1
|
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
|
2
|
Xiao Z, Nian Z, Zhang M, Liu Z, Zhang P, Zhang Z. Single-cell and bulk RNA-sequencing reveal SPP1 and CXCL12 as cell-to-cell communication markers to predict prognosis in lung adenocarcinoma. ENVIRONMENTAL TOXICOLOGY 2024; 39:4610-4622. [PMID: 38622884 DOI: 10.1002/tox.24297] [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: 01/10/2024] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
Lung adenocarcinoma (LUAD) generally presents as an immunosuppressive microenvironment. The characteristics of cell-to-cell communication in the LUAD microenvironment has been unclear. In this study, the LUAD bulk RNA-seq data and single-cell RNA-seq data were retrieved from public dataset. Differential expression genes (DEGs) between LUAD tumor and adjacent non-tumor tissues were calculated by limma algorithm, and then detected by PPI, KEGG, and GO analysis. Cell-cell interactions were explored using the single-cell RNA-seq data. Finally, the first 15 CytoHubba genes were used to establish related pathways and these pathways were used to characterize the immune-related ligands and their receptors in LUAD. Our analyses showed that monocytes or macrophages interact with tissue stem cells and NK cells via SPP1 signaling pathway and tissue stem cells interact with T and B cells via CXCL signaling pathway in different states. Hub genes of SPP1 participated in SPP1 signaling pathway, which was negatively correlated with CD4+ T cell and CD8+ T cell. The expression of SPP1 in LUAD tumor tissues was negatively correlated with the prognosis. While CXCL12 participated in CXCL signaling pathway, which was positively correlated with CD4+ T cell and CD8+ T cell. The role of CXCL12 in LUAD tumor tissues exhibits an opposite effect to that of SPP1. This study reveals that tumor-associated monocytes or macrophages may affect tumor progression. Moreover, the SPP1 and CXCL12 may be the critic genes of cell-to-cell communication in LUAD, and targeting these pathways may provide a new molecular mechanism for the treatment of LUAD.
Collapse
Affiliation(s)
- Zengtuan Xiao
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
- Department of Immunology, Biochemistry and Molecular Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, Tianjin Medical University, Tianjin, China
| | - Zhe Nian
- Department of Immunology, Biochemistry and Molecular Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, Tianjin Medical University, Tianjin, China
| | - Mengzhe Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Zuo Liu
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| | - Zhenfa Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, China
| |
Collapse
|
3
|
AbuQeis I, Zou Y, Ba YC, Teeti AA. Neuroscience of cancer: Research progress and emerging of the field. IBRAIN 2024; 10:305-322. [PMID: 39346791 PMCID: PMC11427805 DOI: 10.1002/ibra.12172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 10/01/2024]
Abstract
Cancer cells immediately expand and penetrate adjoining tissues, as opposed to metastasis, that is the spread of cancer cells through the circulatory or lymphatic systems to more distant places via the invasion process. We found that a lack of studies discussed tumor development with the nervous system, by the aspects of cancer-tissue invasion (biological) and chemical modulation of growth that cascades by releasing neural-related factors from the nerve endings via chemical substances known as neurotransmitters. In this review, we aimed to carefully demonstrate and describe the cancer invasion and interaction with the nervous system, as well as reveal the research progress and the emerging neuroscience of cancer. An initial set of 160 references underwent systematic review and summarization. Through a meticulous screening process, these data were refined, ultimately leading to the inclusion of 98 studies that adhered to predetermined criteria. The outcomes show that one formidable challenge in the realm of cancer lies in its intrinsic heterogeneity and remarkable capacity for rapid adaptation. Despite advancements in genomics and precision medicine, there is still a need to identify new molecular targets. Considering cancer within its molecular and cellular environment, including neural components, is crucial for addressing this challenge. In conclusion, this review provides good referential data for direct, indirect, biological, and chemical interaction for nerve tissue-tumor interaction, suggesting the establishment of new therapy techniques and mechanisms by controlling and modifying neuron networks that supply signals to tumors.
Collapse
Affiliation(s)
- Issam AbuQeis
- Department of Radiology Palestinian Ministry of Health Ramallah Palestine
- Department of Anatomy, Institute of Neuroscience, School of Basic Medicine Kunming Medical University Kunming China
| | - Yu Zou
- Department of Anatomy, Institute of Neuroscience, School of Basic Medicine Kunming Medical University Kunming China
| | - Ying-Chun Ba
- Department of Anatomy, Institute of Neuroscience, School of Basic Medicine Kunming Medical University Kunming China
| | - Abeer A Teeti
- Department of Chemistry, School of Science Hebron University Hebron Palestine
- Department of Epidemiology, School of Public Health Kunming Medical University Kunming China
| |
Collapse
|
4
|
Cong R, Xu R, Ming J, Zhu Z. Construction of a preoperative nomogram model for predicting perineural invasion in advanced gastric cancer. Front Med (Lausanne) 2024; 11:1344982. [PMID: 38912337 PMCID: PMC11190154 DOI: 10.3389/fmed.2024.1344982] [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: 11/27/2023] [Accepted: 05/24/2024] [Indexed: 06/25/2024] Open
Abstract
Objective This study aimed to develop and validate a clinical and imaging-based nomogram for preoperatively predicting perineural invasion (PNI) in advanced gastric cancer. Methods A retrospective cohort of 351 patients with advanced gastric cancer who underwent surgical resection was included. Multivariable logistic regression analysis was conducted to identify independent risk factors for PNI and to construct the nomogram. The performance of the nomogram was assessed using calibration curves, the concordance index (C-index), the area under the curve (AUC), and decision curve analysis (DCA). The disparity in disease-free survival (DFS) between the nomogram-predicted PNI-positive group and the nomogram-predicted PNI-negative group was evaluated using the Log-Rank test and Kaplan-Meier analysis. Results Extramural vascular invasion (EMVI), Borrmann classification, tumor thickness, and the systemic inflammation response index (SIRI) emerged as independent risk factors for PNI. The nomogram model demonstrated a commendable AUC value of 0.838. Calibration curves exhibited excellent concordance, with a C-index of 0.814. DCA indicated that the model provided good clinical net benefit. The DFS of the nomogram-predicted PNI-positive group was significantly lower than that of the nomogram-predicted PNI-negative group (p < 0.001). Conclusion This study successfully developed a preoperative nomogram model that not only effectively predicted PNI in gastric cancer but also facilitated postoperative risk stratification.
Collapse
Affiliation(s)
- Ruochen Cong
- Department of Radiology, Nantong No. 1 People’s Hospital, Nantong, China
| | - Ruonan Xu
- Department of Radiology, Nantong No. 6 People’s Hospital, Nantong, China
| | - Jialei Ming
- Department of Radiology, Nantong No. 1 People’s Hospital, Nantong, China
| | - Zhengqi Zhu
- Department of Radiology, Nantong City Cancer Hospital, Nantong, China
| |
Collapse
|
5
|
Cai Z, Yao H, Chen J, Ahmed AA, Li C, Hu X, Tang X, Jiang C. Schwann cells in pancreatic cancer: Unraveling their multifaceted roles in tumorigenesis and neural interactions. Cancer Lett 2024; 587:216689. [PMID: 38367898 DOI: 10.1016/j.canlet.2024.216689] [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: 12/14/2023] [Revised: 01/15/2024] [Accepted: 01/26/2024] [Indexed: 02/19/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), characterized by heightened neural density, presents a challenging prognosis primarily due to perineural invasion. Recognized for their crucial roles in neural support and myelination, Schwann cells (SCs) significantly influence the process of tumorigenesis. This review succinctly outlines the interplay between PDAC and neural systems, positioning SCs as a nexus in the tumor-neural interface. Subsequently, it delves into the cellular origin and influencers of SCs within the pancreatic tumor microenvironment, emphasizing their multifaceted roles in tumor initiation, progression, and modulation of the neural and immune microenvironment. The discussion encompasses potential therapeutic interventions targeting SCs. Lastly, the review underscores pressing issues, advocating for sustained exploration into the diverse contributions of SCs within the intricate landscape of PDAC, with the aim of enhancing our understanding of their involvement in this complex malignancy.
Collapse
Affiliation(s)
- Zhiwei Cai
- Department of General Surgery, Pancreatobiliary Surgery Center, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, PR China
| | - Hongfei Yao
- Department of General Surgery, Pancreatobiliary Surgery Center, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, PR China
| | - Jiahao Chen
- Department of General Surgery, Pancreatobiliary Surgery Center, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, PR China
| | - Abousalam Abdoulkader Ahmed
- Department of General Surgery, Pancreatobiliary Surgery Center, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, PR China
| | - Chunjing Li
- Department of General Surgery, Pancreatobiliary Surgery Center, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, PR China
| | - Xiao Hu
- Department of General Surgery, Pancreatobiliary Surgery Center, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, PR China
| | - Xiaoyan Tang
- Department of General Surgery, Pancreatobiliary Surgery Center, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, PR China
| | - Chongyi Jiang
- Department of General Surgery, Pancreatobiliary Surgery Center, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, PR China.
| |
Collapse
|
6
|
Giri SS, Tripathi AS, Erkekoğlu P, Zaki MEA. Molecular pathway of pancreatic cancer-associated neuropathic pain. J Biochem Mol Toxicol 2024; 38:e23638. [PMID: 38613466 DOI: 10.1002/jbt.23638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 04/15/2024]
Abstract
The pancreas is a heterocrine gland that has both exocrine and endocrine parts. Most pancreatic cancer begins in the cells that line the ducts of the pancreas and is called pancreatic ductal adenocarcinoma (PDAC). PDAC is the most encountered pancreatic cancer type. One of the most important characteristic features of PDAC is neuropathy which is primarily due to perineural invasion (PNI). PNI develops tumor microenvironment which includes overexpression of fibroblasts cells, macrophages, as well as angiogenesis which can be responsible for neuropathy pain. In tumor microenvironment inactive fibroblasts are converted into an active form that is cancer-associated fibroblasts (CAFs). Neurotrophins they also increase the level of Substance P, calcitonin gene-related peptide which is also involved in pain. Matrix metalloproteases are the zinc-associated proteases enzymes which activates proinflammatory interleukin-1β into its activated form and are responsible for release and activation of Substance P which is responsible for neuropathic pain by transmitting pain signal via dorsal root ganglion. All the molecules and their role in being responsible for neuropathic pain are described below.
Collapse
Affiliation(s)
| | - Alok Shiomurti Tripathi
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, Uttar Pradesh, India
| | - Pınar Erkekoğlu
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Magdi E A Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Zhou Y, Zhang Y, Xu J, Wang Y, Yang Y, Wang W, Gu A, Han B, Shurin GV, Zhong R, Shurin MR, Zhong H. Schwann cell-derived exosomes promote lung cancer progression via miRNA-21-5p. Glia 2024; 72:692-707. [PMID: 38192185 DOI: 10.1002/glia.24497] [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: 09/29/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 01/10/2024]
Abstract
Schwann cells (SCs), the primary glial cells of the peripheral nervous system, which have been identified in many solid tumors, play an important role in cancer development and progression by shaping the tumor immunoenvironment and supporting the development of metastases. Using different cellular, molecular, and genetic approaches with integrated bioinformatics analysis and functional assays, we revealed the role of human SC-derived exosomal miRNAs in lung cancer progression in vitro and in vivo. We found that exosomal miRNA-21 from SCs up-regulated the proliferation, motility, and invasiveness of human lung cancer cells in vitro, which requires functional Rab small GTPases Rab27A and Rab27B in SCs for exosome release. We also revealed that SC exosomal miRNA-21-5p regulated the functional activation of tumor cells by targeting metalloprotease inhibitor RECK in tumor cells. Integrated bioinformatic analyses showed that hsa-miRNA-21-5p is associated with poor prognosis in patients with lung adenocarcinoma and can promote lung cancer progression through multiple signaling pathways including the MAPK, PI3K/Akt, and TNF signaling. Furthermore, in mouse xenograft models, SC exosomes and SC exosomal hsa-miRNA-21-5p augmented human lung cancer cell growth and lymph node metastasis in vivo. Together our data revealed, for the first time, that SC-secreted exosomes and exosomal miRNA-21-5p promoted the proliferation, motility, and spreading of human lung cancer cells in vitro and in vivo. Thus, exosomal miRNA-21 may play an oncogenic role in SC-accelerated progression of lung cancer and this pathway may serve as a new therapeutic target for further evaluation.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianlin Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Wang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Yang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weimin Wang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aiqin Gu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baohui Han
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Galina V Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Runbo Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Hua Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Yoshida A, Nishibata M, Maruyama T, Sunami S, Isono K, Kawamata T. Activation of Transient Receptor Potential Vanilloid 1 Is Involved in Both Pain and Tumor Growth in a Mouse Model of Cancer Pain. Neuroscience 2024; 538:80-92. [PMID: 38157977 DOI: 10.1016/j.neuroscience.2023.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/03/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Activation of calcitonin gene-related peptide (CGRP)-positive sensory neurons in the tumor microenvironment has been shown to be involved in tumor growth. However, how CGRP-positive sensory neurons are activated requires elucidation. In this study, we focused on transient receptor potential vanilloid 1 (TRPV1) and examined the contribution of TRPV1 to tumor growth and cancer pain in a mouse cancer model in which Lewis lung carcinoma was subcutaneously inoculated in the left plantar region. Tumor inoculation gradually increased the volumes of the hind paws of wild type (WT) mice over time, but those of both αCGRP knockout mice and TRPV1 knockout mice were significantly smaller than those of WT mice after tumor inoculation. Both TRPV1 and CGRP are therefore suggested to be involved in tumor growth. In an immunohistochemical study, the percentage of phosphorylated cyclic adenosine monophosphate response element-binding protein (p-CREB)-positive profiles in CGRP-positive dorsal root ganglion (DRG) neurons in WT mice was significantly increased after tumor inoculation. The percentage of p-CREB-positive profiles in CGRP-positive DRG neurons in TRPV1 knockout mice was also increased after tumor inoculation, but was significantly lower than that in WT mice, indicating the contribution of TRPV1 to activation of CGRP-positive DRG neurons. Cancer pain in TRPV1 knockout mice was significantly lower than that in WT mice. In conclusion, TRPV1 is involved in both tumor growth and cancer pain, potentially leading to a novel strategy for the treatment of cancer pain and cancer development. Cancer pain is also suggested to facilitate tumor growth.
Collapse
Affiliation(s)
- Akari Yoshida
- Department of Anesthesiology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan.
| | - Masayuki Nishibata
- Department of Anesthesiology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan
| | - Tomoyuki Maruyama
- Department of Anesthesiology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan
| | - Shogo Sunami
- Department of Anesthesiology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan
| | - Kyoichi Isono
- Laboratory Animal Center, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan
| | - Tomoyuki Kawamata
- Department of Anesthesiology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 640-0012, Japan
| |
Collapse
|
9
|
Wang H, Huo R, He K, Cheng L, Zhang S, Yu M, Zhao W, Li H, Xue J. Perineural invasion in colorectal cancer: mechanisms of action and clinical relevance. Cell Oncol (Dordr) 2024; 47:1-17. [PMID: 37610689 PMCID: PMC10899381 DOI: 10.1007/s13402-023-00857-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND In recent years, the significance of the nervous system in the tumor microenvironment has gained increasing attention. The bidirectional communication between nerves and cancer cells plays a critical role in tumor initiation and progression. Perineural invasion (PNI) occurs when tumor cells invade the nerve sheath and/or encircle more than 33% of the nerve circumference. PNI is a common feature in various malignancies and is associated with tumor invasion, metastasis, cancer-related pain, and unfavorable clinical outcomes. The colon and rectum are highly innervated organs, and accumulating studies support PNI as a histopathologic feature of colorectal cancer (CRC). Therefore, it is essential to investigate the role of nerves in CRC and comprehend the mechanisms of PNI to impede tumor progression and improve patient survival. CONCLUSION This review elucidates the clinical significance of PNI, summarizes the underlying cellular and molecular mechanisms, introduces various experimental models suitable for studying PNI, and discusses the therapeutic potential of targeting this phenomenon. By delving into the intricate interactions between nerves and tumor cells, we hope this review can provide valuable insights for the future development of CRC treatments.
Collapse
Affiliation(s)
- Hao Wang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P.R. China
| | - Ruixue Huo
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P.R. China
| | - Kexin He
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P.R. China
| | - Li Cheng
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P.R. China
| | - Shan Zhang
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Minhao Yu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200217, P.R. China
| | - Wei Zhao
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P.R. China.
| | - Hui Li
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China.
| | - Junli Xue
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P.R. 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
|
Safak O, Wang S, Mota Reyes C, Gürcinar IH, Tokalov S, Cevik NC, Görgülü K, Yilmaz BS, Erdogan E, Ye L, Li Q, Sever EA, Özer S, Süyen G, Friess H, Ceyhan GO, Istvanffy R, Algül H, Demir IE. Dynamics and cytokinic regulation of immune cell infiltration in genetically engineered mouse models of pancreatic cancer dictate the sensitivity to immunotherapy. Cancer Commun (Lond) 2024; 44:178-182. [PMID: 37877813 PMCID: PMC10794007 DOI: 10.1002/cac2.12496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/20/2023] [Accepted: 10/10/2023] [Indexed: 10/26/2023] Open
Affiliation(s)
- Okan Safak
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Shenghan Wang
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Carmen Mota Reyes
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Ibrahim Halil Gürcinar
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Sergey Tokalov
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Nedim Can Cevik
- Department of General Surgery, HPB‐Unit, School of MedicineAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
| | - Kivanc Görgülü
- Comprehensive Cancer Center München, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Bengi Su Yilmaz
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Emre Erdogan
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Linhan Ye
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Qiaolin Li
- Department of Hematology, Oncology and Tumor ImmunologyCharité University Medicine Berlin, Campus Virchow ClinicBerlinBerlinGermany
| | - Elif Arik Sever
- Department of General Surgery, HPB‐Unit, School of MedicineAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
| | - Samed Özer
- Graduate School of Health SciencesAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
| | - Güldal Süyen
- Department of PhysiologyAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Güralp Onur Ceyhan
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
- Department of General Surgery, HPB‐Unit, School of MedicineAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
| | - Rouzanna Istvanffy
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Hana Algül
- Comprehensive Cancer Center München, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
- Department of General Surgery, HPB‐Unit, School of MedicineAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
- Else Kröner Clinician Scientist Professorship for Translational Pancreatic SurgeryMunichGermany
- Neural Influences in Cancer (NIC) International Research ConsortiumMunichGermany
| |
Collapse
|
12
|
Wang X, Istvanffy R, Ye L, Teller S, Laschinger M, Diakopoulos KN, Görgülü K, Li Q, Ren L, Jäger C, Steiger K, Muckenhuber A, Vilne B, Çifcibaşı K, Reyes CM, Yurteri Ü, Kießler M, Gürçınar IH, Sugden M, Yıldızhan SE, Sezerman OU, Çilingir S, Süyen G, Reichert M, Schmid RM, Bärthel S, Oellinger R, Krüger A, Rad R, Saur D, Algül H, Friess H, Lesina M, Ceyhan GO, Demir IE. Phenotype screens of murine pancreatic cancer identify a Tgf-α-Ccl2-paxillin axis driving human-like neural invasion. J Clin Invest 2023; 133:e166333. [PMID: 37607005 PMCID: PMC10617783 DOI: 10.1172/jci166333] [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: 10/20/2022] [Accepted: 08/17/2023] [Indexed: 08/23/2023] Open
Abstract
Solid cancers like pancreatic ductal adenocarcinoma (PDAC), a type of pancreatic cancer, frequently exploit nerves for rapid dissemination. This neural invasion (NI) is an independent prognostic factor in PDAC, but insufficiently modeled in genetically engineered mouse models (GEMM) of PDAC. Here, we systematically screened for human-like NI in Europe's largest repository of GEMM of PDAC, comprising 295 different genotypes. This phenotype screen uncovered 2 GEMMs of PDAC with human-like NI, which are both characterized by pancreas-specific overexpression of transforming growth factor α (TGF-α) and conditional depletion of p53. Mechanistically, cancer-cell-derived TGF-α upregulated CCL2 secretion from sensory neurons, which induced hyperphosphorylation of the cytoskeletal protein paxillin via CCR4 on cancer cells. This activated the cancer migration machinery and filopodia formation toward neurons. Disrupting CCR4 or paxillin activity limited NI and dampened tumor size and tumor innervation. In human PDAC, phospho-paxillin and TGF-α-expression constituted strong prognostic factors. Therefore, we believe that the TGF-α-CCL2-CCR4-p-paxillin axis is a clinically actionable target for constraining NI and tumor progression in PDAC.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Rouzanna Istvanffy
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC) International Research Consortium
| | - Linhan Ye
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Pain Clinic, Department of Anesthesiology, First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Steffen Teller
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Melanie Laschinger
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Kalliope N. Diakopoulos
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Internal Medicine II & Comprehensive Cancer Center Munich, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Kıvanç Görgülü
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Internal Medicine II & Comprehensive Cancer Center Munich, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Qiaolin Li
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lei Ren
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Carsten Jäger
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Comparative Experimental Pathology and Institute of Pathology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Alexander Muckenhuber
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Comparative Experimental Pathology and Institute of Pathology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Baiba Vilne
- Bioinformatics laboratory, Riga Stradins University, Riga, Latvia
| | - Kaan Çifcibaşı
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Carmen Mota Reyes
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC) International Research Consortium
| | - Ümmügülsüm Yurteri
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Maximilian Kießler
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Ibrahim Halil Gürçınar
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Maya Sugden
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | | | - Sümeyye Çilingir
- Department of Physiology, Acibadem Mehmet Ali Aydinlar University, School of Medicine, Istanbul, Turkey
| | - Güldal Süyen
- Department of Physiology, Acibadem Mehmet Ali Aydinlar University, School of Medicine, Istanbul, Turkey
| | - Maximilian Reichert
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Roland M. Schmid
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Stefanie Bärthel
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Institute of Translational Cancer Research (TranslaTUM) and Experimental Cancer Therapy
| | - Rupert Oellinger
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics
| | - Achim Krüger
- Institute of Experimental Oncology and Therapy Research, School of Medicine, Technical University Munich, Munich, Germany
| | - Roland Rad
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics
| | - Dieter Saur
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Institute of Translational Cancer Research (TranslaTUM) and Experimental Cancer Therapy
| | - Hana Algül
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Internal Medicine II & Comprehensive Cancer Center Munich, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC) International Research Consortium
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Marina Lesina
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Internal Medicine II & Comprehensive Cancer Center Munich, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Güralp Onur Ceyhan
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- Neural Influences in Cancer (NIC) International Research Consortium
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC) International Research Consortium
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
- Else Kröner Clinician Scientist Professor for Translational Pancreatic Surgery, Technical University of Munich, Munich, Germany
| |
Collapse
|
13
|
García-Reyes B, Kuzmanov I, Schneider R, Schneiker B, Efferz P, Kalff JC, Wehner S. Glial cell-derived soluble factors increase the metastatic potential of pancreatic adenocarcinoma cells and induce epithelial-to-mesenchymal transition. J Cancer Res Clin Oncol 2023; 149:14315-14327. [PMID: 37572121 PMCID: PMC10590291 DOI: 10.1007/s00432-023-05133-y] [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: 06/26/2023] [Accepted: 07/04/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive types of cancer, characterized by the spreading of highly metastatic cancer cells, including invasion into surrounding nerves and perineural spaces. Nerves, in turn, can invade the tumor tissue and, through the secretion of neurotrophic factors, chemokines, and cytokines, contribute to PDAC progression. However, the contribution of the nerve-associated glial cells to PDAC progression is not well characterized. METHODS Two murine PDAC cell lines were cultured with the conditioned media (CM) of primary enteric glial cells or IMS32 Schwann cells (SCs). Different properties of PDAC cells, such as invasiveness, migratory capacity, and resistance to gemcitabine, were measured by RT-qPCR, microscopy, and MTT assays. Using a neuronal cell line, the observed effects were confirmed to be specific to the glial lineage. RESULTS Compared to the control medium, PDAC cells in the glial cell-conditioned medium showed increased invasiveness and migratory capacity. These cells showed reduced E-cadherin and increased N-cadherin and Vimentin levels, all markers of epithelial-mesenchymal transition (EMT). Primary enteric glial cell CM inhibited the proliferation of PDAC cells but preserved their viability, upregulated transcription factor Snail, and increased their resistance to gemcitabine. The conditioned medium generated from the IMS32 SCs produced comparable results. CONCLUSION Our data suggest that glial cells can increase the metastatic potential of PDAC cells by increasing their migratory capacity and inducing epithelial-to-mesenchymal transition, a re-programming that many solid tumors use to undergo metastasis. Glial cell-conditioned medium also increased the chemoresistance of PDAC cells. These findings may have implications for future therapeutic strategies, such as targeting glial cell-derived factor signaling in PDAC.
Collapse
Affiliation(s)
- Balbina García-Reyes
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
- Mildred Scheel School of Oncology, Aachen Bonn Cologne Düsseldorf (MSSO ABCD), Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Ivan Kuzmanov
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Reiner Schneider
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Bianca Schneiker
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Patrik Efferz
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Jörg C Kalff
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Sven Wehner
- Department of Surgery, Medical Faculty, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
14
|
Shi RJ, Ke BW, Tang YL, Liang XH. Perineural invasion: A potential driver of cancer-induced pain. Biochem Pharmacol 2023; 215:115692. [PMID: 37481133 DOI: 10.1016/j.bcp.2023.115692] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/24/2023]
Abstract
Perineural invasion (PNI) is the process through which tumors invade and interact with nerves. The dynamic changes in the nerves caused by PNI may induce disturbing symptoms. PNI-related cancer pain in neuro-rich tumors has attracted much attention because the occurrence of tumor-induced pain is closely related to the invasion of nerves in the tumor microenvironment. PNI-related pain might indicate the occurrence of PNI, guide the improvement of treatment strategies, and predict the unresectability of tumors and the necessity of palliative care. Although many studies have investigated PNI, its relationship with tumor-induced pain and its common mechanisms have not been summarized thoroughly. Therefore, in this review, we evaluated the relationship between PNI and cancer-associated pain. We showed that PNI is a major cause of cancer-related pain and that this pain can predict the occurrence of PNI. We also elucidated the cellular and molecular mechanisms of PNI-induced pain. Finally, we analyzed the possible targets for alleviating PNI-related pain or combined antitumor and pain management. Our findings might provide new perspectives for improving the treatment of patients with malignant tumors.
Collapse
Affiliation(s)
- Rong-Jia Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery,West China Hospital of Stomatology (Sichuan University), No.14, Sec. 3, Renminnan Road, Chengdu 610041, Sichuan, China
| | - Bo-Wen Ke
- Laboratory of Anesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Pathology, West China Hospital of Stomatology (Sichuan University), No.14, Sec. 3, Renminnan Road, Chengdu 610041, Sichuan, China.
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery,West China Hospital of Stomatology (Sichuan University), No.14, Sec. 3, Renminnan Road, Chengdu 610041, Sichuan, China.
| |
Collapse
|
15
|
Kruglov O, Vats K, Soman V, Tyurin VA, Tyurina YY, Wang J, Williams L, Zhang J, Donahue Carey C, Jaklitsch E, Chandran UR, Bayir H, Kagan VE, Bunimovich YL. Melanoma-associated repair-like Schwann cells suppress anti-tumor T-cells via 12/15-LOX/COX2-associated eicosanoid production. Oncoimmunology 2023; 12:2192098. [PMID: 36998620 PMCID: PMC10044150 DOI: 10.1080/2162402x.2023.2192098] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 04/01/2023] Open
Abstract
Peripheral glia, specifically the Schwann cells (SCs), have been implicated in the formation of the tumor microenvironment (TME) and in cancer progression. However, in vivo and ex vivo analyses of how cancers reprogram SC functions in different organs of tumor-bearing mice are lacking. We generated Plp1-CreERT/tdTomato mice which harbor fluorescently labeled myelinated and non-myelin forming SCs. We show that this model enables the isolation of the SCs with high purity from the skin and multiple other organs. We used this model to study phenotypic and functional reprogramming of the SCs in the skin adjacent to melanoma tumors. Transcriptomic analyses of the peritumoral skin SCs versus skin SCs from tumor-free mice revealed that the former existed in a repair-like state typically activated during nerve and tissue injury. Peritumoral skin SCs also downregulated pro-inflammatory genes and pathways related to protective anti-tumor responses. In vivo and ex vivo functional assays confirmed immunosuppressive activities of the peritumoral skin SCs. Specifically, melanoma-reprogrammed SCs upregulated 12/15-lipoxygenase (12/15-LOX) and cyclooxygenase (COX)-2, and increased production of anti-inflammatory polyunsaturated fatty acid (PUFA) metabolites prostaglandin E2 (PGE2) and lipoxins A4/B4. Inhibition of 12/15-LOX or COX2 in SCs, or EP4 receptor on lymphocytes reversed SC-dependent suppression of anti-tumor T-cell activation. Therefore, SCs within the skin adjacent to melanoma tumors demonstrate functional switching to repair-like immunosuppressive cells with dysregulated lipid oxidation. Our study suggests the involvement of the melanoma-associated repair-like peritumoral SCs in the modulation of locoregional and systemic anti-tumor immune responses.
Collapse
Affiliation(s)
- Oleg Kruglov
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kavita Vats
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Vishal Soman
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Vladimir A. Tyurin
- Center for Free Radical and Antioxidant Health, Department of Environmental Health and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yulia Y. Tyurina
- Center for Free Radical and Antioxidant Health, Department of Environmental Health and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jiefei Wang
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Li’an Williams
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jiying Zhang
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cara Donahue Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Erik Jaklitsch
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Uma R. Chandran
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hülya Bayir
- Center for Free Radical and Antioxidant Health, Department of Environmental Health and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Children’s Neuroscience Institute, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Valerian E. Kagan
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Free Radical and Antioxidant Health, Department of Environmental Health and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Children’s Neuroscience Institute, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Hillman Cancer Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Yuri L. Bunimovich
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Free Radical and Antioxidant Health, Department of Environmental Health and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
- Hillman Cancer Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
16
|
Zhang Y, Sang R, Bao J, Jiang Z, Qian D, Zhou Y, Su W, Wei J, Zhao L, Wei Z, Zhao Y, Shi M, Chen G. Schwann cell-derived CXCL2 contributes to cancer pain by modulating macrophage infiltration in a mouse breast cancer model. Brain Behav Immun 2023; 109:308-320. [PMID: 36754246 DOI: 10.1016/j.bbi.2023.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 01/28/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
Pain is one of the most severe complications affecting the quality of life of cancer patients. Although substantial progress has been made in the diagnosis and treatment of cancer, the neurobiological mechanism of cancer pain is still unclear. In the present study, we identified the critical role of CXC chemokine 2 (CXCL2), released by Schwann cells after being activated by cancer cells, in maintaining cancer-induced macrophage infiltration and the resulting mechanical hypersensitivity and persistent spontaneous nociception. In vitro, Schwann cells cocultured with breast cancer cells exhibited a significant increase in CXCL2 expression; in addition, conditioned medium from Schwann cells activated by breast cancer cells had a similar effect to recombinant CXCL2 in terms of inducing macrophage migration. Targeting CXCL2 signaling by both CXC chemokine receptor 2 (CXCR2) antagonist pharmacological blockade and anti-CXCL2 mAb immunological blockade robustly prevented conditioned medium-induced macrophage migration. In vivo, both application of recombinant CXCL2 and perineural breast cancer cell implantation resulted in mechanical hypersensitivity and persistent spontaneous nociception in mice, along with increased macrophage infiltration into the sciatic nerves. Similar to the in vitro results, inhibition of CXCL2/CXCR2 signaling or conditional knockdown of CXCL2 in sciatic nerve Schwann cells effectively attenuated breast cancer cell-induced mechanical hypersensitivity, persistent spontaneous nociception, and macrophage recruitment in the sciatic nerve. Mechanistically, we found that redox effector factor-1 (Ref-1) secreted by breast cancer cells activated hypoxia inducible factor-1α (HIF-1α) expression and inhibited reactive oxygen species (ROS) production in Schwann cells, ultimately inducing CXCL2 expression in Schwann cells. In brief, the present study expands new insights into cancer pain mechanisms from promising animal models to provide new strategies for the control of cancer pain.
Collapse
Affiliation(s)
- Yonghui Zhang
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China; Department of Thoracic Surgery, Tumor Hospital Affiliated to Nantong University, Nantong 226001, Jiangsu Province, China
| | - Rui Sang
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Jingyin Bao
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Zhihao Jiang
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Danni Qian
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Yi Zhou
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Wenfeng Su
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Jinhuan Wei
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Long Zhao
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Zhongya Wei
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Yayu Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, China
| | - Minxin Shi
- Department of Thoracic Surgery, Tumor Hospital Affiliated to Nantong University, Nantong 226001, Jiangsu Province, China.
| | - Gang Chen
- Basic Medical Research Center, Medical School of Nantong University, Co-Innovation Center of Neuroregeneration, Nantong 226001, Jiangsu Province, China; Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, China; Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China.
| |
Collapse
|
17
|
Chen Z, Fang Y, Jiang W. Important Cells and Factors from Tumor Microenvironment Participated in Perineural Invasion. Cancers (Basel) 2023; 15:1360. [PMID: 36900158 PMCID: PMC10000249 DOI: 10.3390/cancers15051360] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Perineural invasion (PNI) as the fourth way for solid tumors metastasis and invasion has attracted a lot of attention, recent research reported a new point that PNI starts to include axon growth and possible nerve "invasion" to tumors as the component. More and more tumor-nerve crosstalk has been explored to explain the internal mechanism for tumor microenvironment (TME) of some types of tumors tends to observe nerve infiltration. As is well known, the interaction of tumor cells, peripheral blood vessels, extracellular matrix, other non-malignant cells, and signal molecules in TME plays a key role in the occurrence, development, and metastasis of cancer, as to the occurrence and development of PNI. We aim to summarize the current theories on the molecular mediators and pathogenesis of PNI, add the latest scientific research progress, and explore the use of single-cell spatial transcriptomics in this invasion way. A better understanding of PNI may help to understand tumor metastasis and recurrence and will be beneficial for improving staging strategies, new treatment methods, and even paradigm shifts in our treatment of patients.
Collapse
Affiliation(s)
- Zirong Chen
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Anatomy Laboratory of Division of Nose and Cranial Base, Clinical Anatomy Center of Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yan Fang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Anatomy Laboratory of Division of Nose and Cranial Base, Clinical Anatomy Center of Xiangya Hospital, Central South University, Changsha 410008, China
| | - Weihong Jiang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Anatomy Laboratory of Division of Nose and Cranial Base, Clinical Anatomy Center of Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
18
|
Sun C, Ye Y, Tan Z, Liu Y, Li Y, Hu W, Liang K, Egranov SD, Huang LA, Zhang Z, Zhang Y, Yao J, Nguyen TK, Zhao Z, Wu A, Marks JR, Caudle AS, Sahin AA, Gao J, Gammon ST, Piwnica-Worms D, Hu J, Chiao PJ, Yu D, Hung MC, Curran MA, Calin GA, Ying H, Han L, Lin C, Yang L. Tumor-associated nonmyelinating Schwann cell-expressed PVT1 promotes pancreatic cancer kynurenine pathway and tumor immune exclusion. SCIENCE ADVANCES 2023; 9:eadd6995. [PMID: 36724291 PMCID: PMC9891701 DOI: 10.1126/sciadv.add6995] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 01/03/2023] [Indexed: 05/16/2023]
Abstract
One of the major obstacles to treating pancreatic ductal adenocarcinoma (PDAC) is its immunoresistant microenvironment. The functional importance and molecular mechanisms of Schwann cells in PDAC remains largely elusive. We characterized the gene signature of tumor-associated nonmyelinating Schwann cells (TASc) in PDAC and indicated that the abundance of TASc was correlated with immune suppressive tumor microenvironment and the unfavorable outcome of patients with PDAC. Depletion of pancreatic-specific TASc promoted the tumorigenesis of PDAC tumors. TASc-expressed long noncoding RNA (lncRNA) plasmacytoma variant translocation 1 (PVT1) was triggered by the tumor cell-produced interleukin-6. Mechanistically, PVT1 modulated RAF proto-oncogene serine/threonine protein kinase-mediated phosphorylation of tryptophan 2,3-dioxygenase in TASc, facilitating its enzymatic activities in catalysis of tryptophan to kynurenine. Depletion of TASc-expressed PVT1 suppressed PDAC tumor growth. Furthermore, depletion of TASc using a small-molecule inhibitor effectively sensitized PDAC to immunotherapy, signifying the important roles of TASc in PDAC immune resistance.
Collapse
Affiliation(s)
- Chengcao Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Youqiong Ye
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Zhi Tan
- Center for Drug Discovery, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yuan Liu
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Yajuan Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wei Hu
- Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Ke Liang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sergey D. Egranov
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lisa Angela Huang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhao Zhang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Yaohua Zhang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jun Yao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tina K. Nguyen
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zilong Zhao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew Wu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey R. Marks
- Division of Surgical Science, Department of Surgery, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Abigail S. Caudle
- Department of Breast Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aysegul A. Sahin
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Seth T. Gammon
- Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David Piwnica-Worms
- Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian Hu
- Department of Cancer Biology, Division of Basic Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paul J. Chiao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan
- Department of Biotechnology, Asia University, Taichung 413, Taiwan
| | - Michael A. Curran
- The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George A. Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Liuqing Yang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
19
|
D’Alterio C, Giardino A, Scognamiglio G, Butturini G, Portella L, Guardascione G, Frigerio I, Montella M, Gobbo S, Martignoni G, Napolitano V, De Vita F, Tatangelo F, Franco R, Scala S. CXCR4-CXCL12-CXCR7 and PD-1/PD-L1 in Pancreatic Cancer: CXCL12 Predicts Survival of Radically Resected Patients. Cells 2022; 11:3340. [PMID: 36359736 PMCID: PMC9655815 DOI: 10.3390/cells11213340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/15/2022] [Accepted: 10/17/2022] [Indexed: 04/21/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is currently the most deadly cancer. Although characterized by 5-20% of neoplastic cells in the highly fibrotic stroma, immunotherapy is not a valid option in PDAC treatment. As CXCR4-CXCL12 regulates tumor invasion and T-cell access and PD-1/PD-L1 controls immune tolerance, 76 PDACs were evaluated for CXCR4-CXCL12-CXCR7 and PD-1/PD-L1 in the epithelial and stromal component. Neoplastic CXCR4 and CXCL12 discriminated PDACs for recurrence-free survival (RFS), while CXCL12 and CXCR7 discriminated patients for cancer-specific survival (CSS). Interestingly, among patients with radical resection (R0), high tumor CXCR4 clustered patients with worse RFS, high CXCL12 identified poor prognostic patients for both RFS and CSS, while stromal lymphocytic-monocytic PD-L1 associated with improved RFS and CSS. PD-1 was only sporadically expressed (<1%) in focal lymphocyte infiltrate and does not impact prognosis. In multivariate analysis, tumoral CXCL12, perineural invasion, and AJCC lymph node status were independent prognostic factors for RFS; tumoral CXCL12, AJCC Stage, and vascular invasion were independent prognostic factors for CSS. CXCL12's poor prognostic meaning was confirmed in an additional perspective-independent 13 fine-needle aspiration cytology advanced stage-PDACs. Thus, CXCR4-CXCL12 evaluation in PDAC identifies prognostic categories and could orient therapeutic approaches.
Collapse
Affiliation(s)
- Crescenzo D’Alterio
- Microenvironment Molecular Targets, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Alessandro Giardino
- Unit of HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy
| | - Giosuè Scognamiglio
- Pathology Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Giovanni Butturini
- Unit of HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy
| | - Luigi Portella
- Microenvironment Molecular Targets, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Giuseppe Guardascione
- Microenvironment Molecular Targets, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Isabella Frigerio
- Unit of HPB Surgery, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy
| | - Marco Montella
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Stefano Gobbo
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Guido Martignoni
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, 37019 Verona, Italy
| | - Vincenzo Napolitano
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Ferdinando De Vita
- Medical Oncology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Fabiana Tatangelo
- Pathology Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Stefania Scala
- Microenvironment Molecular Targets, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| |
Collapse
|
20
|
Liu Q, Ma Z, Cao Q, Zhao H, Guo Y, Liu T, Li J. Perineural invasion-associated biomarkers for tumor development. Biomed Pharmacother 2022; 155:113691. [PMID: 36095958 DOI: 10.1016/j.biopha.2022.113691] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Perineural invasion (PNI) is the process of neoplastic invasion of peripheral nerves and is considered to be the fifth mode of cancer metastasis. PNI has been detected in head and neck tumors and pancreatic, prostate, bile duct, gastric, and colorectal cancers. It leads to poor prognostic outcomes and high local recurrence rates. Despite the increasing number of studies on PNI, targeted therapeutic modalities have not been proposed. The identification of PNI-related biomarkers would facilitate the non-invasive and early diagnosis of cancers, the establishment of prognostic panels, and the development of targeted therapeutic approaches. In this review, we compile information on the molecular mediators involved in PNI-associated cancers. The expression and prognostic significance of molecular mediators and their receptors in PNI-associated cancers are analyzed, and the possible mechanisms of action of these mediators in PNI are explored, as well as the association of cells in the microenvironment where PNI occurs.
Collapse
Affiliation(s)
- Qi Liu
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Zhiming Ma
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Qian Cao
- Department of Education, The Second Hospital of Jilin University, Changchun 130041, China
| | - Hongyu Zhao
- Gastroenterology and Center of Digestive Endoscopy, The Second Hospital of Jilin University, Changchun 130041, China
| | - Yu Guo
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Tongjun Liu
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China.
| |
Collapse
|
21
|
Cui Zhou D, Jayasinghe RG, Chen S, Herndon JM, Iglesia MD, Navale P, Wendl MC, Caravan W, Sato K, Storrs E, Mo CK, Liu J, Southard-Smith AN, Wu Y, Naser Al Deen N, Baer JM, Fulton RS, Wyczalkowski MA, Liu R, Fronick CC, Fulton LA, Shinkle A, Thammavong L, Zhu H, Sun H, Wang LB, Li Y, Zuo C, McMichael JF, Davies SR, Appelbaum EL, Robbins KJ, Chasnoff SE, Yang X, Reeb AN, Oh C, Serasanambati M, Lal P, Varghese R, Mashl JR, Ponce J, Terekhanova NV, Yao L, Wang F, Chen L, Schnaubelt M, Lu RJH, Schwarz JK, Puram SV, Kim AH, Song SK, Shoghi KI, Lau KS, Ju T, Chen K, Chatterjee D, Hawkins WG, Zhang H, Achilefu S, Chheda MG, Oh ST, Gillanders WE, Chen F, DeNardo DG, Fields RC, Ding L. Spatially restricted drivers and transitional cell populations cooperate with the microenvironment in untreated and chemo-resistant pancreatic cancer. Nat Genet 2022; 54:1390-1405. [PMID: 35995947 PMCID: PMC9470535 DOI: 10.1038/s41588-022-01157-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 07/13/2022] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma is a lethal disease with limited treatment options and poor survival. We studied 83 spatial samples from 31 patients (11 treatment-naïve and 20 treated) using single-cell/nucleus RNA sequencing, bulk-proteogenomics, spatial transcriptomics and cellular imaging. Subpopulations of tumor cells exhibited signatures of proliferation, KRAS signaling, cell stress and epithelial-to-mesenchymal transition. Mapping mutations and copy number events distinguished tumor populations from normal and transitional cells, including acinar-to-ductal metaplasia and pancreatic intraepithelial neoplasia. Pathology-assisted deconvolution of spatial transcriptomic data identified tumor and transitional subpopulations with distinct histological features. We showed coordinated expression of TIGIT in exhausted and regulatory T cells and Nectin in tumor cells. Chemo-resistant samples contain a threefold enrichment of inflammatory cancer-associated fibroblasts that upregulate metallothioneins. Our study reveals a deeper understanding of the intricate substructure of pancreatic ductal adenocarcinoma tumors that could help improve therapy for patients with this disease.
Collapse
Affiliation(s)
- Daniel Cui Zhou
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Reyka G Jayasinghe
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Siqi Chen
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - John M Herndon
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Michael D Iglesia
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Pooja Navale
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, USA
| | - Michael C Wendl
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
- Department of Genetics, Washington University in St Louis, St Louis, MO, USA
- Department of Mathematics, Washington University in St Louis, St Louis, MO, USA
| | - Wagma Caravan
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Kazuhito Sato
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Erik Storrs
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Chia-Kuei Mo
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Jingxian Liu
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Austin N Southard-Smith
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Yige Wu
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Nataly Naser Al Deen
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - John M Baer
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, USA
| | - Robert S Fulton
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Ruiyang Liu
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Catrina C Fronick
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Lucinda A Fulton
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Andrew Shinkle
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Lisa Thammavong
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Houxiang Zhu
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Hua Sun
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Liang-Bo Wang
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Yize Li
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Chong Zuo
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Joshua F McMichael
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Sherri R Davies
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
| | | | - Keenan J Robbins
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Sara E Chasnoff
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
| | - Xiaolu Yang
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Ashley N Reeb
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Otolaryngology-Head & Neck Surgery, Washington University in St Louis, St Louis, MO, USA
| | - Clara Oh
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Mamatha Serasanambati
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Preet Lal
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Rajees Varghese
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Jay R Mashl
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Jennifer Ponce
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Nadezhda V Terekhanova
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Lijun Yao
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Fang Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lijun Chen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Schnaubelt
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rita Jui-Hsien Lu
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Julie K Schwarz
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
- Department of Radiation Oncology, Washington University in St Louis, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, MO, USA
| | - Sidharth V Puram
- Department of Otolaryngology-Head & Neck Surgery, Washington University in St Louis, St Louis, MO, USA
| | - Albert H Kim
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
- Department of Neurological Surgery, Washington University in St Louis, St Louis, MO, USA
| | - Sheng-Kwei Song
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Kooresh I Shoghi
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Ken S Lau
- Department of Cell and Developmental Biology and Epithelial Biology Center, Vanderbilt University School of Medicine, Vanderbilt, TN, USA
| | - Tao Ju
- Department of Computer Science and Engineering, Washington University in St Louis, St Louis, MO, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Deyali Chatterjee
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William G Hawkins
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samuel Achilefu
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Milan G Chheda
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Stephen T Oh
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, USA
| | - William E Gillanders
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Feng Chen
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - David G DeNardo
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA.
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, USA.
| | - Ryan C Fields
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA.
| | - Li Ding
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA.
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA.
- Department of Genetics, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
22
|
Yurteri Ü, Çifcibaşı K, Friess H, Ceyhan GO, Istvanffy R, Demir IE. Schwann Cells in Peripheral Cancers: Bystanders or Promoters? Adv Biol (Weinh) 2022; 6:e2200033. [PMID: 35656739 DOI: 10.1002/adbi.202200033] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/19/2022] [Indexed: 01/28/2023]
Abstract
The tumor microenvironment is subject to intense investigation in terms of its influence on tumorigenesis. Despite the fact that Schwann cells are cancer cells' early interaction partners, investigations on tumor progression and the molecular drivers of carcinogenesis do not place enough emphasis on them. Recent studies have shown that malignant cells and nerves interact on several levels during early carcinogenesis. For instance, the emergence of nerves in cancer, known as cancer neo-neurogenesis, is one important mechanism that contributes to cancer progression. Recent studies on Schwann cells brought the investigation of tumor-nerve interactions to a whole new level. Schwann cells make up the majority of glial cells in the peripheral nervous system, are outstandingly plastic cells, and serve a variety of roles in most organs. All these properties make Schwann cells excellent potential targets for tumor cells to exploit and turn them into promoters of carcinogenesis. In the present review, the distinctive features of Schwann cell-tumor cell interactions and the implications of this interaction on the tumor microenvironment are outlined. Further, this study points out the neglected aspects of Schwann cells in the tumor microenvironment and provides a potential new avenue for future research.
Collapse
Affiliation(s)
- Ümmügülsüm Yurteri
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, 81675, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, 81675, Munich, Germany.,CRC 1321 Modelling and Targeting Pancreatic Cancer, 81675, Munich, Germany
| | - Kaan Çifcibaşı
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, 81675, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, 81675, Munich, Germany.,CRC 1321 Modelling and Targeting Pancreatic Cancer, 81675, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, 81675, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, 81675, Munich, Germany.,CRC 1321 Modelling and Targeting Pancreatic Cancer, 81675, Munich, Germany
| | - Güralp O Ceyhan
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, 34752, Turkey
| | - Rouzanna Istvanffy
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, 81675, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, 81675, Munich, Germany.,CRC 1321 Modelling and Targeting Pancreatic Cancer, 81675, Munich, Germany
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, 81675, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, 81675, Munich, Germany.,CRC 1321 Modelling and Targeting Pancreatic Cancer, 81675, Munich, Germany.,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, 34752, Turkey.,Else Kröner Clinician Scientist Professor for Translational Pancreatic Surgery, 81675, Munich, Germany
| |
Collapse
|
23
|
Santi MD, Zhang M, Salvo E, Asam K, Viet C, Xie T, Amit M, Aouizerat B, Ye Y. Schwann Cells Induce Phenotypic Changes in Oral Cancer Cells. Adv Biol (Weinh) 2022; 6:e2200187. [PMID: 35925609 PMCID: PMC9474679 DOI: 10.1002/adbi.202200187] [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: 07/04/2022] [Revised: 07/18/2022] [Indexed: 01/28/2023]
Abstract
Head and neck cancer (HNC) is the seventh most common cancer worldwide, the majority being oral squamous cell carcinoma. Despite advances in cancer diagnosis and treatment, the survival rate of patients with HNC remains stagnant. The cancer-nerve interaction has been recognized as an important driver of cancer progression. Schwann cells, a type of peripheral glia, have been implicated in promoting cancer cell growth, migration, dispersion, and invasion into the nerve in many cancers. Here, it is demonstrated that the presence of Schwann cells makes oral cancer cells more aggressive by promoting their proliferation, extracellular matrix breakdown, and altering cell metabolism. Furthermore, oral cancer cells became larger, more circular, with more projections and nuclei following co-culturing with Schwann cells. RNA-sequencing analysis in oral cancer cells following exposure to Schwann cells shows corresponding changes in genes involved in the hallmarks of cancer and cell metabolism; the enriched KEGG pathways are spliceosome, RNA transport, cell cycle, axon guidance, signaling pathways regulating pluripotency of stem cells, cAMP signaling, WNT signaling, proteoglycans in cancer and PI3K-Akt signaling. Taken together, these results suggest a significant role for Schwann cells in facilitating oral cancer progression, highlighting their potential as a target to treat oral cancer progression.
Collapse
Affiliation(s)
- Maria Daniela Santi
- Bluestone Center for Clinical Research, College of Dentistry, New York University
- Department of Oral Maxillofacial Surgery, College of Dentistry, New York University
| | - Morgan Zhang
- Bluestone Center for Clinical Research, College of Dentistry, New York University
- Department of Oral Maxillofacial Surgery, College of Dentistry, New York University
| | - Elizabeth Salvo
- Bluestone Center for Clinical Research, College of Dentistry, New York University
- Department of Oral Maxillofacial Surgery, College of Dentistry, New York University
| | - Kesava Asam
- Bluestone Center for Clinical Research, College of Dentistry, New York University
- Department of Oral Maxillofacial Surgery, College of Dentistry, New York University
| | - Chi Viet
- Loma Linda University School of Dentistry, Loma Linda, CA92350
| | - Tongxin Xie
- Head & Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Moran Amit
- Head & Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Bradley Aouizerat
- Bluestone Center for Clinical Research, College of Dentistry, New York University
- Department of Oral Maxillofacial Surgery, College of Dentistry, New York University
| | - Yi Ye
- Bluestone Center for Clinical Research, College of Dentistry, New York University
- Department of Oral Maxillofacial Surgery, College of Dentistry, New York University
| |
Collapse
|
24
|
Skorupan N, Palestino Dominguez M, Ricci SL, Alewine C. Clinical Strategies Targeting the Tumor Microenvironment of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:4209. [PMID: 36077755 PMCID: PMC9454553 DOI: 10.3390/cancers14174209] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 12/04/2022] Open
Abstract
Pancreatic cancer has a complex tumor microenvironment which engages in extensive crosstalk between cancer cells, cancer-associated fibroblasts, and immune cells. Many of these interactions contribute to tumor resistance to anti-cancer therapies. Here, new therapeutic strategies designed to modulate the cancer-associated fibroblast and immune compartments of pancreatic ductal adenocarcinomas are described and clinical trials of novel therapeutics are discussed. Continued advances in our understanding of the pancreatic cancer tumor microenvironment are generating stromal and immune-modulating therapeutics that may improve patient responses to anti-tumor treatment.
Collapse
Affiliation(s)
- Nebojsa Skorupan
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Medical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mayrel Palestino Dominguez
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samuel L. Ricci
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christine Alewine
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
25
|
Chen YF, Wang SY, Le PH, Chen TH, Kuo CJ, Lin CJ, Chou WC, Yeh TS, Hsu JT. Prognostic Significance of Perineural Invasion in Patients with Stage II/III Gastric Cancer Undergoing Radical Surgery. J Pers Med 2022; 12:962. [PMID: 35743747 PMCID: PMC9224547 DOI: 10.3390/jpm12060962] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 12/17/2022] Open
Abstract
The prognostic significance of perineural invasion in patients with gastric cancer (GC) is controversial. This study aimed to determine the prognostic value of perineural invasion in patients with stage II/III GC undergoing radical surgery. A total of 1913 patients with stage II/III GC who underwent curative resection between 1994 and 2015 were recruited. Clinicopathological factors, tumor recurrence patterns, disease-free survival, and cancer-specific survival were compared in terms of perineural invasion. The prognostic factors of disease-free survival and cancer-specific survival were determined using univariate and multivariate analyses. Perineural invasion was found in 57.1% of the patients. Age of <65 years, female sex, large tumor size, upper tumor location, total gastrectomy, advanced tumor invasion depth and nodal involvement, greater metastatic to examined lymph node ratio, undifferentiated tumor, and presence of lymphatic or vascular invasion were significantly associated with perineural invasion. The patients with perineural invasion had higher locoregional/peritoneal recurrence rates than those without. Perineural invasion was independently associated with disease-free survival and cancer-specific survival. In conclusion, perineural invasion positivity is associated with aggressive tumor behaviors and higher locoregional/peritoneal recurrence rates in patients with stage II/III GC undergoing curative surgery. It is an independent unfavorable prognostic factor of disease recurrence and cancer-specific survival.
Collapse
Affiliation(s)
- Yi-Fu Chen
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (Y.-F.C.); (S.-Y.W.); (T.-S.Y.)
| | - Shan-Yu Wang
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (Y.-F.C.); (S.-Y.W.); (T.-S.Y.)
| | - Puo-Hsien Le
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (P.-H.L.); (T.-H.C.); (C.-J.K.); (C.-J.L.)
| | - Tsung-Hsing Chen
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (P.-H.L.); (T.-H.C.); (C.-J.K.); (C.-J.L.)
| | - Chia-Jung Kuo
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (P.-H.L.); (T.-H.C.); (C.-J.K.); (C.-J.L.)
| | - Chun-Jung Lin
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (P.-H.L.); (T.-H.C.); (C.-J.K.); (C.-J.L.)
| | - Wen-Chi Chou
- Department of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan;
| | - Ta-Sen Yeh
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (Y.-F.C.); (S.-Y.W.); (T.-S.Y.)
| | - Jun-Te Hsu
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (Y.-F.C.); (S.-Y.W.); (T.-S.Y.)
| |
Collapse
|
26
|
Szymoński K, Milian-Ciesielska K, Lipiec E, Adamek D. Current Pathology Model of Pancreatic Cancer. Cancers (Basel) 2022; 14:2321. [PMID: 35565450 PMCID: PMC9105915 DOI: 10.3390/cancers14092321] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 02/01/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most aggressive and lethal malignant neoplasms, ranking in seventh place in the world in terms of the incidence of death, with overall 5-year survival rates still below 10%. The knowledge about PC pathomechanisms is rapidly expanding. Daily reports reveal new aspects of tumor biology, including its molecular and morphological heterogeneity, explain complicated "cross-talk" that happens between the cancer cells and tumor stroma, or the nature of the PC-associated neural remodeling (PANR). Staying up-to-date is hard and crucial at the same time. In this review, we are focusing on a comprehensive summary of PC aspects that are important in pathologic reporting, impact patients' outcomes, and bring meaningful information for clinicians. Finally, we show promising new trends in diagnostic technologies that might bring a difference in PC early diagnosis.
Collapse
Affiliation(s)
- Krzysztof Szymoński
- Department of Pathomorphology, Jagiellonian University Medical College, 31-531 Cracow, Poland;
- Department of Pathomorphology, University Hospital, 30-688 Cracow, Poland;
| | | | - Ewelina Lipiec
- M. Smoluchowski Institute of Physics, Jagiellonian University, 30-348 Cracow, Poland;
| | - Dariusz Adamek
- Department of Pathomorphology, Jagiellonian University Medical College, 31-531 Cracow, Poland;
| |
Collapse
|
27
|
Goluba K, Kunrade L, Riekstina U, Parfejevs V. Schwann Cells in Digestive System Disorders. Cells 2022; 11:832. [PMID: 35269454 PMCID: PMC8908985 DOI: 10.3390/cells11050832] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/18/2022] Open
Abstract
Proper functioning of the digestive system is ensured by coordinated action of the central and peripheral nervous systems (PNS). Peripheral innervation of the digestive system can be viewed as intrinsic and extrinsic. The intrinsic portion is mainly composed of the neurons and glia of the enteric nervous system (ENS), while the extrinsic part is formed by sympathetic, parasympathetic, and sensory branches of the PNS. Glial cells are a crucial component of digestive tract innervation, and a great deal of research evidence highlights the important status of ENS glia in health and disease. In this review, we shift the focus a bit and discuss the functions of Schwann cells (SCs), the glial cells of the extrinsic innervation of the digestive system. For more context, we also provide information on the basic findings regarding the function of innervation in disorders of the digestive organs. We find diverse SC roles described particularly in the mouth, the pancreas, and the intestine. We note that most of the scientific evidence concerns the involvement of SCs in cancer progression and pain, but some research identifies stem cell functions and potential for regenerative medicine.
Collapse
Affiliation(s)
| | | | | | - Vadims Parfejevs
- Faculty of Medicine, University of Latvia, House of Science, Jelgavas Str. 3, LV-1004 Riga, Latvia; (K.G.); (L.K.); (U.R.)
| |
Collapse
|
28
|
Schwann Cells in the Tumor Microenvironment: Need More Attention. JOURNAL OF ONCOLOGY 2022; 2022:1058667. [PMID: 35186076 PMCID: PMC8853772 DOI: 10.1155/2022/1058667] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/20/2022] [Indexed: 12/13/2022]
Abstract
The tumor microenvironment (TME), which is composed of various cell components and signaling molecules, plays an important role in the occurrence and progression of tumors and has become the central issue of current cancer research. In recent years, as a part of the TME, the peripheral nervous system (PNS) has attracted increasing attention. Moreover, emerging evidence shows that Schwann cells (SCs), which are the most important glial cells in the PNS, are not simply spectators in the TME. In this review article, we focused on the up-to-date research progress on SCs in the TME and introduced our point of view. In detail, we described that under two main tumor-nerve interaction patterns, perineural invasion (PNI) and tumor innervation, SCs were reprogrammed and acted as important participants. We also investigated the newest mechanisms between the interactions of SCs and tumor cells. In addition, SCs can have profound impacts on other cellular components in the TME, such as immune cells and cancer-associated fibroblasts (CAFs), involving immune regulation, tumor-related pain, and nerve remodeling. Overall, these innovative statements can expand the scope of the TME, help fully understand the significant role of SCs in the tumor-nerve-immune axis, and propose enlightenments to innovate antitumor therapeutic methods and future research.
Collapse
|
29
|
Malik S, Westcott JM, Brekken RA, Burrows FJ. CXCL12 in Pancreatic Cancer: Its Function and Potential as a Therapeutic Drug Target. Cancers (Basel) 2021; 14:cancers14010086. [PMID: 35008248 PMCID: PMC8750050 DOI: 10.3390/cancers14010086] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Pancreatic cancer is a challenging disease to treat effectively. Fibroblasts associated with pancreatic cancer contribute to disease progression by secreting factors that enhance tumor cell survival and help tumor cells avoid detection by the immune system. This overview focuses on a chemokine, CXCL12, produced by cancer-associated fibroblasts and how CXCL12 signaling enhances pancreatic cancer progression by contributing to various hallmarks of cancer including, but not limited to, tumor growth and evasion of immune response. These pro-oncogenic functions of CXCL12 make it an attractive target in pancreatic cancer. We discuss the different approaches in development to therapeutically target CXCL12 and finally propose a novel approach, the use of the farnesyl transferase inhibitor tipifarnib to inhibit CXCL12 expression in pancreatic fibroblasts. Abstract Pancreatic ductal adenocarcinoma (PDAC) is a disease with limited therapeutic options and dismal long-term survival. The unique tumor environment of PDAC, consisting of desmoplastic stroma, immune suppressive cells, and activated fibroblasts, contributes to its resistance to therapy. Activated fibroblasts (cancer-associated fibroblasts and pancreatic stellate cells) secrete chemokines and growth factors that support PDAC growth, spread, chemoresistance, and immune evasion. In this review, we focus on one such chemokine, CXCL12, secreted by the cancer-associated fibroblasts and discuss its contribution to several of the classical hallmarks of PDAC and other tumors. We review the various therapeutic approaches in development to target CXCL12 signaling in PDAC. Finally, we propose an unconventional use of tipifarnib, a farnesyl transferase inhibitor, to inhibit CXCL12 production in PDAC.
Collapse
Affiliation(s)
| | - Jill M. Westcott
- Division of Surgical Oncology, Department of Surgery, and Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Rolf A. Brekken
- Division of Surgical Oncology, Department of Surgery, and Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Correspondence: (R.A.B.); (F.J.B.)
| | - Francis J. Burrows
- Kura Oncology, Inc., San Diego, CA 92130, USA;
- Correspondence: (R.A.B.); (F.J.B.)
| |
Collapse
|
30
|
Jones JO, Moody WM, Shields JD. Microenvironmental modulation of the developing tumour: an immune-stromal dialogue. Mol Oncol 2021; 15:2600-2633. [PMID: 32741067 PMCID: PMC8486574 DOI: 10.1002/1878-0261.12773] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/03/2020] [Accepted: 07/27/2020] [Indexed: 12/17/2022] Open
Abstract
Successful establishment of a tumour relies on a cascade of interactions between cancer cells and stromal cells within an evolving microenvironment. Both immune and nonimmune cellular components are key factors in this process, and the individual players may change their role from tumour elimination to tumour promotion as the microenvironment develops. While the tumour-stroma crosstalk present in an established tumour is well-studied, aspects in the early tumour or premalignant microenvironment have received less attention. This is in part due to the challenges in studying this process in the clinic or in mouse models. Here, we review the key anti- and pro-tumour factors in the early microenvironment and discuss how understanding this process may be exploited in the clinic.
Collapse
Affiliation(s)
- James O. Jones
- MRC Cancer UnitHutchison/MRC Research CentreUniversity of CambridgeCambridgeUK
- Department of OncologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - William M. Moody
- MRC Cancer UnitHutchison/MRC Research CentreUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
31
|
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: 8.0] [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
|
32
|
Xu H, Peng C, Chen XT, Yao YY, Chen LP, Yin Q, Shen W. Chemokine receptor CXCR4 activates the RhoA/ROCK2 pathway in spinal neurons that induces bone cancer pain. Mol Pain 2021; 16:1744806920919568. [PMID: 32349612 PMCID: PMC7227150 DOI: 10.1177/1744806920919568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Chemokine receptor CXCR4 has been found to be associated with spinal neuron and glial cell activation during bone cancer pain. However, the underlying mechanism remains unknown. Furthermore, the RhoA/ROCK2 pathway serves as a downstream pathway activated by CXCR4 during bone cancer pain. We first validated the increase in the expressions of CXCR4, p-RhoA, and p-ROCK2 in the spinal dorsal horn of a well-characterized tumor cell implantation-induced cancer pain rat model and how these expressions contributed to the pain behavior in tumor cell implantation rats. We hypothesized that spinal blockade of the CXCR4-RhoA/ROCK2 pathway is a potential analgesic therapy for cancer pain management. Methods Adult female Sprague–Dawley rats (body weight of 180–220 g) and six- to seven-week old female Sprague–Dawley rats (body weight of 80–90 g) were taken. Ascitic cancer cells were extracted from the rats (body weight of 80–90 g) with intraperitoneally implanted Walker 256 mammary gland carcinoma cells. Walker 256 rat mammary gland carcinoma cells were then injected (tumor cell implantation) into the intramedullary space of the tibia to establish a rat model of bone cancer pain. Results We found increased expressions of CXCR4, p-RhoA, and p-ROCK2 in the neurons in the spinal cord. p-RhoA and p-ROCK2 were co-expressed in the neurons and promoted by overexpressed CXCR4. Intrathecal delivery of CXCR4 inhibitor Plerixafor (AMD3100) or ROCK2 inhibitor Fasudil abrogated tumor cell implantation-induced pain hypersensitivity and tumor cell implantation-induced increase in p-RhoA and p-ROCK2 expressions. Intrathecal injection of stromal-derived factor-1, the principal ligand for CXCR4, accelerated p-RhoA expression in naive rats, which was prevented by postadministration of CXCR4 inhibitor Plerixafor (AMD3100) or ROCK2 inhibitor Fasudil. Conclusions Collectively, the spinal RhoA/ROCK2 pathway could be a critical downstream target for CXCR4-mediated neuronal sensitization and pain hypersensitivity in bone cancer pain, and it may serve as a potent therapeutic target for pain treatment.
Collapse
Affiliation(s)
- Heng Xu
- Department of Pain Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chong Peng
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xue-Tai Chen
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ying-Ying Yao
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Anesthesiology, Xuzhou First People's Hospital, Xuzhou, Jiangsu, China
| | - Li-Ping Chen
- Department of Pain Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qin Yin
- Department of Pain Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wen Shen
- Department of Pain Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
33
|
Abstract
The enteric nervous system (ENS) is the largest division of the peripheral nervous system and closely resembles components and functions of the central nervous system. Although the central role of the ENS in congenital enteric neuropathic disorders, including Hirschsprung disease and inflammatory and functional bowel diseases, is well acknowledged, its role in systemic diseases is less understood. Evidence of a disordered ENS has accumulated in neurodegenerative diseases ranging from amyotrophic lateral sclerosis, Alzheimer disease and multiple sclerosis to Parkinson disease as well as neurodevelopmental disorders such as autism. The ENS is a key modulator of gut barrier function and a regulator of enteric homeostasis. A 'leaky gut' represents the gateway for bacterial and toxin translocation that might initiate downstream processes. Data indicate that changes in the gut microbiome acting in concert with the individual genetic background can modify the ENS, central nervous system and the immune system, impair barrier function, and contribute to various disorders such as irritable bowel syndrome, inflammatory bowel disease or neurodegeneration. Here, we summarize the current knowledge on the role of the ENS in gastrointestinal and systemic diseases, highlighting its interaction with various key players involved in shaping the phenotypes. Finally, current flaws and pitfalls related to ENS research in addition to future perspectives are also addressed.
Collapse
|
34
|
Glia and Orofacial Pain: Progress and Future Directions. Int J Mol Sci 2021; 22:ijms22105345. [PMID: 34069553 PMCID: PMC8160907 DOI: 10.3390/ijms22105345] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/14/2022] Open
Abstract
Orofacial pain is a universal predicament, afflicting millions of individuals worldwide. Research on the molecular mechanisms of orofacial pain has predominately focused on the role of neurons underlying nociception. However, aside from neural mechanisms, non-neuronal cells, such as Schwann cells and satellite ganglion cells in the peripheral nervous system, and microglia and astrocytes in the central nervous system, are important players in both peripheral and central processing of pain in the orofacial region. This review highlights recent molecular and cellular findings of the glia involvement and glia–neuron interactions in four common orofacial pain conditions such as headache, dental pulp injury, temporomandibular joint dysfunction/inflammation, and head and neck cancer. We will discuss the remaining questions and future directions on glial involvement in these four orofacial pain conditions.
Collapse
|
35
|
Xu L, Li C, Hua F, Liu X. The CXCL12/CXCR7 signalling axis promotes proliferation and metastasis in cervical cancer. Med Oncol 2021; 38:58. [DOI: 10.1007/s12032-021-01481-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/13/2021] [Indexed: 02/07/2023]
|
36
|
Majidpoor J, Mortezaee K. Angiogenesis as a hallmark of solid tumors - clinical perspectives. Cell Oncol (Dordr) 2021; 44:715-737. [PMID: 33835425 DOI: 10.1007/s13402-021-00602-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Angiogenesis is a key and early step in tumorigenesis, and is known as a hallmark of solid tumors and a key promoter of tumor recurrence. Unlike normal tissue vessels, the architecture of the tumor vasculature is abnormal, being leaky, tortuous, fragile and blind-ended. Perivascular cells are either detached or absent, causing reduction of vascular integrity, an increase in vessel immaturity, incoherent perfusion, defective functionality and enhanced tumor dissemination and metastasis. The abnormal tumor vasculature along with the defective tumor vessel functionality finally causes bouts of hypoxia and acidity in the tumor microenvironment (TME), further reinvigorating tumor aggression. Interstitial hypertension or high interstitial fluid pressure (IFP) is an outcome of tumor hyper-permeability. High IFP can be a barrier for either effective delivery of anti-cancer drugs toward the TME or accumulation of drugs within the tumor area, thus promoting tumor resistance to therapy. Some tumors do, however, not undergo angiogenesis but instead undergo vessel co-option or vascular mimicry, thereby adding another layer of complexity to cancer development and therapy. CONCLUSIONS Combination of anti-angiogenesis therapy with chemotherapy and particularly with immune checkpoint inhibitors (ICIs) is a promising strategy for a number of advanced cancers. Among the various approaches for targeting tumor angiogenesis, vascular normalization is considered as the most desired method, which allows effective penetration of chemotherapeutics into the tumor area, thus being an appropriate adjuvant to other cancer modalities.
Collapse
Affiliation(s)
- Jamal Majidpoor
- Department of Anatomy, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Keywan Mortezaee
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
37
|
Demir IE, Mota Reyes C. Chemokines: the (un)usual suspects in pancreatic cancer neural invasion. Nat Rev Gastroenterol Hepatol 2021; 18:221-222. [PMID: 32561869 DOI: 10.1038/s41575-020-0329-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany. .,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey. .,CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.
| | - Carmen Mota Reyes
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.,CRC 1321 Modelling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| |
Collapse
|
38
|
TNFα promotes oral cancer growth, pain, and Schwann cell activation. Sci Rep 2021; 11:1840. [PMID: 33469141 PMCID: PMC7815837 DOI: 10.1038/s41598-021-81500-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Oral cancer is very painful and impairs a patient's ability to eat, talk, and drink. Mediators secreted from oral cancer can excite and sensitize sensory neurons inducing pain. Cancer mediators can also activate Schwann cells, the peripheral glia that regulates neuronal function and repair. The contribution of Schwann cells to oral cancer pain is unclear. We hypothesize that the oral cancer mediator TNFα activates Schwann cells, which further promotes cancer progression and pain. We demonstrate that TNFα is overexpressed in human oral cancer tissues and correlates with increased self-reported pain in patients. Antagonizing TNFα reduces oral cancer proliferation, cytokine production, and nociception in mice with oral cancer. Oral cancer or TNFα alone increases Schwann cell activation (measured by Schwann cell proliferation, migration, and activation markers), which can be inhibited by neutralizing TNFα. Cancer- or TNFα-activated Schwann cells release pro-nociceptive mediators such as TNFα and nerve growth factor (NGF). Activated Schwann cells induce nociceptive behaviors in mice, which is alleviated by blocking TNFα. Our study suggests that TNFα promotes cancer proliferation, progression, and nociception at least partially by activating Schwann cells. Inhibiting TNFα or Schwann cell activation might serve as therapeutic approaches for the treatment of oral cancer and associated pain.
Collapse
|
39
|
Saricaoglu ÖC, Teller S, Wang X, Wang S, Stupakov P, Heinrich T, Istvanffy R, Friess H, Ceyhan GO, Demir IE. Localisation analysis of nerves in the mouse pancreas reveals the sites of highest nerve density and nociceptive innervation. Neurogastroenterol Motil 2020; 32:e13880. [PMID: 32406093 DOI: 10.1111/nmo.13880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 03/03/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Neuropathy and neuro-inflammation drive the severe pain and disease progression in human chronic pancreatitis and pancreatic cancer. Mice, especially genetically induced-mouse models, have been increasingly utilized in mechanistic research on pancreatic neuropathy, but the normal "peripheral neurobiology" of the mouse pancreas has not yet been critically compared to human pancreas. METHODS We introduced a standardized tissue-harvesting technique that preserves the anatomic orientation of the mouse pancreas and allows complete sectioning in an anterior to posterior fashion. We applied immunohistochemistry and quantitative colorimetry of all nerves from the whole organ for studying pancreatic neuro-anatomy. KEY RESULTS Nerves in the mouse pancreas appeared as "clusters" of nerve trunks in contrast to singly distributed nerve trunks in the human pancreas. Nerve trunks in the mouse pancreas were exclusively found around intrapancreatic blood vessels, and around lymphoid structures. The majority of nerve trunks were located in the pancreatic head (0.15 ± 0.08% of tissue area) and the anterior/front surface of the corpus/body (0.17 ± 0.27%), thus significantly more than in the tail (0.02 ± 0.02%, P = .006). Nerves in the tail included a higher proportion of nociceptive fibers, but the absolute majority, ie, ca. 70%, of all nociceptive fibers, were localized in the head. Mice heterozygous for Bdnf knockout allele (Bdnf+/- ) exhibited enrichment of nitrergic nerve fibers specifically in the head and corpus. CONCLUSIONS & INFERENCES Neuro-anatomy of the "mesenteric type" mouse pancreas is highly different from the "compact" human pancreas. Studies that aim at reproducing human pancreatic neuro-phenomena in mouse models should pay diligent attention to these anatomic differences.
Collapse
Affiliation(s)
- Ömer Cemil Saricaoglu
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Steffen Teller
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Xiaobo Wang
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Shenghan Wang
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Pavel Stupakov
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Tobias Heinrich
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Rouzanna Istvanffy
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Güralp O Ceyhan
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.,German Cancer Consortium (DKTK), Partner Site, Munich, Germany.,CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany
| |
Collapse
|
40
|
Mejia I, Bodapati S, Chen KT, Díaz B. Pancreatic Adenocarcinoma Invasiveness and the Tumor Microenvironment: From Biology to Clinical Trials. Biomedicines 2020; 8:E401. [PMID: 33050151 PMCID: PMC7601142 DOI: 10.3390/biomedicines8100401] [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: 08/31/2020] [Revised: 09/28/2020] [Accepted: 10/03/2020] [Indexed: 12/18/2022] Open
Abstract
Pancreatic adenocarcinoma (PDAC) originates in the glandular compartment of the exocrine pancreas. Histologically, PDAC tumors are characterized by a parenchyma that is embedded in a particularly prominent stromal component or desmoplastic stroma. The unique characteristics of the desmoplastic stroma shape the microenvironment of PDAC and modulate the reciprocal interactions between cancer and stromal cells in ways that have profound effects in the pathophysiology and treatment of this disease. Here, we review some of the most recent findings regarding the regulation of PDAC cell invasion by the unique microenvironment of this tumor, and how new knowledge is being translated into novel therapeutic approaches.
Collapse
Affiliation(s)
- Isabel Mejia
- Department of Medicine, Division of Medical Hematology Oncology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
| | - Sandhya Bodapati
- College of Osteopathic Medicine, Pacific Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Kathryn T. Chen
- Department of Surgery, Division of Surgical Oncology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
| | - Begoña Díaz
- Department of Medicine, Division of Medical Hematology Oncology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
41
|
Hessmann E, Buchholz SM, Demir IE, Singh SK, Gress TM, Ellenrieder V, Neesse A. Microenvironmental Determinants of Pancreatic Cancer. Physiol Rev 2020; 100:1707-1751. [DOI: 10.1152/physrev.00042.2019] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) belongs to the most lethal solid tumors in humans. A histological hallmark feature of PDAC is the pronounced tumor microenvironment (TME) that dynamically evolves during tumor progression. The TME consists of different non-neoplastic cells such as cancer-associated fibroblasts, immune cells, endothelial cells, and neurons. Furthermore, abundant extracellular matrix components such as collagen and hyaluronic acid as well as matricellular proteins create a highly dynamic and hypovascular TME with multiple biochemical and physical interactions among the various cellular and acellular components that promote tumor progression and therapeutic resistance. In recent years, intensive research efforts have resulted in a significantly improved understanding of the biology and pathophysiology of the TME in PDAC, and novel stroma-targeted approaches are emerging that may help to improve the devastating prognosis of PDAC patients. However, none of anti-stromal therapies has been approved in patients so far, and there is still a large discrepancy between multiple successful preclinical results and subsequent failure in clinical trials. Furthermore, recent findings suggest that parts of the TME may also possess tumor-restraining properties rendering tailored therapies even more challenging.
Collapse
Affiliation(s)
- Elisabeth Hessmann
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Soeren M. Buchholz
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Ihsan Ekin Demir
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Shiv K. Singh
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Thomas M. Gress
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Volker Ellenrieder
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Albrecht Neesse
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| |
Collapse
|
42
|
Zhou Y, Li J, Han B, Zhong R, Zhong H. Schwann cells promote lung cancer proliferation by promoting the M2 polarization of macrophages. Cell Immunol 2020; 357:104211. [PMID: 32977156 DOI: 10.1016/j.cellimm.2020.104211] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/13/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022]
Abstract
The interplay between immune cells and tumor cells determines the fate of tumorigenesis. Targeting the abnormal immune response of tumors has been recently achieved great success in some patients. Emerging evidence demonstrated the nervous system plays vital roles in immune regulation, but if the nervous system affects the immune-tumor response and the possible mechanism involved remain largely unexplored. Here, we report that Schwann cells, the major component of the peripheral nervous system (PNS), induce M2 polarization of macrophages by secreting cytokines and chemokines, and these polarized macrophages promote the proliferation of lung cancer cells. We cocultured peripheral blood mononuclear cells (PBMCs) with Schwann cells or treated PBMCs with the culture supernatant of Schwann cells. We found that both treatments induced M2 polarization of the macrophages in peripheral blood mononuclear cell cultures. We performed a bioinformatic analysis of the transcriptome of Schwann cells and analyzed cytokines and chemokines by ELISAs. We found that Schwann cells secreted high levels of CCL2, CXCL5, CXCL12, and CXCL8. CCL2 promotes the M2 polarization of macrophages. Furthermore, we isolated CD14-positive macrophages that were cocultured with the Schwann cells and treated A549 and H1299 lung cancer cells with these macrophages. We found that the Schwann cell-polarized macrophages increased the proliferation of the lung cancer cells. Our study sheds new light on the involvement of the PNS in the regulation of tumor progression via a "Schwann cell"-"immune cell"-"tumor cell" axis.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jingwen Li
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Baohui Han
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Runbo Zhong
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Hua Zhong
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
43
|
Anbil S, Pigula M, Huang HC, Mallidi S, Broekgaarden M, Baglo Y, De Silva P, Simeone DM, Mino-Kenudson M, Maytin EV, Rizvi I, Hasan T. Vitamin D Receptor Activation and Photodynamic Priming Enables Durable Low-dose Chemotherapy. Mol Cancer Ther 2020; 19:1308-1319. [PMID: 32220968 DOI: 10.1158/1535-7163.mct-19-0791] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/08/2019] [Accepted: 03/12/2020] [Indexed: 12/18/2022]
Abstract
Patients with cancer often confront the decision of whether to continue high-dose chemotherapy at the expense of cumulative toxicities. Reducing the dose of chemotherapy regimens while preserving efficacy is sorely needed to preserve the performance status of these vulnerable patients, yet has not been prioritized. Here, we introduce a dual pronged approach to modulate the microenvironment of desmoplastic pancreatic tumors and enable significant dose deescalation of the FDA-approved chemotherapeutic nanoliposomal irinotecan (nal-IRI) without compromising tumor control. We demonstrate that light-based photodynamic priming (PDP) coupled with vitamin D3 receptor (VDR) activation within fibroblasts increases intratumoral nal-IRI accumulation and suppresses protumorigenic CXCL12/CXCR7 crosstalk. Combined photodynamic and biochemical modulation of the tumor microenvironment enables a 75% dose reduction of nal-IRI while maintaining treatment efficacy, resulting in improved tolerability. Modifying the disease landscape to increase the susceptibility of cancer, via preferentially modulating fibroblasts, represents a promising and relatively underexplored strategy to enable dose deescalation. The approach presented here, using a combination of three clinically available therapies with nonoverlapping toxicities, can be rapidly translated with minimal modification to treatment workflow, and challenges the notion that significant improvements in chemotherapy efficacy can only be achieved at the expense of increased toxicity.
Collapse
Affiliation(s)
- Sriram Anbil
- Long School of Medicine, UT Health San Antonio, San Antonio, Texas.,Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Michael Pigula
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Huang-Chiao Huang
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mans Broekgaarden
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yan Baglo
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pushpamali De Silva
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Diane M Simeone
- Department of Surgery, Langone School of Medicine, New York University, New York, New York
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Edward V Maytin
- Department of Dermatology and Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio
| | - Imran Rizvi
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts. .,Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
44
|
Mortezaee K. CXCL12/CXCR4 axis in the microenvironment of solid tumors: A critical mediator of metastasis. Life Sci 2020; 249:117534. [PMID: 32156548 DOI: 10.1016/j.lfs.2020.117534] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/24/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023]
Abstract
Tumors are dynamic tissue masses, so requiring continuous exposure to the host cells, nurturing them into pave a path for tumor growth and metastasis. C-X-C chemokine ligand 12 (CXCL12)/C-X-C chemokine receptor type 4 (CXCR4) is the key signaling for such aim. Gathering knowledge about the activity within this axis would deepen our insight into the utmost importance this signaling taken to attract and cross-connect multiple cells within the tumor microenvironment (TME) aiming for tumor progression and metastasis. The concept behind this review is to underscore the multi-tasking roles taken by CXCL12/CXCR4 signaling in tumor metastasis, and to also suggest some strategies to target the activities within this axis.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
45
|
Ni T, Huang T, Gu SL, Wang J, Liu Y, Sun X, Wang YD. DRG Neurons Promote Perineural Invasion of Endometrial Cancer via GluR2. J Cancer 2020; 11:2518-2528. [PMID: 32201522 PMCID: PMC7066017 DOI: 10.7150/jca.40055] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/19/2020] [Indexed: 01/28/2023] Open
Abstract
Background: Perineural invasion (PNI) is correlated with negative prognosis in multiple cancers, but its role in endometrial cancer (EC) is still largely unknown; thus, targeted treatment for nerve infiltration is lacking as well. Methods: The interaction between nerve and EC cells were investigated by in vitro neural invasion assay and transwell coculture system. Then the nerve-related receptor gene glutamate ionotropic receptor AMPA type subunit 2 (GRIA2) was detected in EC tissues and cells using PCR array, western blotting, and immunohistochemistry. The role of GluR2 (gene name GRIA2) on EC proliferation, migration and invasion was evaluated by a GluR2 antagonist and shRNA. At the same time, the neurotransmitter effect on GluR2 (glutamate) from the cocultured conditional medium was measured using high-performance liquid chromatography (HPLC). Results: EC cell line Ishikawa (ISK) showed the ability to migrate along neurites in vitro and the numbers of migrated/invaded EC cells in the DRG neuron coculture group were significantly increased. The expression of GluR2 in EC tissue was found to be higher than that in para-carcinoma tissue. After GluR2 antagonist and GluR2 shRNA treatment, the proliferation, migration and invasion of ISK cells was markedly inhibited. Moreover, the ability of DRG neurons to promote the migration and invasion of ISK cells could also be attenuated by downregulation of GluR2, and the concentration of the neurotransmitter glutamate was notably increased in the coculture conditional medium compared to that in the DRG neuron or ISK cells alone. Conclusions: DRG neurons promote metastasis of EC cells via GluR2, which might be a risk factor for PNI in EC. Moreover, the perineural system may promote tumor invasion and metastasis under certain circumstances.
Collapse
Affiliation(s)
- Ting Ni
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, No. 910 Hengshan Road, Shanghai 200030, China
| | - Ting Huang
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, No. 910 Hengshan Road, Shanghai 200030, China
| | - Sheng-Lan Gu
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, No. 910 Hengshan Road, Shanghai 200030, China
| | - Jing Wang
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, No. 910 Hengshan Road, Shanghai 200030, China
| | - Yao Liu
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, No. 910 Hengshan Road, Shanghai 200030, China
| | - Xiao Sun
- Laboratory for Gynecologic Oncology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, No. 910 Hengshan Road, Shanghai 200030, China
| | - Yu-Dong Wang
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, No. 910 Hengshan Road, Shanghai 200030, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China
| |
Collapse
|
46
|
Huang T, Fan Q, Wang Y, Cui Y, Wang Z, Yang L, Sun X, Wang Y. Schwann Cell-Derived CCL2 Promotes the Perineural Invasion of Cervical Cancer. Front Oncol 2020; 10:19. [PMID: 32064233 PMCID: PMC7000531 DOI: 10.3389/fonc.2020.00019] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
Perineural invasion (PNI) has guiding significances for nerve preservation in cervical cancer, but there is no definite marker indicating PNI. Two cervical cancer cell lines (HeLa and ME-180) showed significant abilities to migrate along neurites in vitro and in vivo. Morphological observation revealed that Schwann cells (SC) arrived at the sites of cervical cancer cells before the onset of cancer metastasis. We used high-throughput antibody array to screen the signals mediating the interaction of nerve cells and cancer cells and found the high expression of CCL2 in dorsal root ganglion (DRG). Meanwhile, serum CCL2 showed a notable raise especially in cervical adenocarcinoma. SC-derived CCL2 bound to its receptor CCR2 and promoted the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of cervical cancer cells. In turn, cancer cell-derived signals triggered the expression of metalloproteinases (MMPs) including MMP2, MMP9, and MMP12 in SCs, promoting SCs to dissolve matrix. These data demonstrated that the cancer-nerve crosstalk formed a tumor microenvironment (TME) that facilitated to PNI. We identified the CCL2/CCR2 axis as a potential marker to predict the PNI and affect the nerve preservation for cervical cancer.
Collapse
Affiliation(s)
- Ting Huang
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiong Fan
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Public Health Clinical Center, Female Tumor Reproductive Specialty, Shanghai, China
| | - Yiwei Wang
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunxia Cui
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihua Wang
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Linlin Yang
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Sun
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Public Health Clinical Center, Female Tumor Reproductive Specialty, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Yudong Wang
- Department of Gynecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Shanghai, China.,Shanghai Public Health Clinical Center, Female Tumor Reproductive Specialty, Shanghai, China
| |
Collapse
|
47
|
Han L, Jiang J, Xue M, Qin T, Xiao Y, Wu E, Shen X, Ma Q, Ma J. Sonic hedgehog signaling pathway promotes pancreatic cancer pain via nerve growth factor. Reg Anesth Pain Med 2019; 45:137-144. [PMID: 31792027 DOI: 10.1136/rapm-2019-100991] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/30/2019] [Accepted: 11/03/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Many patients with pancreatic cancer (PC) suffer from abdominal pain and back pain. However, the cause of pain associated with PC is largely unclear. In this study, we tested the potential influence of the sonic hedgehog (sHH) signaling pathway on PC pain. METHODS Substance P (SP) and calcitonin gene-related peptide (CGRP) expression was measured in cultured PC cells and dorsal root ganglions (DRG) by real-time PCR, western blotting analysis and ELISA. Small interfering RNA transfection and plasmid constructs were used to regulate the expression of sHH in the AsPc-1 and Panc-1 cell lines. Pain-related behavior was observed in an orthotopic tumor model in nude mice. RESULTS In this study, the results show that sHH increased the expression of SP and CGRP in DRGs in a concentration and time-dependent manner. Additionally, sHH secretion from PC cells could activate the sHH signaling pathway and, in turn, increase the expression of nerve growth factor (NGF), P75, and TrkA in DRGs. Furthermore, the sHH signaling pathway and NGF/NGF receptor contributed to pain sensitivity in a nude mouse model. CONCLUSION Our results demonstrate that PC pain originates from the sHH signaling pathway, and NGF mediates the pain mechanism via regulating SP and CGRP.
Collapse
Affiliation(s)
- Liang Han
- Department of Hepatobiliary Surgery, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, Shaanxi, China
| | - Jie Jiang
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Mengwen Xue
- Department of Anesthesiology, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, China
| | - Tao Qin
- Department of Hepatobiliary Surgery, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, Shaanxi, China
| | - Ying Xiao
- Department of Hepatobiliary Surgery, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, Shaanxi, China
| | - Erxi Wu
- Department of Surgery, Baylor Scott and White Health, Dallas, Texas, USA.,Department of Neurosurgery, Baylor Scott and White Health, Dallas, Texas, USA
| | - Xin Shen
- Department of Anesthesiology, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, Shaanxi, China
| | - Jiguang Ma
- Department of Anesthesiology, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, China
| |
Collapse
|
48
|
Roger E, Martel S, Bertrand-Chapel A, Depollier A, Chuvin N, Pommier RM, Yacoub K, Caligaris C, Cardot-Ruffino V, Chauvet V, Aires S, Mohkam K, Mabrut JY, Adham M, Fenouil T, Hervieu V, Broutier L, Castets M, Neuzillet C, Cassier PA, Tomasini R, Sentis S, Bartholin L. Schwann cells support oncogenic potential of pancreatic cancer cells through TGFβ signaling. Cell Death Dis 2019; 10:886. [PMID: 31767842 PMCID: PMC6877617 DOI: 10.1038/s41419-019-2116-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/09/2019] [Accepted: 11/05/2019] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the solid tumors with the poorest prognosis. The stroma of this tumor is abundant and composed of extracellular matrix and stromal cells (including cancer-associated fibroblasts and immune cells). Nerve fibers invading this stroma represent a hallmark of PDAC, involved in neural remodeling, which participates in neuropathic pain, cancer cell dissemination and tumor relapse after surgery. Pancreatic cancer-associated neural remodeling is regulated through functional interplays mediated by physical and molecular interactions between cancer cells, nerve cells and surrounding Schwann cells, and other stromal cells. In the present study, we show that Schwann cells (glial cells supporting peripheral neurons) can enhance aggressiveness (migration, invasion, tumorigenicity) of pancreatic cancer cells in a transforming growth factor beta (TGFβ)-dependent manner. Indeed, we reveal that conditioned medium from Schwann cells contains high amounts of TGFβ able to activate the TGFβ-SMAD signaling pathway in cancer cells. We also observed in human PDAC samples that high levels of TGFβ signaling activation were positively correlated with perineural invasion. Secretome analyses by mass spectrometry of Schwann cells and pancreatic cancer cells cultured alone or in combination highlighted the central role of TGFβ in neuro-epithelial interactions, as illustrated by proteomic signatures related to cell adhesion and motility. Altogether, these results demonstrate that Schwann cells are a meaningful source of TGFβ in PDAC, which plays a crucial role in the acquisition of aggressive properties by pancreatic cancer cells.
Collapse
Affiliation(s)
- Elodie Roger
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Sylvie Martel
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Adrien Bertrand-Chapel
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Arnaud Depollier
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Nicolas Chuvin
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Roxane M Pommier
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Karam Yacoub
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Cassandre Caligaris
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Victoire Cardot-Ruffino
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Véronique Chauvet
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Sophie Aires
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Kayvan Mohkam
- Hospices Civils de Lyon, Croix Rousse hospital, Claude-Bernard Lyon 1 University, Department of General Surgery & Liver Transplantation, Lyon, France
| | - Jean-Yves Mabrut
- Hospices Civils de Lyon, Croix Rousse hospital, Claude-Bernard Lyon 1 University, Department of General Surgery & Liver Transplantation, Lyon, France
| | - Mustapha Adham
- Hospices Civils de Lyon, Edouard Herriot hospital, Claude-Bernard Lyon 1 University, Department of General Surgery & Liver Transplantation, Lyon, France
| | - Tanguy Fenouil
- Hospices Civils de Lyon Institute of Pathology EST, CRCL INSERM U1052, University Lyon 1, Lyon, France
| | - Valérie Hervieu
- Hospices Civils de Lyon Institute of Pathology EST, CRCL INSERM U1052, University Lyon 1, Lyon, France
| | - Laura Broutier
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Marie Castets
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Cindy Neuzillet
- Medical Oncology Department, Curie Institute, Versailles Saint-Quentin University, 35 rue Dailly, 92210, Saint Cloud, France
| | - Philippe A Cassier
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France.,Departement d'Oncologie Médicale, Centre Léon Bérard, Lyon, 69008, France
| | - Richard Tomasini
- Aix-Marseille Université, Institut Paoli-Calmettes, INSERM U1068, CNRS UMR 7258, Centre de Recherche en Cancérologie de Marseille, Marseille, France
| | - Stéphanie Sentis
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Laurent Bartholin
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France.
| |
Collapse
|
49
|
Miura T, Mitsunaga S, Ikeda M, Ohno I, Takahashi H, Kuwata T, Ochiai A. Neural Invasion Spreads Macrophage-Related Allodynia via Neural Root in Pancreatic Cancer. Anesth Analg 2019; 126:1729-1738. [PMID: 29570153 DOI: 10.1213/ane.0000000000002803] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Neural invasion (N-inv) induces the neural damage and pain in pancreatic cancer (PCa). Benign nerve injury evokes allodynia through neuroinflammation in the neural root, which might be seen in PCa. Macrophages have the potential to release excitatory cytokines after nerve injury and so may play a role in the generation of chronic neuropathic pain. The aim of this study is to represent N-inv-induced allodynia in patients with PCa and to characterize allodynia-related neuroinflammation as macrophage accumulation on dorsal root ganglion (DRG) in the N-inv animal model (N-inv model). METHODS Treatment-naïve patients with advanced PCa with no opioid use were enrolled in the clinical study. To evaluate allodynia, the current perception threshold on epigastric skin and pain score from questionnaire were measured. The association between the degrees of radiological N-inv and allodynia was evaluated. In the animal experiments, we used the N-inv model, which is established by the inoculation of the human PCa cell line into the left sciatic nerve of mice and mimics the invasion behavior of human PCa. The change of sensation was weekly measured at right hind paw, and the expressions of mRNA and protein were investigated on DRG at 6 weeks in the N-inv and sham models. The effect of macrophage depletion using liposome-encapsulated clodronate (Lp-CLD) was evaluated in the N-inv model. Tumor size and the degree of macrophage accumulation on DRG or around the tumor were investigated. RESULTS In the clinical study, 43 patients were analyzed. The threshold of epigastric skin at 2000 Hz touch and pressure sensation was decreased in patients with severe N-inv, compared to patients without severe N-inv. Patients with severe N-inv showed a high pain score. In the animal experiments, the N-inv model decreased the threshold of right hind paw at 5 and 6 weeks. The macrophage-related gene expression and F4/80-positive macrophages were increased in the left DRG. Lp-CLD-induced macrophage depletion induced an increase of the threshold in the right hind paw and a decrease of CD206-positive macrophages accumulation in the left DRG. Lp-CLD had no effect for tumor size. CONCLUSIONS The present study first showed that the N-inv-induced allodynia was spread in patients with PCa and in the N-inv model. Allodynia was related to the amount of macrophages at DRG in the N-inv model. The neuroinflammation may be a target for researching the N-inv-induced pain mechanism and developing novel analgesics.
Collapse
Affiliation(s)
- Tomofumi Miura
- From the Division of Biomarker Discovery, Exploratory Oncology Research and Clinical Trial Center.,Departments of Palliative Medicine
| | - Shuichi Mitsunaga
- From the Division of Biomarker Discovery, Exploratory Oncology Research and Clinical Trial Center.,Hepatobiliary and Pancreatic Oncology
| | | | | | | | - Takeshi Kuwata
- Pathology and Clinical Laboratories, National Cancer Hospital East, Kashiwa, Japan
| | - Atsushi Ochiai
- From the Division of Biomarker Discovery, Exploratory Oncology Research and Clinical Trial Center
| |
Collapse
|
50
|
Saloman JL, Albers KM, Cruz-Monserrate Z, Davis BM, Edderkaoui M, Eibl G, Epouhe AY, Gedeon JY, Gorelick FS, Grippo PJ, Groblewski GE, Husain SZ, Lai KK, Pandol SJ, Uc A, Wen L, Whitcomb DC. Animal Models: Challenges and Opportunities to Determine Optimal Experimental Models of Pancreatitis and Pancreatic Cancer. Pancreas 2019; 48:759-779. [PMID: 31206467 PMCID: PMC6581211 DOI: 10.1097/mpa.0000000000001335] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
At the 2018 PancreasFest meeting, experts participating in basic research met to discuss the plethora of available animal models for studying exocrine pancreatic disease. In particular, the discussion focused on the challenges currently facing the field and potential solutions. That meeting culminated in this review, which describes the advantages and limitations of both common and infrequently used models of exocrine pancreatic disease, namely, pancreatitis and exocrine pancreatic cancer. The objective is to provide a comprehensive description of the available models but also to provide investigators with guidance in the application of these models to investigate both environmental and genetic contributions to exocrine pancreatic disease. The content covers both nongenic and genetically engineered models across multiple species (large and small). Recommendations for choosing the appropriate model as well as how to conduct and present results are provided.
Collapse
Affiliation(s)
- Jami L. Saloman
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA
| | - Kathryn M. Albers
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition; Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Brian M. Davis
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA
| | - Mouad Edderkaoui
- Basic and Translational Pancreas Research, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA
| | - Ariel Y. Epouhe
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA
| | - Jeremy Y. Gedeon
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA
| | - Fred S. Gorelick
- Department of Internal Medicine, Section of Digestive Diseases & Department of Cell Biology Yale University School of Medicine; Veterans Affairs Connecticut Healthcare, West Haven, CT
| | - Paul J. Grippo
- Department of Medicine, Division of Gastroenterology and Hepatology, UI Cancer Center, University of Illinois at Chicago, Chicago, IL
| | - Guy E. Groblewski
- Department of Nutritional Sciences, University of Wisconsin, Madison, WI
| | | | - Keane K.Y. Lai
- Department of Pathology (National Medical Center), Department of Molecular Medicine (Beckman Research Institute), and Comprehensive Cancer Center, City of Hope, Duarte, CA
| | - Stephen J. Pandol
- Department of Surgery, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA
| | - Aliye Uc
- Stead Family Department of Pediatrics, University of Iowa, Stead Family Children’s Hospital, Iowa City, IA
| | - Li Wen
- Department of Pediatrics, Stanford University, Palo Alto, CA
| | | |
Collapse
|