1
|
Wang YH, Yang X, Liu CC, Wang X, Yu KD. Unraveling the peripheral nervous System's role in tumor: A Double-edged Sword. Cancer Lett 2025; 611:217451. [PMID: 39793755 DOI: 10.1016/j.canlet.2025.217451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
The peripheral nervous system (PNS) includes all nerves outside the brain and spinal cord, comprising various cells like neurons and glial cells, such as schwann and satellite cells. The PNS is increasingly recognized for its bidirectional interactions with tumors, exhibiting both pro- and anti-tumor effects. Our review delves into the complex mechanisms underlying these interactions, highlighting recent findings that challenge the conventional understanding of PNS's role in tumorigenesis. We emphasize the contradictory results in the literature and propose novel perspectives on how these discrepancies can be resolved. By focusing on the PNS's influence on tumor initiation, progression, and microenvironment remodeling, we provide a comprehensive analysis that goes beyond the structural description of the PNS. Our review suggests that a deeper comprehension of the PNS-tumor crosstalk is pivotal for developing targeted anticancer strategies. We conclude by emphasizing the need for future research to unravel the intricate dynamics of the PNS in cancer, which may lead to innovative diagnostic tools and therapeutic approaches.
Collapse
Affiliation(s)
- Yan-Hao Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| | - Xuan Yang
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, 030000, PR China
| | - Cui-Cui Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| | - Xin Wang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Ke-Da Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China.
| |
Collapse
|
2
|
Li X, Ye C, Wang M, Kwan P, Tian X, Zhang Y. Crosstalk Between the Nervous System and Colorectal Cancer. Neurosci Bull 2025; 41:93-106. [PMID: 38879846 PMCID: PMC11748644 DOI: 10.1007/s12264-024-01238-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/22/2024] [Indexed: 01/05/2025] Open
Abstract
The nervous system is the dominant regulatory system in the human body. The traditional theory is that tumors lack innervation. However, an increasing number of studies have shown complex bidirectional interactions between tumors and the nervous system. Globally, colorectal cancer (CRC) is the third most common cancer. With the rise of tumor neuroscience, the role of nervous system imbalances in the occurrence and development of CRC has attracted increasing amounts of attention. However, there are still many gaps in the research on the interactions and mechanisms involved in the nervous system in CRC. This article systematically reviews emerging research on the bidirectional relationships between the nervous system and CRC, focusing on the following areas: (1) Effects of the nervous system on colon cancer. (2) Effects of CRC on the nervous system. (3) Treatment of CRC associated with the nervous system.
Collapse
Affiliation(s)
- Xi Li
- Jining Medical University, Jining, 272000, China
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Chunshui Ye
- Department of Gastrointestinal Surgery, Jining No. 1 People's Hospital, Jining, 272000, China
| | - Min Wang
- Department of Neurology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Patrick Kwan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China.
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.
- Department of Neurology, Alfred Health, Melbourne, VIC, 3004, Australia.
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, 3004, Australia.
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China.
| | - Yanke Zhang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, 272000, China.
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
3
|
Jiang L, Cai S, Weng Z, Zhang S, Jiang SH. Peripheral, central, and chemotherapy-induced neuropathic changes in pancreatic cancer. Trends Neurosci 2024:S0166-2236(24)00242-X. [PMID: 39730257 DOI: 10.1016/j.tins.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/27/2024] [Accepted: 11/27/2024] [Indexed: 12/29/2024]
Abstract
In pancreatic cancer, significant alterations occur in the local nervous system, including axonogenesis, neural remodeling, perineural invasion, and perineural neuritis. Pancreatic cancer can impact the central nervous system (CNS) through cancer cell-intrinsic factors or systemic factors, particularly in the context of cancer cachexia. These peripheral and central neuropathic changes exert substantial influence on cancer initiation and progression. Moreover, chemotherapy-induced neuropathy is common in pancreatic cancer, causing peripheral nerve damage and cognitive dysfunction. Targeting the crosstalk between pancreatic cancer and the nervous system, either peripherally or centrally, holds promise in cancer treatment, pain relief, and improved quality of life. Here, we summarize recent findings on the molecular mechanisms behind these neuropathic changes in pancreatic cancer and discuss potential intervention strategies.
Collapse
Affiliation(s)
- Luju Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Shuqi Cai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Zheqi Weng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Shan Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Shu-Heng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| |
Collapse
|
4
|
Gao J, Liu J, Lu J, Zhang X, Zhang W, Li Q, Cai J, Li M, Gan Y, Tang Y, Wu S. SKAP1 Expression in Cancer Cells Enhances Colon Tumor Growth and Impairs Cytotoxic Immunity by Promoting Neutrophil Extracellular Trap Formation via the NFATc1/CXCL8 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403430. [PMID: 39269257 PMCID: PMC11538704 DOI: 10.1002/advs.202403430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/15/2024] [Indexed: 09/15/2024]
Abstract
The mechanisms underlying the development and progression of colon cancer are not fully understood. Herein, Src kinase associated phosphoprotein 1 (SKAP1), an immune cell adaptor, is identified as a novel colon cancer-related gene. SKAP1 expression is significantly increased in colon cancer cells. High SKAP1 levels are independently predictive of poor survival in patients with colon cancer. Notably, SKAP1 expression in colon cancer cells exerted a significant tumor-promoting effect in vivo rather than in vitro. Screening of tumor-infiltrating immune cells revealed the involvement of neutrophils in SKAP1-induced colon tumor promotion. Enhanced formation of neutrophil extracellular traps (NETs) is found to be a key downstream event that contributed to the pro-tumor role of SKAP1. In colon cancer cells, SKAP1 increased the expression of C-X-C motif chemokine ligand 8 (CXCL8) via nuclear factor of activated T cells c1 (NFATc1). The blockade of CXCL8 or NFATc1 largely attenuated neutrophil infiltration, NET formation, and tumor promotion induced by SKAP1. Furthermore, inhibiting SKAP1-induced NET significantly enhanced the antitumor efficiency of adoptive natural killer cell therapy in colon tumor models. In conclusion, SKAP1 significantly promotes colon cancer growth via the cancer cell/neutrophil NFATc1/CXCL8/NET axis, suggesting that SKAP1 is a potential target for colon cancer therapy.
Collapse
Affiliation(s)
- Jian Gao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200032China
| | - Jun Liu
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| | - Jilin Lu
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| | - Xiaofei Zhang
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| | - Wei Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200032China
| | - Qian Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200032China
| | - Jiayi Cai
- Clinical Research UnitRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Mengjun Li
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| | - Yu Gan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200032China
| | - Yifan Tang
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| | - Shuangjie Wu
- Department of General SurgeryHuashan Hospital (Hongqiao Campus)Fudan UniversityShanghai201107China
| |
Collapse
|
5
|
Rangel-Sosa MM, Mann F, Chauvet S. Pancreatic Schwann cell reprogramming supports cancer-associated neuronal remodeling. Glia 2024; 72:1840-1861. [PMID: 38961612 DOI: 10.1002/glia.24586] [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: 12/20/2023] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
The peripheral nervous system is a key regulator of cancer progression. In pancreatic ductal adenocarcinoma (PDAC), the sympathetic branch of the autonomic nervous system inhibits cancer development. This inhibition is associated with extensive sympathetic nerve sprouting in early pancreatic cancer precursor lesions. However, the underlying mechanisms behind this process remain unclear. This study aimed to investigate the roles of pancreatic Schwann cells in the structural plasticity of sympathetic neurons. We examined the changes in the number and distribution of Schwann cells in a transgenic mouse model of PDAC and in a model of metaplastic pancreatic lesions induced by chronic inflammation. Schwann cells proliferated and expanded simultaneously with new sympathetic nerve sprouts in metaplastic/neoplastic pancreatic lesions. Sparse genetic labeling showed that individual Schwann cells in these lesions had a more elongated and branched structure than those under physiological conditions. Schwann cells overexpressed neurotrophic factors, including glial cell-derived neurotrophic factor (GDNF). Sympathetic neurons upregulated the GDNF receptors and exhibited enhanced neurite growth in response to GDNF in vitro. Selective genetic deletion of Gdnf in Schwann cells completely blocked sympathetic nerve sprouting in metaplastic pancreatic lesions in vivo. This study demonstrated that pancreatic Schwann cells underwent adaptive reprogramming during early cancer development, supporting a protective antitumor neuronal response. These finding could help to develop new strategies to modulate cancer associated neural plasticity.
Collapse
Affiliation(s)
| | - Fanny Mann
- Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | | |
Collapse
|
6
|
Cait J, Winder CB, Mason GJ. How much 'enrichment' is enough for laboratory rodents? A systematic review and meta-analysis re-assessing the impact of well-resourced cages on morbidity and mortality. Appl Anim Behav Sci 2024; 278:106361. [DOI: 10.1016/j.applanim.2024.106361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
7
|
Zhang H, Yang Y, Cao Y, Guan J. Effects of chronic stress on cancer development and the therapeutic prospects of adrenergic signaling regulation. Biomed Pharmacother 2024; 175:116609. [PMID: 38678960 DOI: 10.1016/j.biopha.2024.116609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
Long-term chronic stress is an important factor in the poor prognosis of cancer patients. Chronic stress reduces the tissue infiltration of immune cells in the tumor microenvironment (TME) by continuously activating the adrenergic signaling, inhibits antitumor immune response and tumor cell apoptosis while also inducing epithelial-mesenchymal transition (EMT) and tumor angiogenesis, promoting tumor invasion and metastasis. This review first summarizes how adrenergic signaling activates intracellular signaling by binding different adrenergic receptor (AR) heterodimers. Then, we focused on reviewing adrenergic signaling to regulate multiple functions of immune cells, including cell differentiation, migration, and cytokine secretion. In addition, the article discusses the mechanisms by which adrenergic signaling exerts pro-tumorigenic effects by acting directly on the tumor itself. It also highlights the use of adrenergic receptor modulators in cancer therapy, with particular emphasis on their potential role in immunotherapy. Finally, the article reviews the beneficial effects of stress intervention measures on cancer treatment. We think that enhancing the body's antitumor response by adjusting adrenergic signaling can enhance the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, The Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100091, China; Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| | - Yuwei Yang
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Yan Cao
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Jingzhi Guan
- Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| |
Collapse
|
8
|
Yang F, Hua Q, Zhu X, Xu P. Surgical stress induced tumor immune suppressive environment. Carcinogenesis 2024; 45:185-198. [PMID: 38366618 DOI: 10.1093/carcin/bgae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/25/2024] [Accepted: 02/14/2024] [Indexed: 02/18/2024] Open
Abstract
Despite significant advances in cancer treatment over the decades, surgical resection remains a prominent management approach for solid neoplasms. Unfortunately, accumulating evidence suggests that surgical stress caused by tumor resection may potentially trigger postoperative metastatic niche formation. Surgical stress not only activates the sympathetic-adrenomedullary axis and hypothalamic-pituitary-adrenocortical axis but also induces hypoxia and hypercoagulable state. These adverse factors can negatively impact the immune system by downregulating immune effector cells and upregulating immune suppressor cells, which contribute to the colonization and progression of postoperative tumor metastatic niche. This review summarizes the effects of surgical stress on four types of immune effector cells (neutrophils, macrophages, natural killer cells and cytotoxic T lymphocytes) and two types of immunosuppressive cells (regulatory T cells and myeloid-derived suppressor cells), and discusses the immune mechanisms of postoperative tumor relapse and progression. Additionally, relevant therapeutic strategies to minimize the pro-tumorigenic effects of surgical stress are elucidated.
Collapse
Affiliation(s)
- Fan Yang
- Department of Anesthesiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Research Center for Neuro-Oncology Interaction, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qing Hua
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaoyan Zhu
- Department of Physiology, Navy Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Pingbo Xu
- Department of Anesthesiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Research Center for Neuro-Oncology Interaction, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| |
Collapse
|
9
|
Wang S, Gong X, Xiao F, Yang Y. Recent advances in host-focused molecular tools for investigating host-gut microbiome interactions. Front Microbiol 2024; 15:1335036. [PMID: 38605718 PMCID: PMC11007152 DOI: 10.3389/fmicb.2024.1335036] [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: 11/08/2023] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Microbial communities in the human gut play a significant role in regulating host gene expression, influencing a variety of biological processes. To understand the molecular mechanisms underlying host-microbe interactions, tools that can dissect signaling networks are required. In this review, we discuss recent advances in molecular tools used to study this interplay, with a focus on those that explore how the microbiome regulates host gene expression. These tools include CRISPR-based whole-body genetic tools for deciphering host-specific genes involved in the interaction process, Cre-loxP based tissue/cell-specific gene editing approaches, and in vitro models of host-derived organoids. Overall, the application of these molecular tools is revolutionizing our understanding of how host-microbiome interactions contribute to health and disease, paving the way for improved therapies and interventions that target microbial influences on the host.
Collapse
Affiliation(s)
- Siyao Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| | - Xu Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| | - Fei Xiao
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yun Yang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| |
Collapse
|
10
|
Xu W, Liu J, Zhang J, Lu J, Guo J. Tumor microenvironment crosstalk between tumors and the nervous system in pancreatic cancer: Molecular mechanisms and clinical perspectives. Biochim Biophys Acta Rev Cancer 2024; 1879:189032. [PMID: 38036106 DOI: 10.1016/j.bbcan.2023.189032] [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: 07/06/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) exhibits the highest incidence of perineural invasion among all solid tumors. The intricate interplay between tumors and the nervous system plays an important role in PDAC tumorigenesis, progression, recurrence, and metastasis. Various clinical symptoms of PDAC, including anorexia and cancer pain, have been linked to aberrant neural activity, while the presence of perineural invasion is a significant prognostic indicator. The use of conventional neuroactive drugs and neurosurgical interventions for PDAC patients is on the rise. An in-depth exploration of tumor-nervous system crosstalk has revealed novel therapeutic strategies for mitigating PDAC progression and effectively relieving symptoms. In this comprehensive review, we elucidate the regulatory functions of tumor-nervous system crosstalk, provide a succinct overview of the relationship between tumor-nervous system dialogue and clinical symptomatology, and deliberate the current research progress and forthcoming avenues of neural therapy for PDAC.
Collapse
Affiliation(s)
- Wenchao Xu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianzhou Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianlu Zhang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Jun Lu
- Department of General Surgery, Peking University Third Hospital, Beijing 100730, China
| | - Junchao Guo
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
11
|
Huang FF, Cui WH, Ma LY, Chen Q, Liu Y. Crosstalk of nervous and immune systems in pancreatic cancer. Front Cell Dev Biol 2023; 11:1309738. [PMID: 38099290 PMCID: PMC10720593 DOI: 10.3389/fcell.2023.1309738] [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: 10/08/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
Pancreatic cancer is a highly malignant tumor known for its extremely low survival rate. The combination of genetic disorders within pancreatic cells and the tumor microenvironment contributes to the emergence and progression of this devastating disease. Extensive research has shed light on the nature of the microenvironmental cells surrounding the pancreatic cancer, including peripheral nerves and immune cells. Peripheral nerves release neuropeptides that directly target pancreatic cancer cells in a paracrine manner, while immune cells play a crucial role in eliminating cancer cells that have not evaded the immune response. Recent studies have revealed the intricate interplay between the nervous and immune systems in homeostatic condition as well as in cancer development. In this review, we aim to summarize the function of nerves in pancreatic cancer, emphasizing the significance to investigate the neural-immune crosstalk during the advancement of this malignant cancer.
Collapse
Affiliation(s)
- Fei-Fei Huang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wen-Hui Cui
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lan-Yue Ma
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qi Chen
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| | - Yang Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
12
|
Luo Q, Zheng J, Fan B, Liu J, Liao W, Zhang X. Enriched environment attenuates ferroptosis after cerebral ischemia/reperfusion injury by regulating iron metabolism. Brain Res Bull 2023; 203:110778. [PMID: 37812906 DOI: 10.1016/j.brainresbull.2023.110778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/20/2023] [Accepted: 10/06/2023] [Indexed: 10/11/2023]
Abstract
Preventing neuronal death after ischemic stroke (IS) is crucial for neuroprotective treatment, yet current management options are limited. Enriched environment (EE) is an effective intervention strategy that promotes the recovery of neurological function after cerebral ischemia/reperfusion (I/R) injury. Ferroptosis has been identified as one of the mechanisms of neuronal death during IS, and inhibiting ferroptosis can reduce cerebral I/R injury. Our previous research has demonstrated that EE reduced ferroptosis by inhibiting lipid peroxidation, but the underlying mechanism still needs to be investigated. This study aims to explore the potential molecular mechanisms by which EE modulates iron metabolism to reduce ferroptosis. The experimental animals were randomly divided into four groups based on the housing environment and the procedure the animals received: the sham-operated + standard environment (SSE) group, the sham-operated + enriched environment (SEE) group, the ischemia/reperfusion + standard environment (ISE) group, and the ischemia/reperfusion + enriched environment (IEE) group. The results showed that EE reduced IL-6 expression during cerebral I/R injury, hence reducing JAK2-STAT3 pathway activation and hepcidin expression. Reduced hepcidin expression led to decreased DMT1 expression and increased FPN1 expression in neurons, resulting in lower neuronal iron levels and alleviated ferroptosis. In addition, EE also reduced the expression of TfR1 in neurons. Our research suggested that EE played a neuroprotective role by modulating iron metabolism and reducing neuronal ferroptosis after cerebral I/R injury, which might be achieved by inhibiting inflammatory response and down-regulating hepcidin expression.
Collapse
Affiliation(s)
- Qihang Luo
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Zheng
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bin Fan
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jingying Liu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Weijing Liao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xin Zhang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
13
|
Liu T, Li J, Li Q, Liang Y, Gao J, Meng Z, Li P, Yao M, Gu J, Tu H, Gan Y. Environmental eustress promotes liver regeneration through the sympathetic regulation of type 1 innate lymphoid cells to increase IL-22 in mice. Hepatology 2023; 78:136-149. [PMID: 36631003 DOI: 10.1097/hep.0000000000000239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 12/02/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND AIMS The liver has the unique ability of regeneration, which is extremely important for restoring homeostasis after liver injury. Although clinical observations have revealed an association between psychological stress and the liver, whether stress has a causal influence on the liver regeneration remains markedly less defined. APPROACH AND RESULTS Rearing rodents in an enriched environment (EE) can induce eustress or positive psychological stress. Herein, EE-induced eustress was found to significantly enhance the ability of liver regeneration after partial hepatectomy or carbon tetrachloride-induced liver injury based on the more rapid restoration of liver/body weight ratio and the significantly increased number of proliferating hepatocytes in EE mice. Mechanistically, the cytokine array revealed that IL-22 was markedly increased in the regenerating liver in response to EE. Blockade of IL-22 signaling abrogated the enhanced liver regeneration induced by EE. Group 1 innate lymphoid cells (ILCs), including type 1 ILCs (ILC1s), have been identified as the major sources of IL-22 in the regenerating liver. EE housing led to a rapid accumulation of hepatic ILC1s after partial hepatectomy and the EE-induced enhancement of liver regeneration and elevation of IL-22 was nearly eliminated in ILC1-deficient Tbx21-/- mice. Chemical sympathectomy or blockade of β-adrenergic signaling also abolished the effect of EE on ILC1s and attenuated the enhanced liver regeneration of EE-housed mice. CONCLUSION The study findings support the brain-liver axis and suggest that environment-induced eustress promotes liver regeneration through the sympathetic nerve/ILC1/IL-22 axis.
Collapse
Affiliation(s)
- Tingting Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiyi Liang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Gao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zihong Meng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinyang Gu
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Transplantation, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Tu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Gan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Canali MM, Guyot M, Simon T, Daoudlarian D, Chabry J, Panzolini C, Petit-Paitel A, Hypolite N, Nicolas S, Bourdely P, Schmid-Antomarchi H, Schmid-Alliana A, Soria J, Karimdjee Soilihi B, Hofman P, Prevost-Blondel A, Kato M, Mougneau E, Glaichenhaus N, Blancou P. Environmental signals perceived by the brain abate pro-metastatic monocytes by dampening glucocorticoids receptor signaling. Cancer Cell Int 2023; 23:15. [PMID: 36726173 PMCID: PMC9893572 DOI: 10.1186/s12935-023-02855-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 01/19/2023] [Indexed: 02/03/2023] Open
Abstract
While positive social-behavioral factors predict longer survival in cancer patients, the underlying mechanisms are unknown. Since tumor metastasis are the major cancer mortality factor, we investigated how an enriched environment (EE) conductive to enhanced sensory, cognitive and motor stimulation impact metastatic progression in lungs following intravasation in the circulation. We find that mice housed in EE exhibited reduced number of lung metastatic foci compared to control mice housed in a standard environment (SE). Compared to SE mice, EE mice increased lung inflammation as early as 4 days after circulating tumor cells extravasation. The impact of environmental signals on lung metastasis is independent of adrenergic receptors signaling. By contrast, we find that serum corticosterone levels are lower in EE mice and that glucocorticoid receptor (GR) antagonist reduces the number of lung metastasis in SE mice. In addition, the difference of the number of lung metastasis between SE and EE mice is abolished when inflammatory monocytes are rendered deficient in GR signaling. This decreased GR signaling in inflammatory monocytes of SE mice results in an exacerbated inflammatory profile in the lung. Our study shows that not only EE reduces late stages of metastatic progression in lungs but disclose a novel anti-tumor mechanism whereby GR-dependent reprogramming of inflammatory monocytes can inhibit metastatic progression in lungs. Moreover, while inflammatory monocytes have been shown to promote cancer progression, they also have an anti-tumor effect, suggesting that their role is more complex than currently thought.
Collapse
Affiliation(s)
- María Magdalena Canali
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Mélanie Guyot
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Thomas Simon
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Douglas Daoudlarian
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Joelle Chabry
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Clara Panzolini
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Agnès Petit-Paitel
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Nicolas Hypolite
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Sarah Nicolas
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Pierre Bourdely
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Heidy Schmid-Antomarchi
- grid.460782.f0000 0004 4910 6551Université Côte d’Azur, CNRS, INSERM, Valrose Biology Institute, 28 Avenue de Valombrose, Nice, France
| | - Annie Schmid-Alliana
- grid.460782.f0000 0004 4910 6551Université Côte d’Azur, CNRS, INSERM, Valrose Biology Institute, 28 Avenue de Valombrose, Nice, France
| | - Javier Soria
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Babou Karimdjee Soilihi
- grid.460782.f0000 0004 4910 6551Université Côte d’Azur, CNRS, INSERM, Valrose Biology Institute, 28 Avenue de Valombrose, Nice, France ,Polyclinique Saint Jean, Cagnes sur mer, France
| | - Paul Hofman
- grid.410528.a0000 0001 2322 4179Laboratory of Clinical and Experimental Pathology and Biobank, Nice University Hospital, Nice, France ,grid.460782.f0000 0004 4910 6551Research Institute on Cancer and Aging, Université Côte d’Azur, CNRS, INSERM, 28 Avenue de Valombrose, Nice, France
| | - Armelle Prevost-Blondel
- grid.462098.10000 0004 0643 431XUniversité Paris Descartes, CNRS, INSERM, Institut Cochin, 22 rue Méchain, 75014 Paris, France
| | - Masashi Kato
- grid.27476.300000 0001 0943 978XDepartment of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Evelyne Mougneau
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Nicolas Glaichenhaus
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Philippe Blancou
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| |
Collapse
|
15
|
Yoon M, Lee HK, Park EY, Kim JH, Lee JH, Kim YS, Kim HJ, Kim H, Yoo CW, Lee S, Hong EK, Kim TH, Kim TS, Seo SS, Kang S, Chang SJ, Shin HJ, Uong TNT, Lee S, Kim JY. Randomized multicenter phase II trial of prophylactic irradiation of para-aortic lymph nodes in advanced cervical cancer according to tumor hypoxia: Korean Radiation Oncology Group (KROG 07-01) study. Int J Cancer 2022; 151:2182-2194. [PMID: 35751421 DOI: 10.1002/ijc.34190] [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: 12/15/2021] [Revised: 05/24/2022] [Accepted: 06/13/2022] [Indexed: 11/09/2022]
Abstract
We conducted a prospective phase II study on whether extended-field irradiation (EFI) confers survival benefits depending on hypoxic markers in locally advanced uterine cervical cancer (LAUCC). RNA-seq was performed to identify immune and hypoxic gene signatures. A total of 288 patients were randomized to either EFI or pelvic radiotherapy (PRT). All patients completed chemoradiotherapy. Overall, significantly higher 5-year para-aortic recurrence free survival (PARFS) rate occurred in EFI (97.6%) than in PRT group (87.2%), with marginal tendency to improve disease-free survival (DFS; 78% vs 70%, P = .066). Subgroup analyses were performed based on carbonic anhydrase 9 (CA9)-only positive, CA9/hypoxia-inducible factor (HIF) double positive and CA9 negative. In the CA9-only positive, EFI successfully increased 5-year PARFS (100% vs 76.4%, P = .010), resulting in significantly improved long-term DFS (85.7% vs 54.7%, P = .023) compared to the PRT, while there was no such benefit of EFI in the CA9/HIFs double positive. RNA-seq analysis identified distinct immunehigh subgroup with negative correlation with hypoxia gene signatures (R = -.37, P < .01), which showed a higher 5-year DFS than the immunelow (P = .032). Hypoxia-related genes were upregulated in the CA9/HIFs double positive compared to CA9 negative (P < .05). Only 17.4% of patients in CA9-negative group showed immunelow signatures, while 40.0% of patients in the double-positive group exhibited immunelow signatures. In conclusion, EFI improved PARFS significantly in all patients, but therapeutic efficacy of EFI in terms of improved DFS was solely observed in CA9-only positive LAUCC, and not in CA9/HIFs double-positive subgroup. RNA-seq analysis suggested that hypoxia-induced immunosuppression may be related to treatment resistance in LAUCC.
Collapse
Affiliation(s)
- Meesun Yoon
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Gwangju, South Korea
| | - Hyo Kyung Lee
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Eun Young Park
- Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Jin Hee Kim
- Department of Radiation Oncology, Dongsan Medical Center, Keimyung University School of Medicine, Daegu, South Korea
| | - Jong Hoon Lee
- Department of Radiation Oncology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University, Seoul, South Korea
| | - Young Seok Kim
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, South Korea
| | - Hak Jae Kim
- Department of Radiation Oncology, Seoul National University, College of Medicine, Seoul, South Korea
| | - Hunjung Kim
- Department of Radiation Oncology, Inha University Hospital, Inha University, School of Medicine, Incheon, South Korea
| | - Chong Woo Yoo
- Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Sun Lee
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, South Korea
| | - Eun Kyung Hong
- Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Tae Hyun Kim
- Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Tae-Sung Kim
- Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Sang-Soo Seo
- Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Sokbom Kang
- Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Suk-Joon Chang
- Gynecologic Cancer Center, Department of Obstetrics and Gynecology, Ajou University School of Medicine, Suwon, South Korea
| | - Hye Jin Shin
- Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Tung Nguyen Thanh Uong
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Gwangju, South Korea
| | - Semin Lee
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Joo-Young Kim
- Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| |
Collapse
|
16
|
Investigational Microbiological Therapy for Glioma. Cancers (Basel) 2022; 14:cancers14235977. [PMID: 36497459 PMCID: PMC9736089 DOI: 10.3390/cancers14235977] [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: 10/09/2022] [Revised: 11/27/2022] [Accepted: 11/29/2022] [Indexed: 12/07/2022] Open
Abstract
Glioma is the most common primary malignancy of the central nervous system (CNS), and 50% of patients present with glioblastoma (GBM), which is the most aggressive type. Currently, the most popular therapies are progressive chemotherapy and treatment with temozolomide (TMZ), but the median survival of glioma patients is still low as a result of the emergence of drug resistance, so we urgently need to find new therapies. A growing number of studies have shown that the diversity, bioactivity, and manipulability of microorganisms make microbial therapy a promising approach for cancer treatment. However, the many studies on the research progress of microorganisms and their derivatives in the development and treatment of glioma are scattered, and nobody has yet provided a comprehensive summary of them. Therefore, in this paper, we review the research progress of microorganisms and their derivatives in the development and treatment of glioma and conclude that it is possible to treat glioma by exogenous microbial therapies and targeting the gut-brain axis. In this article, we discuss the prospects and pressing issues relating to these therapies with the aim of providing new ideas for the treatment of glioma.
Collapse
|
17
|
Wu Y, Zhou L, Zhang X, Yang X, Niedermann G, Xue J. Psychological distress and eustress in cancer and cancer treatment: Advances and perspectives. SCIENCE ADVANCES 2022; 8:eabq7982. [PMID: 36417542 PMCID: PMC9683699 DOI: 10.1126/sciadv.abq7982] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 10/03/2022] [Indexed: 05/31/2023]
Abstract
Facing cancer diagnosis, patients with cancer are prone to psychological stress and consequent psychological disorders. The association between psychological stress and cancer has long been a subject of high interest. To date, preclinical studies have gradually uncovered the promotive effects of psychological distress on tumor hallmarks. In contrast, eustress may exert suppressive effects on tumorigenesis and beneficial effects on tumor treatment, which brings a practicable means and psychosocial perspective to cancer treatment. However, the underlying mechanisms remain incompletely understood. Here, by focusing on the hypothalamic-pituitary-adrenal axis and the sympathetic nervous system, as well as stress-related crucial neurotransmitters and hormones, we highlight the effects of distress and eustress on tumorigenesis, the tumor microenvironment, and tumor treatment. We also discuss the findings of clinical studies on stress management in patients with cancer. Last, we summarize questions that remain to be addressed and provide suggestions for future research directions.
Collapse
Affiliation(s)
- Yuanjun Wu
- Thoracic Oncology Ward, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Laiyan Zhou
- Thoracic Oncology Ward, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuanwei Zhang
- Thoracic Oncology Ward, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xue Yang
- Thoracic Oncology Ward, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Gabriele Niedermann
- Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, Freiburg, Germany, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg, Germany
| | - Jianxin Xue
- Thoracic Oncology Ward, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
18
|
An Overview of the Molecular Cues and Their Intracellular Signaling Shared by Cancer and the Nervous System: From Neurotransmitters to Synaptic Proteins, Anatomy of an All-Inclusive Cooperation. Int J Mol Sci 2022; 23:ijms232314695. [PMID: 36499024 PMCID: PMC9739679 DOI: 10.3390/ijms232314695] [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: 10/18/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022] Open
Abstract
We propose an overview of the molecular cues and their intracellular signaling involved in the crosstalk between cancer and the nervous system. While "cancer neuroscience" as a field is still in its infancy, the relation between cancer and the nervous system has been known for a long time, and a huge body of experimental data provides evidence that tumor-nervous system connections are widespread. They encompass different mechanisms at different tumor progression steps, are multifaceted, and display some intriguing analogies with the nervous system's physiological processes. Overall, we can say that many of the paradigmatic "hallmarks of cancer" depicted by Weinberg and Hanahan are affected by the nervous system in a variety of manners.
Collapse
|
19
|
Gupta P, Hodgman CF, Alvarez-Florez C, Schadler KL, Markofski MM, O’Connor DP, LaVoy EC. Comparison of three exercise interventions with and without gemcitabine treatment on pancreatic tumor growth in mice: No impact on tumor infiltrating lymphocytes. Front Physiol 2022; 13:1039988. [PMID: 36479351 PMCID: PMC9720271 DOI: 10.3389/fphys.2022.1039988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/03/2022] [Indexed: 10/06/2023] Open
Abstract
Exercise has been shown to slow pancreatic tumor growth, but whether exercise interventions of differing volume or intensity yield differential effects on tumor outcomes is unknown. In this study, we compared three exercise training interventions implemented with and without chemotherapy on pancreatic tumor growth in mice. Methods: Male C57BL/6 mice (6-8 weeks old) were subcutaneously inoculated with pancreatic ductal adenocarcinoma tumor cells (PDAC 4662). Upon tumor detection, mice received gemcitabine 15 mg/kg intraperitoneally 3 days/week and were assigned to exercise: high volume continuous exercise (HVCE), low volume continuous exercise (LVCE), high intensity interval training (HIIT), or sedentary (SED). HVCE ran at 12 m/min for 45 min and LVCE for 15 min, 5 days/week. HIIT ran 1-min at 20 m/min, followed by 1-min walking at 8 m/min for 20 total intervals, 3 days/week. SED did not run. Additional sets of inoculated mice were assigned to the exercise interventions but did not receive gemcitabine. Tumor volume was measured every other day for 2 weeks; tumor-infiltrating lymphocytes were assessed by flow cytometry 3-week post-inoculation. Results: Tumor growth did not differ between groups that received gemcitabine (F(3, 34) = 1.487; p = 0.235; η2 = 0.116). In contrast, tumor growth differed between groups not provided gemcitabine (F(3,14) = 3.364; p = 0.049, η2 = 0.419), with trends for slower growth in LVCE than SED (p = 0.088) and HIIT (p = 0.084). Groups did not differ in tumor infiltrating lymphocytes. Conclusion: Contrary to our hypotheses, the exercise interventions compared here did not further reduce pancreatic tumor growth beyond that provided by gemcitabine. However, in mice not receiving gemcitabine, there was a trend for reduced tumor growth in LVCE.
Collapse
Affiliation(s)
- Priti Gupta
- Department of Health and Human Performance, University of Houston, Houston, TX, United States
| | - Charles F. Hodgman
- Department of Health and Human Performance, University of Houston, Houston, TX, United States
| | - Claudia Alvarez-Florez
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, MD Anderson Cancer Center, Houston, TX, United States
| | - Keri L. Schadler
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Melissa M. Markofski
- Department of Health and Human Performance, University of Houston, Houston, TX, United States
| | - Daniel P. O’Connor
- Department of Health and Human Performance, University of Houston, Houston, TX, United States
| | - Emily C. LaVoy
- Department of Health and Human Performance, University of Houston, Houston, TX, United States
| |
Collapse
|
20
|
Gola M, Sejda A, Godlewski J, Cieślak M, Starzyńska A. Neural Component of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:5246. [PMID: 36358664 PMCID: PMC9657005 DOI: 10.3390/cancers14215246] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/04/2022] [Accepted: 10/25/2022] [Indexed: 10/15/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive primary malignancy of the pancreas, with a dismal prognosis and limited treatment options. It possesses a unique tumor microenvironment (TME), generating dense stroma with complex elements cross-talking with each other to promote tumor growth and progression. Diversified neural components makes for not having a full understanding of their influence on its aggressive behavior. The aim of the study was to summarize and integrate the role of nerves in the pancreatic tumor microenvironment. The role of autonomic nerve fibers on PDAC development has been recently studied, which resulted in considering the targeting of sympathetic and parasympathetic pathways as a novel treatment opportunity. Perineural invasion (PNI) is commonly found in PDAC. As the severity of the PNI correlates with a poorer prognosis, new quantification of this phenomenon, distinguishing between perineural and endoneural invasion, could feature in routine pathological examination. The concepts of cancer-related neurogenesis and axonogenesis in PDAC are understudied; so, further research in this field may be warranted. A better understanding of the interdependence between the neural component and cancer cells in the PDAC microenvironment could bring new nerve-oriented treatment options into clinical practice and improve outcomes in patients with pancreatic cancer. In this review, we aim to summarize and integrate the current state of knowledge and future challenges concerning nerve-cancer interactions in PDAC.
Collapse
Affiliation(s)
- Michał Gola
- Department of Human Histology and Embryology, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Aleksandra Sejda
- Department of Pathomorphology and Forensic Medicine, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 18 Żołnierska Street, 10-561 Olsztyn, Poland
| | - Janusz Godlewski
- Department of Human Histology and Embryology, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Małgorzata Cieślak
- Department of Pathomorphology and Forensic Medicine, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 18 Żołnierska Street, 10-561 Olsztyn, Poland
| | - Anna Starzyńska
- Department of Oral Surgery, Medical University of Gdańsk, 7 Dębinki Street, 80-211 Gdańsk, Poland
| |
Collapse
|
21
|
Liu Y, Tian S, Ning B, Huang T, Li Y, Wei Y. Stress and cancer: The mechanisms of immune dysregulation and management. Front Immunol 2022; 13:1032294. [PMID: 36275706 PMCID: PMC9579304 DOI: 10.3389/fimmu.2022.1032294] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Advances in the understanding of psychoneuroimmunology in the past decade have emphasized the notion that stress and cancer are interlinked closely. Durable chronic stress accelerated tumorigenesis and progression, which is unfavorable for clinical outcomes of cancer patients. Available evidence has provided unprecedented knowledge about the role and mechanisms of chronic stress in carcinogenesis, the most well-known one is dysfunction of the hypothalamus-pituitary-adrenal (HPA) axis and the sympathetic nervous system (SNS). With abnormal activation of neuroendocrine system, stress-related hormones contribute to increased oncogenes expression, exacerbated chronic inflammation and impaired immunologic function. In addition, accumulating studies have demonstrated that diverse stress interventions including pharmacological approaches, physical exercises and psychological relaxation have been administered to assist in mental disorders reduction and life quality improvement in cancer patients. In this review, we systematically summarize the connection and mechanisms in the stress-immune-cancer axis identified by animal and clinical studies, as well as conclude the effectiveness and deficiencies of existing stress management strategies.
Collapse
Affiliation(s)
- Yixin Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Sheng Tian
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Biao Ning
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Tianhe Huang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yi Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Cong H, Gao J, Wang Q, Du M, Li H, Li Q, Li J, Liang Y, Zhao D, Yang H, Gan Y, Tu H. Increased Expression of Mitochondrial UQCRC1 in Pancreatic Cancer Impairs Antitumor Immunity of Natural Killer Cells via Elevating Extracellular ATP. Front Oncol 2022; 12:872017. [PMID: 35769718 PMCID: PMC9234308 DOI: 10.3389/fonc.2022.872017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most lethal malignancies characterized by a highly immunosuppressive tumor microenvironment (TME). Previously, we have reported that ubiquinol-cytochrome c reductase core protein I (UQCRC1), a key component of mitochondrial complex III, is generally upregulated in PC and produces extracellular ATP (eATP) to promote PC progression. Here, we sought to investigate whether the oncogenic property of UQCRC1 is generated through its effects on natural killer (NK) cells in the TME. We found that UQCRC1 overexpression in PC cells inhibited cytotoxicity of NK cells, as well as the infiltration of NK cells toward PC, whereas knockdown of UQCRC1 enhanced the cytotoxicity and chemotaxis of NK cells. Adoptive NK cell therapy in the subcutaneous mouse model and CIBERSORTx analysis with human PC specimens confirmed UQCRC1 elicited immunosuppressive effects on NK cells. Such UQCRC1-induced impairment of NK cells was mediated by eATP and its metabolite adenosine via P2Y11R and A2AR, respectively. Mechanistically, we found the UQCRC1/eATP axis reduced the expression of chemokine CCL5 in cancer cells and altered the balance of activating receptor DNAM-1 and inhibitory receptor CD96 on NK-92MI cells, resulting in decreased chemotaxis and exhausted phenotype of NK-92MI cells. Taken together, our study provides the evidence to support a novel mechanism by which energy metabolism change in cancer cells remodels the TME and impedes NK cell surveillance. It also suggests that targeting UQCRC1 may be a potential combined strategy for PC immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Yu Gan
- *Correspondence: Hong Tu, ; Yu Gan,
| | - Hong Tu
- *Correspondence: Hong Tu, ; Yu Gan,
| |
Collapse
|
23
|
Bai PY, Chen SQ, Jia DL, Pan LH, Liu CB, Liu J, Luo W, Yang Y, Sun MY, Wan NF, Rong WW, Sun AJ, Ge JB. Environmental eustress improves postinfarction cardiac repair via enhancing cardiac macrophage survival. SCIENCE ADVANCES 2022; 8:eabm3436. [PMID: 35476440 PMCID: PMC9045726 DOI: 10.1126/sciadv.abm3436] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 03/10/2022] [Indexed: 05/24/2023]
Abstract
Macrophages play a vital role in cardiac repair following myocardial infarction (MI). An enriched environment (EE) is involved in the regulation of macrophage-related activities and disease progression; however, whether EE affects the phenotype and function of macrophages to improve postinfarction cardiac repair remains unknown. In this study, we found that EE improved cardiac function, decreased mortality, and ameliorated adverse ventricular remodeling in mice after MI, with these outcomes closely related to the increased survival of Ly6Clow macrophages and their CCR2-MHCIIlow subsets. EE increased the expression of brain-derived neurotrophic factor (BDNF) in the hypothalamus, leading to higher circulating levels of BDNF, which, in turn, regulated the cardiac macrophages. BDNF bound to tropomyosin receptor kinase B to activate downstream ERK1/2 and AKT pathways, promoting macrophage survival. These findings demonstrate that EE optimizes postinfarction cardiac repair and highlights the significance of EE as a previously unidentified strategy for impeding adverse ventricular remodeling.
Collapse
Affiliation(s)
- Pei-Yuan Bai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Si-Qin Chen
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Dai-Le Jia
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Li-Hong Pan
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chao-Bao Liu
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Wei Luo
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yang Yang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ma-Yu Sun
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nai-Fu Wan
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wu-Wei Rong
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ai-Jun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jun-Bo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Guillot J, Dominici C, Lucchesi A, Nguyen HTT, Puget A, Hocine M, Rangel-Sosa MM, Simic M, Nigri J, Guillaumond F, Bigonnet M, Dusetti N, Perrot J, Lopez J, Etzerodt A, Lawrence T, Pudlo P, Hubert F, Scoazec JY, van de Pavert SA, Tomasini R, Chauvet S, Mann F. Sympathetic axonal sprouting induces changes in macrophage populations and protects against pancreatic cancer. Nat Commun 2022; 13:1985. [PMID: 35418199 PMCID: PMC9007988 DOI: 10.1038/s41467-022-29659-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 03/23/2022] [Indexed: 01/06/2023] Open
Abstract
Neuronal nerve processes in the tumor microenvironment were highlighted recently. However, the origin of intra-tumoral nerves remains poorly known, in part because of technical difficulties in tracing nerve fibers via conventional histological preparations. Here, we employ three-dimensional (3D) imaging of cleared tissues for a comprehensive analysis of sympathetic innervation in a murine model of pancreatic ductal adenocarcinoma (PDAC). Our results support two independent, but coexisting, mechanisms: passive engulfment of pre-existing sympathetic nerves within tumors plus an active, localized sprouting of axon terminals into non-neoplastic lesions and tumor periphery. Ablation of the innervating sympathetic nerves increases tumor growth and spread. This effect is explained by the observation that sympathectomy increases intratumoral CD163+ macrophage numbers, which contribute to the worse outcome. Altogether, our findings provide insights into the mechanisms by which the sympathetic nervous system exerts cancer-protective properties in a mouse model of PDAC.
Collapse
Affiliation(s)
| | | | | | - Huyen Thi Trang Nguyen
- Aix Marseille Univ, CNRS, IBDM, Marseille, France
- University of Science and Technology of Hanoi (USTH), VAST, 18 Hoang Quoc Viet, Hanoi, Vietnam
| | | | | | | | - Milesa Simic
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Jérémy Nigri
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Fabienne Guillaumond
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Martin Bigonnet
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Nelson Dusetti
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Jimmy Perrot
- Department of Anatomopathology, Lyon Sud University Hospital, Hospices Civils de Lyon, Lyon, France
| | - Jonathan Lopez
- Department of Biochemistry and Molecular Biology, Lyon Sud University Hospital, Hospices Civils de Lyon, Lyon, France
- Faculty of Medicine Lyon-Est, Lyon 1 University, Université de Lyon, Lyon, France
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR5286, Lyon, France
| | - Anders Etzerodt
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
- Department of Biomedecine, Aarhus University, Aarhus, Denmark
| | - Toby Lawrence
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Pierre Pudlo
- Aix Marseille Univ, CNRS, Centrale Marseille, I2M, Marseille, France
| | - Florence Hubert
- Aix Marseille Univ, CNRS, Centrale Marseille, I2M, Marseille, France
| | - Jean-Yves Scoazec
- Department of Pathology, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Richard Tomasini
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | | | - Fanny Mann
- Aix Marseille Univ, CNRS, IBDM, Marseille, France.
| |
Collapse
|
25
|
The Nervous System Contributes to the Tumorigenesis and Progression of Human Digestive Tract Cancer. J Immunol Res 2022; 2022:9595704. [PMID: 35295188 PMCID: PMC8920690 DOI: 10.1155/2022/9595704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/09/2022] [Accepted: 02/14/2022] [Indexed: 11/17/2022] Open
Abstract
Tumors of the gastrointestinal tract are one of the highest incidences of morbidity and mortality in humans. Recently, a growing number of researchers have indicated that nerve fibers and nerve signals participate in tumorigenesis. The current overarching view based on the responses to therapy revealed that tumors are partly promoted by the tumor microenvironment (TME), endogenous oncogenic factors, and complex systemic processes. Homeostasis of the neuroendocrine-immune axis (NEI axis) maintains a healthy in vivo environment in humans, and dysfunction of the axis contributes to various cancers, including the digestive tract. Interestingly, nerves might promote tumor development via multiple mechanisms, including perineural invasion (PNI), central level regulation, NEI axis effect, and neurotransmitter induction. This review focuses on the association between digestive tumors and nerve regulation, including PNI, the NEI axis, stress, and neurotransmitters, as well as on the potential clinical application of neurotherapy, aiming to provide a new perspective on the management of digestive cancers.
Collapse
|
26
|
de León-Guerrero SD, Salazar-León J, Meza-Sosa KF, Valle-Garcia D, Aguilar-León D, Pedraza-Alva G, Pérez-Martínez L. An enriched environment reestablishes metabolic homeostasis by reducing obesity-induced inflammation. Dis Model Mech 2022; 15:274225. [PMID: 35112705 PMCID: PMC9227715 DOI: 10.1242/dmm.048936] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 12/09/2021] [Indexed: 11/20/2022] Open
Abstract
Obesity can lead to chronic inflammation in different tissues, generating insulin and leptin resistance and alterations in glucose and lipid metabolism, favoring the development of degenerative diseases, including type II diabetes. Congruently, the inflammatory signaling inhibition prevents the development of obesity and restores insulin sensitivity. Via the enhancement of central nervous system activity, an enriched environment (EE) has beneficial effects on learning and memory as well as on immune cell functions and inflammation in different disease models. Here, we explored whether an EE can restore energy balance in obese mice that previously presented metabolic alterations. We discovered that an EE improved glucose metabolism, increased insulin signaling in liver, and reduced hepatic steatosis and inflammation, and increased lipolysis and browning in the white adipose tissue of high-fat diet (HFD)-fed mice. Finally, we found reduced inflammatory signaling and increased anorexigenic signaling in the hypothalamus of HFD-fed mice exposed to an EE. These data indicate that an EE is able to restore the metabolic imbalance caused by HFD feeding. Thus, we propose EE as a novel therapeutic approach for treating obesity-related metabolic alterations. This article has an associated First Person interview with the first author of the paper. Summary: A series of physiological, histochemical and molecular analyses reveal that enriched environment decreases inflammation in adipose tissue and in hypothalamus, re-establishing glucose metabolism in metabolically compromised mice.
Collapse
Affiliation(s)
- Sol Díaz de León-Guerrero
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| | - Jonathan Salazar-León
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| | - Karla F Meza-Sosa
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| | - David Valle-Garcia
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| | - Diana Aguilar-León
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Tlalpan, Ciudad de México, CP 14000, México
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| |
Collapse
|
27
|
Liu HM, Ma LL, Li C, Cao B, Jiang Y, Han L, Xu R, Lin J, Zhang D. The molecular mechanism of chronic stress affecting the occurrence and development of breast cancer and potential drug therapy. Transl Oncol 2021; 15:101281. [PMID: 34875482 PMCID: PMC8652015 DOI: 10.1016/j.tranon.2021.101281] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022] Open
Abstract
According to the 2020 data released by the International Agency for Research on Cancer, breast cancer has surpassed lung cancer as the world's most newly diagnosed first-time cancer. Compared with patients with other types of cancer, those with breast cancer experience greater mental stress and more severe psychological impacts because of the life-threatening diagnosis, physical changes, treatment side effects, and family and social life dysfunctions. These usually manifest as anxiety, depression, nervousness, and insomnia, all of which elicit stress responses. Particularly under chronic stress, the continuous release of neurotransmitters from the neuroendocrine system can have a highly profound impact on the occurrence and prognosis of breast cancer. However, because of the complex mechanisms underlying chronic stress and the variability in individual tolerance, evidence of the role of chronic stress in the occurrence and evolution of breast cancer remains unclear. This article reviewed previous research on the correlation between chronic stress and the occurrence and development of breast cancer, particularly the molecular mechanism through which chronic stress promotes breast cancer via neurotransmitters secreted by the nervous system. We also review the progress in the development of potential drugs or blockers for the treatment of breast cancer by targeting the neuroendocrine system.
Collapse
Affiliation(s)
- Hui-Min Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, No. 1066 Avenue Liutai, Chengdu 611137, China
| | - Le-le Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, No. 1066 Avenue Liutai, Chengdu 611137, China
| | - Chunyu Li
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bo Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, No. 1066 Avenue Liutai, Chengdu 611137, China; National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yifang Jiang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Road Shierqiao, Chengdu 610072, China
| | - Li Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, No. 1066 Avenue Liutai, Chengdu 611137, China
| | - Runchun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, No. 1066 Avenue Liutai, Chengdu 611137, China.
| | - Junzhi Lin
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Road Shierqiao, Chengdu 610072, China.
| | - Dingkun Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, No. 1066 Avenue Liutai, Chengdu 611137, China.
| |
Collapse
|
28
|
Tian W, Liu Y, Cao C, Zeng Y, Pan Y, Liu X, Peng Y, Wu F. Chronic Stress: Impacts on Tumor Microenvironment and Implications for Anti-Cancer Treatments. Front Cell Dev Biol 2021; 9:777018. [PMID: 34869378 PMCID: PMC8640341 DOI: 10.3389/fcell.2021.777018] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic stress is common among cancer patients due to the psychological, operative, or pharmaceutical stressors at the time of diagnosis or during the treatment of cancers. The continuous activations of the hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic nervous system (SNS), as results of chronic stress, have been demonstrated to take part in several cancer-promoting processes, such as tumorigenesis, progression, metastasis, and multi-drug resistance, by altering the tumor microenvironment (TME). Stressed TME is generally characterized by the increased proportion of cancer-promoting cells and cytokines, the reduction and malfunction of immune-supportive cells and cytokines, augmented angiogenesis, enhanced epithelial-mesenchymal transition, and damaged extracellular matrix. For the negative effects that these alterations can cause in terms of the efficacies of anti-cancer treatments and prognosis of patients, supplementary pharmacological or psychotherapeutic strategies targeting HPA, SNS, or psychological stress may be effective in improving the prognosis of cancer patients. Here, we review the characteristics and mechanisms of TME alterations under chronic stress, their influences on anti-cancer therapies, and accessory interventions and therapies for stressed cancer patients.
Collapse
Affiliation(s)
- Wentao Tian
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China
| | - Yi Liu
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Chenghui Cao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China
| | - Yue Zeng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yue Pan
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaohan Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yurong Peng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fang Wu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Cancer Mega-Data Intelligent Application and Engineering Research Centre, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
29
|
Eckerling A, Ricon-Becker I, Sorski L, Sandbank E, Ben-Eliyahu S. Stress and cancer: mechanisms, significance and future directions. Nat Rev Cancer 2021; 21:767-785. [PMID: 34508247 DOI: 10.1038/s41568-021-00395-5] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/23/2021] [Indexed: 02/08/2023]
Abstract
The notion that stress and cancer are interlinked has dominated lay discourse for decades. More recent animal studies indicate that stress can substantially facilitate cancer progression through modulating most hallmarks of cancer, and molecular and systemic mechanisms mediating these effects have been elucidated. However, available clinical evidence for such deleterious effects is inconsistent, as epidemiological and stress-reducing clinical interventions have yielded mixed effects on cancer mortality. In this Review, we describe and discuss specific mediating mechanisms identified by preclinical research, and parallel clinical findings. We explain the discrepancy between preclinical and clinical outcomes, through pointing to experimental strengths leveraged by animal studies and through discussing methodological and conceptual obstacles that prevent clinical studies from reflecting the impacts of stress. We suggest approaches to circumvent such obstacles, based on targeting critical phases of cancer progression that are more likely to be stress-sensitive; pharmacologically limiting adrenergic-inflammatory responses triggered by medical procedures; and focusing on more vulnerable populations, employing personalized pharmacological and psychosocial approaches. Recent clinical trials support our hypothesis that psychological and/or pharmacological inhibition of excess adrenergic and/or inflammatory stress signalling, especially alongside cancer treatments, could save lives.
Collapse
Affiliation(s)
- Anabel Eckerling
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Itay Ricon-Becker
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Liat Sorski
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Elad Sandbank
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shamgar Ben-Eliyahu
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
30
|
Sakama S, Kurusu K, Morita M, Oizumi T, Masugata S, Oka S, Yokomizo S, Nishimura M, Morioka T, Kakinuma S, Shimada Y, Nakamura AJ. An Enriched Environment Alters DNA Repair and Inflammatory Responses After Radiation Exposure. Front Immunol 2021; 12:760322. [PMID: 34745135 PMCID: PMC8570081 DOI: 10.3389/fimmu.2021.760322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/08/2021] [Indexed: 11/22/2022] Open
Abstract
After the Fukushima Daiichi Nuclear Power Plant accident, there is growing concern about radiation-induced carcinogenesis. In addition, living in a long-term shelter or temporary housing due to disasters might cause unpleasant stress, which adversely affects physical and mental health. It's been experimentally demonstrated that "eustress", which is rich and comfortable, has beneficial effects for health using mouse models. In a previous study, mice raised in the enriched environment (EE) has shown effects such as suppression of tumor growth and enhancement of drug sensitivity during cancer treatment. However, it's not yet been evaluated whether EE affects radiation-induced carcinogenesis. Therefore, to evaluate whether EE suppresses a radiation-induced carcinogenesis after radiation exposure, in this study, we assessed the serum leptin levels, radiation-induced DNA damage response and inflammatory response using the mouse model. In brief, serum and tissues were collected and analyzed over time in irradiated mice after manipulating the raising environment during the juvenile or adult stage. To assess the radiation-induced DNA damage response, we performed immunostaining for phosphorylated H2AX which is a marker of DNA double-strand break. Focusing on the polarization of macrophages in the inflammatory reaction that has an important role in carcinogenesis, we performed analysis using tissue immunofluorescence staining and RT-qPCR. Our data confirmed that EE breeding before radiation exposure improved the responsiveness to radiation-induced DNA damage and basal immunity, further suppressing the chronic inflammatory response, and that might lead to a reduction of the risk of radiation-induced carcinogenesis.
Collapse
Affiliation(s)
- Sae Sakama
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Keisuke Kurusu
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Mayu Morita
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Takashi Oizumi
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Shinya Masugata
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Shohei Oka
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Shinya Yokomizo
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Mayumi Nishimura
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Takamitsu Morioka
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Yoshiya Shimada
- Executive Director, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Asako J. Nakamura
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| |
Collapse
|
31
|
Liu J, Zheng J, Xu Y, Cao W, Wang J, Wang B, Zhao L, Zhang X, Liao W. Enriched Environment Attenuates Pyroptosis to Improve Functional Recovery After Cerebral Ischemia/Reperfusion Injury. Front Aging Neurosci 2021; 13:717644. [PMID: 34646128 PMCID: PMC8504677 DOI: 10.3389/fnagi.2021.717644] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/01/2021] [Indexed: 12/16/2022] Open
Abstract
Enriched environment (EE) is a complex containing social, cognitive, and motor stimuli. Exposure to EE can promote functional recovery after ischemia/reperfusion (I/R) injury. However, the underlying mechanisms remained unclear. Pyroptosis has recently been identified and demonstrated a significant role in ischemic stroke. The purpose of this study was to explore the effect of EE on neuronal pyroptosis after cerebral I/R injury. In the current study, middle cerebral artery occlusion/reperfusion (MCAO/R) was applied to establish the cerebral I/R injury model. Behavior tests including the modified Neurological Severity Scores (mNSS) and the Morris Water Maze (MWM) were performed. The infarct volume was evaluated by Nissl staining. To evaluate the levels of pyroptosis-related proteins, the levels of GSDMD-N and nod-like receptor protein 1/3 (NLRP1/3) inflammasome-related proteins were examined. The mRNA levels of IL-1β and IL-18 were detected by Quantitative Real-Time PCR (qPCR). The secretion levels of IL-1β and IL-18 were analyzed by ELISA. Also, the expression of p65 and p-p65 were detected. The results showed that EE treatment improved functional recovery, reduced infarct volume, attenuated neuronal pyroptosis after cerebral I/R injury. EE treatment also suppressed the activities of NLRP1/NLRP3 inflammasomes. These may be affected by inhabiting the NF-κB p65 signaling pathway. Our findings suggested that neuronal pyroptosis was probably the neuroprotective mechanism that EE treatment rescued neurological deficits after I/R injury.
Collapse
Affiliation(s)
- Jingying Liu
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Zheng
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yang Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenyue Cao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jinchen Wang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Biru Wang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Linyao Zhao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Zhang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weijing Liao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
32
|
Liu C, Yang Y, Chen C, Li L, Li J, Wang X, Chu Q, Qiu L, Ba Q, Li X, Wang H. Environmental eustress modulates β-ARs/CCL2 axis to induce anti-tumor immunity and sensitize immunotherapy against liver cancer in mice. Nat Commun 2021; 12:5725. [PMID: 34593796 PMCID: PMC8484272 DOI: 10.1038/s41467-021-25967-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 09/01/2021] [Indexed: 01/22/2023] Open
Abstract
Although psycho-social stress is a well-known factor that contributes to the development of cancer, it remains largely unclear whether and how environmental eustress influences malignant diseases and regulates cancer-related therapeutic responses. Using an established eustress model, we demonstrate that mice living in an enriched environment (EE) are protected from carcinogen-induced liver neoplasia and transplantable syngeneic liver tumors, owning to a CD8+ T cell-dependent tumor control. We identify a peripheral Neuro-Endocrine-Immune pathway in eustress, including Sympathetic nervous system (SNS)/β-adrenergic receptors (β-ARs)/CCL2 that relieves tumor immunosuppression and overcomes PD-L1 resistance to immunotherapy. Notably, EE activates peripheral SNS and β-ARs signaling in tumor cells and tumor infiltrated myeloid cells, leading to suppression of CCL2 expression and activation of anti-tumor immunity. Either blockade of CCL2/CCR2 or β-AR signaling in EE mice lose the tumor protection capability. Our study reveales that environmental eustress via EE stimulates anti-tumor immunity, resulting in more efficient tumor control and a better outcome of immunotherapy.
Collapse
Affiliation(s)
- Chaobao Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yang Yang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Cheng Chen
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ling Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jingquan Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaonan Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qiao Chu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lin Qiu
- CAS Key Laboratory of Nutrition, Metabolism and Food safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qian Ba
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoguang Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,CAS Key Laboratory of Nutrition, Metabolism and Food safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
33
|
Wang J, Chen Y, Li X, Zou X. Perineural Invasion and Associated Pain Transmission in Pancreatic Cancer. Cancers (Basel) 2021; 13:4594. [PMID: 34572820 PMCID: PMC8467801 DOI: 10.3390/cancers13184594] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/10/2021] [Accepted: 09/10/2021] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the cancers with the highest incidence of perineural invasion (PNI), which often indicates a poor prognosis. Aggressive tumor cells invade nerves, causing neurogenic inflammation; the tumor microenvironment also induces nerves to undergo a series of structural and functional reprogramming. In turn, neurons and the surrounding glial cells promote the development of pancreatic cancer through autocrine and/or paracrine signaling. In addition, hyperalgesia in PDAC patients implies alterations of pain transmission in the peripheral and central nervous systems. Currently, the studies on this topic are relatively limited. This review will elaborate on the mechanisms of tumor-neural interactions and its possible relationship with pain from several aspects that have been focused on in recent years.
Collapse
Affiliation(s)
| | | | | | - Xiaoping Zou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China; (J.W.); (Y.C.); (X.L.)
| |
Collapse
|
34
|
Mormino A, Bernardini G, Cocozza G, Corbi N, Passananti C, Santoni A, Limatola C, Garofalo S. Enriched Environment Cues Suggest a New Strategy to Counteract Glioma: Engineered rAAV2-IL-15 Microglia Modulate the Tumor Microenvironment. Front Immunol 2021; 12:730128. [PMID: 34552593 PMCID: PMC8450436 DOI: 10.3389/fimmu.2021.730128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/13/2021] [Indexed: 12/16/2022] Open
Abstract
Several types of cancer grow differently depending on the environmental stimuli they receive. In glioma, exposure to an enriched environment (EE) increases the overall survival rate of tumor-bearing mice, acting on the cells that participate to define the tumor microenvironment. In particular, environmental cues increase the microglial production of interleukin (IL)-15 which promotes a pro-inflammatory (antitumor) phenotype of microglia and the cytotoxic activity of natural killer (NK) cells, counteracting glioma growth, thus representing a virtuous mechanism of interaction between NK cells and microglia. To mimic the effect of EE on glioma, we investigated the potential of creating engineered microglia as the source of IL-15 in glioma. We demonstrated that microglia modified with recombinant adeno-associated virus serotype 2 (rAAV2) carrying IL-15 (rAAV2-IL-15), to force the production of IL-15, are able to increase the NK cells viability in coculture. Furthermore, the intranasal delivery of rAAV2-IL-15 microglia triggered the interplay with NK cells in vivo, enhancing NK cell recruitment and pro-inflammatory microglial phenotype in tumor mass of glioma-bearing mice, and ultimately counteracted tumor growth. This approach has a high potential for clinical translatability, highlighting the therapeutic efficacy of forced IL-15 production in microglia: the delivery of engineered rAAV2-IL-15 microglia to boost the immune response paves the way to design a new perspective therapy for glioma patients.
Collapse
Affiliation(s)
- Alessandro Mormino
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Giovanni Bernardini
- Department of Molecular Medicine, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University, Rome, Italy
| | - Germana Cocozza
- Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Nicoletta Corbi
- Department of Molecular Medicine, CNR-Institute of Molecular Biology and Pathology, Sapienza University, Rome, Italy
| | - Claudio Passananti
- Department of Molecular Medicine, CNR-Institute of Molecular Biology and Pathology, Sapienza University, Rome, Italy
| | - Angela Santoni
- Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
| | - Cristina Limatola
- Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University, Rome, Italy
| | - Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| |
Collapse
|
35
|
Liang Y, Li H, Gan Y, Tu H. Shedding Light on the Role of Neurotransmitters in the Microenvironment of Pancreatic Cancer. Front Cell Dev Biol 2021; 9:688953. [PMID: 34395421 PMCID: PMC8363299 DOI: 10.3389/fcell.2021.688953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/13/2021] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy with a 5-year survival rate of less than 8%. The fate of PC is determined not only by the malignant behavior of the cancer cells, but also by the surrounding tumor microenvironment (TME), consisting of various cellular (cancer cells, immune cells, stromal cells, endothelial cells, and neurons) and non-cellular (cytokines, neurotransmitters, and extracellular matrix) components. The pancreatic TME has the unique characteristic of exhibiting increased neural density and altered microenvironmental concentration of neurotransmitters. The neurotransmitters, produced by both neuron and non-neuronal cells, can directly regulate the biological behavior of PC cells via binding to their corresponding receptors on tumor cells and activating the intracellular downstream signals. On the other hand, the neurotransmitters can also communicate with other cellular components such as the immune cells in the TME to promote cancer growth. In this review, we will summarize the pleiotropic effects of neurotransmitters on the initiation and progression of PC, and particularly discuss the emerging mechanisms of how neurotransmitters influence the innate and adaptive immune responses in the TME in an autocrine or paracrine manner. A better understanding of the interplay between neurotransmitters and the immune cells in the TME might facilitate the development of new effective therapies for PC.
Collapse
Affiliation(s)
| | | | - Yu Gan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Tu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Xiao R, Ali S, Caligiuri MA, Cao L. Enhancing Effects of Environmental Enrichment on the Functions of Natural Killer Cells in Mice. Front Immunol 2021; 12:695859. [PMID: 34394087 PMCID: PMC8355812 DOI: 10.3389/fimmu.2021.695859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/14/2021] [Indexed: 01/02/2023] Open
Abstract
The environment of an organism can convey a powerful influence over its biology. Environmental enrichment (EE), as a eustress model, has been used extensively in neuroscience to study neurogenesis and brain plasticity. EE has also been used as an intervention for the treatment and prevention of neurological and psychiatric disorders with limited clinical application. By contrast, the effects of EE on the immune system are relatively less investigated. Recently, accumulating evidence has demonstrated that EE can robustly impact immune function. In this review, we summarize the major components of EE, the impact of EE on natural killer (NK) cells, EE's immunoprotective roles in cancer, and the underlying mechanisms of EE-induced NK cell regulation. Moreover, we discuss opportunities for translational application based on insights from animal research of EE-induced NK cell regulation.
Collapse
Affiliation(s)
- Run Xiao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, United States
| | - Seemaab Ali
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, United States
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, United States
| | - Michael A. Caligiuri
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center and the Beckman Research Institute, Los Angeles, CA, United States
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, United States
| |
Collapse
|
37
|
Bergin SM, Xiao R, Huang W, Judd CRT, Liu X, Mansour AG, Queen N, Widstrom KJ, Caligiuri MA, Cao L. Environmental activation of a hypothalamic BDNF-adipocyte IL-15 axis regulates adipose-natural killer cells. Brain Behav Immun 2021; 95:477-488. [PMID: 33989745 PMCID: PMC8493653 DOI: 10.1016/j.bbi.2021.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/21/2021] [Accepted: 05/09/2021] [Indexed: 12/21/2022] Open
Abstract
Physical and social environments influence immune homeostasis within adipose tissue, yet the mechanisms remain poorly defined. We report that an enriched environment (EE) housing modulates the immune cell population in white adipose tissue of mice including an increase in the abundance of natural killer (NK) cells. EE upregulates the expression of IL-15 and its receptor IL-15Rα specifically within mature adipocytes. Mechanistically, we show that hypothalamic brain-derived neurotrophic factor (BDNF) upregulates IL-15 production in adipocytes via sympathetic β-adrenergic signaling. Overexpressing BDNF mediated by recombinant adeno-associated virus (rAAV) vector in the hypothalamus expands adipose NK cells. Conversely, inhibition of hypothalamic BDNF signaling via gene transfer of a dominant negative TrkB receptor suppresses adipose NK cells. In white adipose tissue, overexpression of IL-15 using an adipocyte-specific rAAV vector stimulates adipose NK cells and inhibits the progression of subcutaneous melanoma, whereas local IL-15 knockdown blocks the EE effect. These results suggest that bio-behavioral factors regulate adipose NK cells via a hypothalamic BDNF-sympathoneural-adipocyte IL-15 axis. Targeting this pathway may have therapeutic significance for cancer.
Collapse
Affiliation(s)
- Stephen M Bergin
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, United States; Medical Scientist Training Program, College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Run Xiao
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, United States; Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Wei Huang
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, United States; Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - C Ryan T Judd
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, United States
| | - Xianglan Liu
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, United States; Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Anthony G Mansour
- Department of Hematological Malignancies and Stem Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, United States
| | - Nicholas Queen
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, United States; Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Kyle J Widstrom
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, United States; Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Michael A Caligiuri
- Department of Hematological Malignancies and Stem Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, United States.
| | - Lei Cao
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, United States; Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
38
|
Carvalho-Paulo D, Bento Torres Neto J, Filho CS, de Oliveira TCG, de Sousa AA, dos Reis RR, dos Santos ZA, de Lima CM, de Oliveira MA, Said NM, Freitas SF, Sosthenes MCK, Gomes GF, Henrique EP, Pereira PDC, de Siqueira LS, de Melo MAD, Guerreiro Diniz C, Magalhães NGDM, Diniz JAP, Vasconcelos PFDC, Diniz DG, Anthony DC, Sherry DF, Brites D, Picanço Diniz CW. Microglial Morphology Across Distantly Related Species: Phylogenetic, Environmental and Age Influences on Microglia Reactivity and Surveillance States. Front Immunol 2021; 12:683026. [PMID: 34220831 PMCID: PMC8250867 DOI: 10.3389/fimmu.2021.683026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/31/2021] [Indexed: 12/16/2022] Open
Abstract
Microglial immunosurveillance of the brain parenchyma to detect local perturbations in homeostasis, in all species, results in the adoption of a spectrum of morphological changes that reflect functional adaptations. Here, we review the contribution of these changes in microglia morphology in distantly related species, in homeostatic and non-homeostatic conditions, with three principal goals (1): to review the phylogenetic influences on the morphological diversity of microglia during homeostasis (2); to explore the impact of homeostatic perturbations (Dengue virus challenge) in distantly related species (Mus musculus and Callithrix penicillata) as a proxy for the differential immune response in small and large brains; and (3) to examine the influences of environmental enrichment and aging on the plasticity of the microglial morphological response following an immunological challenge (neurotropic arbovirus infection). Our findings reveal that the differences in microglia morphology across distantly related species under homeostatic condition cannot be attributed to the phylogenetic origin of the species. However, large and small brains, under similar non-homeostatic conditions, display differential microglial morphological responses, and we argue that age and environment interact to affect the microglia morphology after an immunological challenge; in particular, mice living in an enriched environment exhibit a more efficient immune response to the virus resulting in earlier removal of the virus and earlier return to the homeostatic morphological phenotype of microglia than it is observed in sedentary mice.
Collapse
Affiliation(s)
- Dario Carvalho-Paulo
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - João Bento Torres Neto
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
- Faculdade de Fisioterapia e Terapia Ocupacional, Universidade Federal do Pará, Belém, Brazil
| | - Carlos Santos Filho
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Thais Cristina Galdino de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Aline Andrade de Sousa
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Renata Rodrigues dos Reis
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Zaire Alves dos Santos
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Camila Mendes de Lima
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Marcus Augusto de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Nivin Mazen Said
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Sinara Franco Freitas
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Marcia Consentino Kronka Sosthenes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Giovanni Freitas Gomes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Ediely Pereira Henrique
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação Ciência e Tecnologia do Pará, Bragança, Brazil
| | - Patrick Douglas Côrrea Pereira
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação Ciência e Tecnologia do Pará, Bragança, Brazil
| | - Lucas Silva de Siqueira
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação Ciência e Tecnologia do Pará, Bragança, Brazil
| | - Mauro André Damasceno de Melo
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação Ciência e Tecnologia do Pará, Bragança, Brazil
| | - Cristovam Guerreiro Diniz
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação Ciência e Tecnologia do Pará, Bragança, Brazil
| | - Nara Gyzely de Morais Magalhães
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação Ciência e Tecnologia do Pará, Bragança, Brazil
| | | | - Pedro Fernando da Costa Vasconcelos
- Dep. de Arbovirologia e Febres Hemorrágicas, Instituto Evandro Chagas, Belém, Brazil
- Departamento de Patologia, Universidade do Estado do Pará, Belém, Brazil
| | - Daniel Guerreiro Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Brazil
| | | | - David Francis Sherry
- Department of Psychology, Advanced Facility for Avian Research, University of Western Ontario, London, ON, Canada
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| |
Collapse
|
39
|
Chai ZT, Zhang XP, Ao JY, Zhu XD, Wu MC, Lau WY, Sun HC, Cheng SQ. AXL Overexpression in Tumor-Derived Endothelial Cells Promotes Vessel Metastasis in Patients With Hepatocellular Carcinoma. Front Oncol 2021; 11:650963. [PMID: 34123800 PMCID: PMC8191462 DOI: 10.3389/fonc.2021.650963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/28/2021] [Indexed: 12/20/2022] Open
Abstract
Portal vein tumor thrombus (PVTT) is one of the most serious forms of hepatocellular carcinoma (HCC) vessel metastasis and has a poor survival rate. However, the molecular mechanism of PVTT has not yet been elucidated. In this study, the molecular mechanism of AXL expressed in tumor-derived endothelial cells (TECs) in vessel metastasis was investigated. High AXL expression was observed in TECs, but not in the tumor cells of HCC patients with PVTT and this was associated with poor overall survival (OS) and disease-free survival (DFS). AXL overexpression was positively associated with CD 31 expression both in vitro and in vivo. AXL promoted the cell proliferation, tube formation, and migration of both TECs and normal endothelial cells (NECs). High expression of AXL in TECs promoted the cell migration, but not the proliferation of HCC cells. Further studies demonstrated that AXL promoted cell migration and tube formation through activation of the PI3K/AKT/SOX2/DKK-1 axis. AXL overexpression in HUVECs promoted tumor growth and liver or vessel metastasis of HCC in xenograft nude mice, which could be counteracted by treatment with R428, an AXL inhibitor. R428 reduced tumor growth and CD 31 expression in HCC in PDX xenograft nude mice. Therefore, AXL over-expression in TECs promotes vessel metastasis of HCC, which indicates that AXL in TECs could be a potential therapeutic target in HCC patients with PVTT.
Collapse
Affiliation(s)
- Zong-Tao Chai
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Xiu-Ping Zhang
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jian-Yang Ao
- Department of Biliary Surgery I, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Xiao-Dong Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai, China
| | - Meng-Chao Wu
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Wan Yee Lau
- Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China
| | - Hui-Chuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai, China
| | - Shu-Qun Cheng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
40
|
Norepinephrine Enhances Aerobic Glycolysis and May Act as a Predictive Factor for Immunotherapy in Gastric Cancer. J Immunol Res 2021; 2021:5580672. [PMID: 33855088 PMCID: PMC8019630 DOI: 10.1155/2021/5580672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/22/2021] [Accepted: 03/08/2021] [Indexed: 11/17/2022] Open
Abstract
Methods Monoamine neurotransmitters were detected in gastric cancer tissue and paired normal tissue, and The Cancer Genome Atlas was used to identify differentially expressed norepinephrine-degrading and synthetic enzymes. Quantitative real-time PCR and the Seahorse assay were used to determine the effect of norepinephrine on gastric cancer cell glycolysis. MAOA expression in cancer tissues was analyzed by immunohistochemistry and was compared with the patient SUVmax value of PET-CT and other clinicopathological characteristics. Results The norepinephrine levels were markedly high in gastric cancer tissue, while the norepinephrine-degrading enzymes MAOA and MAOB showed low expression. High norepinephrine levels were associated with activated glycolysis. The MAOA or MAOB expression levels in tumor tissue were closely correlated with the patient SUV max values of PET-CT and immunotherapy evaluation indices, such as PD-L1 and the microsatellite status. Conclusions Norepinephrine shows relatively higher expression in gastric cancer tissue than in normal tissue, and its expression level is associated with the glycolysis levels in patients. The norepinephrine-degrading enzymes MAOA and MAOB have significant expression differences in cancer and normal tissue, and their missing or low expression may predict immune therapy outcomes for gastric cancer patients. High norepinephrine levels with metabolic abnormalities may be more suitable for metabolic targeted therapy or immunotherapy.
Collapse
|
41
|
Mansour AG, Xiao R, Bergin SM, Huang W, Chrislip LA, Zhang J, Ali S, Queen NJ, Caligiuri MA, Cao L. Enriched environment enhances NK cell maturation through hypothalamic BDNF in male mice. Eur J Immunol 2021; 51:557-566. [PMID: 33169371 DOI: 10.1002/eji.201948358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/05/2020] [Accepted: 11/05/2019] [Indexed: 12/15/2022]
Abstract
Macroenvironmental factors, including a patient's physical and social environment, play a role in cancer risk and progression. Our previous preclinical studies have shown that the enriched environment (EE) confers anti-obesity and anti-cancer phenotypes that are associated with enhanced adaptive immunity and are mediated by brain-derived neurotrophic factor (BDNF). Natural killer (NK) cells have anti-cancer and anti-viral properties, and their absence or depletion is associated with inferior clinical outcomes. In this study, we investigated the effects of EE on NK cell maturation following their depletion. Mice living in EE displayed a higher proportion of NK cells in the spleen, bone marrow, and blood, compared to those living in the standard environment (SE). EE enhanced NK cell maturation in the spleen and was associated with upregulation of BDNF expression in the hypothalamus. Hypothalamic BDNF overexpression reproduced the EE effects on NK cell maturation in secondary lymphoid tissues. Conversely, hypothalamic BDNF knockdown blocked the EE modulation on NK cell maturation. Our results demonstrate that a bio-behavior intervention enhanced NK cell maturation and was mediated at least in part by hypothalamic BDNF.
Collapse
Affiliation(s)
- Anthony G Mansour
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center and the Beckman Research Institute, Los Angeles, CA, 91010, USA.,The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, 43210, USA
| | - Run Xiao
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, 43210, USA.,Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Stephen M Bergin
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, 43210, USA
| | - Wei Huang
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, 43210, USA.,Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Logan A Chrislip
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, 43210, USA.,Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center and the Beckman Research Institute, Los Angeles, CA, 91010, USA
| | - Seemaab Ali
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, 43210, USA.,Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Nicholas J Queen
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, 43210, USA.,Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Michael A Caligiuri
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center and the Beckman Research Institute, Los Angeles, CA, 91010, USA
| | - Lei Cao
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, 43210, USA.,Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
42
|
Hu D, Shen W, Gong C, Fang C, Yao C, Zhu X, Wang L, Zhao C, Zhu S. Grain-sized moxibustion promotes NK cell antitumour immunity by inhibiting adrenergic signalling in non-small cell lung cancer. J Cell Mol Med 2021; 25:2900-2908. [PMID: 33506637 PMCID: PMC7957214 DOI: 10.1111/jcmm.16320] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/05/2021] [Accepted: 01/09/2021] [Indexed: 12/30/2022] Open
Abstract
Lung cancer is the leading cause of cancer‐related death worldwide, and non–small cell lung cancer (NSCLC) accounts for 85% of lung cancer diagnoses. As an ancient therapy, moxibustion has been used to treat cancer‐related symptoms in clinical practice. However, its antitumour effect on NSCLC remains largely unexplored. In the present study, a Lewis lung cancer (LLC) xenograft tumour model was established, and grain‐sized moxibustion (gMoxi) was performed at the acupoint of Zusanli (ST36). Flow cytometry and RNA sequencing (RNA‐Seq) were used to access the immune cell phenotype, cytotoxicity and gene expression. PK136, propranolol and epinephrine were used for natural killer (NK) cell depletion, β‐adrenoceptor blockade and activation, respectively. Results showed that gMoxi significantly inhibited LLC tumour growth. Moreover, gMoxi significantly increased the proportion, infiltration and activation of NK cells, whereas it did not affect CD4+ and CD8+ T cells. NK cell depletion reversed gMoxi‐mediated tumour regression. LLC tumour RNA‐Seq indicated that these effects might be related to the inhibition of adrenergic signalling. Surely, β‐blocker propranolol clearly inhibited LLC tumour growth and promoted NK cells, and gMoxi no longer increased tumour regression and promoted NK cells after propranolol treatment. Epinephrine could inhibit NK cell activity, and gMoxi significantly inhibited tumour growth and promoted NK cells after epinephrine treatment. These results demonstrated that gMoxi could promote NK cell antitumour immunity by inhibiting adrenergic signalling, suggesting that gMoxi could be used as a promising therapeutic regimen for the treatment of NSCLC, and it had a great potential in NK cell–based cancer immunotherapy.
Collapse
Affiliation(s)
- Dan Hu
- School of Acupuncture, Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiming Shen
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyuan Gong
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Fang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Yao
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaowen Zhu
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lixin Wang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Zhao
- School of Acupuncture, Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shiguo Zhu
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
43
|
Anderson G, Maes M. Mitochondria and immunity in chronic fatigue syndrome. Prog Neuropsychopharmacol Biol Psychiatry 2020; 103:109976. [PMID: 32470498 DOI: 10.1016/j.pnpbp.2020.109976] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
It is widely accepted that the pathophysiology and treatment of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) could be considerably improved. The heterogeneity of ME/CFS and the confusion over its classification have undoubtedly contributed to this, although this would seem a consequence of the complexity of the array of ME/CFS presentations and high levels of diverse comorbidities. This article reviews the biological underpinnings of ME/CFS presentations, including the interacting roles of the gut microbiome/permeability, endogenous opioidergic system, immune cell mitochondria, autonomic nervous system, microRNA-155, viral infection/re-awakening and leptin as well as melatonin and the circadian rhythm. This details not only relevant pathophysiological processes and treatment options, but also highlights future research directions. Due to the complexity of interacting systems in ME/CFS pathophysiology, clarification as to its biological underpinnings is likely to considerably contribute to the understanding and treatment of other complex and poorly managed conditions, including fibromyalgia, depression, migraine, and dementia. The gut and immune cell mitochondria are proposed to be two important hubs that interact with the circadian rhythm in driving ME/CFS pathophysiology.
Collapse
Affiliation(s)
- G Anderson
- CRC Scotland & London, Eccleston Square, London, UK.
| | - M Maes
- Dept Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Dept Psychiatry, Medical University Plovdiv, Plovdiv, Bulgaria.; IMPACT Research Center, Deakin University, Geelong, Australia
| |
Collapse
|
44
|
Jiang SH, Zhang XX, Hu LP, Wang X, Li Q, Zhang XL, Li J, Gu JR, Zhang ZG. Systemic Regulation of Cancer Development by Neuro-Endocrine-Immune Signaling Network at Multiple Levels. Front Cell Dev Biol 2020; 8:586757. [PMID: 33117814 PMCID: PMC7561376 DOI: 10.3389/fcell.2020.586757] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022] Open
Abstract
The overarching view of current tumor therapies simplifies cancer to a cell-biology problem in which neoplasms are caused solely by malignant cells and the exploration of carcinogenesis and tumor progression largely focuses on somatic mutations and other genetic abnormalities of cancer cells. The limited therapeutic response indicates that cancer is driven not only by endogenous oncogenic factors and reciprocal interactions within the tumor microenvironment, but also by complex systemic processes. Homeostasis is the fundamental premise of health, and is maintained by systemic regulation of neuro-endocrine-immune axis. Cancer is also a systemic disease that manifested by dysfunction of the nervous, endocrine, and immune systems. Multiple axes of regulation exist in cancer, including central-, organ-, and microenvironment-level manipulation. At each specific regulatory level, the tridirectional communication among the nervous, endocrine, and immune factors transmit flexible signaling to induce proliferation, invasion, reprogrammed metabolism, therapeutic resistance, and other malignant phenotypes of cancer cells, resulting in the extremely poor prognosis of this lethal disease. Understanding this coordinated signaling network will enable the development of new approaches for cancer treatment via behavioral and pharmacological interventions.
Collapse
Affiliation(s)
- Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Xin Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Ren Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
45
|
Hassan QN, Queen NJ, Cao L. Regulation of aging and cancer by enhanced environmental activation of a hypothalamic-sympathoneural-adipocyte axis. Transl Cancer Res 2020; 9:5687-5699. [PMID: 33134111 PMCID: PMC7595574 DOI: 10.21037/tcr.2020.02.39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/11/2020] [Indexed: 12/20/2022]
Abstract
Social and environmental factors impact cancer and energy balance profoundly. Years ago, our lab established the existence of a novel brain-fat interaction we termed the "hypothalamic-sympathoneural-adipocyte (HSA) axis", through which complex environmental stimuli provided by an enriched environment regulate body composition, energy balance, and development of cancer. We have spent a significant portion of the past decade to further characterize the broad health benefits of an enriched environment (for example, leanness, enhanced immune function, and cancer resistance), and to identify mediators in the brain and periphery along the HSA axis. This review summarizes our recent work regarding the interface between endocrinology, immunology, cancer biology, aging, and neuroscience. We will discuss the interplay between these systemic phenomena and how the HSA axis can be targeted for regulation of cancer and aging.
Collapse
Affiliation(s)
- Quais N. Hassan
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Medical Scientist Training Program, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Nicholas J. Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
46
|
Mravec B, Tibensky M, Horvathova L. Stress and cancer. Part II: Therapeutic implications for oncology. J Neuroimmunol 2020; 346:577312. [PMID: 32652364 DOI: 10.1016/j.jneuroim.2020.577312] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023]
Abstract
Accumulated evidence has confirmed the ability of stress to promote the induction and progression of cancer (for review see Stress and cancer. Part I: Mechanisms mediating the effect of stressors on cancer). In support of this, data from clinical trials utilizing approaches that reduce stress-related signaling have shown prolonged survival of cancer patients. Therefore, the question has arisen as to how we can utilize this knowledge in the daily treatment of cancer patients. The main aim of this review is to critically analyze data from studies utilizing psychotherapy or treatment by β-blockers on the survival of cancer patients. Because these approaches, especially treatment by β-blockers, have been routinely used in clinical practice for decades in the treatment of non-cancer patients, their wider introduction into oncology might be realized in the near future.
Collapse
Affiliation(s)
- Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Slovakia; Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Miroslav Tibensky
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Slovakia
| | - Lubica Horvathova
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
47
|
Regulation of natural killer cell activity by glucocorticoids, serotonin, dopamine, and epinephrine. Cell Mol Immunol 2020; 17:705-711. [PMID: 32503998 PMCID: PMC7331581 DOI: 10.1038/s41423-020-0477-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023] Open
Abstract
The immune system and the nervous system are highly complex organs composed of various different cells that must interact with each other for proper function of the system. This communication can be mediated by soluble factors. The factors released by the nervous system (neurotransmitters) differ from those released by the immune system (cytokines). Nevertheless, the nervous and immune systems can influence each other’s activity because immune cells express neurotransmitter receptors, and neurons express cytokine receptors. Moreover, immune cells can synthesize and release neurotransmitters themselves, thus using neurotransmitter-mediated pathways via autocrine and paracrine mechanisms. Natural killer (NK) cells are innate lymphocytes that are important for early and effective immune reactions against infections and cancer. Many studies have shown the strong influence of stress and the nervous system on NK cell activity. This phenomenon may be one reason why chronic stress leads to a higher incidence of infections and cancer. Here, we review the effects of neuroendocrine factors on the different activities of NK cells. Understanding the effects of neuroendocrine factors on NK cell activities during physiological and pathophysiological conditions may result in novel therapeutic strategies to enhance NK cell functions against tumors.
Collapse
|
48
|
Jiang W, Li Y, Wei W, Li JW, Li L, Zhang C, Zhang SQ, Kong GY, Li ZF. Spleen contributes to restraint stress induced hepatocellular carcinoma progression. Int Immunopharmacol 2020; 83:106420. [PMID: 32203905 DOI: 10.1016/j.intimp.2020.106420] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/08/2020] [Accepted: 03/16/2020] [Indexed: 12/17/2022]
Abstract
The spleen is the largest secondary immune organ and plays a critical role in the progression of tumor. Psychological stress promotes tumor progression through inhibiting antitumor immune. However, the role of spleen in tumor progression induced by stress is unclear. Here, we showed that restraint stress promoted tumor growth, increased the percentage of CD11b+Gr-1+ MDSC while decreased the percentages of CD3-NK1.1+ NK and CD3+NK1.1+ NKT in the tumor tissues. Restraint stress decreased the percentages of CD3+CD4+ T lymphocytes and CD3+CD8+ T lymphocytes while increased the percentage of CD11b+Gr-1+ MDSC in the blood of tumor-bearing mice. Restraint stress increased the percentages of CD3+CD4+ T lymphocytes, CD3+CD8+ T lymphocytes, CD4+PD1+ T lymphocytes and CD8+PD1+ T lymphocytes while decreased the percentage of CD11b+Gr-1+ MDSC in the spleen of tumor-bearing mice. Interestingly, splenectomy inhibited tumor growth and attenuated the changes of CD3+CD4+ T lymphocytes, CD3+CD8+ T lymphocytes, and CD11b+Gr-1+ MDSC in blood induced by chronic restraint stress. Finally, splenectomy blocked the increases of CD11b+Gr-1+ MDSC but did not attenuate the decreases of CD3-NK1.1+ NK and CD3+NK1.1+ NKT in tumor tissue induced by chronic stress. Together, these data indicate that chronic restraint stress promotes hepatocellular carcinoma growth and suppresses the antitumor immunity of tumor-bearing mice. Splenectomy could inhibit tumor growth and partly block the decrease of antitumor immune activity induced by stress.
Collapse
MESH Headings
- Animals
- Bone Marrow/immunology
- Cell Line, Tumor
- Disease Progression
- Immune Tolerance
- Liver Neoplasms, Experimental/etiology
- Liver Neoplasms, Experimental/immunology
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/psychology
- Male
- Mice
- Mice, Inbred C57BL
- Myeloid-Derived Suppressor Cells/immunology
- Myeloid-Derived Suppressor Cells/metabolism
- Restraint, Physical/adverse effects
- Spleen/immunology
- Splenectomy
- Stress, Psychological/complications
- Stress, Psychological/immunology
- Stress, Psychological/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Wei Jiang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yu Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of thoracic surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wei Wei
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiang-Wei Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of thoracic surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chen Zhang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shu-Qun Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guang-Yao Kong
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zong-Fang Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Environment and Disease-Related Gene, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
49
|
Yin B, Jiang H, Liu X, Guo SW. Enriched Environment Decelerates the Development of Endometriosis in Mouse. Reprod Sci 2020; 27:1423-1435. [PMID: 32318984 DOI: 10.1007/s43032-019-00117-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022]
Abstract
We tested the hypothesis that enriched environment (EE), consisting of enlarged space, and increased physical activity and social interactions, hinders the development of endometriosis through attenuated adrenergic signaling, enhanced autophagy, and reduced leptin levels. Two mouse experiments were performed. In Experiment 1, 40 female Balb/C mice were randomly divided into four equal-sized groups, the SE (standard environment), EE, p-EE (EE instituted after endometriosis induction), and the d-EE (SE housing but received uterine fragments from EE donors) groups. Housing intervention was initiated 3 weeks before the induction of endometriosis and continued for 3 weeks after induction. In Experiment 2, 20 female mice were randomly divided into SE and EE groups, and the plasma leptin levels were measured. We measured lesion weight and hotplate latency and performed Masson trichrome staining as well as immunohistochemistry analysis of β2 adrenergic receptor (ADRB2), dopamine receptor D2 (DRD2), vascular endothelial growth factor (VEGF), and microtubule-associated protein light chain 3 (LC3). We found that EE reduced the lesion weight by 40.8% as compared with SE mice, but the reduction in p-EE and d-EE mice did not reach statistical significance. EE significantly reduced staining levels of ADRB2 and VEGF as well as the extent of lesional fibrosis but increased staining levels of LC3 and DRD2 in lesions as compared with the SE group. EE mice had reduced plasma leptin levels as compared with SE mice. Thus, EE decelerates the development of endometriosis and fibrogenesis and improved generalized hyperalgesia, possibly through increased DRD2 expression but decreased expression of ADRB2 and VEGF as well as enhanced autophagy and reduced leptin level.
Collapse
Affiliation(s)
- Bo Yin
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, 200011, China
| | - Hongyuan Jiang
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, 200011, China
| | - Xishi Liu
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, 200011, China. .,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China.
| | - Sun-Wei Guo
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, 200011, China. .,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
50
|
D'Alessandro G, Antonangeli F, Marrocco F, Porzia A, Lauro C, Santoni A, Limatola C. Gut microbiota alterations affect glioma growth and innate immune cells involved in tumor immunosurveillance in mice. Eur J Immunol 2020; 50:705-711. [PMID: 32034922 PMCID: PMC7216943 DOI: 10.1002/eji.201948354] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/30/2019] [Accepted: 02/05/2020] [Indexed: 12/28/2022]
Abstract
Glioma is a CNS tumor with few therapeutic options. Recently, host microbiota has been involved in the immune modulation of different tumors, but no data are available on the possible effects of the gut–immune axis on brain tumors. Here, we investigated the effect of gut microbiota alteration in a syngeneic (GL261) mouse model of glioma, treating mice with two antibiotics (ABX) and evaluating the effects on tumor growth, microbe composition, natural killer (NK) cells and microglia phenotype. We report that ABX treatment (i) altered the intestinal microbiota at family level, (ii) reduced cytotoxic NK cell subsets, and (iii) altered the expression of inflammatory and homeostatic proteins in microglia. All these findings could contribute to the increased growth of intracranial glioma that was observed after ABX treatment. These results demonstrate that chronic ABX administration alters microbiota composition and contributes to modulate brain immune state paving the way to glioma growth.
Collapse
Affiliation(s)
- Giuseppina D'Alessandro
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.,IRCCS Neuromed, Pozzilli, IS, Italy
| | - Fabrizio Antonangeli
- Department of Molecular Medicine, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University, Rome, Italy
| | - Francesco Marrocco
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.,Center for Life Nanoscience-IIT@Sapienza, Rome, Italy
| | - Alessandra Porzia
- Department of Molecular Medicine, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University, Rome, Italy
| | - Clotilde Lauro
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Angela Santoni
- IRCCS Neuromed, Pozzilli, IS, Italy.,Department of Molecular Medicine, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University, Rome, Italy
| | - Cristina Limatola
- IRCCS Neuromed, Pozzilli, IS, Italy.,Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University, Rome, Italy
| |
Collapse
|