collapse
|
32
|
Garcia E, Picciotto S, Neophytou AM, Bradshaw PT, Balmes JR, Eisen EA. Lung cancer mortality and exposure to synthetic metalworking fluid and biocides: controlling for the healthy worker survivor effect. Occup Environ Med 2018; 75:730-735. [PMID: 29743185 PMCID: PMC11460203 DOI: 10.1136/oemed-2017-104812] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 04/06/2018] [Accepted: 04/21/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Synthetic metalworking fluids (MWFs), widely used to cool and lubricate industrial machining and grinding operations, have been linked with increased risk of several cancers. Estimates of their relation with lung cancer, however, are inconsistent. Controlling for the healthy worker survivor effect, we examined the relations between lung cancer mortality and exposure to synthetic MWF, as well as to biocides added to water-based fluids to control microbial growth, in a cohort of autoworkers. Biocides served as a marker for endotoxin, which has reported antitumour effects, and were hypothesised to be the reason prior studies found reduced lung cancer risk associated with exposure to synthetic fluids. METHODS Using the parametric g-formula, we estimated risk ratios (RRs) comparing cumulative lung cancer mortality under no intervention with what would have occurred under hypothetical interventions reducing exposure to zero (ie, a ban) separately for two exposures: synthetic fluids and biocides. We also specified an intervention on synthetic MWF and biocides simultaneously to estimate joint effects. RESULTS Under a synthetic MWF ban, we observed decreased lung cancer mortality risk at age 86, RR=0.96 (0.91-1.01), but when we also intervened to ban biocides, the RR increased to 1.03 (0.95-1.11). A biocide-only ban increased lung cancer mortality (RR=1.07 (1.00-1.16)), with slightly larger RR in younger ages. CONCLUSIONS Findings suggest a modest positive association for synthetic MWF with lung cancer mortality, contrary to the negative associations reported in earlier studies. Biocide exposure, however, was inversely associated with risk of lung cancer mortality.
Collapse
Affiliation(s)
- Erika Garcia
- Environmental Health Sciences Division, School of Public Health, University of California, Berkeley, California, USA
| | - Sally Picciotto
- Environmental Health Sciences Division, School of Public Health, University of California, Berkeley, California, USA
| | - Andreas M Neophytou
- Environmental Health Sciences Division, School of Public Health, University of California, Berkeley, California, USA
| | - Patrick T Bradshaw
- Epidemiology Division, School of Public Health, University of California, Berkeley, California, USA
| | - John R Balmes
- Environmental Health Sciences Division, School of Public Health, University of California, Berkeley, California, USA
- Department of Medicine, Division of Occupational and Environmental Medicine, University of California, San Francisco, California, USA
| | - Ellen A Eisen
- Environmental Health Sciences Division, School of Public Health, University of California, Berkeley, California, USA
| |
Collapse
|
33
|
Shetab Boushehri MA, Lamprecht A. TLR4-Based Immunotherapeutics in Cancer: A Review of the Achievements and Shortcomings. Mol Pharm 2018; 15:4777-4800. [DOI: 10.1021/acs.molpharmaceut.8b00691] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, D-53121 Bonn, Germany
- PEPITE EA4267, Univ. Bourgonge Franch-Comte, 25030 Besançon, France
| |
Collapse
|
34
|
Nowak-Sliwinska P, Alitalo K, Allen E, Anisimov A, Aplin AC, Auerbach R, Augustin HG, Bates DO, van Beijnum JR, Bender RHF, Bergers G, Bikfalvi A, Bischoff J, Böck BC, Brooks PC, Bussolino F, Cakir B, Carmeliet P, Castranova D, Cimpean AM, Cleaver O, Coukos G, Davis GE, De Palma M, Dimberg A, Dings RPM, Djonov V, Dudley AC, Dufton NP, Fendt SM, Ferrara N, Fruttiger M, Fukumura D, Ghesquière B, Gong Y, Griffin RJ, Harris AL, Hughes CCW, Hultgren NW, Iruela-Arispe ML, Irving M, Jain RK, Kalluri R, Kalucka J, Kerbel RS, Kitajewski J, Klaassen I, Kleinmann HK, Koolwijk P, Kuczynski E, Kwak BR, Marien K, Melero-Martin JM, Munn LL, Nicosia RF, Noel A, Nurro J, Olsson AK, Petrova TV, Pietras K, Pili R, Pollard JW, Post MJ, Quax PHA, Rabinovich GA, Raica M, Randi AM, Ribatti D, Ruegg C, Schlingemann RO, Schulte-Merker S, Smith LEH, Song JW, Stacker SA, Stalin J, Stratman AN, Van de Velde M, van Hinsbergh VWM, Vermeulen PB, Waltenberger J, Weinstein BM, Xin H, Yetkin-Arik B, Yla-Herttuala S, Yoder MC, Griffioen AW. Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis 2018; 21:425-532. [PMID: 29766399 PMCID: PMC6237663 DOI: 10.1007/s10456-018-9613-x] [Citation(s) in RCA: 455] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.
Collapse
Affiliation(s)
- Patrycja Nowak-Sliwinska
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, University of Lausanne, Rue Michel-Servet 1, CMU, 1211, Geneva 4, Switzerland.
- Translational Research Center in Oncohaematology, University of Geneva, Geneva, Switzerland.
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Elizabeth Allen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
| | - Andrey Anisimov
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Alfred C Aplin
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Hellmut G Augustin
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - David O Bates
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - R Hugh F Bender
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
- Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Andreas Bikfalvi
- Angiogenesis and Tumor Microenvironment Laboratory (INSERM U1029), University Bordeaux, Pessac, France
| | - Joyce Bischoff
- Vascular Biology Program and Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Barbara C Böck
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - Peter C Brooks
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Federico Bussolino
- Department of Oncology, University of Torino, Turin, Italy
- Candiolo Cancer Institute-FPO-IRCCS, 10060, Candiolo, Italy
| | - Bertan Cakir
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Anca M Cimpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Ondine Cleaver
- Department of Molecular Biology, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - George Coukos
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine and Dalton Cardiovascular Center, Columbia, MO, USA
| | - Michele De Palma
- School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Emily Couric Cancer Center, The University of Virginia, Charlottesville, VA, USA
| | - Neil P Dufton
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute, Leuven, Belgium
| | | | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London, UK
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bart Ghesquière
- Metabolomics Expertise Center, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, Metabolomics Expertise Center, KU Leuven, Leuven, Belgium
| | - Yan Gong
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Adrian L Harris
- Molecular Oncology Laboratories, Oxford University Department of Oncology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Nan W Hultgren
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | | | - Melita Irving
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Robert S Kerbel
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Jan Kitajewski
- Department of Physiology and Biophysics, University of Illinois, Chicago, IL, USA
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hynda K Kleinmann
- The George Washington University School of Medicine, Washington, DC, USA
| | - Pieter Koolwijk
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Elisabeth Kuczynski
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | | | - Juan M Melero-Martin
- Department of Cardiac Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Roberto F Nicosia
- Department of Pathology, University of Washington, Seattle, WA, USA
- Pathology and Laboratory Medicine Service, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Agnes Noel
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Jussi Nurro
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Tatiana V Petrova
- Department of oncology UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund, Sweden
| | - Roberto Pili
- Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| | - Jeffrey W Pollard
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Mark J Post
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Paul H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Department Surgery, LUMC, Leiden, The Netherlands
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine, National Council of Scientific and Technical Investigations (CONICET), Buenos Aires, Argentina
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Anna M Randi
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
- National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Curzio Ruegg
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Steven A Stacker
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre and The Sir Peter MacCallum, Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Jimmy Stalin
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Amber N Stratman
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Maureen Van de Velde
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Victor W M van Hinsbergh
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Peter B Vermeulen
- HistoGeneX, Antwerp, Belgium
- Translational Cancer Research Unit, GZA Hospitals, Sint-Augustinus & University of Antwerp, Antwerp, Belgium
| | - Johannes Waltenberger
- Medical Faculty, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hong Xin
- University of California, San Diego, La Jolla, CA, USA
| | - Bahar Yetkin-Arik
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Seppo Yla-Herttuala
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
35
|
Aspirin Disrupts the Crosstalk of Angiogenic and Inflammatory Cytokines between 4T1 Breast Cancer Cells and Macrophages. Mediators Inflamm 2018; 2018:6380643. [PMID: 30034291 PMCID: PMC6035832 DOI: 10.1155/2018/6380643] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/21/2018] [Accepted: 05/15/2018] [Indexed: 02/08/2023] Open
Abstract
The tumor microenvironment is rich in multiple cell types that influence tumor development. Macrophages infiltrate tumors, where they are the most abundant immune cell population and secrete a number of cytokines. Aspirin acts as a chemopreventive agent against cancer development. This study investigated whether aspirin regulates crosstalk between breast cancer cells and macrophages. To study these interactions in a tumor microenvironment, a conditioned media was employed using 4T1 breast cancer cells cultured in RAW 264.7 cell-conditioned medium (RAW-CM), and a cocultured model of both cells was used. When 4T1 cells were cultured in the RAW-CM, there were increases in cell viability and secretion of the cytokines VEGF, PAI-1, TNF-α, and IL-6. Treatment with aspirin inhibited 4T1 cell growth and migration and MCP-1, PAI-1, and IL-6 production. In the coculture of both cells, aspirin inhibited secretion of MCP-1, IL-6, and TGF-β. Furthermore, aspirin significantly decreased the M2 macrophage marker CD206, but increased M1 marker CD11c expression. In summary, aspirin treatment inhibited the crosstalk of 4T1 and RAW 264.7 cells through regulation of angiogenic and inflammatory mediator production and influenced the M1/M2 macrophage subtype. This highlighted that aspirin suppresses the tumor favorable microenvironment and could be a promising agent against triple-negative breast cancer.
Collapse
|
36
|
Müller E, Christopoulos PF, Halder S, Lunde A, Beraki K, Speth M, Øynebråten I, Corthay A. Toll-Like Receptor Ligands and Interferon-γ Synergize for Induction of Antitumor M1 Macrophages. Front Immunol 2017; 8:1383. [PMID: 29123526 PMCID: PMC5662546 DOI: 10.3389/fimmu.2017.01383] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/06/2017] [Indexed: 01/01/2023] Open
Abstract
Tumor-associated macrophages may either promote or suppress tumor growth depending on their activation status. Interferon-γ (IFN-γ) has been identified as a key factor for inducing tumoricidal M1 phenotype in macrophages. However, it remains unclear whether IFN-γ is sufficient or if additional stimuli are required. Here, we tested IFN-γ and a panel of toll-like receptor (TLR) agonists for the ability to activate murine macrophages toward a tumoricidal M1 phenotype. The following TLR ligands were used: TLR1/TLR2 agonist Pam3CSK4, TLR2/TLR6 agonist lipotechoic acid, TLR3 agonist poly(I:C), TLR4 agonist lipopolysaccharide (LPS), TLR5 agonist flagellin, TLR7 agonist CL264, and TLR9 agonist CpG. We used an in vitro growth inhibition assay to measure both cytotoxic and cytostatic activity of mouse macrophages against Lewis lung carcinoma (LLC) and MOPC315 plasmacytoma tumor cells. Production of nitric oxide (NO) and cytokines by activated macrophages was quantified. We found that IFN-γ alone was not able to render macrophages tumoricidal. Similarly, macrophage activation with single TLR agonists was inefficient. In sharp contrast, IFN-γ was shown to synergize with TLR agonists for induction of macrophage tumoricidal activity and production of both NO and pro-inflammatory cytokines (TNF-α, IL-12p40, and IL-12p70). Furthermore, IFN-γ was shown to suppress macrophage IL-10 secretion induced by TLR agonists. NO production was necessary for macrophage tumoricidal activity. We conclude that two signals from the microenvironment are required for optimal induction of antitumor M1 macrophage phenotype. Combination treatment with IFN-γ and TLR agonists may offer new avenues for macrophage-based cancer immunotherapy.
Collapse
Affiliation(s)
- Elisabeth Müller
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Panagiotis F Christopoulos
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Sanjib Halder
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Anna Lunde
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Kahsai Beraki
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Martin Speth
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Inger Øynebråten
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Alexandre Corthay
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| |
Collapse
|
37
|
Hofmann JN, Shiels MS, Friesen MC, Kemp TJ, Chaturvedi AK, Lynch CF, Parks CG, Pinto LA, Hildesheim A, Alavanja MCR, Beane Freeman LE. Industrial hog farming is associated with altered circulating immunological markers. Occup Environ Med 2017; 75:212-217. [PMID: 29055885 DOI: 10.1136/oemed-2017-104519] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 10/06/2017] [Accepted: 10/12/2017] [Indexed: 12/25/2022]
Abstract
OBJECTIVES The previously observed inverse association between hog farming and risk of lung cancer in the Agricultural Health Study (AHS) has been attributed to endotoxin exposure, the levels of which are particularly high in industrial hog confinement facilities. We conducted an investigation to explore the potential biological mechanisms underlying this association, as well as other immunological changes associated with hog farming. METHODS Serum immune marker levels were measured using a multiplexed bead-based assay in 61 active hog farmers and 61 controls matched on age, phlebotomy date and raising cattle. Both groups comprised non-smoking male AHS participants from Iowa. We compared natural log-transformed marker levels between hog farmers and controls using multivariate linear regression models. RESULTS Circulating levels of macrophage-derived chemokine (CCL22), a chemokine previously implicated in lung carcinogenesis, were reduced among hog farmers (17% decrease; 95% CI -28% to -4%), in particular for those with the largest operations (>6000 hogs: 26% decrease; 95% CI -39% to -10%; ptrend=0.002). We also found that hog farmers had elevated levels of other immune markers, including macrophage inflammatory protein-3 alpha (MIP-3A/CCL20; 111% increase, 95% CI 19% to 273%), basic fibroblast growth factor (FGF-2; 93% increase, 95% CI 10% to 240%) and soluble interleukin-4 receptor (12% increase, 95% CI 1% to 25%), with particularly strong associations for MIP-3A/CCL20 and FGF-2 in winter. CONCLUSIONS These results provide insights into potential immunomodulatory mechanisms through which endotoxin or other exposures associated with hog farming may influence lung cancer risk, and warrant further investigation with more detailed bioaerosol exposure assessment.
Collapse
Affiliation(s)
- Jonathan N Hofmann
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Meredith S Shiels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Melissa C Friesen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Troy J Kemp
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, USA
| | - Anil K Chaturvedi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Christine G Parks
- Department of Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Ligia A Pinto
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, USA
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Michael C R Alavanja
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Laura E Beane Freeman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
38
|
Tual S, Lemarchand C, Boulanger M, Dalphin JC, Rachet B, Marcotullio E, Velten M, Guizard AV, Clin B, Baldi I, Lebailly P. Exposure to Farm Animals and Risk of Lung Cancer in the AGRICAN Cohort. Am J Epidemiol 2017; 186:463-472. [PMID: 28830081 DOI: 10.1093/aje/kwx125] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022] Open
Abstract
Epidemiologic studies have found lower risks of lung cancer in farmers. However, little is known about the types of agricultural activities concerned. In the Agriculture and Cancer cohort, we assessed the relationship between animal farming and lung cancer by investigating the types of animals, tasks, and timing of exposure. Analyses included 170,834 participants from the Agriculture and Cancer (AGRICAN) cohort in France. Incident lung cancers were identified through linkage with cancer registries from enrollment (2005-2007) to 2011. A Cox model, adjusting for pack-years of cigarette smoking, was used to calculate hazard ratios and 95% confidence intervals. Lung cancer risk was inversely related to duration of exposure to cattle (≥40 years: hazard ratio = 0.60, 95% confidence interval: 0.41, 0.89; P for trend < 0.01) and to horse farming (≥20 years: hazard ratio = 0.64, 95% confidence interval: 0.35, 1.17; P for trend = 0.09), especially for adenocarcinomas, but not with poultry or pig farming. More pronounced decreased risks were reported among individuals who had cared for animals, undertaken milking, and who had been exposed to cattle in infancy. Our study provides strong evidence of an inverse association between lung cancer and cattle and horse farming. Further research is warranted to identify the etiologic protective agents and biological mechanisms.
Collapse
|
39
|
Bauer AK, Upham BL, Rondini EA, Tennis MA, Velmuragan K, Wiese D. Toll-like receptor expression in human non-small cell lung carcinoma: potential prognostic indicators of disease. Oncotarget 2017; 8:91860-91875. [PMID: 29190881 PMCID: PMC5696147 DOI: 10.18632/oncotarget.19463] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 06/02/2017] [Indexed: 12/13/2022] Open
Abstract
Introduction Lung cancer remains the highest cause of cancer mortality worldwide. Toll-like receptors (TLR) are innate immune receptors that have both pro- and anti-tumorigenic properties. Based on findings from epidemiological studies and in rodents, we hypothesized that elevated TLR expression would be a positive prognostic indicator of disease in non-small cell lung carcinoma patients. Results Higher mRNA expression of TLR1-3 and 5-8 were significantly associated with increased overall survival (OS) when analyzed individually or as a group in both non-small cell lung carcinoma (NSCLC) patients and in the adenocarcinoma (ADC) subtype. Significant co-expression of many TLR combinations in ADC patients were also observed via RNA sequencing. Immunostaining demonstrated TLR4 and 8 significantly correlated in tumor tissue, similar to RNA. Methods We used kmplot.com to perform a meta-analysis on mRNA expression of TLR1-10 to determine any significant associations with OS in NSCLC and the ADC subtype. cBioportal was also used simultaneously to assess co-expression in TLR1-10 in ADC patients via RNA sequencing and to identify any molecular alterations. Lastly, immunostaining for a subset of TLRs was conducted on ADC patients. Conclusions Expression of innate immune receptors TLR1-10 is associated with improved survival outcomes in NSCLC. Thus, further evaluation of their predictive capacity and therapeutic utility is warranted.
Collapse
Affiliation(s)
- Alison K Bauer
- Department of Environmental and Occupational Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brad L Upham
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI 48824, USA
| | - Elizabeth A Rondini
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, USA
| | - Meredith A Tennis
- Department of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kalpana Velmuragan
- Department of Environmental and Occupational Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David Wiese
- McLaren Regional Medical Center, Flint, MI, 48532, USA
| |
Collapse
|
40
|
Tual S, Silverman DT, Koutros S, Blair A, Sandler DP, Lebailly P, Andreotti G, Hoppin JA, Freeman LEB. Use of Dieselized Farm Equipment and Incident Lung Cancer: Findings from the Agricultural Health Study Cohort. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:611-8. [PMID: 26452295 PMCID: PMC4858397 DOI: 10.1289/ehp.1409238] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 10/05/2015] [Indexed: 05/28/2023]
Abstract
BACKGROUND Diesel exhaust is a known lung carcinogen. Farmers use a variety of dieselized equipment and thus may be at increased risk of lung cancer, but farm exposures such as endotoxins may also be protective for lung cancer. OBJECTIVES We evaluated the relative risk of incident lung cancer, including histological subtype, from enrollment (1993-1997) to 2010-2011 in relation to farm equipment use in the Agricultural Health Study (AHS), a prospective cohort study of pesticide applicators and spouses in Iowa and North Carolina, USA. METHODS Farm equipment use was reported by 21,273 farmers and 29,840 spouses. Rate ratios (RRs) were estimated separately for farmers and spouses with Poisson regression models adjusted for smoking and other confounders. We conducted stratified analyses by exposure to animals or stored grain, a surrogate for endotoxin exposure. RESULTS Daily diesel tractor use (vs. no use) was positively associated with lung cancer in farmers (RR = 1.48; 95% CI: 0.87, 2.50; 35 exposed, 32 unexposed cases), particularly adenocarcinoma (RR = 3.39; 95% CI: 1.23, 9.33; 12 exposed, 7 unexposed cases). The association of adenocarcinoma with daily (vs. low/no) use of diesel tractors was stronger for farmers with no animal or stored grain exposures (RR = 6.23; 95% CI: 2.25, 17.25; 5 exposed, 18 unexposed cases) than among farmers with these exposures (RR = 1.19; 95% CI: 0.51, 2.79; 7 exposed, 27 unexposed cases) (p-interaction = 0.05). CONCLUSIONS This study provides preliminary evidence of an increased risk of lung adenocarcinoma among daily drivers of diesel tractors and suggests that exposure to endotoxins may modify the impact of diesel exposure on lung cancer risk. Confirmation of these findings with more exposed cases and more detailed exposure information is warranted. CITATION Tual S, Silverman DT, Koutros S, Blair A, Sandler DP, Lebailly P, Andreotti G, Hoppin JA, Beane Freeman LE. 2016. Use of dieselized farm equipment and incident lung cancer: findings from the Agricultural Health Study Cohort. Environ Health Perspect 124:611-618; http://dx.doi.org/10.1289/ehp.1409238.
Collapse
Affiliation(s)
- Séverine Tual
- INSERM, UMR 1086 Cancers et Préventions, Caen, France
- Université de Caen, Normandie, Caen, France
- Centre de Lutte Contre le Cancer François Baclesse, Caen, France
| | - Debra T. Silverman
- Division of Cancer Epidemiology and Genetics, Occupational and Environmental Epidemiology Branch, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Rockville, Maryland, USA
| | - Stella Koutros
- Division of Cancer Epidemiology and Genetics, Occupational and Environmental Epidemiology Branch, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Rockville, Maryland, USA
| | - Aaron Blair
- Division of Cancer Epidemiology and Genetics, Occupational and Environmental Epidemiology Branch, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Rockville, Maryland, USA
| | - Dale P. Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, DHHS, Research Triangle, North Carolina, USA
| | - Pierre Lebailly
- INSERM, UMR 1086 Cancers et Préventions, Caen, France
- Université de Caen, Normandie, Caen, France
- Centre de Lutte Contre le Cancer François Baclesse, Caen, France
| | - Gabriella Andreotti
- Division of Cancer Epidemiology and Genetics, Occupational and Environmental Epidemiology Branch, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Rockville, Maryland, USA
| | - Jane A. Hoppin
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Laura E. Beane Freeman
- Division of Cancer Epidemiology and Genetics, Occupational and Environmental Epidemiology Branch, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Rockville, Maryland, USA
| |
Collapse
|
41
|
Alexander CM, Xiong KN, Velmurugan K, Xiong J, Osgood RS, Bauer AK. Differential innate immune cell signatures and effects regulated by toll-like receptor 4 during murine lung tumor promotion. Exp Lung Res 2016; 42:154-73. [PMID: 27093379 PMCID: PMC5506691 DOI: 10.3109/01902148.2016.1164263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Tumor promotion is an early and critical stage during lung adenocarcinoma (ADC). We previously demonstrated that Tlr4 mutant mice were more susceptible to butylated hydroxytoluene (BHT)-induced pulmonary inflammation and tumor promotion in comparison to Tlr4-sufficient mice. Our study objective was to elucidate the underlying differences in Tlr4 mutant mice in innate immune cell populations, their functional responses, and the influence of these cellular differences on ADC progenitor (type II) cells following BHT-treatment. BALB (Tlr4-sufficient) and C.C3-Tlr4(Lps-d)/J (BALB(Lpsd); Tlr4 mutant) mice were treated with BHT (promoter) followed by bronchoalveolar lavage (BAL) and flow cytometry processing on the lungs. ELISAs, Club cell enrichment, macrophage function, and RNA isolation were also performed. Bone marrow-derived macrophages (BMDM) co-cultured with a type II cell line were used for wound healing assays. Innate immune cells significantly increased in whole lung in BHT-treated BALB(Lpsd) mice compared to BALB mice. BHT-treated BALB(Lpsd) mice demonstrated enhanced macrophage functionality, increased epithelial wound closure via BMDMs, and increased Club cell number in BALB(Lpsd) mice, all compared to BALB BHT-treated mice. Cytokine/chemokine (Kc, Mcp1) and growth factor (Igf1) levels also significantly differed among the strains and within macrophages, gene expression, and cell surface markers collectively demonstrated a more plastic phenotype in BALB(Lpsd) mice. Therefore, these correlative studies suggest that distinct innate immune cell populations are associated with the differences observed in the Tlr4-mutant model. Future studies will investigate the macrophage origins and the utility of the pathways identified herein as indicators of immune system deficiencies and lung tumorigenesis.
Collapse
Affiliation(s)
- Carla-Maria Alexander
- a Department of Environmental and Occupational Health , Colorado School of Public Health , University of Colorado at Denver-Anschutz Medical Campus , Aurora , Colorado , USA
| | - Ka-Na Xiong
- a Department of Environmental and Occupational Health , Colorado School of Public Health , University of Colorado at Denver-Anschutz Medical Campus , Aurora , Colorado , USA
| | - Kalpana Velmurugan
- a Department of Environmental and Occupational Health , Colorado School of Public Health , University of Colorado at Denver-Anschutz Medical Campus , Aurora , Colorado , USA
| | - Julie Xiong
- a Department of Environmental and Occupational Health , Colorado School of Public Health , University of Colorado at Denver-Anschutz Medical Campus , Aurora , Colorado , USA
| | - Ross S Osgood
- b Department of Pharmaceutical Sciences , School of Pharmacy , University of Colorado Denver , Aurora , Colorado , USA
| | - Alison K Bauer
- a Department of Environmental and Occupational Health , Colorado School of Public Health , University of Colorado at Denver-Anschutz Medical Campus , Aurora , Colorado , USA
| |
Collapse
|
42
|
Jing S, Chai W, Guo G, Zhang X, Dai J, Yan LJ. Comparison of antioxidant and antiproliferative activity between Kunlun Chrysanthemum flowers polysaccharides (KCCP) and fraction PII separated by column chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1019:169-177. [DOI: 10.1016/j.jchromb.2016.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 01/04/2016] [Accepted: 01/05/2016] [Indexed: 01/03/2023]
|
43
|
Olesch C, Sha W, Angioni C, Sha LK, Açaf E, Patrignani P, Jakobsson PJ, Radeke HH, Grösch S, Geisslinger G, von Knethen A, Weigert A, Brüne B. MPGES-1-derived PGE2 suppresses CD80 expression on tumor-associated phagocytes to inhibit anti-tumor immune responses in breast cancer. Oncotarget 2016; 6:10284-96. [PMID: 25871398 PMCID: PMC4496355 DOI: 10.18632/oncotarget.3581] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 02/13/2015] [Indexed: 01/04/2023] Open
Abstract
Prostaglandin E2 (PGE2) favors multiple aspects of tumor development and immune evasion. Therefore, microsomal prostaglandin E synthase (mPGES-1/-2), is a potential target for cancer therapy. We explored whether inhibiting mPGES-1 in human and mouse models of breast cancer affects tumor-associated immunity. A new model of breast tumor spheroid killing by human PBMCs was developed. In this model, tumor killing required CD80 expression by tumor-associated phagocytes to trigger cytotoxic T cell activation. Pharmacological mPGES-1 inhibition increased CD80 expression, whereas addition of PGE2, a prostaglandin E2 receptor 2 (EP2) agonist, or activation of signaling downstream of EP2 reduced CD80 expression. Genetic ablation of mPGES-1 resulted in markedly reduced tumor growth in PyMT mice. Macrophages of mPGES-1−/− PyMT mice indeed expressed elevated levels of CD80 compared to their wildtype counterparts. CD80 expression in tumor-spheroid infiltrating mPGES-1−/− macrophages translated into antigen-specific cytotoxic T cell activation. In conclusion, mPGES-1 inhibition elevates CD80 expression by tumor-associated phagocytes to restrict tumor growth. We propose that mPGES-1 inhibition in combination with immune cell activation might be part of a therapeutic strategy to overcome the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Catherine Olesch
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Weixiao Sha
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Carlo Angioni
- Institute of Clinical Pharmacology/ZAFES, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Lisa Katharina Sha
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Elias Açaf
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Paola Patrignani
- Department of Neuroscience, Imaging and Clinical Sciences and Center of Excellence on Aging (CeSI), "G. d'Annunzio" University, Chieti, Italy
| | - Per-Johan Jakobsson
- Department of Medicine, Rheumatology Research Unit, Karolinska Institutet, Stockholm, Sweden
| | - Heinfried H Radeke
- Pharmazentrum Frankfurt/ZAFES, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Sabine Grösch
- Institute of Clinical Pharmacology/ZAFES, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology/ZAFES, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Andreas von Knethen
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Occupational exposures in the agricultural industry are associated with numerous lung diseases, including chronic obstructive pulmonary disease, asthma, hypersensitivity pneumonitis, lung cancer, and interstitial lung diseases. Efforts are ongoing to ascertain contributing factors to these negative respiratory outcomes and improve monitoring of environmental factors leading to disease. In this review, recently published studies investigating the deleterious effects of occupational exposures in the agricultural industry are discussed. RECENT FINDINGS Occupational exposures to numerous agricultural environment aerosols, including pesticides, fungi, and bacteria are associated with impaired respiratory function and disease. Increases in certain farming practices, including mushroom and greenhouse farming, present new occupational exposure concerns. Improved detection methods may provide opportunities to better monitor safe exposure levels to known lung irritants. SUMMARY In the agricultural industry, occupational exposures to organic and inorganic aerosols lead to increased risk for lung disease among workers. Increased awareness of respiratory risks and improved monitoring of agricultural environments are necessary to limit pulmonary health risks to exposed populations.
Collapse
Affiliation(s)
| | - Kristina L. Bailey
- VA Nebraska Western Iowa Health Care System, Omaha, NE
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy Division, Omaha, NE
| |
Collapse
|
45
|
Shin JM, Gwak JW, Kamarajan P, Fenno JC, Rickard AH, Kapila YL. Biomedical applications of nisin. J Appl Microbiol 2016; 120:1449-65. [PMID: 26678028 DOI: 10.1111/jam.13033] [Citation(s) in RCA: 354] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/20/2015] [Accepted: 12/07/2015] [Indexed: 12/16/2022]
Abstract
Nisin is a bacteriocin produced by a group of Gram-positive bacteria that belongs to Lactococcus and Streptococcus species. Nisin is classified as a Type A (I) lantibiotic that is synthesized from mRNA and the translated peptide contains several unusual amino acids due to post-translational modifications. Over the past few decades, nisin has been used widely as a food biopreservative. Since then, many natural and genetically modified variants of nisin have been identified and studied for their unique antimicrobial properties. Nisin is FDA approved and generally regarded as a safe peptide with recognized potential for clinical use. Over the past two decades the application of nisin has been extended to biomedical fields. Studies have reported that nisin can prevent the growth of drug-resistant bacterial strains, such as methicillin-resistant Staphylococcus aureus, Streptococcus pneumoniae, Enterococci and Clostridium difficile. Nisin has now been shown to have antimicrobial activity against both Gram-positive and Gram-negative disease-associated pathogens. Nisin has been reported to have anti-biofilm properties and can work synergistically in combination with conventional therapeutic drugs. In addition, like host-defence peptides, nisin may activate the adaptive immune response and have an immunomodulatory role. Increasing evidence indicates that nisin can influence the growth of tumours and exhibit selective cytotoxicity towards cancer cells. Collectively, the application of nisin has advanced beyond its role as a food biopreservative. Thus, this review will describe and compare studies on nisin and provide insight into its future biomedical applications.
Collapse
Affiliation(s)
- J M Shin
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - J W Gwak
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - P Kamarajan
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - J C Fenno
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - A H Rickard
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Y L Kapila
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
46
|
Noe G. A hybrid fever therapy for increased tumor selectivity. Med Hypotheses 2016; 89:63-4. [PMID: 26968911 DOI: 10.1016/j.mehy.2016.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 02/01/2016] [Accepted: 02/03/2016] [Indexed: 12/12/2022]
Abstract
Lipopolysaccharide (LPS), mediator of the Coley and Shwartzman fever therapies, is a potent endotoxin capable of inducing sepsis at moderate intravenous doses, which currently limits its experimental and clinical use. By exploiting elevated G-CSF concentrations and neutrophil numbers within tumors, it may be possible to selectively trigger a sepsis-like syndrome in malignant tissues using only moderate hyperthermia and low-dose LPS, leaving normal organs unaffected. Analogous to the organ failure that can result from severe sepsis, repeated application of the proposed procedure could lead to tumor necrosis and regression. Biomarkers including heat shock proteins and circulating endothelial cells, and intravital microscopy of intravascular NETs, microthrombi, and bleeding in tumors can be used to test predicted consequences of this therapy. The two components of this hybrid approach, hyperthermia and acute but mild endotoxemia, could provide a tumor-specific therapeutic modality with relatively mild side effects, and therefore permit repeated application over an extended treatment period.
Collapse
Affiliation(s)
- Garry Noe
- Physics Department, Virginia Wesleyan College, 1584 Wesleyan Drive, Norfolk, VA 23502, United States.
| |
Collapse
|
47
|
Woappi Y, Singh OV. Commensals and Foodborne Pathogens can Arbitrate Epithelial-carcinogenesis. ACTA ACUST UNITED AC 2016; 15. [PMID: 31456935 PMCID: PMC6711482 DOI: 10.9734/bmrj/2016/26690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Major shifts in intestinal commensal bacteria often result in changes in CD4+ T lymphocyte populations, leading to an influx of Th17 cells, chronic inflammation, and eventually cancer. Consequently, the inappropriate propagation of certain commensal species in the gut has been associated with mucosal inflammatory diseases and cancer development. Recent experiments investigating the relationships between food-borne pathogens, enteric bacteria, and cancer have exposed the ability of certain bacterial species to significantly reduce tumor size and tumor progression in mice. In similar studies, pro-inflammatory Th17 and Th1 cells were at times found present along with anti-inflammatory Treg populations in the intestinal mucosa. This antitumor response was mediated by a balanced production of pro- and anti-inflammatory cytokines, resulting in a controlled threshold of mucosal immunity largely moderated by CD4+ T lymphocyte populations, through a dendritic cell-dependent pathway. These findings provide new evidence that certain species of bacteria can help manage subcutaneous tumor development by calibrating mucosal and, in some instances, systemic thresholds of innate and adaptive immunity.
Collapse
Affiliation(s)
- Yvon Woappi
- Division of Biological and Health Sciences, University of Pittsburgh, Bradford, PA-16701, USA
| | - Om V Singh
- Division of Biological and Health Sciences, University of Pittsburgh, Bradford, PA-16701, USA
| |
Collapse
|
48
|
CYP-epoxygenase metabolites of docosahexaenoic acid protect HL-1 cardiac cells against LPS-induced cytotoxicity Through SIRT1. Cell Death Discov 2015; 1. [PMID: 27182450 PMCID: PMC4864499 DOI: 10.1038/cddiscovery.2015.54] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Bacterial LPS is an environmental toxin capable of promoting various cardiac complications. Current evidence suggests that LPS-induced myocardial dysfunction emerges as a consequence of compromised quality of cardiac mitochondria. Docosahexaenoic acid (DHA, 22:6n3) is an n-3 polyunsaturated fatty acid (PUFA), which produces a broad spectrum of intrinsic physiological effects including regulation of cell survival and death mechanisms. Although, numerous studies revealed fundamentally beneficial effects of DHA on cardiovascular system, it remains unknown whether these effects were produced by DHA or one of its possibly more potent metabolites. Emerging evidence indicates that cytochrome P450 (CYP) epoxygenase metabolites of DHA, epoxydocosapentaenoic acids (EDPs), produce more potent biological activity compared to its precursor DHA. In this study, we investigated whether DHA and its metabolite 19,20-EDP could protect HL-1 cardiac cells against LPS-induced cytotoxicity. We provide evidence that exogenously added or DHA-derived EDPs promote mitochondrial biogenesis and function in HL-1 cardiac cells. Our results illustrate the CYP epoxygenase metabolite of DHA, 19,20-EDP, confers extensive protection to HL-1 cardiac cells against LPS-induced cytotoxicity via activation of SIRT1.
Collapse
|
49
|
Ahmadi SF, Streja E, Zahmatkesh G, Streja D, Kashyap M, Moradi H, Molnar MZ, Reddy U, Amin AN, Kovesdy CP, Kalantar-Zadeh K. Reverse Epidemiology of Traditional Cardiovascular Risk Factors in the Geriatric Population. J Am Med Dir Assoc 2015; 16:933-9. [PMID: 26363864 PMCID: PMC4636955 DOI: 10.1016/j.jamda.2015.07.014] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 07/21/2015] [Indexed: 01/08/2023]
Abstract
Traditional risk factors of cardiovascular death in the general population, including body mass index (BMI), serum cholesterol, and blood pressure (BP), are also found to relate to outcomes in the geriatric population, but in an opposite direction. Some degrees of elevated BMI, serum cholesterols, and BP are reportedly associated with lower, instead of higher, risk of death among the elderly. This phenomenon is termed "reverse epidemiology" or "risk factor paradox" (such as obesity paradox) and is also observed in a variety of chronic disease states such as end-stage renal disease requiring dialysis, chronic heart failure, rheumatoid arthritis, and AIDS. Several possible causes are hypothesized to explain this risk factor reversal: competing short-term and long-term killers, improved hemodynamic stability in the obese, adipokine protection against tumor necrosis factor-α, lipoprotein protection against endotoxins, and lipophilic toxin sequestration by the adipose tissue. It is possible that the current thresholds for intervention and goal levels for such traditional risk factors as BMI, serum cholesterol, and BP derived based on younger populations do not apply to the elderly, and that new levels for such risk factors should be developed for the elderly population. Reverse epidemiology of conventional cardiovascular risk factors may have a bearing on the management of the geriatric population, thus it deserves further attention.
Collapse
Affiliation(s)
- Seyed-Foad Ahmadi
- Division of Nephrology and Hypertension, Harold Simmons Center for Kidney Disease Research and Epidemiology, University of California Irvine Medical Center, Orange, CA; Department of Population Health and Disease Prevention, Program in Public Health, University of California Irvine, Irvine, CA
| | - Elani Streja
- Division of Nephrology and Hypertension, Harold Simmons Center for Kidney Disease Research and Epidemiology, University of California Irvine Medical Center, Orange, CA; Nephrology Section, Department of Medicine, Veterans Affairs Long Beach Healthcare System, Long Beach, CA
| | - Golara Zahmatkesh
- Division of Nephrology and Hypertension, Harold Simmons Center for Kidney Disease Research and Epidemiology, University of California Irvine Medical Center, Orange, CA
| | - Dan Streja
- Department of Medicine, Providence Medical Institute, West Hills, CA
| | - Moti Kashyap
- Nephrology Section, Department of Medicine, Veterans Affairs Long Beach Healthcare System, Long Beach, CA
| | - Hamid Moradi
- Division of Nephrology and Hypertension, Harold Simmons Center for Kidney Disease Research and Epidemiology, University of California Irvine Medical Center, Orange, CA
| | - Miklos Z Molnar
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN
| | - Uttam Reddy
- Division of Nephrology and Hypertension, Harold Simmons Center for Kidney Disease Research and Epidemiology, University of California Irvine Medical Center, Orange, CA
| | - Alpesh N Amin
- Department of Medicine, University of California Irvine Medical Center, Orange, CA
| | - Csaba P Kovesdy
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN; Memphis Veterans Affairs Medical Center, Memphis, TN
| | - Kamyar Kalantar-Zadeh
- Division of Nephrology and Hypertension, Harold Simmons Center for Kidney Disease Research and Epidemiology, University of California Irvine Medical Center, Orange, CA; Department of Population Health and Disease Prevention, Program in Public Health, University of California Irvine, Irvine, CA; Nephrology Section, Department of Medicine, Veterans Affairs Long Beach Healthcare System, Long Beach, CA; Department of Epidemiology, UCLA School of Public Health, Los Angeles, CA.
| |
Collapse
|
50
|
Dor-On E, Solomon B. Targeting glioblastoma via intranasal administration of Ff bacteriophages. Front Microbiol 2015; 6:530. [PMID: 26074908 PMCID: PMC4445050 DOI: 10.3389/fmicb.2015.00530] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 05/14/2015] [Indexed: 01/29/2023] Open
Abstract
Bacteriophages (phages) are ubiquitous viruses that control the growth and diversity of bacteria. Although they have no tropism to mammalian cells, accumulated evidence suggests that phages are not neutral to the mammalian macro-host and can promote immunomodulatory and anti-tumorigenic activities. Here we demonstrate that Ff phages that do not display any proteins or peptides could inhibit the growth of subcutaneous glioblastoma tumors in mice and that this activity is mediated in part by lipopolysaccharide molecules attached to their virion. Using the intranasal route, a non-invasive approach to deliver therapeutics directly to the CNS, we further show that phages rapidly accumulate in the brains of mice and could attenuate progression of orthotopic glioblastoma. Taken together, this study provides new insight into phages non-bacterial activities and demonstrates the feasibility of delivering Ff phages intranasally to treat brain malignancies.
Collapse
Affiliation(s)
- Eyal Dor-On
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel-Aviv University Tel-Aviv, Israel
| | - Beka Solomon
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel-Aviv University Tel-Aviv, Israel
| |
Collapse
|