1
|
Bi J, Witt E, McGovern MK, Cafi AB, Swetha Tunuguntla SN, Cotoia AT, Raygoza Garay JA, Balk KR, Boge L, Hatfield S, Courtney R, Du J, Chan CHF, Huang Y, Voltarelli VA, Smith MG, Mailloux A, Bosch DE, Tift MS, Otterbein LE, Traverso G, Byrne JD. Potentiating the effect of immunotherapy in pancreatic cancer using gas-entrapping materials. Biomaterials 2025; 317:123097. [PMID: 39799699 PMCID: PMC11788032 DOI: 10.1016/j.biomaterials.2025.123097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Immune checkpoint inhibitors (ICIs) show limited success in treating pancreatic ductal adenocarcinoma (PDAC), largely due to immune evasion mechanisms, including downregulating expression of major histocompatibility complex class I (MHC-I). Our retrospective analysis demonstrated that smoking - a state of elevated CO exposure - is correlated with increased MHC I expression in pancreatic tumors. Here we tested our hypothesis that introducing exogenous CO augments the anti-cancer effects of immunotherapy. To evaluate this influence, we created a novel oral delivery system for CO, termed CO-Gas-entrapping materials (GeMs), utilizing Food and Drug Administration Generally Recognized as Safe components. In vitro studies demonstrated that CO exposure increased MHC-I and PD-L1 gene and protein expression in various pancreatic cancer cell lines, as well as enhancing T-cell migration and recruitment. In vivo studies showed increased T-cell infiltration in PDAC allograft tumors treated with the combination therapy as confirmed by flow cytometry and immunohistochemical analysis. Further, CO-GeMs combined with ICIs significantly suppressed tumor growth in multiple PDAC mouse models, including subcutaneous and hepatic metastatic allograft models. These findings suggest that CO enhances immune recognition to potentiate the efficacy of ICIs in PDAC. Thus, our CO-GeMs offer a safe, effective method for systemic CO delivery to treat cancer, representing a promising adjunct to immunotherapy in PDAC and potentially other malignancies.
Collapse
Affiliation(s)
- Jianling Bi
- Women and Children's Hospital of Chongqing Medical University, Chongqing, 400015, China; Chongqing Health Center for Women and Children, Chongqing, 400015, China; Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA; Department of Radiation Oncology, University of Iowa, Iowa City, IA, 52242, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242, USA
| | - Emily Witt
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA; Department of Radiation Oncology, University of Iowa, Iowa City, IA, 52242, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242, USA
| | - Megan K McGovern
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242, USA; Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Arielle B Cafi
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA; Department of Radiation Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Sri Naga Swetha Tunuguntla
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA; Department of Radiation Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Alicia T Cotoia
- University of North Carolina at Wilmington, Wilmington, NC, 28403, USA
| | | | - Kyle R Balk
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA; Department of Radiation Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Lilly Boge
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA; Department of Radiation Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Samual Hatfield
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242, USA; Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Ryan Courtney
- Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Juan Du
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA; Department of Radiation Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Carlos H F Chan
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242, USA; Division of Surgical Oncology, Department of Surgery, University of Iowa, Iowa City, IA, 52242, USA
| | - Yi Huang
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242, USA
| | - Vanessa A Voltarelli
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| | - Matthew G Smith
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, 52242, USA
| | - Adam Mailloux
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, 52242, USA
| | - Dustin E Bosch
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242, USA; Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Michael S Tift
- University of North Carolina at Wilmington, Wilmington, NC, 28403, USA
| | - Leo E Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| | - Giovanni Traverso
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02139, USA
| | - James D Byrne
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA; Department of Radiation Oncology, University of Iowa, Iowa City, IA, 52242, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
2
|
Ahn GJ, Lee S, Lee SJ, Cha YS. Internal Malignancy Risk After Carbon Monoxide Poisoning: A Nationwide Population-Based Cohort Study. J Clin Med 2025; 14:937. [PMID: 39941608 PMCID: PMC11818198 DOI: 10.3390/jcm14030937] [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: 11/15/2024] [Revised: 01/22/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Background: We aimed to investigate the association between acute carbon monoxide (CO) poisoning and the risk of internal malignancies, including hematologic malignancies. Methods: The study population was derived from the National Health Insurance Service (NHIS) database of Korea between 2002 and 2022. Adults diagnosed with CO poisoning and controls were included. Demographics, socioeconomic statuses, lifestyle factors, and comorbidity profiles of participants were retrieved from the NHIS database. Covariates potentially associated with disease outcomes were selected based on the available literature and biological plausibility, balanced between the two cohorts using inverse probability of treatment weighting, and applied to adjust multivariable models. Results: Overall, 42,874 patients with CO poisoning and 905,285 controls were included; both cohorts comprised 44.3% females. The mean age of the CO poisoning and controls was 51.5 and 50.9 years, respectively. Patients with CO poisoning had a 1.02-fold increase in the overall risk of malignancy (a 1.03-fold increase in solid organ malignancies and a 0.71-fold decrease in hematologic malignancies) compared with controls. The risk of internal malignancy was increased in the oral cavity (adjusted hazard ratio, 1.33; 95% confidence intervals, 1.19-1.49), lungs (1.39; 1.33-1.46), bone (1.68; 1.23-2.30), cervix (1.32; 95% CI, 1.17-1.49), and kidneys (1.14; 1.04-1.24). Conversely, the risk of internal malignancies was decreased in the thorax (0.59; 0.45-0.77), anus (0.14; 0.06-0.34), uterus (0.71; 0.60-0.82), ovaries (0.59; 0.45-0.77), prostate (0.89; 0.84-0.95), Hodgkin lymphoma (0.35; 0.20-0.61), non-Hodgkin lymphoma (0.67; 0.59-0.75), and multiple myeloma (0.36; 0.30-0.43). Conclusions: CO poisoning was associated with the development of internal malignancies.
Collapse
Affiliation(s)
- Gyo Jin Ahn
- Department of Emergency Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
| | - Solam Lee
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
| | - Seok Jeong Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Yong Sung Cha
- Department of Emergency Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
- Research Institute of Hyperbaric Medicine and Science, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| |
Collapse
|
3
|
Xue XP, Sheng Y, Ren QQ, Xu SM, Li M, Liu ZX, Lu CH. Inhibition of ATP1V6G3 prompts hepatic stellate cell senescence with reducing ECM by activating Notch1 pathway to alleviate hepatic fibrosis. Tissue Cell 2024; 91:102554. [PMID: 39316936 DOI: 10.1016/j.tice.2024.102554] [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: 06/22/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024]
Abstract
Liver fibrosis is characterized by an excessive reparative response to various etiological factors, with the activated hepatic stellate cells (aHSCs) leading to extracellular matrix (ECM) accumulation. Senescence is a stable growth arrest, and the senescence of aHSCs is associated with the degradation of ECM and the regression of hepatic fibrosis, making it a promising approach for managing hepatic fibrosis. The role and specific mechanisms by which V-Type Proton ATPase Subunit G 3 (ATP6V1G3) influences senescence in activated HSCs during liver fibrosis remain unclear. Our preliminary results reveal upregulation of ATP6V1G3 in both human fibrotic livers and murine liver fibrosis models. Additionally, ATP6V1G3 inhibition induced senescence in aHSCs in vitro. Moreover, suppressing Notch1 reversed the senescence caused by ATP6V1G3 inhibition in HSCs. Thus, targeting ATP6V1G3, which appears to drive HSCs senescence through the Notch1 pathway, emerges as a potential therapeutic strategy for hepatic fibrosis.
Collapse
Affiliation(s)
- Xiao-Pei Xue
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Department Gastroenterology, Rugao Hospital of traditional Chinese Medicine, Nantong 226500, China
| | - Yu Sheng
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Qi-Qi Ren
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Shi-Meng Xu
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Min Li
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Zhao-Xiu Liu
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Cui-Hua Lu
- Department Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
4
|
Sun M, Wang T, Zhu Y, Ling F, Bai J, Tang C. Gas immnuo-nanomedicines fight cancers. Biomed Pharmacother 2024; 180:117595. [PMID: 39476762 DOI: 10.1016/j.biopha.2024.117595] [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: 07/23/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 11/14/2024] Open
Abstract
Certain gas molecules, including hydrogen (H2), nitric oxide (NO), carbon monoxide (CO), hydrogen sulfide (H2S), oxygen (O2) and sulfur dioxide (SO2) exhibit significant biological functionalities that can modulate the immune response. Strategies pertaining to gas-based immune therapy have garnered considerable attention in recent years. Nevertheless, delivering various gas molecules precisely into tumors, which leads to enhanced anti-tumor immunotherapeutic effect, is still a main challenge. The advent of gas treatment modality with desirable immunotherapeutic efficiency has been made possible by the rapid development of nanotechnology, which even derives the concept of the gas immnuo-nanomedicines (GINMs). In light of the fact, we herein aim to furnish a cutting-edge review on the latest progress of GINMs. The underlying mechanisms of action for several gases utilized in cancer immunotherapy are initially outlined. Additionally, it provides a succinct overview of the current clinical landscape of gas therapy, and introduces GINMs specifically designed for cancer treatment based on immunotherapeutic principles across multiple strategies. Last but not least, we address the challenges and opportunities associated with GINMs, exploring the potential future developments and clinical applications of this innovative approach.
Collapse
Affiliation(s)
- Mengchi Sun
- Huzhou Key Laboratory of Translational Medicine, Department of Hepatopancreatobiliary Surgery, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; College of Art and Science, Northeast Agricultural University, Harbin, Heilongjiang, China.
| | - Tianye Wang
- Department of General Surgery, The First Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yinmei Zhu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Feng Ling
- Huzhou Key Laboratory of Translational Medicine, Department of Hepatopancreatobiliary Surgery, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Jingwen Bai
- College of Art and Science, Northeast Agricultural University, Harbin, Heilongjiang, China.
| | - Chengwu Tang
- Huzhou Key Laboratory of Translational Medicine, Department of Hepatopancreatobiliary Surgery, First affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China.
| |
Collapse
|
5
|
Chen M, Xu T, Song L, Sun T, Xu Z, Zhao Y, Du P, Xiong L, Yang Z, Jing J, Shi H. Nanotechnology based gas delivery system: a "green" strategy for cancer diagnosis and treatment. Theranostics 2024; 14:5461-5491. [PMID: 39310098 PMCID: PMC11413789 DOI: 10.7150/thno.98884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/17/2024] [Indexed: 09/25/2024] Open
Abstract
Gas therapy, a burgeoning clinical treatment modality, has garnered widespread attention to treat a variety of pathologies in recent years. The advent of nanoscale gas drug therapy represents a novel therapeutic strategy, particularly demonstrating immense potential in the realm of oncology. This comprehensive review navigates the landscape of gases endowed with anti-cancer properties, including hydrogen (H2), carbon monoxide (CO), carbon dioxide (CO2), nitric oxide (NO), oxygen (O2), sulfur dioxide (SO2), hydrogen sulfide (H2S), ozone (O3), and heavier gases. The selection of optimal delivery vectors is also scrutinized in this review to ensure the efficacy of gaseous agents. The paper highlights the importance of engineering stimulus-responsive delivery systems that enable precise and targeted gas release, thereby augmenting the therapeutic efficiency of gas therapy. Additionally, the review examines the synergistic potential of integrating gas therapy with conventional treatments such as starvation therapy, ultrasound (US) therapy, chemotherapy, radiotherapy (RT), and photodynamic therapy (PDT). It also discusses the burgeoning role of advanced multimodal and US imaging in enhancing the precision of gas therapy applications. The insights presented are pivotal in the strategic development of nanomedicine platforms designed for the site-specific delivery of therapeutic gases, heralding a new era in cancer therapeutics.
Collapse
Affiliation(s)
- Meixu Chen
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China, 610041
| | - Tianyue Xu
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China, 610041
| | - Linlin Song
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China, 610041
- Department of Ultrasound & Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041
| | - Ting Sun
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China, 610041
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041
| | - Zihan Xu
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China, 610041
| | - Yujie Zhao
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China, 610041
| | - Peixin Du
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China, 610041
| | - Ling Xiong
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China, 610041
| | - Zhankun Yang
- College of Chemical Engineering, Shijiazhuang University, Shijiazhuang, Hebei, China, 050035
| | - Jing Jing
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China, 610041
| | - Hubing Shi
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China, 610041
| |
Collapse
|
6
|
Zaid A, Ariel A. Harnessing anti-inflammatory pathways and macrophage nano delivery to treat inflammatory and fibrotic disorders. Adv Drug Deliv Rev 2024; 207:115204. [PMID: 38342241 DOI: 10.1016/j.addr.2024.115204] [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: 07/30/2023] [Revised: 12/08/2023] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
Targeting specific organs and cell types using nanotechnology and sophisticated delivery methods has been at the forefront of applicative biomedical sciences lately. Macrophages are an appealing target for immunomodulation by nanodelivery as they are heavily involved in various aspects of many diseases and are highly plastic in their nature. Their continuum of functional "polarization" states has been a research focus for many years yielding a profound understanding of various aspects of these cells. The ability of monocyte-derived macrophages to metamorphose from pro-inflammatory to reparative and consequently to pro-resolving effectors has raised significant interest in its therapeutic potential. Here, we briefly survey macrophages' ontogeny and various polarization phenotypes, highlighting their function in the inflammation-resolution shift. We review their inducing mediators, signaling pathways, and biological programs with emphasis on the nucleic acid sensing-IFN-I axis. We also portray the polarization spectrum of macrophages and the characteristics of their transition between different subtypes. Finally, we highlighted different current drug delivery methods for targeting macrophages with emphasis on nanotargeting that might lead to breakthroughs in the treatment of wound healing, bone regeneration, autoimmune, and fibrotic diseases.
Collapse
Affiliation(s)
- Ahmad Zaid
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel
| | - Amiram Ariel
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel.
| |
Collapse
|
7
|
Afsar A, Zhang L. Putative Molecular Mechanisms Underpinning the Inverse Roles of Mitochondrial Respiration and Heme Function in Lung Cancer and Alzheimer's Disease. BIOLOGY 2024; 13:185. [PMID: 38534454 DOI: 10.3390/biology13030185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
Mitochondria are the powerhouse of the cell. Mitochondria serve as the major source of oxidative stress. Impaired mitochondria produce less adenosine triphosphate (ATP) but generate more reactive oxygen species (ROS), which could be a major factor in the oxidative imbalance observed in Alzheimer's disease (AD). Well-balanced mitochondrial respiration is important for the proper functioning of cells and human health. Indeed, recent research has shown that elevated mitochondrial respiration underlies the development and therapy resistance of many types of cancer, whereas diminished mitochondrial respiration is linked to the pathogenesis of AD. Mitochondria govern several activities that are known to be changed in lung cancer, the largest cause of cancer-related mortality worldwide. Because of the significant dependence of lung cancer cells on mitochondrial respiration, numerous studies demonstrated that blocking mitochondrial activity is a potent strategy to treat lung cancer. Heme is a central factor in mitochondrial respiration/oxidative phosphorylation (OXPHOS), and its association with cancer is the subject of increased research in recent years. In neural cells, heme is a key component in mitochondrial respiration and the production of ATP. Here, we review the role of impaired heme metabolism in the etiology of AD. We discuss the numerous mitochondrial effects that may contribute to AD and cancer. In addition to emphasizing the significance of heme in the development of both AD and cancer, this review also identifies some possible biological connections between the development of the two diseases. This review explores shared biological mechanisms (Pin1, Wnt, and p53 signaling) in cancer and AD. In cancer, these mechanisms drive cell proliferation and tumorigenic functions, while in AD, they lead to cell death. Understanding these mechanisms may help advance treatments for both conditions. This review discusses precise information regarding common risk factors, such as aging, obesity, diabetes, and tobacco usage.
Collapse
Affiliation(s)
- Atefeh Afsar
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Li Zhang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
8
|
Jeong EJ, Kim C, Lee YC, Rhim T, Lee SK, Lee KY. Tumor-specific cytolysis by peptide-conjugated echogenic polymer micelles. Biomed Pharmacother 2024; 172:116272. [PMID: 38354570 DOI: 10.1016/j.biopha.2024.116272] [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: 11/24/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/16/2024] Open
Abstract
Interest in multifunctional polymer nanoparticles for targeted delivery of anti-cancer drugs has grown significantly in recent years. In this study, tumor-targeting echogenic polymer micelles were prepared from poly(ethylene glycol) methyl ether-alkyl carbonate (mPEG-AC) derivatives, and their potential in cancer therapy was assessed. Various mPEG derivatives with carbonate linkages were synthesized via an alkyl halide reaction between mPEG and alkyl chloroformate. Micelle formation using polymer amphiphiles in aqueous media and the subsequent carbon dioxide (CO2) gas generation from the micelles was confirmed. Their ability to target neuroblastoma was substantially enhanced by incorporating the rabies virus glycoprotein (RVG) peptide. RVG-modified gas-generating micelles significantly inhibited tumor growth in a tumor-bearing mouse model owing to CO2 gas generation within tumor cells and resultant cytolytic effects, showing minimal side effects. The development of multifunctional polymer micelles may offer a promising therapeutic approach for various diseases, including cancer.
Collapse
Affiliation(s)
- Eun Ju Jeong
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea
| | - Choonggu Kim
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea
| | - Yun-Chan Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea
| | - Taiyoun Rhim
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea; Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul 04763, the Republic of Korea.
| | - Sang-Kyung Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea; Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul 04763, the Republic of Korea.
| | - Kuen Yong Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, the Republic of Korea; Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul 04763, the Republic of Korea.
| |
Collapse
|
9
|
Li Z, Wang Y, Liu M, Pan Y, Ni Z, Min Q, Wang B, Ke H, Ji X. Reactive Oxygen Species-Activated Metal-Free Carbon Monoxide Prodrugs for Targeted Cancer Treatment. J Med Chem 2023; 66:14583-14596. [PMID: 37909153 DOI: 10.1021/acs.jmedchem.3c01056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Carbon monoxide has shown promise as a therapeutic agent against cancers. Reactive oxygen species (ROS)-activated CO prodrugs are highly demanded for targeted cancer treatment but remain sporadic. In addition, little attention is on how the release rate affects CO's biological effects. Herein, we describe a new type of ROS-activated metal-free CO prodrug, which releases CO with tunable release rates in response to multiple ROS and exhibits very pronounced tumor suppression effects in a mouse 4t1 breast tumor model. Importantly, for the first time, we observe both in vitro and in vivo that CO release rate has a direct impact on its antiproliferative potency and a correlation between release rate and antiproliferative activity is observed. In aggregates, our results not only deliver ROS-sensitive CO prodrugs for cancer treatment but also represent a promising starting point for further in-depth studies of how CO release kinetics affect anticancer activity.
Collapse
Affiliation(s)
- Zhang Li
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Yongming Wang
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Miao Liu
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Yiyao Pan
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Zihui Ni
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Qingqiang Min
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - Hengte Ke
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| | - Xingyue Ji
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215021, China
| |
Collapse
|
10
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
11
|
Oza PP, Kashfi K. The Triple Crown: NO, CO, and H 2S in cancer cell biology. Pharmacol Ther 2023; 249:108502. [PMID: 37517510 PMCID: PMC10529678 DOI: 10.1016/j.pharmthera.2023.108502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023]
Abstract
Nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S) are three endogenously produced gases with important functions in the vasculature, immune defense, and inflammation. It is increasingly apparent that, far from working in isolation, these three exert many effects by modulating each other's activity. Each gas is produced by three enzymes, which have some tissue specificities and can also be non-enzymatically produced by redox reactions of various substrates. Both NO and CO share similar properties, such as activating soluble guanylate cyclase (sGC) to increase cyclic guanosine monophosphate (cGMP) levels. At the same time, H2S both inhibits phosphodiesterase 5A (PDE5A), an enzyme that metabolizes sGC and exerts redox regulation on sGC. The role of NO, CO, and H2S in the setting of cancer has been quite perplexing, as there is evidence for both tumor-promoting and pro-inflammatory effects and anti-tumor and anti-inflammatory activities. Each gasotransmitter has been found to have dual effects on different aspects of cancer biology, including cancer cell proliferation and apoptosis, invasion and metastasis, angiogenesis, and immunomodulation. These seemingly contradictory actions may relate to each gas having a dual effect dependent on its local flux. In this review, we discuss the major roles of NO, CO, and H2S in the context of cancer, with an effort to highlight the dual nature of each gas in different events occurring during cancer progression.
Collapse
Affiliation(s)
- Palak P Oza
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA; Graduate Program in Biology, City University of New York Graduate Center, New York 10091, USA.
| |
Collapse
|
12
|
Chai J, Zhu J, Tian Y, Yang K, Luan J, Wang Y. Carbon monoxide therapy: a promising strategy for cancer. J Mater Chem B 2023; 11:1849-1865. [PMID: 36786000 DOI: 10.1039/d2tb02599j] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cancer is one of the acute life-threatening diseases endangering the whole of humanity. The treatment modalities for cancer are various. However, in most cases, a single treatment choice provides multiple side effects, poor targeting, and ineffective treatment. In recent years, the physiological regulatory function of carbon monoxide (CO) in the cancer process has been reported gradually, and CO-related nano-drugs have been explored. It shows better application prospects in cancer treatment and provides new ideas for treatment. The present review introduces the pathophysiological role of CO. The recent advances in cancer therapy, such as CO-mediated gas therapy, combined application of CO chemotherapy, photodynamic therapy (PDT), photothermal therapy (PTT), and immunotherapy, are described. Current challenges and future developments in CO-based treatment are also discussed. This review provides comprehensive information on recent advances in CO therapy and also some valuable guidance for promoting the progress of gas therapy nanomedicine.
Collapse
Affiliation(s)
- Jingjing Chai
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Junfei Zhu
- China-Japan Friendship Hospital, No. 2 Sakura East Street, Chaoyang District, Beijing, China
| | - Yu Tian
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Kui Yang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Jiajie Luan
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| |
Collapse
|
13
|
Zhang X, Wang X, Hou L, Xu Z, Liu Y, Wang X. Nanoparticles overcome adaptive immune resistance and enhance immunotherapy via targeting tumor microenvironment in lung cancer. Front Pharmacol 2023; 14:1130937. [PMID: 37033636 PMCID: PMC10080031 DOI: 10.3389/fphar.2023.1130937] [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: 12/23/2022] [Accepted: 03/16/2023] [Indexed: 04/11/2023] Open
Abstract
Lung cancer is one of the common malignant cancers worldwide. Immune checkpoint inhibitor (ICI) therapy has improved survival of lung cancer patients. However, ICI therapy leads to adaptive immune resistance and displays resistance to PD-1/PD-L1 blockade in lung cancer, leading to less immune response of lung cancer patients. Tumor microenvironment (TME) is an integral tumor microenvironment, which is involved in immunotherapy resistance. Nanomedicine has been used to enhance the immunotherapy in lung cancer. In this review article, we described the association between TME and immunotherapy in lung cancer. We also highlighted the importance of TME in immunotherapy in lung cancer. Moreover, we discussed how nanoparticles are involved in regulation of TME to improve the efficacy of immunotherapy, including Nanomedicine SGT-53, AZD1080, Nanomodulator NRF2, Cisplatin nanoparticles, Au@PG, DPAICP@ME, SPIO NP@M-P, NBTXR3 nanoparticles, ARAC nanoparticles, Nano-DOX, MS NPs, Nab-paclitaxel, GNPs-hPD-L1 siRNA. Furthermore, we concluded that targeting TME by nanoparticles could be helpful to overcome resistance to PD-1/PD-L1 blockade in lung cancer.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xuemei Wang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Lijian Hou
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Zheng Xu
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yu’e Liu
- School of Medicine, Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, Tongji University, Shanghai, China
| | - Xueju Wang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
- *Correspondence: Xueju Wang,
| |
Collapse
|
14
|
Canesin G, Feldbrügge L, Wei G, Janovicova L, Janikova M, Csizmadia E, Ariffin J, Hedblom A, Herbert ZT, Robson SC, Celec P, Swanson KD, Nasser I, Popov YV, Wegiel B. Heme oxygenase-1 mitigates liver injury and fibrosis via modulation of LNX1/Notch1 pathway in myeloid cells. iScience 2022; 25:104983. [PMID: 36093061 PMCID: PMC9450142 DOI: 10.1016/j.isci.2022.104983] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/01/2022] [Accepted: 08/16/2022] [Indexed: 01/12/2023] Open
Abstract
Activation of resident macrophages (Mϕ) and hepatic stellate cells is a key event in chronic liver injury. Mice with heme oxygenase-1 (HO-1; Hmox1)-deficient Mϕ (LysM-Cre:Hmox1 flfl ) exhibit increased inflammation, periportal ductular reaction, and liver fibrosis following bile duct ligation (BDL)-induced liver injury and increased pericellular fibrosis in NASH model. RiboTag-based RNA-sequencing profiling of hepatic HO-1-deficient Mϕ revealed dysregulation of multiple genes involved in lipid and amino acid metabolism, regulation of oxidative stress, and extracellular matrix turnover. Among these genes, ligand of numb-protein X1 (LNX1) expression is strongly suppressed in HO-1-deficient Mϕ. Importantly, HO-1 and LNX1 were expressed by hepatic Mϕ in human biliary and nonbiliary end-stage cirrhosis. We found that Notch1 expression, a downstream target of LNX1, was increased in LysM-Cre:Hmox1 flfl mice. In HO-1-deficient Mϕ treated with heme, transient overexpression of LNX1 drives M2-like Mϕ polarization. In summary, we identified LNX1/Notch1 pathway as a downstream target of HO-1 in liver fibrosis.
Collapse
Affiliation(s)
- Giacomo Canesin
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Linda Feldbrügge
- Charité – Universitätsmedizin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, 13353 Berlin, Germany,Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Guangyan Wei
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, 510080 Guangzhou, China
| | - Lubica Janovicova
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Institute of Molecular Biomedicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia
| | - Monika Janikova
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Institute of Molecular Biomedicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia
| | - Eva Csizmadia
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Juliana Ariffin
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Andreas Hedblom
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Zachary T. Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Simon C. Robson
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Peter Celec
- Institute of Molecular Biomedicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia
| | - Kenneth D. Swanson
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Imad Nasser
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yury V. Popov
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Corresponding author
| | - Barbara Wegiel
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Corresponding author
| |
Collapse
|
15
|
Chen J, Chen D, Chen J, Shen T, Jin T, Zeng B, Li L, Yang C, Mu Z, Deng H, Cai X. An all-in-one CO gas therapy-based hydrogel dressing with sustained insulin release, anti-oxidative stress, antibacterial, and anti-inflammatory capabilities for infected diabetic wounds. Acta Biomater 2022; 146:49-65. [PMID: 35500813 DOI: 10.1016/j.actbio.2022.04.043] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 12/16/2022]
Abstract
To effectively treat diabetic wounds, the development of versatile medical dressings that can long-term regulate blood glucose and highly effective anti-oxidative stress, antibacterial and anti-inflammatory are critical. Here, an all-in-one CO gas-therapy-based versatile hydrogel dressing (ICOQF) was developed via the dynamic Schiff base reaction between the amino groups on quaternized chitosan (QCS) and the aldehyde groups on benzaldehyde-terminated F108 (F108-CHO) micelles. CORM-401 (an oxidant-sensitive CO-releasing molecules) was encapsulated in the hydrophobic core of F108-CHO micelles and insulin was loaded in the three-dimensional network structure of ICOQF. The dynamic Schiff base bonds not only endowed ICOQF with good tissue adhesion, injectability and self-healing, but also gave it sustained and controllable insulin release ability. In addition, ICOQF could quickly generate CO in inflamed wound tissue by consuming reactive oxygen species. The generated CO could effectively anti-oxidative stress by activating the expression of heme oxygenase; antibacterial by inducing the rupture of bacterial cell membranes and mitochondrial dysfunction and inhibiting the synthesis of adenosine triphosphate; and anti-inflammatory by inhibiting the proliferation of activated macrophages and promoting the polarization of the M1 phenotype to the M2 phenotype. Due to these outstanding properties, ICOQF significantly promoted the healing of STZ-induced MRSA-infected diabetic wounds accompanied by good biocompatibility. This study clearly shows that ICOQF is a versatile hydrogel dressing with great application potential for the management of diabetic wounds. STATEMENT OF SIGNIFICANCE: The development of some versatile hydrogel dressings that can not only provide a prolonged and controlled insulin release property but also utilize a non-antibiotic treatment modality for highly effective antibacterial, anti-inflammatory, and anti-oxidative stress effects is vital for the successful treatment of diabetic wounds. Herein, we developed an all-in-one CO gas-therapy-based versatile hydrogel dressing (ICOQF) with sustained and controllable insulin release abilities. Moreover, ICOQF could not only quickly release CO in the inflamed wound tissue by consumption of reactive oxygen species but also utilize the generated CO to highly effectively anti-oxidative stress, antibacterial, and anti-inflammatory. ICOQF therapy substantially promoted the healing of STZ-induced MRSA-infected diabetic wounds. Overall, this work provides a multifunctional hydrogel dressing for the management of diabetic wounds.
Collapse
|
16
|
Salihi A, Al-Naqshabandi MA, Khudhur ZO, Housein Z, Hama HA, Abdullah RM, Hussen BM, Alkasalias T. Gasotransmitters in the tumor microenvironment: Impacts on cancer chemotherapy (Review). Mol Med Rep 2022; 26:233. [PMID: 35616143 PMCID: PMC9178674 DOI: 10.3892/mmr.2022.12749] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/07/2022] [Indexed: 11/23/2022] Open
Abstract
Nitric oxide, carbon monoxide and hydrogen sulfide are three endogenous gasotransmitters that serve a role in regulating normal and pathological cellular activities. They can stimulate or inhibit cancer cell proliferation and invasion, as well as interfere with cancer cell responses to drug treatments. Understanding the molecular pathways governing the interactions between these gases and the tumor microenvironment can be utilized for the identification of a novel technique to disrupt cancer cell interactions and may contribute to the conception of effective and safe cancer therapy strategies. The present review discusses the effects of these gases in modulating the action of chemotherapies, as well as prospective pharmacological and therapeutic interfering approaches. A deeper knowledge of the mechanisms that underpin the cellular and pharmacological effects, as well as interactions, of each of the three gases could pave the way for therapeutic treatments and translational research.
Collapse
Affiliation(s)
- Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region 44001, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region 44002, Iraq
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, SE-17165 Stockholm, Sweden
| | - Mohammed A. Al-Naqshabandi
- Department of Clinical Biochemistry, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Zhikal Omar Khudhur
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Kurdistan Region 44001, Iraq
| | - Zjwan Housein
- Department of Medical Laboratory Technology, Technical Health and Medical College, Erbil Polytechnique University, Erbil, Kurdistan Region 44002, Iraq
| | - Harmand A. Hama
- Department of Biology, Faculty of Education, Tishk International University, Erbil, Kurdistan Region 44002, Iraq
| | - Ramyar M. Abdullah
- College of Medicine, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Twana Alkasalias
- General Directorate of Scientific Research Center, Salahaddin University-Erbil, Erbil, Kurdistan Region 44002, Iraq
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| |
Collapse
|
17
|
Krukowska K, Magierowski M. Carbon monoxide (CO)/heme oxygenase (HO)-1 in gastrointestinal tumors pathophysiology and pharmacology - possible anti- and pro-cancer activities. Biochem Pharmacol 2022; 201:115058. [PMID: 35490732 DOI: 10.1016/j.bcp.2022.115058] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 11/02/2022]
Abstract
Gastrointestinal (GI) tract cancers pose a significant pharmacological challenge for researchers in terms of the discovery of molecular agents and the development of targeted therapies. Although many ongoing clinical trials have brought new perspectives, there is still a lack of successful long-term treatment. Several novel pharmacological and molecular agents are being studied in the prevention and treatment of GI cancers. On the other hand, pharmacological tools designed to release an endogenous gaseous mediator, carbon monoxide (CO), were shown to prevent the gastric mucosa against various types of injuries and exert therapeutic properties in the treatment of GI pathologies. In this review, we summarized the current evidence on the role of CO and heme oxygenase 1 (HO-1) as a CO producing enzyme in the pathophysiology of GI tumors. We focused on a beneficial role of HO-1 and CO in biological systems and common pathological conditions. We further discussed the complex and ambiguous function of the HO-1/CO pathway in cancer cells with a special emphasis on molecular and cellular pro-cancerous and anti-cancer mechanisms. We also focused on the role that HO-1/CO plays in GI cancers, especially within upper parts such as esophagus or stomach.
Collapse
Affiliation(s)
- Kinga Krukowska
- Cellular Engineering and Isotope Diagnostics Laboratory, Department of Physiology, Jagiellonian University Medical College, Poland
| | - Marcin Magierowski
- Cellular Engineering and Isotope Diagnostics Laboratory, Department of Physiology, Jagiellonian University Medical College, Poland.
| |
Collapse
|
18
|
Cui Q, Liang XL, Wang JQ, Zhang JY, Chen ZS. Therapeutic implication of carbon monoxide in drug resistant cancers. Biochem Pharmacol 2022; 201:115061. [PMID: 35489394 DOI: 10.1016/j.bcp.2022.115061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/14/2022]
Abstract
Drug resistance is the major obstacle that undermines effective cancer treatment. Recently, the application of gas signaling molecules, e.g., carbon monoxide (CO), in overcoming drug resistance has gained significant attention. Growing evidence showed that CO could inhibit mitochondria respiratory effect and glycolysis, two major ATP production pathways in cancer cells, and suppress angiogenesis and inhibit the activity of cystathionine β-synthase that is important in regulating cancer cells homeostasis, leading to synergistic effects when combined with cisplatin, doxorubicin, or phototherapy, etc. in certain resistant cancer cells. In the current review, we attempted to have a summary of these research conducted in the past decade using CO in treating drug resistant cancers, and have a detailed interpretation of the underlying mechanisms. The critical challenges will be discussed and potential solutions will also be provided. The information collected in this work will hopefully evoke more effects in using CO for the treatment of drug resistant cancers.
Collapse
Affiliation(s)
- Qingbin Cui
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiao-Lan Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Jian-Ye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; Institute for Biotechnology, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|
19
|
Moayedi-Nia S, Pasquet R, Siemiatycki J, Koushik A, Ho V. Occupational Exposures and Lung Cancer Risk-An Analysis of the CARTaGENE Study. J Occup Environ Med 2022; 64:295-304. [PMID: 35019894 DOI: 10.1097/jom.0000000000002481] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To determine the associations between prevalent occupational agents and lung cancer risk. METHODS A case-cohort design (ncases= 147; nsub-cohort= 1,032) was nested within the CARTaGENE prospective cohort study. The Canadian Job Exposure Matrix was used to determine the probability of exposure to 27 agents in participants' longest-held jobs. Multivariable logistic regression with robust variance estimators was used to determine the associations between each agent and lung cancer risk while adjusting for established lung cancer risk factors. RESULTS Increased lung cancer risk was observed among those exposed to ashes, calcium sulfate, formaldehyde, cooking fumes, alkanes, aliphatic aldehydes, and cleaning agents. Lower lung cancer risk was found among participants exposed to carbon monoxide and polycyclic aromatic hydrocarbons from petroleum. CONCLUSION Our findings support the role of several occupational agents, for which we have limited knowledge, in contributing to lung cancer risk.
Collapse
Affiliation(s)
- Saeedeh Moayedi-Nia
- Department of Social and Preventive Medicine, University of Montreal, Montreal, Quebec, Canada (Ms Moayedi-Nia, Dr Siemiatycki, Dr Koushik, Dr Ho); and Health Innovation and Evaluation Hub, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada (Dr Pasquet, Dr Siemiatycki, Dr Koushik, Dr Ho)
| | | | | | | | | |
Collapse
|
20
|
Ren X, Wang Y, Jia L, Guo X, He X, Zhao Z, Gao D, Yang Z. Intelligent Nanomedicine Approaches Using Medical Gas-Mediated Multi-Therapeutic Modalities Against Cancer. J Biomed Nanotechnol 2022; 18:24-49. [PMID: 35180898 DOI: 10.1166/jbn.2022.3224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The emerging area of gas-mediated cancer treatment has received widespread attention in the medical community. Featuring unique physical, chemical, and biological properties, nanomaterials can facilitate the delivery and controllable release of medicinal gases at tumor sites, and also serve as ideal platforms for the integration of other therapeutic modalities with gas therapy to augment cancer therapeutic efficacy. This review presents an overview of anti-cancer mechanisms of several therapeutic gases: nitric oxide (NO), hydrogen sulfide (H₂S), carbon monoxide (CO), oxygen (O₂), and hydrogen (H₂). Controlled release behaviors of gases under different endogenous and exogenous stimuli are also briefly discussed, followed by their synergistic effects with different therapeutic modes. Moreover, the potential challenges and future prospects regarding gas therapy based on nanomaterials are also described, aiming to facilitate the advancement of gas therapeutic nanomedicine in new frontiers for highly efficient cancer treatment.
Collapse
Affiliation(s)
- Xuechun Ren
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ying Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Liangliang Jia
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiaoqing Guo
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinyu He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhipeng Zhao
- School of Physical Education, Xizang Minzu University, Xianyang, 712000, Shaanxi, China
| | - Di Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
21
|
Barrett JA, Li Z, Garcia JV, Wein E, Zheng D, Hunt C, Ngo L, Sepunaru L, Iretskii AV, Ford PC. Redox-mediated carbon monoxide release from a manganese carbonyl-implications for physiological CO delivery by CO releasing moieties. ROYAL SOCIETY OPEN SCIENCE 2021; 8:211022. [PMID: 34804570 PMCID: PMC8580448 DOI: 10.1098/rsos.211022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/07/2021] [Indexed: 05/05/2023]
Abstract
The dynamics of hydrogen peroxide reactions with metal carbonyls have received little attention. Given reports that therapeutic levels of carbon monoxide are released in hypoxic tumour cells upon manganese carbonyls reactions with endogenous H2O2, it is critical to assess the underlying CO release mechanism(s). In this context, a quantitative mechanistic investigation of the H2O2 oxidation of the water-soluble model complex fac-[Mn(CO)3(Br)(bpCO2)]2-, (A, bpCO2 2- = 2,2'-bipyridine-4,4'-dicarboxylate dianion) was undertaken under physiologically relevant conditions. Characterizing such pathways is essential to evaluating the viability of redox-mediated CO release as an anti-cancer strategy. The present experimental studies demonstrate that approximately 2.5 equivalents of CO are released upon H2O2 oxidation of A via pH-dependent kinetics that are first-order both in [A] and in [H2O2]. Density functional calculations were used to evaluate the key intermediates in the proposed reaction mechanisms. These pathways are discussed in terms of their relevance to physiological CO delivery by carbon monoxide releasing moieties.
Collapse
Affiliation(s)
- Jacob A. Barrett
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Zhi Li
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - John V. Garcia
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Emily Wein
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Dongyun Zheng
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Camden Hunt
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Loc Ngo
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Lior Sepunaru
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Alexei V. Iretskii
- Department of Chemistry and Environmental Sciences, Lake Superior State University, Sault Sainte Marie, MI 49783, USA
| | - Peter C. Ford
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
22
|
Jing YZ, Li SJ, Sun ZJ. Gas and gas-generating nanoplatforms in cancer therapy. J Mater Chem B 2021; 9:8541-8557. [PMID: 34608920 DOI: 10.1039/d1tb01661j] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gas therapy is the usage of certain gases with special therapeutic effects for the treatment of diseases. Hydrogen (H2), nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S) acting as gas signalling molecules are representative gases in cancer therapy. They act directly on mitochondria or nuclei to lead to cell apoptosis. They can also alleviate immuno-suppression in the tumour microenvironment and promote phenotype conversion of tumour-associated macrophages. Moreover, the combination of gas therapy and other traditional therapy methods can reduce side effects and improve therapeutic efficacy. Here, we discuss the roles of NO, CO, H2S and H2 in cancer biology. Considering the rapidly developing nanotechnology, gas-generating nanoplatforms which can achieve targeted delivery and controlled release were also discussed. Finally, we highlight the current challenges and future opportunities of gas-based cancer therapy.
Collapse
Affiliation(s)
- Yuan-Zhe Jing
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P. R. China.
| | - Shu-Jin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P. R. China.
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P. R. China. .,Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P. R. China
| |
Collapse
|
23
|
Luu Hoang KN, Anstee JE, Arnold JN. The Diverse Roles of Heme Oxygenase-1 in Tumor Progression. Front Immunol 2021; 12:658315. [PMID: 33868304 PMCID: PMC8044534 DOI: 10.3389/fimmu.2021.658315] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/15/2021] [Indexed: 12/16/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). HO-1 is expressed across a range of cancers and has been demonstrated to promote tumor progression through a variety of mechanisms. HO-1 can be expressed in a variety of cells within the tumor microenvironment (TME), including both the malignant tumor cells as well as stromal cell populations such as macrophages, dendritic cells and regulatory T-cells. Intrinsically to the cell, HO-1 activity provides antioxidant, anti-apoptotic and cytoprotective effects via its catabolites as well as clearing toxic intracellular heme. However, the catabolites of heme degradation can also diffuse outside of the cell to extrinsically modulate the wider TME, influencing cellular functionality and biological processes which promote tumor progression, such as facilitating angiogenesis and metastasis, as well as promoting anti-inflammation and immune suppression. Pharmacological inhibition of HO-1 has been demonstrated to be a promising therapeutic approach to promote anti-tumor immune responses and inhibit metastasis. However, these biological functions might be context, TME and cell type-dependent as there is also conflicting reports for HO-1 activity facilitating anti-tumoral processes. This review will consider our current understanding of the role of HO-1 in cancer progression and as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Kim Ngan Luu Hoang
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Joanne E Anstee
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - James N Arnold
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
24
|
Chen RJ, Lee YH, Chen TH, Chen YY, Yeh YL, Chang CP, Huang CC, Guo HR, Wang YJ. Carbon monoxide-triggered health effects: the important role of the inflammasome and its possible crosstalk with autophagy and exosomes. Arch Toxicol 2021; 95:1141-1159. [PMID: 33554280 DOI: 10.1007/s00204-021-02976-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/04/2021] [Indexed: 12/18/2022]
Abstract
Carbon monoxide (CO) has long been known as a "silent killer" because of its ability to bind hemoglobin (Hb), leading to reduced oxygen carrying capacity of Hb, which is the main cause of CO poisoning (COP) in humans. Emerging studies suggest that mitochondria is a key target of CO action that can impact key biological processes, including apoptosis, cellular proliferation, inflammation, and autophagy. Despite its toxicity at high concentrations, CO also exhibits cyto- and tissue-protective effects at low concentrations in animal models of organ injury and disease. Specifically, CO modulates the production of pro- or anti-inflammatory cytokines and mediators by regulating the NLRP3 inflammasome. Given that human diseases are strongly associated with inflammation, a deep understanding of the exact mechanism is helpful for treatment. Autophagic factors and inflammasomes interact in various situations, including inflammatory disease, and exosomes might function as the bridge between the inflammasome and autophagy activation. Thus, the interplay among autophagy, mitochondrial dysfunction, exosomes, and the inflammasome may play pivotal roles in the health effects of CO. In this review, we summarize the latest research on the beneficial and toxic effects of CO and their underlying mechanisms, focusing on the important role of the inflammasome and its possible crosstalk with autophagy and exosomes. This knowledge may lead to the development of new therapies for inflammation-related diseases and is essential for the development of new therapeutic strategies and biomarkers of COP.
Collapse
Affiliation(s)
- Rong-Jane Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Hsuan Lee
- Department of Cosmeceutics, China Medical University, Taichung, Taiwan
| | - Tzu-Hao Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan.,Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Yu-Ying Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Ya-Ling Yeh
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Chien-Cheng Huang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan.,Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan.,Department of Senior Services, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - How-Ran Guo
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan. .,Department of Occupational and Environmental Medicine, National Cheng Kung University Hospital, Tainan, Taiwan. .,Occupational Safety, Health and Medicine Research Center, National Cheng Kung University Hospital, Tainan, Taiwan.
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70428, Taiwan. .,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
25
|
Impact of carbon monoxide poisoning on the risk of breast cancer. Sci Rep 2020; 10:20450. [PMID: 33235264 PMCID: PMC7687884 DOI: 10.1038/s41598-020-77371-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 10/28/2020] [Indexed: 11/20/2022] Open
Abstract
Carbon monoxide (CO) is a toxic gas and an endogenous signaling molecule. Some studies involving cell lines have revealed the potential antibreast cancer effects of CO. Data on such effects in humans, however, are limited. Thus, we conducted a study on patients with CO poisoning (COP) to evaluate the effects of CO on the risk of breast cancer. We identified female patients who were diagnosed with COP over the period of 2002 and 2009 from the Nationwide Poisoning Database of Taiwan. For comparison, we selected females without COP from the National Health Insurance Research Database. Participants in the COP and comparison cohorts were matched on the index year, age, monthly income, and geographic region of residence at a 1:6 ratio. We followed up the two cohorts until the end of 2014 and compared their risks of developing breast cancer. We included 7053 participants with COP and 42,318 participants without COP. Participants with COP were at a lower risk of developing breast cancer than those without COP (0.7% vs. 1.0%, p < 0.001). Cox proportional hazard regression analyses revealed that COP was associated with a hazard ratio of 0.67 (95% confidence interval [95% CI] 0.50–0.90) for breast cancer after we adjusted for age, monthly income, geographic region, and comorbidities of hypertension, diabetes, and hyperlipidemia. Our result provides evidence for the potential protective effects of CO against breast cancer in humans. Further studies that directly evaluate the potential effects are warranted.
Collapse
|
26
|
Harnessing carbon monoxide-releasing platforms for cancer therapy. Biomaterials 2020; 255:120193. [DOI: 10.1016/j.biomaterials.2020.120193] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/19/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022]
|
27
|
Yang X, de Caestecker M, Otterbein LE, Wang B. Carbon monoxide: An emerging therapy for acute kidney injury. Med Res Rev 2020. [PMID: 31820474 DOI: 10.1012/med.21650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Treating acute kidney injury (AKI) represents an important unmet medical need both in terms of the seriousness of this medical problem and the number of patients. There is also a large untapped market opportunity in treating AKI. Over the years, there has been much effort in search of therapeutics with minimal success. However, over the same time period, new understanding of the underlying pathobiology and molecular mechanisms of kidney injury have undoubtedly helped the search for new therapeutics. Along this line, carbon monoxide (CO) has emerged as a promising therapeutic agent because of its demonstrated cytoprotective, and immunomodulatory effects. CO has also been shown to sensitize cancer, but not normal cells, to chemotherapy. This is particularly important in treating cisplatin-induced AKI, a common clinical problem that develops in patients receiving cisplatin therapies for a number of different solid organ malignancies. This review will examine and make the case that CO be developed into a therapeutic agent against AKI.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| | - Mark de Caestecker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leo E Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| |
Collapse
|
28
|
Zhou Y, Chen X, Cao J, Gao H. Overcoming the biological barriers in the tumor microenvironment for improving drug delivery and efficacy. J Mater Chem B 2020; 8:6765-6781. [PMID: 32315375 DOI: 10.1039/d0tb00649a] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The delivery of drugs to tumors by nanoparticles is a rapidly growing field. However, the complex tumor microenvironment (TME) barriers greatly hinder drug delivery to tumors. In this study, we first summarized the barriers in TME, including anomalous vasculature, rigid extracellular matrix, hypoxia, acidic pH, irregular enzyme level, altered metabolism pathway and immunosuppressive conditions. To overcome these barriers, many strategies have been developed, such as modulating TME, active targeting by ligand modification and biomimetic strategies, and TME-responsive drug delivery strategies to improve nanoparticle penetration, cellular uptake and drug release. Although extensive progress has been achieved, there are still many challenges, which are discussed in the last section. Overall, we carefully discuss the landscape of TME, development for improving drug delivery, and challenges that need to be further addressed.
Collapse
Affiliation(s)
- Yang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| | | | | | | |
Collapse
|
29
|
Ho YJ, Li JP, Fan CH, Liu HL, Yeh CK. Ultrasound in tumor immunotherapy: Current status and future developments. J Control Release 2020; 323:12-23. [PMID: 32302759 DOI: 10.1016/j.jconrel.2020.04.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 12/24/2022]
Abstract
Immunotherapy has considerable potential in eliminating cancers by activating the host's own immune system, while the thermal and mechanical effects of ultrasound have various applications in tumor therapy. Hyperthermia, ablation, histotripsy, and microbubble stable/inertial cavitation can alter the tumor microenvironment to enhance immunoactivation to inhibit tumor growth. Microbubble cavitation can increase vessel permeability and thereby improve the delivery of immune cells, cytokines, antigens, and antibodies to tumors. Violent microbubble cavitation can disrupt tumor cells and efficiently expose them to numerous antigens so as to promote the maturity of antigen-presenting cells and subsequent adaptive immune-cell activation. This review provides an overview and compares the mechanisms of ultrasound-induced immune modulation for peripheral and brain tumor therapy, even degenerative brain diseases therapy. The possibility of reversing tumors to an immunoactive microenvironment by utilizing the cavitation of microbubbles loaded with therapeutic gases is also proposed as another potential pathway for immunotherapy. Finally, we disuss the challenges and opportunities of ultrasound in immunotherapy for future development.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Ju-Pi Li
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan; School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, Chang-Gung University, Taoyuan 333, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan.
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan.
| |
Collapse
|
30
|
Fiorito V, Chiabrando D, Petrillo S, Bertino F, Tolosano E. The Multifaceted Role of Heme in Cancer. Front Oncol 2020; 9:1540. [PMID: 32010627 PMCID: PMC6974621 DOI: 10.3389/fonc.2019.01540] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
Heme, an iron-containing porphyrin, is of vital importance for cells due to its involvement in several biological processes, including oxygen transport, energy production and drug metabolism. Besides these vital functions, heme also bears toxic properties and, therefore, the amount of heme inside the cells must be tightly regulated. Similarly, heme intake from dietary sources is strictly controlled to meet body requirements. The multifaceted nature of heme renders it a best candidate molecule exploited/controlled by tumor cells in order to modulate their energetic metabolism, to interact with the microenvironment and to sustain proliferation and survival. The present review summarizes the literature on heme and cancer, emphasizing the importance to consider heme as a prominent player in different aspects of tumor onset and progression.
Collapse
Affiliation(s)
- Veronica Fiorito
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Deborah Chiabrando
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Sara Petrillo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Francesca Bertino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Emanuela Tolosano
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| |
Collapse
|
31
|
Yang X, de Caestecker M, Otterbein LE, Wang B. Carbon monoxide: An emerging therapy for acute kidney injury. Med Res Rev 2019; 40:1147-1177. [PMID: 31820474 DOI: 10.1002/med.21650] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/31/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022]
Abstract
Treating acute kidney injury (AKI) represents an important unmet medical need both in terms of the seriousness of this medical problem and the number of patients. There is also a large untapped market opportunity in treating AKI. Over the years, there has been much effort in search of therapeutics with minimal success. However, over the same time period, new understanding of the underlying pathobiology and molecular mechanisms of kidney injury have undoubtedly helped the search for new therapeutics. Along this line, carbon monoxide (CO) has emerged as a promising therapeutic agent because of its demonstrated cytoprotective, and immunomodulatory effects. CO has also been shown to sensitize cancer, but not normal cells, to chemotherapy. This is particularly important in treating cisplatin-induced AKI, a common clinical problem that develops in patients receiving cisplatin therapies for a number of different solid organ malignancies. This review will examine and make the case that CO be developed into a therapeutic agent against AKI.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| | - Mark de Caestecker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leo E Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia
| |
Collapse
|
32
|
Yan H, Du J, Zhu S, Nie G, Zhang H, Gu Z, Zhao Y. Emerging Delivery Strategies of Carbon Monoxide for Therapeutic Applications: from CO Gas to CO Releasing Nanomaterials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1904382. [PMID: 31663244 DOI: 10.1002/smll.201904382] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/08/2019] [Indexed: 06/10/2023]
Abstract
Carbon monoxide (CO) therapy has emerged as a hot topic under exploration in the field of gas therapy as it shows the promise of treating various diseases. Due to the gaseous property and the high affinity for human hemoglobin, the main challenges of administrating medicinal CO are the lack of target selectivity as well as the toxic profile at relatively high concentrations. Although abundant CO releasing molecules (CORMs) with the capacity to deliver CO in biological systems have been developed, several disadvantages related to CORMs, including random diffusion, poor solubility, potential toxicity, and lack of on-demand CO release in deep tissue, still confine their practical use. Recently, the advent of versatile nanomedicine has provided a promising chance for improving the properties of naked CORMs and simultaneously realizing the therapeutic applications of CO. This review presents a brief summarization of the emerging delivery strategies of CO based on nanomaterials for therapeutic application. First, an introduction covering the therapeutic roles of CO and several frequently used CORMs is provided. Then, recent advancements in the synthesis and application of versatile CO releasing nanomaterials are elaborated. Finally, the current challenges and future directions of these important delivery strategies are proposed.
Collapse
Affiliation(s)
- Haili Yan
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, P. R. China
| | - Jiangfeng Du
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, P. R. China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Guangjun Nie
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Hui Zhang
- College of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, P. R. China
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yuliang Zhao
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| |
Collapse
|
33
|
Fang J, Islam R, Islam W, Yin H, Subr V, Etrych T, Ulbrich K, Maeda H. Augmentation of EPR Effect and Efficacy of Anticancer Nanomedicine by Carbon Monoxide Generating Agents. Pharmaceutics 2019; 11:pharmaceutics11070343. [PMID: 31315251 PMCID: PMC6680399 DOI: 10.3390/pharmaceutics11070343] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 11/16/2022] Open
Abstract
One obstacle to the successful delivery of nanodrugs into solid tumors is the heterogeneity of an enhanced permeability and retention (EPR) effect as a result of occluded or embolized tumor blood vessels. Therefore, the augmentation of the EPR effect is critical for satisfactory anticancer nanomedicine. In this study, we focused on one vascular mediator involved in the EPR effect, carbon monoxide (CO), and utilized two CO generating agents, one is an extrinsic CO donor (SMA/CORM2 micelle) and another is an inducer of endogenous CO generation via heme oxygenase-1 (HO-1) induction that is carried out using pegylated hemin. Both agents generated CO selectively in solid tumors, which resulted in an enhanced EPR effect and a two- to three-folds increased tumor accumulation of nanodrugs. An increase in drug accumulation in the normal tissue did not occur with the treatment of CO generators. In vivo imaging also clearly indicated a more intensified fluorescence of macromolecular nanoprobe in solid tumors when combined with these CO generators. Consequently, the combination of CO generators with anticancer nanodrugs resulted in an increased anticancer effect in the different transplanted solid tumor models. These findings strongly warrant the potential application of these CO generators as EPR enhancers in order to enhance tumor detection and therapy using nanodrugs.
Collapse
Affiliation(s)
- Jun Fang
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan.
| | - Rayhanul Islam
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Waliul Islam
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
- Biodynamics Research Foundation, Kumamoto 862-0954, Japan
| | - Hongzhuan Yin
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
- Department of General Surgery, Sheng Jing Hospital, China Medical University, Shenyang 110011, China
| | - Vladimir Subr
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague 16206, Czech Republic
| | - Tomas Etrych
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague 16206, Czech Republic
| | - Karel Ulbrich
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague 16206, Czech Republic
| | - Hiroshi Maeda
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan.
- Biodynamics Research Foundation, Kumamoto 862-0954, Japan.
| |
Collapse
|
34
|
Li Q, Liu Q, Cheng W, Wei H, Jiang W, E F, Yu Y, Jin J, Zou C. Heme Oxygenase-1 Inhibits Tumor Metastasis Mediated by Notch1 Pathway in Murine Mammary Carcinoma. Oncol Res 2019; 27:643-651. [PMID: 30764900 PMCID: PMC7848234 DOI: 10.3727/096504018x15415906335771] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Heme oxygenase-1 (HO-1) plays an important role in the progression of several malignancies including breast cancer. However, its role in breast cancer metastasis is still ambiguous. In this study, we observed the effect of HO-1 on mouse mammary carcinoma metastasis using the in vivo tumor metastasis model. Our results revealed that overexpression of HO-1 strongly inhibits the lung metastasis of 4T1 cells. In in vitro analysis, associated indices for epithelial-mesenchymal transition (EMT), migration, and proliferation of 4T1 cells were evaluated. The results show that HO-1 inhibits EMT, migration, and proliferation of 4T1 cells. In addition, the Notch1/Slug pathway is found to mediate an antimetastasis role of HO-1 in mouse mammary carcinoma. In conclusion, since HO-1/Notch1/Slug axis plays an important role in breast cancer metastasis, induction of HO-1 could be used as a potential therapeutic strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Qiang Li
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
- †Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | - Qi Liu
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Wanpeng Cheng
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Huiyan Wei
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Wenqian Jiang
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Fang E
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Yuan Yu
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Jianfeng Jin
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Chaoxia Zou
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
- ‡Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medicine Sciences, Harbin, P.R. China
| |
Collapse
|
35
|
He ZC, Li XY, Guo YL, Ma D, Fang Q, Ren LL, Zhang ZY, Wang W, Yu ZY, Zhao P, Wang JS. Heme oxygenase-1 attenuates the inhibitory effect of bortezomib against the APRIL-NF-κB-CCL3 signaling pathways in multiple myeloma cells: Corelated with bortezomib tolerance in multiple myeloma. J Cell Biochem 2019; 120:6972-6987. [PMID: 30368867 DOI: 10.1002/jcb.27879] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/21/2018] [Indexed: 01/24/2023]
Abstract
Osteoclasts (OCs) play an essential role in bone destruction in patients with multiple myeloma (MM). Bortezomib can ameliorate bone destruction in patients with MM, but advanced MM often resists bortezomib. We studied the molecular mechanisms of bortezomib tolerance in MM. The expression of the MM-related genes in newly diagnosed patients with MM and normal donors were studied. C-C motif chemokine ligand 3 (CCL3) is a cytokine involved in the differentiation of OCs, and its expression is closely related to APRIL (a proliferation-inducing ligand). We found that bortezomib treatment inhibited APRIL and CCL3. But the heme oxygenase-1 (HO-1) activator hemin attenuated the inhibitory effects of bortezomib on APRIL and CCL3. We induced mononuclear cells to differentiate into OCs, and the enzyme-linked immunosorbent assay showed that the more OCs differentiated, the higher the levels CCL3 secretions detected. Animal experiments showed that hemin promoted MM cell infiltration in mice. The weight and survival rate of tumor mice were associated with HO-1 expression. Immunohistochemical staining showed that HO-1, APRIL, and CCL3 staining were positively stained in the tumor infiltrating sites. Then, MM cells were transfected with L-HO-1/si-HO-1 expression vectors and cultured with an nuclear factor (NF)-kappa B (κB) pathway inhibitor, QNZ. The results showed that HO-1 was the upstream gene of APRIL, NF-κB, and CCL3. We showed that HO-1 could attenuate the inhibitory effect of bortezomib against the APRIL-NF-κB-CCL3 signaling pathways in MM cells, and the tolerance of MM cells to bortezomib and the promotion of bone destruction are related to HO-1.
Collapse
Affiliation(s)
- Zheng C He
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xin Y Li
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yong L Guo
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Dan Ma
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qin Fang
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ling L Ren
- Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhao Y Zhang
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Weili Wang
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zheng Y Yu
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Peng Zhao
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ji S Wang
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
36
|
Ni D, Xiao Z, Zhong B, Feng X. Multiple Human-Behaviour Indicators for Predicting Lung Cancer Mortality with Support Vector Machine. Sci Rep 2018; 8:16596. [PMID: 30413734 PMCID: PMC6226432 DOI: 10.1038/s41598-018-34945-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/27/2018] [Indexed: 11/25/2022] Open
Abstract
Lung cancer is still one of the most common causes of death around the world, while there is overwhelming evidence that the environment and lifestyle factors are predominant causes of most sporadic cancers. However, when applying human-behaviour indicators to the prediction of cancer mortality (CM), we are often caught in a dilemma with inadequate sample size. Thus, this study extracted 30 human-behaviour indicators of seven categories (air pollution, tobacco smoking & alcohol consumption, socioeconomic status, food structure, working culture, medical level, and demographic structure) from Organization for Economic Cooperation and Development Database and World Health Organization Mortality Database for 13 countries (1998-2013), and employed Support Vector Machine (SVM) to examine the weights of 30 indicators across the 13 countries and the power for predicting lung CM for the years between 2014-2016. The weights of different human-behaviour indicators indicate that every country has its own lung cancer killers, that is, the human-behaviour indicators are country specific; Moreover, SVM has an excellent power in predicting their lung CM. The average accuracy in prediction offered by SVM can be as high as 96.08% for the 13 countries tested between 2014 and 2016.
Collapse
Affiliation(s)
- Du Ni
- School of Economics and Business Administration, Chongqing University, Chongqing, 400044, PR China
| | - Zhi Xiao
- School of Economics and Business Administration, Chongqing University, Chongqing, 400044, PR China.
| | - Bo Zhong
- College of Mathematics and Statistics, Chongqing University, Chongqing, 400044, PR China
| | - Xiaodong Feng
- School of Economics and Business Administration, Chongqing University, Chongqing, 400044, PR China
| |
Collapse
|
37
|
Kim DH, Yoon HJ, Cha YN, Surh YJ. Role of heme oxygenase-1 and its reaction product, carbon monoxide, in manifestation of breast cancer stem cell-like properties: Notch-1 as a putative target. Free Radic Res 2018; 52:1336-1347. [DOI: 10.1080/10715762.2018.1473571] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Do-Hee Kim
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Hyo-Jin Yoon
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Young-Nam Cha
- Department of Pharmacology and Toxicology, College of Medicine, Inha University, Incheon, South Korea
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
- Cancer Research Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
38
|
Gao C, Liang X, Guo Z, Jiang BP, Liu X, Shen XC. Diiron Hexacarbonyl Complex Induces Site-Specific Release of Carbon Monoxide in Cancer Cells Triggered by Endogenous Glutathione. ACS OMEGA 2018; 3:2683-2689. [PMID: 30023846 PMCID: PMC6044757 DOI: 10.1021/acsomega.8b00052] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/22/2018] [Indexed: 06/08/2023]
Abstract
In this study, we have evaluated a water-soluble, nontarget reagent and a carrier-free diiron hexacarbonyl complex, [Fe2{μ-SCH2CH(OH)CH2(OH)}2(CO)6] (TG-FeCORM), that can induce the site-specific release of carbon monoxide (CO) in cancer cells triggered by endogenous glutathione (GSH). The releasing rate of CO was dependent on the amount of endogenous GSH. Being the amount of endogenous GSH higher in cancer cells than in normal cells, the CO-releasing rate resulted faster in cancer cells. Moreover, the anti-inflammatory properties related to the intracellular CO release of TG-FeCORM were also confirmed in the living HeLa cells.
Collapse
Affiliation(s)
- Cunji Gao
- State
Key Laboratory for Chemistry and Molecular Engineering of Medicinal
Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China
| | - Xiaohua Liang
- State
Key Laboratory for Chemistry and Molecular Engineering of Medicinal
Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China
| | - Zhengxi Guo
- State
Key Laboratory for Chemistry and Molecular Engineering of Medicinal
Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China
| | - Bang-Ping Jiang
- State
Key Laboratory for Chemistry and Molecular Engineering of Medicinal
Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China
| | - Xiaoming Liu
- College
of Biological, Chemical Sciences and Engineering, Jiaxing University, Jiaxing 314001, P. R. China
| | - Xing-Can Shen
- State
Key Laboratory for Chemistry and Molecular Engineering of Medicinal
Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China
| |
Collapse
|
39
|
Vanella L, Barbagallo I, Tibullo D, Forte S, Zappalà A, Li Volti G. The non-canonical functions of the heme oxygenases. Oncotarget 2018; 7:69075-69086. [PMID: 27626166 PMCID: PMC5356613 DOI: 10.18632/oncotarget.11923] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/05/2016] [Indexed: 11/25/2022] Open
Abstract
Heme oxygenase (HO) isoforms catalyze the conversion of heme to carbon monoxide (CO) and biliverdin with a concurrent release of iron, which can drive the synthesis of ferritin for iron sequestration. Most of the studies so far were directed at evaluating the protective effect of these enzymes because of their ability to generate antioxidant and antiapoptotic molecules such as CO and bilirubin. Recent evidences are suggesting that HO may possess other important physiological functions, which are not related to its enzymatic activity and for which we would like to introduce for the first time the term “non canonical functions”. Recent evidence suggest that both HO isoforms may form protein-protein interactions (i.e. cytochrome P450, adiponectin, CD91) thus serving as chaperone-like protein. In addition, truncated HO-1 isoform was localized in the nuclear compartment under certain experimental conditions (i.e. excitotoxicity, hypoxia) regulating the activity of important nuclear transcription factors (i.e. Nrf2) and DNA repair. In the present review, we discuss three potential signaling mechanisms that we refer to as the non-canonical functions of the HO isoforms: protein-protein interaction, intracellular compartmentalization, and extracellular secretion. The aim of the present review is to describe each of this mechanism and all the aspects warranting additional studies in order to unravel all the functions of the HO system.
Collapse
Affiliation(s)
- Luca Vanella
- Department of Drug Sciences, University of Catania, Catania, Italy
| | | | - Daniele Tibullo
- Division of Haematology, AOU "Policlinico - Vittorio Emanuele", University of Catania, Catania, Italy
| | - Stefano Forte
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Istituto Oncologico del Mediterraneo Ricerca srl Viagrande, Catania, Italy
| | - Agata Zappalà
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,EuroMediterranean Institute of Science and Technology, Palermo, Italy
| |
Collapse
|
40
|
Hegedűs C, Kovács K, Polgár Z, Regdon Z, Szabó É, Robaszkiewicz A, Forman HJ, Martner A, Virág L. Redox control of cancer cell destruction. Redox Biol 2018; 16:59-74. [PMID: 29477046 PMCID: PMC5842284 DOI: 10.1016/j.redox.2018.01.015] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/25/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023] Open
Abstract
Redox regulation has been proposed to control various aspects of carcinogenesis, cancer cell growth, metabolism, migration, invasion, metastasis and cancer vascularization. As cancer has many faces, the role of redox control in different cancers and in the numerous cancer-related processes often point in different directions. In this review, we focus on the redox control mechanisms of tumor cell destruction. The review covers the tumor-intrinsic role of oxidants derived from the reduction of oxygen and nitrogen in the control of tumor cell proliferation as well as the roles of oxidants and antioxidant systems in cancer cell death caused by traditional anticancer weapons (chemotherapeutic agents, radiotherapy, photodynamic therapy). Emphasis is also put on the role of oxidants and redox status in the outcome following interactions between cancer cells, cytotoxic lymphocytes and tumor infiltrating macrophages.
Collapse
Affiliation(s)
- Csaba Hegedűs
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Katalin Kovács
- MTA-DE Cell Biology and Signaling Research Group, Debrecen, Hungary
| | - Zsuzsanna Polgár
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Regdon
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Szabó
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Agnieszka Robaszkiewicz
- Department of General Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Henry Jay Forman
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Anna Martner
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - László Virág
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE Cell Biology and Signaling Research Group, Debrecen, Hungary.
| |
Collapse
|
41
|
Ayudhya TI, Pellechia PJ, Dingra NN. ROS-mediated carbon monoxide and drug release from drug-conjugated carboxyboranes. Dalton Trans 2018; 47:538-543. [DOI: 10.1039/c7dt03581k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dual nature of amine carboxyboranes for combined CO and drug delivery is facilitated by ROS.
Collapse
Affiliation(s)
- T. I. Ayudhya
- Department of Chemistry
- University of Alaska
- Anchorage
- USA
| | - P. J. Pellechia
- Department of Chemistry and Biochemistry
- University of South Carolina
- Columbia
- USA
| | - N. N. Dingra
- Department of Chemistry
- University of Alaska
- Anchorage
- USA
| |
Collapse
|
42
|
Roca H, Jones JD, Purica MC, Weidner S, Koh AJ, Kuo R, Wilkinson JE, Wang Y, Daignault-Newton S, Pienta KJ, Morgan TM, Keller ET, Nör JE, Shea LD, McCauley LK. Apoptosis-induced CXCL5 accelerates inflammation and growth of prostate tumor metastases in bone. J Clin Invest 2017; 128:248-266. [PMID: 29202471 DOI: 10.1172/jci92466] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 10/17/2017] [Indexed: 12/31/2022] Open
Abstract
During tumor progression, immune system phagocytes continually clear apoptotic cancer cells in a process known as efferocytosis. However, the impact of efferocytosis in metastatic tumor growth is unknown. In this study, we observed that macrophage-driven efferocytosis of prostate cancer cells in vitro induced the expression of proinflammatory cytokines such as CXCL5 by activating Stat3 and NF-κB(p65) signaling. Administration of a dimerizer ligand (AP20187) triggered apoptosis in 2 in vivo syngeneic models of bone tumor growth in which apoptosis-inducible prostate cancer cells were either coimplanted with vertebral bodies, or inoculated in the tibiae of immunocompetent mice. Induction of 2 pulses of apoptosis correlated with increased infiltration of inflammatory cells and accelerated tumor growth in the bone. Apoptosis-induced tumors displayed elevated expression of the proinflammatory cytokine CXCL5. Likewise, CXCL5-deficient mice had reduced tumor progression. Peripheral blood monocytes isolated from patients with bone metastasis of prostate cancer were more efferocytic compared with normal controls, and CXCL5 serum levels were higher in metastatic prostate cancer patients relative to patients with localized prostate cancer or controls. Altogether, these findings suggest that the myeloid phagocytic clearance of apoptotic cancer cells accelerates CXCL5-mediated inflammation and tumor growth in bone, pointing to CXCL5 as a potential target for cancer therapeutics.
Collapse
Affiliation(s)
- Hernan Roca
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Jacqueline D Jones
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Marta C Purica
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Savannah Weidner
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Amy J Koh
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Robert Kuo
- Department of Chemical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - John E Wilkinson
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yugang Wang
- Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Stephanie Daignault-Newton
- Department of Biostatistics, Center for Cancer Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Kenneth J Pienta
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Todd M Morgan
- Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Evan T Keller
- Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Jacques E Nör
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - Laurie K McCauley
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
43
|
Li Volti G, Avola R, Vinciguerra M. Commentary: Fasting-Mimicking Diet Reduces HO-1 to Promote T Cell-Mediated Tumor Cytotoxicity. Front Oncol 2017. [PMID: 28626728 PMCID: PMC5455696 DOI: 10.3389/fonc.2017.00116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Roberto Avola
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Manlio Vinciguerra
- Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno, Czechia
| |
Collapse
|
44
|
Germline Proliferation Is Regulated by Somatic Endocytic Genes via JNK and BMP Signaling in Drosophila. Genetics 2017; 206:189-197. [PMID: 28315838 PMCID: PMC5419469 DOI: 10.1534/genetics.116.196535] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 03/06/2017] [Indexed: 12/14/2022] Open
Abstract
Signals derived from the microenvironment contribute greatly to tumorigenesis . The underlying mechanism requires thorough investigation. Here, we use Drosophila testis as a model system to address this question, taking the advantage of the ease to distinguish germline and somatic cells and to track the cell numbers. In an EMS mutagenesis screen, we identified Rab5, a key factor in endocytosis, for its nonautonomous role in germline proliferation. The disruption of Rab5 in somatic cyst cells, which escort the development of germline lineage, induced the overproliferation of underdifferentiated but genetically wild-type germ cells. We demonstrated that this nonautonomous effect was mediated by the transcriptional activation of Dpp [the fly homolog of bone morphogenetic protein (BMP)] by examining the Dpp-reporter expression and knocking down Dpp to block germline overgrowth. Consistently, the protein levels of Bam, the germline prodifferentiation factor normally accumulated in the absence of BMP/Dpp signaling, decreased in the overproliferating germ cells. Further, we discovered that the JNK signaling pathway operated between Rab5 and Dpp, because simultaneously inhibiting the JNK pathway and Rab5 in cyst cells prevented both dpp transcription and germline tumor growth. Additionally, we found that multiple endocytic genes, such as avl, TSG101, Vps25, or Cdc42, were required in the somatic cyst cells to restrict germline amplification. These findings indicate that when the endocytic state of the surrounding cells is impaired, genetically wild-type germ cells overgrow. This nonautonomous model of tumorigenesis provides a simple system to dissect the relation between tumor and its niche.
Collapse
|