201
|
Hu S, Zhang J, Guo G, Zhang L, Dai J, Gao Y. Comprehensive analysis of GSEC/miR-101-3p/SNX16/PAPOLG axis in hepatocellular carcinoma. PLoS One 2022; 17:e0267117. [PMID: 35482720 PMCID: PMC9049542 DOI: 10.1371/journal.pone.0267117] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 04/03/2022] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignancies. A growing number of studies have shown that competitive endogenous RNA (ceRNA) regulatory networks might play important roles during HCC process. The present study aimed to identify a regulatory axis of the ceRNA network associated with the development of HCC. The roles of SNX16 and PAPOLG in HCC were comprehensively analyzed using bioinformatics tools. Subsequently, the “mRNA-miRNA-lncRNA” model was then used to predict the upstream miRNAs and lncRNAs of SNX16 and PAPOLG using the miRNet database, and the miRNAs with low expression and good prognosis in HCC and the lncRNAs with high expression and poor prognosis in HCC were screened by differential expression and survival analysis. Finally, the risk-prognosis models of ceRNA network axes were constructed by univariate and multifactorial Cox proportional risk analysis, and the immune correlations of ceRNA network axes were analyzed using the TIMER and GEPIA database. In this study, the relevant ceRNA network axis GSEC/miR-101-3p/SNX16/PAPOLG with HCC prognosis was constructed, in which GSEC, SNX16, and PAPOLG were highly expressed in HCC with poor prognosis, while miR-101-3p was lowly expressed in HCC with good prognosis. The risk-prognosis model predicted AUC of 0.691, 0.623, and 0.626 for patient survival at 1, 3, and 5 years, respectively. Immuno-infiltration analysis suggested that the GSEC/miR-101-3p/SNX16/PAPOLG axis might affect macrophage polarization. The GSEC/miR-101-3p/SNX16/PAPOLG axis of the ceRNA network axis might be an important factor associated with HCC prognosis and immune infiltration.
Collapse
Affiliation(s)
- Shangshang Hu
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China
| | - Jinyan Zhang
- School of Life Science, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, China
| | - Guoqing Guo
- School of Life Science, Bengbu Medical College, Bengbu, China
| | - Li Zhang
- School of Life Science, Bengbu Medical College, Bengbu, China
| | - Jing Dai
- School of Life Science, Bengbu Medical College, Bengbu, China
| | - Yu Gao
- School of Life Science, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, China
- * E-mail:
| |
Collapse
|
202
|
Lawther AJ, Phillips AJK, Chung NC, Chang A, Ziegler AI, Debs S, Sloan EK, Walker AK. Disrupting circadian rhythms promotes cancer-induced inflammation in mice. Brain Behav Immun Health 2022; 21:100428. [PMID: 35199050 PMCID: PMC8851215 DOI: 10.1016/j.bbih.2022.100428] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 02/09/2023] Open
Abstract
Disruption of circadian rhythms occurs in rotating shift-work, jetlag, and in individuals with irregular sleep schedules. Circadian disruption is known to alter inflammatory responses and impair immune function. However, there is limited understanding of how circadian disruption modulates cancer-induced inflammation. Inflammation is a hallmark of cancer and is linked to worse prognosis and impaired brain function in cancer patients. Here, we investigated the effect of circadian disruption on cancer-induced inflammation in an orthotopic breast cancer model. Using a validated chronic jetlag protocol that advances the light-cycle by 8 h every 2 days to disrupt circadian rhythms, we found that circadian disruption alters cancer-induced inflammation in a tissue-specific manner, increasing inflammation in the body and brain while decreasing inflammation within the tumor tissue. Circadian disruption did not affect inflammation in mice without tumors, suggesting that the impact of circadian disruption may be particularly detrimental in the context of underlying inflammatory conditions, such as cancer. Importantly, circadian disruption did not affect tumor burden, suggesting that increased inflammation was not a result of increased cancer progression. Overall, these findings identify the importance of healthy circadian rhythms for limiting cancer-induced inflammation. Circadian disruption enhances cancer-induced inflammation in the body and brain. The profile of inflammatory cytokines altered by circadian disruption is tissue specific. Changes in inflammatory profiles by circadian disruption are not due to enhanced tumor burden.
Collapse
Affiliation(s)
- Adam J Lawther
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia
| | - Andrew J K Phillips
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Ni-Chun Chung
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Aeson Chang
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Alexandra I Ziegler
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Sophie Debs
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, VIC, 3002, Australia
| | - Adam K Walker
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia.,Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,School of Psychiatry, University of New South Wales, Kensington, NSW, 2033, Australia
| |
Collapse
|
203
|
Zhou J, Liu L, Hu X, Feng R, Zhao N, Zhang L, Hu W, Zhang J, Huang S, Liu L, Li W, Shan Y, Jin J. Matrix metalloproteinase-21 promotes metastasis via increasing the recruitment and M2 polarization of macrophages in HCC. Cancer Sci 2022; 114:423-435. [PMID: 35398966 PMCID: PMC9899621 DOI: 10.1111/cas.15368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 03/30/2022] [Accepted: 04/03/2022] [Indexed: 12/24/2022] Open
Abstract
MMP-21 is a newly identified member of the matrix metalloproteinase family and has been reported to regulate both embryonic development and tumor progression. However, the roles of MMP-21 in hemofiltrate C-C chemokine (HCC) remain largely unclear. In this study, we used western blot, qPCR and immunohistochemistry (IHC) to determine the upregulation of MMP-21 in HCC tissues, and showed that the increase in MMP-21 was associated with vascular invasion and poor prognosis. Although changing levels of MMP-21 in HCC cell lines had no significant effect on cell migration or invasion abilities in in vitro transwell tests, both IHC analysis and in vivo mouse models proved that upregulated MMP-21 promoted metastasis. Functional enrichments of MMP-21 using The Cancer Genome Atlas (TCGA) data suggested that MMP-21 might regulate metastasis via macrophages. Further experiments proved that MMP-21 enhanced macrophage recruitment by increasing CCL-14 levels and promoted M2-type polarization of macrophage by elevating the expression of CSF-1 and FGF-1. Taken together, this study revealed that MMP-21 controlled the tumor microenvironment remodeling and functional regulation of macrophages to regulate HCC metastasis.
Collapse
Affiliation(s)
- Jiangfan Zhou
- Department of Interventional RadiologyThe First Affiliated HospitalWenzhou Medical UniversityWenzhouChina
| | - Li Liu
- Key Laboratory of Laboratory MedicineMinistry of EducationSchool of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouChina
| | - Xudong Hu
- Key Laboratory of Laboratory MedicineMinistry of EducationSchool of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouChina
| | - Rong Feng
- Key Laboratory of Laboratory MedicineMinistry of EducationSchool of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouChina
| | - Niannian Zhao
- Key Laboratory of Laboratory MedicineMinistry of EducationSchool of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouChina
| | - Li Zhang
- Key Laboratory of Laboratory MedicineMinistry of EducationSchool of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouChina
| | - Wenhao Hu
- Department of Interventional RadiologyThe First Affiliated HospitalWenzhou Medical UniversityWenzhouChina
| | - Jian Zhang
- Department of Interventional RadiologyThe First Affiliated HospitalWenzhou Medical UniversityWenzhouChina
| | - Shiyong Huang
- Department of Interventional RadiologyThe First Affiliated HospitalWenzhou Medical UniversityWenzhouChina
| | - Lin Liu
- Key Laboratory of Laboratory MedicineMinistry of EducationSchool of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouChina
| | - Wei Li
- Key Laboratory of Laboratory MedicineMinistry of EducationSchool of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouChina
| | - Yunfeng Shan
- Department of Hepatobiliary SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhouChina
| | - Jing Jin
- Institute of Glycobiological EngineeringZhejiang Provincial Key Laboratory of Medical GeneticsSchool of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhouChina
| |
Collapse
|
204
|
Dawood AF, Alzamil NM, Hewett PW, Momenah MA, Dallak M, Kamar SS, Abdel Kader DH, Yassin H, Haidara MA, Maarouf A, Al-Ani B. Metformin Protects against Diabetic Cardiomyopathy: An Association between Desmin-Sarcomere Injury and the iNOS/mTOR/TIMP-1 Fibrosis Axis. Biomedicines 2022; 10:984. [PMID: 35625721 PMCID: PMC9139128 DOI: 10.3390/biomedicines10050984] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/09/2022] [Accepted: 04/20/2022] [Indexed: 02/05/2023] Open
Abstract
The intermediate filament protein desmin is essential for maintaining the structural integrity of sarcomeres, the fundamental unit of cardiac muscle. Diabetes mellitus (DM) can cause desmin to become dysregulated, following episodes of nitrosative stress, through the activation of the iNOS/mTOR/TIMP-1 pathway, thereby stimulating collagen deposition in the myocardium. In this study, type 2 diabetes mellitus (T2DM) was induced in rats. One group of animals was pre-treated with metformin (200 mg/kg) prior to diabetes induction and subsequently kept on metformin until sacrifice at week 12. Cardiac injuries developed in the diabetic rats as demonstrated by a significant (p < 0.0001) inhibition of desmin immunostaining, profound sarcomere ultrastructural alterations, substantial damage to the left ventricular tissue, collagen deposition, and abnormal ECG recordings. DM also significantly induced the cardiac expression of inducible nitric oxide synthase (iNOS), mammalian target of rapamycin (mTOR), and the profibrogenic biomarker tissue inhibitor of metalloproteinase-1 (TIMP-1). The expression of all these markers was significantly inhibited by metformin. In addition, a significant (p < 0.0001) correlation between desmin tissue levels/sarcomere damage and glycated hemoglobin, heart rate, iNOS, mTOR, and fibrosis was observed. These findings demonstrate an association between damage of the cardiac contractile unit—desmin and sarcomere—and the iNOS/mTOR/TIMP-1/collagen axis of fibrosis in T2DM-induced cardiomyopathy, with metformin exhibiting beneficial cardiovascular pleiotropic effects.
Collapse
Affiliation(s)
- Amal F. Dawood
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
- Department of Physiology, Kasr al-Aini Faculty of Medicine, Cairo University, Cairo 12613, Egypt
| | - Norah M. Alzamil
- Department of Clinical Science, Family Medicine, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Peter W. Hewett
- Institute of Cardiovascular Sciences, College of Medicine and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Maha A. Momenah
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Mohammad Dallak
- Department of Physiology, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia;
| | - Samaa S. Kamar
- Department of Medical Histology, Kasr al-Aini Faculty of Medicine, Cairo University, Cairo 12613, Egypt; (S.S.K.); (D.H.A.K.)
| | - Dina H. Abdel Kader
- Department of Medical Histology, Kasr al-Aini Faculty of Medicine, Cairo University, Cairo 12613, Egypt; (S.S.K.); (D.H.A.K.)
| | - Hanaa Yassin
- Department of Physiology, Kasr al-Aini Faculty of Medicine, Cairo University, Cairo 12613, Egypt
| | - Mohamed A. Haidara
- Department of Physiology, Kasr al-Aini Faculty of Medicine, Cairo University, Cairo 12613, Egypt
| | - Amro Maarouf
- Department of Clinical Biochemistry, Birmingham Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B9 5SS, UK;
| | - Bahjat Al-Ani
- Department of Physiology, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia;
| |
Collapse
|
205
|
Rahat MA. Mini-Review: Can the Metastatic Cascade Be Inhibited by Targeting CD147/EMMPRIN to Prevent Tumor Recurrence? Front Immunol 2022; 13:855978. [PMID: 35418981 PMCID: PMC8995701 DOI: 10.3389/fimmu.2022.855978] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/07/2022] [Indexed: 12/05/2022] Open
Abstract
Solid tumors metastasize very early in their development, and once the metastatic cell is lodged in a remote organ, it can proliferate to generate a metastatic lesion or remain dormant for long periods. Dormant cells represent a real risk for future tumor recurrence, but because they are typically undetectable and insensitive to current modalities of treatment, it is difficult to treat them in time. We describe the metastatic cascade, which is the process that allows tumor cells to detach from the primary tumor, migrate in the tissue, intravasate and extravasate the lymphatics or a blood vessel, adhere to a remote tissue and eventually outgrow. We focus on the critical enabling role of the interactions between tumor cells and immune cells, especially macrophages, in driving the metastatic cascade, and on those stages that can potentially be targeted. In order to prevent the metastatic cascade and tumor recurrence, we would need to target a molecule that is involved in all of the steps of the process, and evidence is brought to suggest that CD147/EMMPRIN is such a protein and that targeting it blocks metastasis and prevents tumor recurrence.
Collapse
Affiliation(s)
- Michal A Rahat
- Immunotherapy Laboratory, Carmel Medical Center, Haifa, Israel.,Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
206
|
Abstract
Much of the current research into immune escape from cancer is focused on molecular and cellular biology, an area of biophysics that is easily overlooked. A large number of immune drugs entering the clinic are not effective for all patients. Apart from the molecular heterogeneity of tumors, the biggest reason for this may be that knowledge of biophysics has not been considered, and therefore an exploration of biophysics may help to address this challenge. To help researchers better investigate the relationship between tumor immune escape and biophysics, this paper provides a brief overview on recent advances and challenges of the biophysical factors and strategies by which tumors acquire immune escape and a comprehensive analysis of the relevant forces acting on tumor cells during immune escape. These include tumor and stromal stiffness, fluid interstitial pressure, shear stress, and viscoelasticity. In addition, advances in biophysics cannot be made without the development of detection tools, and this paper also provides a comprehensive summary of the important detection tools available at this stage in the field of biophysics.
Collapse
Affiliation(s)
- Maonan Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Hui Jiang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiaohui Liu
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xuemei Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
207
|
Zhang P, Liu G, Hu J, Chen S, Wang B, Peng P, Yu X, Guo D. Tenascin-C can Serve as an Indicator for the Immunosuppressive Microenvironment of Diffuse Low-Grade Gliomas. Front Immunol 2022; 13:824586. [PMID: 35371015 PMCID: PMC8966496 DOI: 10.3389/fimmu.2022.824586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/25/2022] [Indexed: 12/15/2022] Open
Abstract
Purpose The development and progression of glioma are associated with the tumor immune microenvironment. Diffuse low-grade gliomas (LGGs) with higher immunosuppressive microenvironment tend to have a poorer prognosis. The study aimed to find a biological marker that can reflect the tumor immune microenvironment status and predict prognosis of LGGs. Methods The target gene tenascin-C (TNC) was obtained by screening the Chinese Glioma Genome Atlas (CGGA) and the Cancer Genome Atlas (TCGA) databases. Then samples of LGGs were collected for experimental verification with immunohistochemistry, immunofluorescence, immunoblotting, quantitative real-time PCR. ELISA was employed to determine the content of TNC in serum and examine its relationship with the tumor immune microenvironment. Eventually, the sensitivity of immunotherapy was predicted on the basis of the content of TNC in LGGs. Results In the high-TNC subgroup, the infiltration of immunosuppressive cells was increased (MDSC: r=0.4721, Treg: r=0.3154, etc.), and immune effector cells were decreased [NKT, γδT, etc. (p<0.05)], immunosuppressive factors were elevated [TGF-β, IL10, etc. (p<0.05)], immunostimulatory factors, such as NKG2D, dropped (p<0.05), hypoxia scores increased (p<0.001), and less benefit from immunotherapy (p<0.05). Serum TNC level could be used to assess the status of tumor immune microenvironment in patients with grade II (AUC=0.8571; 95% CI: 0.6541-1.06) and grade III (AUC=0.8333; 95% CI: 0.6334-1.033) glioma. Conclusions Our data suggested that TNC could serve as an indicator for the immunosuppressive microenvironment status and the prognosis of LGGs. Moreover, it could also act as a predictor for the effect of immunotherapy on LGG patients.
Collapse
Affiliation(s)
- Po Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guohao Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinyang Hu
- Department of Neurosurgery, The People's Hospital of China Three Gorges University, Yichang, China
| | - Sui Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baofeng Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Peng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjiang Yu
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongsheng Guo
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
208
|
Tu B, Gao Y, Sun F, Shi M, Huang Y. Lipid Metabolism Regulation Based on Nanotechnology for Enhancement of Tumor Immunity. Front Pharmacol 2022; 13:840440. [PMID: 35392570 PMCID: PMC8980325 DOI: 10.3389/fphar.2022.840440] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/07/2022] [Indexed: 11/26/2022] Open
Abstract
The hallmarks of cancer include dysregulated metabolism and immune evasion. As a basic way of metabolism, lipid metabolism is reprogrammed for the rapid energy and nutrient supply in the occurrence and development of tumors. Lipid metabolism alterations that occur in the tumor microenvironment (TME) affect the antitumor responses of immune cells and cause immune evasion. Therefore, targeting lipid metabolism in the TME for enhancing the antitumor effect of immune cells is a promising direction for cancer treatment. Cancer nanomedicine has great potential in regulating tumor metabolism and tumor immunity. This review summarizes the nanotechnology-based strategies for lipid metabolism regulation in the TME for enhanced anticancer immune responses.
Collapse
Affiliation(s)
- Bin Tu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yanrong Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Feifei Sun
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Mingjie Shi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, China.,NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai, China.,School of Advanced Study, Institute of Natural Medicine and Health Product, Taizhou University, Taizhou, China
| |
Collapse
|
209
|
Kamiya N, Ishikawa Y, Kotani K, Hatakeyama S, Matsumura M. Monocyte-to-Lymphocyte Ratio in the Diagnosis of Lymphoma in Adult Patients. Int J Gen Med 2022; 15:4221-4226. [PMID: 35480988 PMCID: PMC9035440 DOI: 10.2147/ijgm.s357468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/06/2022] [Indexed: 12/22/2022] Open
Abstract
Background Lymphomas, including Hodgkin lymphoma and non-Hodgkin lymphoma, are one of the differentials for peripheral lymphadenopathy and are difficult to diagnose clinically. Biopsy is essential for diagnosing lymphoma, although it is invasive. Non-invasive methods are required to identify patients with suspected lymphoma who should undergo a biopsy. The relevance of the monocyte-to-lymphocyte ratio has recently been reported to be a useful diagnostic marker in children with lymphoma and a prognostic marker of various other diseases. This study aimed to determine the relevance of the monocyte-to-lymphocyte ratio in the diagnosis of lymphoma in adults. Methods The study included 246 adult outpatients (median age of 49.0 years) presenting with peripheral lymphadenopathy. The final diagnosis was determined by reviewing the medical records. We categorized all patients into either the lymphoma group or the non-lymphoma group. The lymphoma group included patients who underwent biopsy and were diagnosed with lymphoma by histopathology, while the non-lymphoma group included those diagnosed with disease excluding lymphoma. The monocyte-to-lymphocyte ratios were compared between the two groups. Results Of the participants, 33 (13.4%) were assigned to the lymphoma group. The median age of the lymphoma and non-lymphoma groups were 67.0 years (interquartile range [IQR] 55.5–75.5 years) and 46.0 years (IQR 36.0–61.0 years), respectively. The lymphocyte and monocyte levels showed no significant differences between the two groups individually. Nonetheless, the monocyte-to-lymphocyte ratio was significantly higher in the lymphoma group (median, 0.36; IQR, 0.24–0.73) than in the non-lymphoma group (median, 0.29; IQR, 0.21–0.43; P = 0.022), independent of lymph node diameter ≥ 1 cm and C-reactive protein levels. Conclusion This study suggests that the monocyte-to-lymphocyte ratio can be a helpful diagnostic marker for lymphoma in adults with peripheral lymphadenopathy when the etiology is unclear even after a medical interview and physical examination.
Collapse
Affiliation(s)
- Naoko Kamiya
- Division of General Internal Medicine, Jichi Medical University Hospital, Shimotsuke-shi, Tochigi, Japan
- Correspondence: Naoko Kamiya, Division of General Internal Medicine, Jichi Medical University Hospital, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan, Tel +81-285-58-7498, Fax +81-285-40-5160, Email
| | - Yukiko Ishikawa
- Division of General Internal Medicine, Jichi Medical University Hospital, Shimotsuke-shi, Tochigi, Japan
| | - Kazuhiko Kotani
- Division of Community and Family Medicine, Center for Community Medicine, Jichi Medical University, Shimotsuke-shi, Tochigi, Japan
| | - Shuji Hatakeyama
- Division of General Internal Medicine, Jichi Medical University Hospital, Shimotsuke-shi, Tochigi, Japan
- Division of Infectious Diseases, Jichi Medical University Hospital, Shimotsuke-shi, Tochigi, Japan
| | - Masami Matsumura
- Division of General Internal Medicine, Jichi Medical University Hospital, Shimotsuke-shi, Tochigi, Japan
| |
Collapse
|
210
|
Podlesnaya PA, Kovaleva OV, Petrenko AA, Grachev AN. The Mechanism of the Development of Macrophage Tolerance in Tumor Microenvironment. Bull Exp Biol Med 2022; 172:653-657. [PMID: 35352254 DOI: 10.1007/s10517-022-05449-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Indexed: 11/27/2022]
Abstract
Bacteria forming the resident microbiome of the tumor are an integral component of its microenvironment. The interaction of the tumor microbiome with the tumor or tumor stromal cells is not well understood. We hypothesized that bacteria in the tumor microenvironment induce macrophage tolerance. Macrophage tolerance is a phenomenon of macrophage inability to respond to a repetitive inflammatory stimulus, which leads to a loss of cytotoxic activity. We studied the development of macrophage tolerance under the influence of bacteria and cytokines of the tumor microenvironment in vitro. It was found that the macrophage tolerance in the tumor stroma can develop in response to bacterial cell wall components and inflammatory factors. The acquired tolerance is inability of macrophages to produce proinflammatory cytokines TNFα, IL-1β, and MCP-1 and activation of the production of immunosuppressive IL-10.
Collapse
Affiliation(s)
- P A Podlesnaya
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - O V Kovaleva
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A A Petrenko
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A N Grachev
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
211
|
Yang H, Yan M, Li W, Xu L. SIRPα and PD1 expression on tumor-associated macrophage predict prognosis of intrahepatic cholangiocarcinoma. J Transl Med 2022; 20:140. [PMID: 35317832 PMCID: PMC8939174 DOI: 10.1186/s12967-022-03342-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/06/2022] [Indexed: 02/08/2023] Open
Abstract
Background The phagocytosis checkpoints of CD47/SIRPα, PD1/PDL1, CD24/SIGLEC10, and MHC/LILRB1 have shown inhibited phagocytosis of macrophages in distinct tumors. However, phagocytosis checkpoints and their therapeutic significance remain largely unknown in intrahepatic cholangiocarcinoma (ICC) patients. Methods We analyzed sequencing data from the Cancer Genome Atlas (TCGA) and identified differently expressed genes between tumors and para‐tumors. Then, we investigated the expression of CD68, SIRPα, PD1, and SIGLEC10 by IHC in 81 ICC patients, and the clinical significance of these markers with different risk factors was also measured. Results Tumor infiltration immune cells analysis from the TCGA data revealed that macrophages significantly increased. Further analysis showed that M0 macrophages were significantly higher and M2 macrophages were significantly lower in ICC compared with paracancerous tissues, while there was no significant difference in M1 macrophages. We then examined some of M1 and M2 markers, and we found that M1 markers (iNOS, TNF, IL12A, and B) increased, while M2 markers (ARG1 and CD206) decreased in ICCs compared with paracancerous tissues. Furthermore, the expression of CD68, SIRPα, PD1, and SIGLEC10 increased significantly, but LILRB1 expression did not. We also examined the expression of CD68, SIRPα, PD1, and SIGLEC10 in 81 ICC patients by IHC, which revealed a similar expression pattern to that which emerged from the TCGA data. Upon analyzing the correlation between these markers and the progression of ICC patients, we found that the high expression of CD68, SIRPα, and PD1 are correlated with poor progression among ICC patients, while SIGLEC10 shows no correlation. More SIRPα+ or PD1+ TAMs were observed in the tumor tissues of ICC patients with HBV infections compared to non‐HBV‐infected patients. Multivariate analysis indicated that SIRPα and PD1 expression are independent indicators of ICC patient prognosis. Conclusion Hyperactivated CD47/SIRPα and PD1/PD‐L1 signals in CD68+ TAMs in tumor tissues are negative prognostic markers for ICCs after resection. Furthermore, anti-CD47 in combination with anti-PD1 or CD47/PD1 bispecific antibody (BsAb) may represent promising treatments for ICC. Further studies are also required in the future to confirmed our findings.
Collapse
Affiliation(s)
- Hui Yang
- Department of Gastroenterology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Meimei Yan
- Department of Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Linping Xu
- Department of Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
212
|
Exosomal non-coding RNAs: Emerging roles in bilateral communication between cancer cells and macrophages. Mol Ther 2022; 30:1036-1053. [PMID: 34864204 PMCID: PMC8899606 DOI: 10.1016/j.ymthe.2021.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/28/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) is a dynamic network of cellular organization that comprises diverse cell types and significantly contributes to cancer development. As pivotal immune stromal cells in the TME, macrophages are extensively heterogeneous and exert both antitumor and protumor functions. Exosomes are nanosized extracellular membranous vesicles with diameters between 30 and 150 nm. By transferring multiple bioactive substances such as proteins, lipids, and nucleic acids, exosomes play an important role in the communication between cells. Recently, growing evidence has demonstrated that non-coding RNAs (ncRNAs) are enriched in exosomes and that exosomal ncRNAs are involved in the crosstalk between cancer cells and macrophages. Furthermore, circulating exosomal ncRNAs can be detected in biofluids, serving as promising noninvasive biomarkers for the early diagnosis and prognostic prediction of cancer. Exosome-based therapies are emerging as potent strategies that can be utilized to alleviate tumor progression. Herein, the present knowledge of exosomal ncRNAs and their vital roles in regulating the interplay between cancer cells and macrophages, as well as their clinical applications are summarized.
Collapse
|
213
|
Kim S, Lee CH, Yeo JY, Hwang KW, Park SY. Immunostimulatory activity of stem bark of Kalopanax pictus in RAW 264.7 macrophage. J Herb Med 2022. [DOI: 10.1016/j.hermed.2021.100504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
214
|
Sleep Apnoea and Cancer Risk: Where Are We Now? Respir Med Res 2022; 81:100905. [DOI: 10.1016/j.resmer.2022.100905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 11/19/2022]
|
215
|
Lepucki A, Orlińska K, Mielczarek-Palacz A, Kabut J, Olczyk P, Komosińska-Vassev K. The Role of Extracellular Matrix Proteins in Breast Cancer. J Clin Med 2022; 11:jcm11051250. [PMID: 35268340 PMCID: PMC8911242 DOI: 10.3390/jcm11051250] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/16/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix is a structure composed of many molecules, including fibrillar (types I, II, III, V, XI, XXIV, XXVII) and non-fibrillar collagens (mainly basement membrane collagens: types IV, VIII, X), non-collagenous glycoproteins (elastin, laminin, fibronectin, thrombospondin, tenascin, osteopontin, osteonectin, entactin, periostin) embedded in a gel of negatively charged water-retaining glycosaminoglycans (GAGs) such as non-sulfated hyaluronic acid (HA) and sulfated GAGs which are linked to a core protein to form proteoglycans (PGs). This highly dynamic molecular network provides critical biochemical and biomechanical cues that mediate the cell–cell and cell–matrix interactions, influence cell growth, migration and differentiation and serve as a reservoir of cytokines and growth factors’ action. The breakdown of normal ECM and its replacement with tumor ECM modulate the tumor microenvironment (TME) composition and is an essential part of tumorigenesis and metastasis, acting as key driver for malignant progression. Abnormal ECM also deregulate behavior of stromal cells as well as facilitating tumor-associated angiogenesis and inflammation. Thus, the tumor matrix modulates each of the classically defined hallmarks of cancer promoting the growth, survival and invasion of the cancer. Moreover, various ECM-derived components modulate the immune response affecting T cells, tumor-associated macrophages (TAM), dendritic cells and cancer-associated fibroblasts (CAF). This review article considers the role that extracellular matrix play in breast cancer. Determining the detailed connections between the ECM and cellular processes has helped to identify novel disease markers and therapeutic targets.
Collapse
Affiliation(s)
- Arkadiusz Lepucki
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
| | - Kinga Orlińska
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
| | - Aleksandra Mielczarek-Palacz
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.M.-P.); (J.K.)
| | - Jacek Kabut
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.M.-P.); (J.K.)
| | - Pawel Olczyk
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
- Correspondence:
| | - Katarzyna Komosińska-Vassev
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland;
| |
Collapse
|
216
|
Fang F, Zhang T, Li Q, Chen X, Jiang F, Shen X. The tumor immune-microenvironment in gastric cancer. TUMORI JOURNAL 2022; 108:541-551. [PMID: 35196917 DOI: 10.1177/03008916211070051] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS AND BACKGROUND The tumor microenvironment significantly influences malignant behavior and progression. Many components are involved in the tumor microenvironment, including extracellular matrix, stromal cells, immune and inflammatory cells, as well as cytokines that promote tumor development with complex interactions through the exchange of molecular information. It is now known that tumor immune escape may be influenced by the tumor microenvironment. The aim of this work is to conduct a review of the tumor immune-microenvironment in gastric cancer. METHODS We review the current knowledge of several immune cells involved in the gastric tumor microenvironment. In addition, a brief description of immunotherapy strategies for gastric cancer is also reviewed. CONCLUSIONS Among immune cell populations, lymphocytes, macrophages, dendritic cells and myeloid-derived suppressor cells are revealed to make the difference in promoting or suppressing gastric tumorigenesis, either directly or indirectly, via regulating the immune responses. Understanding these interactions in detail within the tumor immune-microenvironment will contribute to unraveling new therapeutic targets.
Collapse
Affiliation(s)
- Fujin Fang
- Key Laboratory of Environmental Medical Engineering and Education Ministry, School of Public Health, Southeast University, Nanjing, Jiangsu, China.,Department of Preventive Medicine, School of Public Health, Southeast University, Nanjing, China
| | - Tiantian Zhang
- Department of Clinical Laboratory, The Third People's Hospital of Bengbu, Bengbu, China
| | - Qiong Li
- Key Laboratory of Environmental Medical Engineering and Education Ministry, School of Public Health, Southeast University, Nanjing, Jiangsu, China.,Department of Preventive Medicine, School of Public Health, Southeast University, Nanjing, China
| | - Xiaowei Chen
- Key Laboratory of Environmental Medical Engineering and Education Ministry, School of Public Health, Southeast University, Nanjing, Jiangsu, China.,Department of Preventive Medicine, School of Public Health, Southeast University, Nanjing, China
| | - Fei Jiang
- Key Laboratory of Environmental Medical Engineering and Education Ministry, School of Public Health, Southeast University, Nanjing, Jiangsu, China.,Department of Preventive Medicine, School of Public Health, Southeast University, Nanjing, China
| | - Xiaobing Shen
- Key Laboratory of Environmental Medical Engineering and Education Ministry, School of Public Health, Southeast University, Nanjing, Jiangsu, China.,Department of Preventive Medicine, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
217
|
Jin Y, Kang Y, Wang M, Wu B, Su B, Yin H, Tang Y, Li Q, Wei W, Mei Q, Hu G, Lukacs-Kornek V, Li J, Wu K, Yuan X, Wang W. Targeting polarized phenotype of microglia via IL6/JAK2/STAT3 signaling to reduce NSCLC brain metastasis. Signal Transduct Target Ther 2022; 7:52. [PMID: 35194016 PMCID: PMC8864012 DOI: 10.1038/s41392-022-00872-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 12/27/2022] Open
Abstract
Tumor-associated macrophages have emerged as crucial factors for metastases. Microglia are indispensable components of the brain microenvironment and play vital roles in brain metastasis (BM). However, the underlying mechanism of how activated microglia promote brain metastasis of non-small cell lung cancer (NSCLC) remains elusive. Here, we purified cell lines with brain-metastatic tropism and employed a co-culture system to reveal their communication with microglia. By single-cell RNA-sequencing and transcriptome difference analysis, we identified IL6 as the key regulator in brain-metastatic cells (A549-F3) to induce anti-inflammatory microglia via JAK2/STAT3 signaling, which in turn promoted the colonization process in metastatic A549-F3 cells. In our clinical samples, patients with higher levels of IL6 in serum showed higher propensity for brain metastasis. Additionally, the TCGA (The Cancer Genome Atlas) data revealed that NSCLC patients with a lower level of IL6 had a longer overall survival time compared to those with a higher level of IL6. Overall, our data indicate that the targeting of IL6/JAK2/STAT3 signaling in activated microglia may be a promising new approach for inhibiting brain metastasis in NSCLC patients.
Collapse
Affiliation(s)
- Yu Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yalin Kang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Bili Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Beibei Su
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Han Yin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yang Tang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Qianxia Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Wenjie Wei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Qi Mei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Veronika Lukacs-Kornek
- Institute of Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Jian Li
- Institute of Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Kongming Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
218
|
Habanjar O, Diab-Assaf M, Caldefie-Chezet F, Delort L. The Impact of Obesity, Adipose Tissue, and Tumor Microenvironment on Macrophage Polarization and Metastasis. BIOLOGY 2022; 11:339. [PMID: 35205204 PMCID: PMC8869089 DOI: 10.3390/biology11020339] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/19/2022] [Accepted: 02/15/2022] [Indexed: 12/11/2022]
Abstract
Tumor metastasis is a major cause of death in cancer patients. It involves not only the intrinsic alterations within tumor cells, but also crosstalk between these cells and components of the tumor microenvironment (TME). Tumorigenesis is a complex and dynamic process, involving the following three main stages: initiation, progression, and metastasis. The transition between these stages depends on the changes within the extracellular matrix (ECM), in which tumor and stromal cells reside. This matrix, under the effect of growth factors, cytokines, and adipokines, can be morphologically altered, degraded, or reorganized. Many cancers evolve to form an immunosuppressive TME locally and create a pre-metastatic niche in other tissue sites. TME and pre-metastatic niches include myofibroblasts, immuno-inflammatory cells (macrophages), adipocytes, blood, and lymphatic vascular networks. Several studies have highlighted the adipocyte-macrophage interaction as a key driver of cancer progression and dissemination. The following two main classes of macrophages are distinguished: M1 (pro-inflammatory/anti-tumor) and M2 (anti-inflammatory/pro-tumor). These cells exhibit distinct microenvironment-dependent phenotypes that can promote or inhibit metastasis. On the other hand, obesity in cancer patients has been linked to a poor prognosis. In this regard, tumor-associated adipocytes modulate TME through the secretion of inflammatory mediators, which modulate and recruit tumor-associated macrophages (TAM). Hereby, this review describes the cellular and molecular mechanisms that link inflammation, obesity, and cancer. It provides a comprehensive overview of adipocytes and macrophages in the ECM as they control cancer initiation, progression, and invasion. In addition, it addresses the mechanisms of tumor anchoring and recruitment for M1, M2, and TAM macrophages, specifically highlighting their origin, classification, polarization, and regulatory networks, as well as their roles in the regulation of angiogenesis, invasion, metastasis, and immunosuppression, specifically highlighting the role of adipocytes in this process.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, ECREIN, f-63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie moléculaire et anticancéreuse, Faculté des Sciences II, Université libanaise Fanar, Beyrouth 1500, Liban;
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, ECREIN, f-63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, ECREIN, f-63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| |
Collapse
|
219
|
Pavliuk-Karachevtseva A, Mihalik J, Biel R, Rybárová S, Hodorová I. Chosen Antioxidant Enzymes GPx4 and GPx8 in Human Colorectal Carcinoma: Study of the Slovak Population. Medicina (B Aires) 2022; 58:medicina58020298. [PMID: 35208621 PMCID: PMC8875726 DOI: 10.3390/medicina58020298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/08/2022] [Accepted: 02/13/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Objectives: Nowadays colorectal carcinoma (CRC) is one of the most common causes of death in patients with malignant neoplasms worldwide. Our work aimed to determine the possible involvement of glutathione peroxidases 4 and 8 (GPx4 and GPx8) in this specific tumor process. Materials and Methods: The expression of GPx4 and GPx8 in 58 specimens of human colorectal cancer tissues and normal tissues was detected by the indirect immunohistochemical method under a light microscope. Statistical analysis was done by Chi-squared test. Histological findings were compared with data such as gender, age, tumor grade, histotype and lymph nodes alteration. Results: In all specimens of healthy tissue the presence of both, GPx4 and GPx8, was detected in the cytoplasm of epithelial cells. On the other hand, a positive immunohistochemical reaction against GPx4 only in 41.4% and against GPx8 only in 29.3% of human colorectal adenocarcinoma specimens were observed. Any significant difference between the presence of GPx and the age, the gender of the patient, tumor grade, histotype of cancer and the lesion of regional lymph nodes has not been detected. Conclusions: Our foundation could mean, that GPx4 and GPx8 have no important role in CRC pathogenesis, but the loss of these enzymes probably indicates a serious pathological process ongoing in the large intestine. To our knowledge, this is the first paper describing GPx8 presence in human colorectal carcinoma.
Collapse
Affiliation(s)
- Andriana Pavliuk-Karachevtseva
- Department of Anatomy, Medical Faculty, Šafárik University, Šrobárova 2, 041 83 Košice, Slovakia; (A.P.-K.); (J.M.); (S.R.)
| | - Jozef Mihalik
- Department of Anatomy, Medical Faculty, Šafárik University, Šrobárova 2, 041 83 Košice, Slovakia; (A.P.-K.); (J.M.); (S.R.)
| | - Róbert Biel
- Department of Clinical Oncology 2, East-Slovakian Oncological Institute, Rastislavova 43, 041 91 Košice, Slovakia;
| | - Silvia Rybárová
- Department of Anatomy, Medical Faculty, Šafárik University, Šrobárova 2, 041 83 Košice, Slovakia; (A.P.-K.); (J.M.); (S.R.)
| | - Ingrid Hodorová
- Department of Anatomy, Medical Faculty, Šafárik University, Šrobárova 2, 041 83 Košice, Slovakia; (A.P.-K.); (J.M.); (S.R.)
- Correspondence:
| |
Collapse
|
220
|
2-methylpyridine-1-ium-1-sulfonate modifies tumor-derived exosome mediated macrophage polarization: Relevance to the tumor microenvironment. Int Immunopharmacol 2022; 106:108581. [PMID: 35149296 DOI: 10.1016/j.intimp.2022.108581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 11/05/2022]
Abstract
The compound "2-methylpyridine-1-ium-1-sulfonate" (MPS) is the active constituent of Allium hirtifolium Boiss. bulbs with potent anti-angiogenic and anti-cancer activities. Tumor microenvironment (TME) plays a key role in tumor progression via tumor derived exosome (TEX) mediated polarization of M2 type tumor associated macrophages (TAMs). In this study, we explored direct and colorectal cancer (CRC) exosome-mediated impacts of MPS on macrophage polarization to find out whether MPS could modify TEX in favor of anti-tumor M1-like macrophage polarization. After MPS isolation and characterization, first its direct anti-cancer effects were evaluated on HT-29 cells. Then, TEX were isolated from untreated (C-TEX) and MPS-treated (MPS-TEX) HT-29 cells. THP-1 M0 macrophages were incubated with MPS, C-TEX and MPS-TEX. Macrophage polarization was evaluated by flow cytometry, ELISA and gene expression analysis of several M1- and M2-related markers. MPS induced apoptosis and cell cycle arrest and reduced the migration ability of HT-29 cells. C-TEX polarized M0 macrophages toward a mixed M1-/M2-like phenotype with a high predominance of M2-like cells. Macrophage treatment with MPS was associated with the induction of M1-like phenotype. Also, MPS was demonstrated to ameliorate TEX-mediated effects in favor of M1-like polarization. In conclusion, our study addresses for the first time, the potential capability of MPS in skewing macrophages toward an anti-cancer M1-like phenotype both directly and in a TEX-dependent manner. Thus, in addition to its direct anti-cancer effects, this compound could also modify TME in favor of tumor eradication via its direct and TEX-mediated effects on macrophage polarization as a novel anti-cancer mechanism.
Collapse
|
221
|
Song Y, Huang Y, Zhou F, Ding J, Zhou W. Macrophage-targeted nanomedicine for chronic diseases immunotherapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.08.090] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
222
|
Kuntzel T, Bagnard D. Manipulating Macrophage/Microglia Polarization to Treat Glioblastoma or Multiple Sclerosis. Pharmaceutics 2022; 14:344. [PMID: 35214076 PMCID: PMC8877500 DOI: 10.3390/pharmaceutics14020344] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 12/24/2022] Open
Abstract
Macrophages and microglia are implicated in several diseases with divergent roles in physiopathology. This discrepancy can be explained by their capacity to endorse different polarization states. Theoretical extremes of these states are called M1 and M2. M1 are pro-inflammatory, microbicidal, and cytotoxic whereas M2 are anti-inflammatory, immunoregulatory cells in favor of tumor progression. In pathological states, these polarizations are dysregulated, thus restoring phenotypes could be an interesting treatment approach against diseases. In this review, we will focus on compounds targeting macrophages and microglia polarization in two very distinctive pathologies: multiple sclerosis and glioblastoma. Multiple sclerosis is an inflammatory disease characterized by demyelination and axon degradation. In this case, macrophages and microglia endorse a M1-like phenotype inducing inflammation. Promoting the opposite M2-like polarization could be an interesting treatment strategy. Glioblastoma is a brain tumor in which macrophages and microglia facilitate tumor progression, spreading, and angiogenesis. They are part of the tumor associated macrophages displaying an anti-inflammatory phenotype, thereby inhibiting anti-tumoral immunity. Re-activating them could be a method to limit and reduce tumor progression. These two pathologies will be used to exemplify that targeting the polarization of macrophages and microglia is a promising approach with a broad spectrum of applications deserving more attention.
Collapse
Affiliation(s)
- Thomas Kuntzel
- UMR7242 Biotechnology and Cell Signaling, Centre National de la Recherche Scientifique, Strasbourg Drug Discovery and Development Institute (IMS), University of Strasbourg, 67400 Illkirch-Graffenstaden, France;
| | - Dominique Bagnard
- UMR7242 Biotechnology and Cell Signaling, Centre National de la Recherche Scientifique, Strasbourg Drug Discovery and Development Institute (IMS), University of Strasbourg, 67400 Illkirch-Graffenstaden, France;
- Ecole Supérieure de Biotechnologie de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
223
|
Meng X, Wang X, Jiang C, Zhang S, Cheng S. Correlation analysis of lymphocyte-monocyte ratio with pathological complete response and clinical prognosis of neoadjuvant chemotherapy in patients with breast cancer. Transl Oncol 2022; 18:101355. [PMID: 35121221 PMCID: PMC8818569 DOI: 10.1016/j.tranon.2022.101355] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/13/2022] [Accepted: 01/23/2022] [Indexed: 01/12/2023] Open
Abstract
We evaluated the impact of LMR on pCR and prognosis in breast cancer patients. High LMR predicts increased pCR in HER2(+) breast cancer patients. High LMR predicts a better prognosis in neoadjuvant chemotherapy patients. LMR is an economical and easy detection index for patients.
Purpose Inflammation plays an important role in tumor proliferation, metastasis, and chemotherapy resistance. Peripheral blood lymphocyte-monocyte ratio (LMR) has been reported to be closely associated with the prognosis of many tumors, such as certain hematologic malignancies and gastric cancer. However, the association in breast cancer is still not clear. This study investigated the relationship between LMR with pathological complete response and clinical prognosis of neoadjuvant chemotherapy in patients with breast cancer, to provide convenient and accurate predictive indicators for pathological complete response (pCR) and prognosis. Methods The clinicopathological data of 192 female breast cancer patients who received neoadjuvant chemotherapy and surgery in Harbin Medical University Tumor Hospital from January 2013 to August 2017 were retrospectively analyzed. Blood lymphocytes and monocytes were obtained by peripheral venous punctures. Results Compared with the low LMR group, pCR was more easily obtained in the high LMR group (P=0.020); Subgroup analysis showed that patients with the high LMR and HER-2(+) group were more likely to obtain pCR (P=0.011).Univariate andmultivariate results showed that the overall survival (OS) and disease free survival (DFS) of the high LMR group were longer than that of the low LMR group. Conclusion LMR and HER-2 status are correlated with pCR of neoadjuvant chemotherapy in breast cancer patients and are independent predictors of pCR after neoadjuvant chemotherapy in breast cancer patients. Meanwhile, both LMR and T stage of tumor are independent prognostic factors of breast cancer patients, with good predictive value.
Collapse
Affiliation(s)
- Xiangyu Meng
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Xueying Wang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Cong Jiang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Shuai Zhang
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Shaoqiang Cheng
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China.
| |
Collapse
|
224
|
Tumor-Associated Macrophages in Hepatocellular Carcinoma Pathogenesis, Prognosis and Therapy. Cancers (Basel) 2022; 14:cancers14010226. [PMID: 35008390 PMCID: PMC8749970 DOI: 10.3390/cancers14010226] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/31/2021] [Accepted: 01/02/2022] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) constitutes a major health burden, accounting for >80% of primary liver cancers globally. Inflammation has come into the spotlight as a hallmark of cancer, and it is evident that tumor-associated inflammation drives the involvement of monocytes in tumor growth and metastasis. Tumor-associated macrophages (TAMs) actively participate in tumor-related inflammation, representing the main type of inflammatory cells in the tumor microenvironment, setting the crosstalk between tumor and stromal cells. Infiltrating TAMs exert either anti-tumorigenic (M1) or pro-tumorigenic (M2) functions. In most solid human tumors, increased TAM infiltration has been associated with enhanced tumor growth and metastasis, while other studies showcase that under certain conditions, TAMs exhibit cytotoxic and tumoricidal activity, inhibiting the progression of cancer. In this review, we summarize the current evidence on the role of macrophages in the pathogenesis and progression of HCC and we highlight their potential utilization in HCC prognosis and therapy. Abstract Hepatocellular carcinoma (HCC) constitutes a major health burden globally, and it is caused by intrinsic genetic mutations acting in concert with a multitude of epigenetic and extrinsic risk factors. Cancer induces myelopoiesis in the bone marrow, as well as the mobilization of hematopoietic stem and progenitor cells, which reside in the spleen. Monocytes produced in the bone marrow and the spleen further infiltrate tumors, where they differentiate into tumor-associated macrophages (TAMs). The relationship between chronic inflammation and hepatocarcinogenesis has been thoroughly investigated over the past decade; however, several aspects of the role of TAMs in HCC development are yet to be determined. In response to certain stimuli and signaling, monocytes differentiate into macrophages with antitumor properties, which are classified as M1-like. On the other hand, under different stimuli and signaling, the polarization of macrophages shifts towards an M2-like phenotype with a tumor promoting capacity. M2-like macrophages drive tumor growth both directly and indirectly, via the suppression of cytotoxic cell populations, including CD8+ T cells and NK cells. The tumor microenvironment affects the response to immunotherapies. Therefore, an enhanced understanding of its immunobiology is essential for the development of next-generation immunotherapies. The utilization of various monocyte-centered anticancer treatment modalities has been under clinical investigation, selectively targeting and modulating the processes of monocyte recruitment, activation and migration. This review summarizes the current evidence on the role of TAMs in HCC pathogenesis and progression, as well as in their potential involvement in tumor therapy, shedding light on emerging anticancer treatment methods targeting monocytes.
Collapse
|
225
|
Stephen B, Hajjar J. Immune System in Action. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1342:1-43. [PMID: 34972961 DOI: 10.1007/978-3-030-79308-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Tumor exists as a complex network of structures with an ability to evolve and evade the host immune surveillance mechanism. The immune milieu which includes macrophages, dendritic cells, natural killer cells, neutrophils, mast cells, B cells, and T cells is found in the core, the invasive margin, or the adjacent stromal or lymphoid component of the tumor. The immune infiltrate is heterogeneous and varies within a patient and between patients of the same tumor histology. The location, density, functionality, and the crosstalk between the immune cells in the tumor microenvironment influence the nature of immune response, prognosis, and treatment outcomes in cancer patients. Therefore, an understanding of the characteristics of the immune cells and their role in tumor immune surveillance is of paramount importance to identify immune targets and to develop novel immune therapeutics in the war against cancer. In this chapter, we provide an overview of the individual components of the human immune system and the translational relevance of predictive biomarkers.
Collapse
Affiliation(s)
- Bettzy Stephen
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Joud Hajjar
- Assistant Professor, Service Chief of Adult Allergy & Immunology, Division of Immunology, Allergy & Retrovirology, Baylor College of Medicine and Texas Children' Hospital, Houston, TX, USA
| |
Collapse
|
226
|
Abstract
The intimate involvement of pathogens with the heightened risk for developing certain cancers is an area of research that has captured a great deal of attention over the last 10 years. One firmly established paradigm that highlights this aspect of disease progression is in the instance of Helicobacter pylori infection and the contribution it makes in elevating the risk for developing gastric cancer. Whilst the molecular mechanisms that pinpoint the contribution that this microorganism inflicts towards host cells during gastric cancer initiation have come into greater focus, another picture that has also emerged is one that implicates the host's immune system, and the chronic inflammation that can arise therefrom, as being a central contributory factor in disease progression. Consequently, when taken with the underlying role that the extracellular matrix plays in the development of most cancers, and how this dynamic can be modulated by proteases expressed from the tumor or inflammatory cells, a complex and detailed relationship shared between the individual cellular components and their surroundings is coming into focus. In this review article, we draw attention to the emerging role played by the cathepsin proteases in modulating the stage-specific progression of Helicobacter pylori-initiated gastric cancer and the underlying immune response, while highlighting the therapeutic significance of this dynamic and how it may be amenable for novel intervention strategies within a basic research or clinical setting.
Collapse
|
227
|
Arora L, Kalia M, Pal D. Role of macrophages in cancer progression and targeted immunotherapies. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 135:281-311. [PMID: 37061335 DOI: 10.1016/bs.apcsb.2022.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The vast complexity of the tumor microenvironment (TME) aggrandizes the underlying principles responsible for immune escape, therapy resistance, and treatment failure. The stromal and immune cell population circumjacent to the tumor cells affects the cancer cell cycle leading to tumor progression. Tumor-associated macrophages (TAMs) exhibiting a unique M2 polarization state constitute a significant portion of the TME. They serve as tumor suppressors at early stages and tumor promoters at advanced stages by governing various microenvironmental cues. TAMs secreted various pro-tumoral cytokines, chemokines, and matrix metalloproteases are known to regulate the different cell cycle molecules including checkpoint inhibitors in cancer cells leading to cell cycle progression with faulty cellular components. Moreover, TAMs are well-known immunosuppressors and thereby facilitating the tumor cells' evasion from immune recognition. This chapter will describe the interaction between TAMs and tumor cells, the involvement of TAMs in the regulation of cancer cell progression by controlling cell cycle checkpoints or molecular pathways, and current TAM-based therapies, including restriction of TAM recruitment, anti-survival strategies, or switching polarity. Moreover, this chapter will also emphasize recently developed drug targets and CAR-macrophage cell therapy that restricts tumor progression.
Collapse
|
228
|
Cytokines in Renal Cell Carcinoma: A Step Towards Earlier Detection and Targeted Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1374:63-72. [DOI: 10.1007/5584_2021_700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
229
|
Szulc-Kielbik I, Kielbik M. Tumor-Associated Macrophages: Reasons to Be Cheerful, Reasons to Be Fearful. EXPERIENTIA SUPPLEMENTUM (2012) 2022; 113:107-140. [PMID: 35165862 DOI: 10.1007/978-3-030-91311-3_4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor microenvironment (TME) is a complex and constantly evolving entity that consists not only of cancer cells, but also of resident host cells and immune-infiltrating cells, among which macrophages are significant components, due to their diversity of functions through which they can influence the immune response against tumor cells. Macrophages present in tumor environment are termed as tumor-associated macrophages (TAMs). They are strongly plastic cells, and depending on the TME stimuli (i.e., cytokines, chemokines), TAMs polarize to antitumoral (M1-like TAMs) or protumoral (M2-like TAMs) phenotype. Both types of TAMs differ in the surface receptors' expression, activation of intracellular signaling pathways, and ability of production and various metabolites release. At the early stage of tumor formation, TAMs are M1-like phenotype, and they are able to eliminate tumor cells, i.e., by reactive oxygen species formation or by presentation of cancer antigens to other effector immune cells. However, during tumor progression, TAMs M2-like phenotype is dominating. They mainly contribute to angiogenesis, stromal remodeling, enhancement of tumor cells migration and invasion, and immunosuppression. This wide variety of TAMs' functions makes them an excellent subject for use in developing antitumor therapies which mainly is based on three strategies: TAMs' elimination, reprograming, or recruitment inhibition.
Collapse
Affiliation(s)
| | - Michal Kielbik
- Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland.
| |
Collapse
|
230
|
Malengier-Devlies B, Metzemaekers M, Wouters C, Proost P, Matthys P. Neutrophil Homeostasis and Emergency Granulopoiesis: The Example of Systemic Juvenile Idiopathic Arthritis. Front Immunol 2021; 12:766620. [PMID: 34966386 PMCID: PMC8710701 DOI: 10.3389/fimmu.2021.766620] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are key pathogen exterminators of the innate immune system endowed with oxidative and non-oxidative defense mechanisms. More recently, a more complex role for neutrophils as decision shaping cells that instruct other leukocytes to fine-tune innate and adaptive immune responses has come into view. Under homeostatic conditions, neutrophils are short-lived cells that are continuously released from the bone marrow. Their development starts with undifferentiated hematopoietic stem cells that pass through different immature subtypes to eventually become fully equipped, mature neutrophils capable of launching fast and robust immune responses. During severe (systemic) inflammation, there is an increased need for neutrophils. The hematopoietic system rapidly adapts to this increased demand by switching from steady-state blood cell production to emergency granulopoiesis. During emergency granulopoiesis, the de novo production of neutrophils by the bone marrow and at extramedullary sites is augmented, while additional mature neutrophils are rapidly released from the marginated pools. Although neutrophils are indispensable for host protection against microorganisms, excessive activation causes tissue damage in neutrophil-rich diseases. Therefore, tight regulation of neutrophil homeostasis is imperative. In this review, we discuss the kinetics of neutrophil ontogenesis in homeostatic conditions and during emergency myelopoiesis and provide an overview of the different molecular players involved in this regulation. We substantiate this review with the example of an autoinflammatory disease, i.e. systemic juvenile idiopathic arthritis.
Collapse
Affiliation(s)
- Bert Malengier-Devlies
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mieke Metzemaekers
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Carine Wouters
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.,Division of Pediatric Rheumatology, University Hospitals Leuven, Leuven, Belgium.,European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) at University Hospital Leuven, Leuven, Belgium
| | - Paul Proost
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
231
|
Li C, Qiu S, Jin K, Zheng X, Zhou X, Jin D, Xu B, Jin X. Tumor-derived microparticles promote the progression of triple-negative breast cancer via PD-L1-associated immune suppression. Cancer Lett 2021; 523:43-56. [PMID: 34601021 DOI: 10.1016/j.canlet.2021.09.039] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/01/2021] [Accepted: 09/27/2021] [Indexed: 02/08/2023]
Abstract
Membrane vesicles, including exosomes and microparticles (MPs), serve to package and transfer the cellular cargo during inter/extracellular communication, which is of great interest in cancer development, especially in the dissemination of signal transduction-associated traits from donor cells to recipient cells. Although increasing evidence suggests that microparticles (MPs) contribute to the development of cancer, their unique characteristics remain to be exploited. Here, we examined the secretion of MPs in tumor tissues from triple-negative breast cancer (TNBC) patients and found that the tumor cells could release MPs loaded with immune checkpoint molecular programmed cell death ligand 1 (PD-L1), especially in patients treated with traditional clinical interventions, such as chemotherapy and radiotherapy. These PD-L1-loading MPs contribute to the suppressive immune microenvironment, eventually resulting in the tumor progression in TNBC. Mechanically, we proved that PD-L1-loading MPs could suppress the activation and function of functional cluster of differentiation CD8+ T cells. Meanwhile, the PD-L1-loading MPs could mediate the differentiation of macrophages toward the immune-suppressive M2 phenotype via the activation of the TANK-binding kinase 1 (TBK1)/signal transducer and activator of transcription 6 (STAT6) signal and suppression of the serine-threonine kinase (AKT)/mammalian target of rapamycin (mTOR) signal. Given the increasing MP production induced by traditional clinical interventions, we further combined chemotherapy with the PD-L1 inhibitor atezolizumab (ATZ) to efficiently abrogate the immunosuppression caused by the PD-L1-loading MPs. Therefore, our study unveils the mechanism by which tumor cells systemically evade immune surveillance by releasing the PD-L1-loading MPs, and provides new insights into clinical TNBC immunotherapy.
Collapse
Affiliation(s)
- Cong Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuannanli, Chaoyang District, 100021, Beijing, China
| | - Shi Qiu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Kun Jin
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xiaonan Zheng
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xianghong Zhou
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Di Jin
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuannanli, Chaoyang District, 100021, Beijing, China.
| | - Xun Jin
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
232
|
Cortesi M, Zanoni M, Pirini F, Tumedei MM, Ravaioli S, Rapposelli IG, Frassineti GL, Bravaccini S. Pancreatic Cancer and Cellular Senescence: Tumor Microenvironment under the Spotlight. Int J Mol Sci 2021; 23:ijms23010254. [PMID: 35008679 PMCID: PMC8745092 DOI: 10.3390/ijms23010254] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 01/10/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has one of the most dismal prognoses of all cancers due to its late manifestation and resistance to current therapies. Accumulating evidence has suggested that the malignant behavior of this cancer is mainly influenced by the associated strongly immunosuppressive, desmoplastic microenvironment and by the relatively low mutational burden. PDAC develops and progresses through a multi-step process. Early in tumorigenesis, cancer cells must evade the effects of cellular senescence, which slows proliferation and promotes the immune-mediated elimination of pre-malignant cells. The role of senescence as a tumor suppressor has been well-established; however, recent evidence has revealed novel pro-tumorigenic paracrine functions of senescent cells towards their microenvironment. Understanding the interactions between tumors and their microenvironment is a growing research field, with evidence having been provided that non-tumoral cells composing the tumor microenvironment (TME) influence tumor proliferation, metabolism, cell death, and therapeutic resistance. Simultaneously, cancer cells shape a tumor-supportive and immunosuppressive environment, influencing both non-tumoral neighboring and distant cells. The overall intention of this review is to provide an overview of the interplay that occurs between senescent and non-senescent cell types and to describe how such interplay may have an impact on PDAC progression. Specifically, the effects and the molecular changes occurring in non-cancerous cells during senescence, and how these may contribute to a tumor-permissive microenvironment, will be discussed. Finally, senescence targeting strategies will be briefly introduced, highlighting their potential in the treatment of PDAC.
Collapse
Affiliation(s)
- Michela Cortesi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.Z.); (F.P.); (M.M.T.); (S.R.); (S.B.)
- Correspondence:
| | - Michele Zanoni
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.Z.); (F.P.); (M.M.T.); (S.R.); (S.B.)
| | - Francesca Pirini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.Z.); (F.P.); (M.M.T.); (S.R.); (S.B.)
| | - Maria Maddalena Tumedei
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.Z.); (F.P.); (M.M.T.); (S.R.); (S.B.)
| | - Sara Ravaioli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.Z.); (F.P.); (M.M.T.); (S.R.); (S.B.)
| | - Ilario Giovanni Rapposelli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (I.G.R.); (G.L.F.)
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (I.G.R.); (G.L.F.)
| | - Sara Bravaccini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.Z.); (F.P.); (M.M.T.); (S.R.); (S.B.)
| |
Collapse
|
233
|
Davidsson S, Carlsson J, Greenberg L, Wijkander J, Söderquist B, Erlandsson A. Cutibacterium acnes Induces the Expression of Immunosuppressive Genes in Macrophages and is Associated with an Increase of Regulatory T-Cells in Prostate Cancer. Microbiol Spectr 2021; 9:e0149721. [PMID: 34937192 PMCID: PMC8694172 DOI: 10.1128/spectrum.01497-21] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/30/2021] [Indexed: 12/26/2022] Open
Abstract
Tumors and infectious agents both benefit from an immunosuppressive environment. Cutibacterium acnes (C. acnes) is a bacterium in the normal skin microbiota, which has the ability to survive intracellularly in macrophages and is significantly more common in prostate cancer tissue compared with normal prostate tissue. This study investigated if prostate cancer tissue culture positive for C. acnes has a higher infiltration of regulatory T-cells (Tregs) and if macrophages stimulated with C. acnes induced the expression of immunosuppressive genes that could be linked to an increase of Tregs in prostate cancer. Real-time PCR and enzyme-linked immunosorbent spot assay (ELISA) were used to examine the expression of immunosuppressive genes in human macrophages stimulated in vitro with C. acnes, and associations between the presence of C. acnes and infiltration of Tregs were investigated by statistically analyzing data generated in two previous studies. The in vitro results demonstrated that macrophages stimulated with C. acnes significantly increased their expression of PD-L1, CCL17, and CCL18 mRNA and protein (p <0.05). In the cohort, Tregs in tumor stroma and tumor epithelia were positively associated with the presence of C. acnes (P = 0.0004 and P = 0.046, respectively). Since the macrophages stimulated with C. acnes in vitro increased the expression of immunosuppressive genes, and prostate cancer patients with prostatic C. acnes infection had higher infiltration of Tregs than their noninfected counterparts, we suggest that C. acnes may contribute to an immunosuppressive tumor environment that is vital for prostate cancer progression. IMPORTANCE In an immune suppressive tumor microenvironment constituted by immunosuppressive cells and immunosuppressive mediators, tumors may improve their ability to give rise to a clinically relevant cancer. In the present study, we found that C. acnes might contribute to an immunosuppressive environment by recruiting Tregs and by increasing the expression of immunosuppressive mediators such as PD-L1, CCL17, and CCL18. We believe that our data add support to the hypothesis of a contributing role of C. acnes in prostate cancer development. If established that C. acnes stimulates prostate cancer progression it may open up avenues for targeted prostate cancer treatment.
Collapse
Affiliation(s)
- Sabina Davidsson
- Department of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Jessica Carlsson
- Department of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Larry Greenberg
- Department of Environmental and Life Sciences/Biology, Faculty of Health, Science and Technology, Karlstad University, Karlstad, Sweden
| | - Jonny Wijkander
- Department of Health Sciences, Faculty of Health, Science and Technology, Karlstad University, Karlstad, Sweden
| | - Bo Söderquist
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ann Erlandsson
- Department of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Environmental and Life Sciences/Biology, Faculty of Health, Science and Technology, Karlstad University, Karlstad, Sweden
| |
Collapse
|
234
|
Zhu X, Wang X, Gong Y, Deng J. E-cadherin on epithelial-mesenchymal transition in thyroid cancer. Cancer Cell Int 2021; 21:695. [PMID: 34930256 PMCID: PMC8690896 DOI: 10.1186/s12935-021-02344-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/15/2021] [Indexed: 02/08/2023] Open
Abstract
Thyroid carcinoma is a common malignant tumor of endocrine system and head and neck. Recurrence, metastasis and high malignant expression after routine treatment are serious clinical problems, so it is of great significance to explore its mechanism and find action targets. Epithelial-mesenchymal transition (EMT) is associated with tumor malignancy and invasion. One key change in tumour EMT is low expression of E-cadherin. Therefore, this article reviews the expression of E-cadherin in thyroid cancers (TC), discuss the potential mechanisms involved, and outline opportunities to exploit E-cadherin on regulating the occurrence of EMT as a critical factor in cancer therapeutics.
Collapse
Affiliation(s)
- Xiaoyu Zhu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Jing'an District, Shanghai, 200040, China
| | - Xiaoping Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Jing'an District, Shanghai, 200040, China.
| | - Yifei Gong
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Jing'an District, Shanghai, 200040, China
| | - Junlin Deng
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Road, Jing'an District, Shanghai, 200040, China
| |
Collapse
|
235
|
Lamplugh Z, Fan Y. Vascular Microenvironment, Tumor Immunity and Immunotherapy. Front Immunol 2021; 12:811485. [PMID: 34987525 PMCID: PMC8720970 DOI: 10.3389/fimmu.2021.811485] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 11/29/2021] [Indexed: 01/01/2023] Open
Abstract
Immunotherapy holds great promise for treating cancer. Nonetheless, T cell-based immunotherapy of solid tumors has remained challenging, largely due to the lack of universal tumor-specific antigens and an immunosuppressive tumor microenvironment (TME) that inhibits lymphocyte infiltration and activation. Aberrant vascularity characterizes malignant solid tumors, which fuels the formation of an immune-hostile microenvironment and induces tumor resistance to immunotherapy, emerging as a crucial target for adjuvant treatment in cancer immunotherapy. In this review, we discuss the molecular and cellular basis of vascular microenvironment-mediated tumor evasion of immune responses and resistance to immunotherapy, with a focus on vessel abnormality, dysfunctional adhesion, immunosuppressive niche, and microenvironmental stress in tumor vasculature. We provide an overview of opportunities and challenges related to these mechanisms. We also propose genetic programming of tumor endothelial cells as an alternative approach to recondition the vascular microenvironment and to overcome tumor resistance to immunotherapy.
Collapse
Affiliation(s)
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
236
|
Wang X, Wu Y, Gu J, Xu J. Tumor-associated macrophages in lung carcinoma: From mechanism to therapy. Pathol Res Pract 2021; 229:153747. [PMID: 34952424 DOI: 10.1016/j.prp.2021.153747] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/09/2022]
Abstract
Tumor-associated macrophages (TAMs), which could be classified into the classical (M1-like) and alternatively activated (M2-like) phenotype, were considered to be important tumor-promoting components in lung cancer microenvironment. Several studies reported that TAMs in lung tumor islet or stroma are usually correlated with poor prognosis. Further studies showed that TAMs could promote the initiation of tumor cells, inhibit antitumor immune responses, and stimulate tumor angiogenesis and subsequently tumor metastasis of lung carcinoma. Currently, TAMs have been considered as penitential targets of lung cancer. This review summarizes from the fundamental information of TAMs to the its role in metastasis and present evidence for TAMs as a potential target of cancer therapy.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yining Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jiahui Gu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jian Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; National Key Clinical Department of Laboratory Medicine, Nanjing 210029, China.
| |
Collapse
|
237
|
Persistent Cryptosporidium parvum Infection Leads to the Development of the Tumor Microenvironment in an Experimental Mouse Model: Results of a Microarray Approach. Microorganisms 2021; 9:microorganisms9122569. [PMID: 34946170 PMCID: PMC8704780 DOI: 10.3390/microorganisms9122569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 12/01/2022] Open
Abstract
Cryptosporidium spp. are enteric protozoa parasites that infect a variety of vertebrate hosts. These parasites are capable of inducing life-threatening gastrointestinal disease in immunocompromised individuals. With the rising epidemiological evidence of the occurrence of Cryptosporidium infections in humans with digestive cancer, the tumorigenic potential of the parasite has been speculated. In this regard, Cryptosporidium parvum has been reported to induce digestive adenocarcinoma in a rodent model of chronic cryptosporidiosis. However, the processes by which the parasite could induce this carcinogenesis are still unknown. Therefore, the transcriptomes of C. parvum infected ileo-cecal regions of mice developing tumors were analyzed in the current study. For the first time, downregulation of the expression of α-defensin, an anti-microbial target of the parasite in response to C. parvum infection was observed in the transformed tissues. This phenomenon has been speculated to be the result of resistance of C. parvum to the host defense through the upregulated expression of interferon γ-stimulated genes. The inflammatory response generated as result of attenuated expression of anti-microbial peptides highlights the role of immune evasion in the C. parvum-induced tumorigenesis. The study has also succeeded in the characterization of the tumor microenvironment (TME) which is characterized by the presence of cancer associated fibroblasts, myeloid-derived suppressor cells, tumor-associated macrophages and extracellular matrix components. Identification of immune suppressor cells and accumulation of pro-inflammatory mediators speculates that chronic inflammation induced by persistent C. parvum infection assists in development of an immunosuppressive tumor microenvironment.
Collapse
|
238
|
Dizman N, Buchbinder EI. Cancer Therapy Targeting CD47/SIRPα. Cancers (Basel) 2021; 13:cancers13246229. [PMID: 34944850 PMCID: PMC8699673 DOI: 10.3390/cancers13246229] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 01/01/2023] Open
Abstract
Simple Summary The interaction between cluster of differentiation 47 (CD47) on cancer cells and signal regulatory protein alpha (SIRPα) on immune cells, such as macrophages and dendritic cells, generates a “don’t eat me” signal. This is a common mechanism that provides cancer cells an escape from the innate immune system. Several therapeutics directed to CD47 or SIRPα have entered early clinical trials in recent years. In this article, we review the role of CD47/SIRPα axis in cancer, and summarize the literature on the efficacy and safety of therapeutics targeting CD47 or SIRPα. We also discuss the future implementation of these therapeutics in the treatments of various cancer types. Abstract In the past decade, the field of cancer immunotherapy has rapidly advanced, establishing a crucial role for immune checkpoint blockers in the treatment of a variety of cancer types. In parallel with these remarkable clinical developments, further efforts have focused on ways of unleashing adaptive immune responses against cancer. CD47, a cell surface molecule overexpressed by several cancer types that facilitates immune escape from macrophages, dendritic cells and natural killer cells, and its ligand SIRPα, have emerged as potential therapeutic targets. A number of agents directed to CD47/SIRPα have been developed and demonstrated preclinical activity. Early phase clinical trials are investigating CD47/SIRPα directed agents with available data, suggesting safety and preliminary activity. Herein, we provide an overview of the mechanistic rationale of targeting CD47/SIRPα axis and associated clinical evidence.
Collapse
Affiliation(s)
- Nazli Dizman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Elizabeth I. Buchbinder
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA
- Correspondence:
| |
Collapse
|
239
|
Pimenta DB, Varela VA, Datoguia TS, Caraciolo VB, Lopes GH, Pereira WO. The Bone Marrow Microenvironment Mechanisms in Acute Myeloid Leukemia. Front Cell Dev Biol 2021; 9:764698. [PMID: 34869355 PMCID: PMC8639599 DOI: 10.3389/fcell.2021.764698] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022] Open
Abstract
Bone marrow (BM) is a highly complex tissue that provides important regulatory signals to orchestrate hematopoiesis. Resident and transient cells occupy and interact with some well characterized niches to produce molecular and cellular mechanisms that interfere with differentiation, migration, survival, and proliferation in this microenvironment. The acute myeloid leukemia (AML), the most common and severe hematological neoplasm in adults, arises and develop in the BM. The osteoblastic, vascular, and reticular niches provide surface co-receptors, soluble factors, cytokines, and chemokines that mediate important functions on hematopoietic cells and leukemic blasts. There are some evidences of how AML modify the architecture and function of these three BM niches, but it has been still unclear how essential those modifications are to maintain AML development. Basic studies and clinical trials have been suggesting that disturbing specific cells and molecules into the BM niches might be able to impair leukemia competencies. Either through niche-specific molecule inhibition alone or in combination with more traditional drugs, the bone marrow microenvironment is currently considered the potential target for new strategies to treat AML patients. This review describes the cellular and molecular constitution of the BM niches under healthy and AML conditions, presenting this anatomical compartment by a new perspective: as a prospective target for current and next generation therapies.
Collapse
Affiliation(s)
- Débora Bifano Pimenta
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Vanessa Araujo Varela
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Tarcila Santos Datoguia
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Victória Bulcão Caraciolo
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Gabriel Herculano Lopes
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Welbert Oliveira Pereira
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
240
|
Chen H, Sun Q, Zhang C, She J, Cao S, Cao M, Zhang N, Adiila AV, Zhong J, Yao C, Wang Y, Xia H, Lan L. Identification and Validation of CYBB, CD86, and C3AR1 as the Key Genes Related to Macrophage Infiltration of Gastric Cancer. Front Mol Biosci 2021; 8:756085. [PMID: 34950700 PMCID: PMC8688826 DOI: 10.3389/fmolb.2021.756085] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) is rampant around the world. Most of the GC cases are detected in advanced stages with poor prognosis. The identification of marker genes for early diagnosis is of great significance. Studying the tumor environment is helpful to acknowledge the process of tumorigenesis, development, and metastasis. Twenty-two kinds of immune cells were calculated by CIBERSORT from Gene Expression Omnibus (GEO) database. Subsequently, higher infiltration of macrophages M0 was discovered in GC compared with normal tissues. WGCNA was utilized to construct the network and then identify key modules and genes related to macrophages in TCGA. Finally, 18 hub genes were verified. In the PPI bar chart, the top 3 genes were chosen as hub genes involved in most pathways. On the TIMER and THPA websites, it is verified that the expression levels of CYBB, CD86, and C3AR1 genes in tumor tissues were higher than those in normal tissues. These genes may work as biomarkers or targets for accurate diagnosis and treatment of GC in the future. Our findings may be a new strategy for the treatment of GC.
Collapse
Affiliation(s)
- Haiyan Chen
- Institute for Cancer Research, School of Basic Medical Science of Xi’an Jiaotong University, Xi’an, China
| | - Qi Sun
- Department of Pathology, School of Basic Medical Sciences and Sir Run Run Hospital and Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Junjun She
- Department of High Talent and General Surgery and Center for Gut Microbiome Research and Med-X Institute, the First Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Shuai Cao
- Department of Orthopedics, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Meng Cao
- Institute for Cancer Research, School of Basic Medical Science of Xi’an Jiaotong University, Xi’an, China
| | - Nana Zhang
- Institute for Cancer Research, School of Basic Medical Science of Xi’an Jiaotong University, Xi’an, China
| | - Ayarick Vivian Adiila
- Institute for Cancer Research, School of Basic Medical Science of Xi’an Jiaotong University, Xi’an, China
| | - Jinjin Zhong
- Institute for Cancer Research, School of Basic Medical Science of Xi’an Jiaotong University, Xi’an, China
| | - Chengyun Yao
- Jiangsu Cancer Hospital and the Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yili Wang
- Institute for Cancer Research, School of Basic Medical Science of Xi’an Jiaotong University, Xi’an, China
| | - Hongping Xia
- Department of Pathology, School of Basic Medical Sciences and Sir Run Run Hospital and Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China
- Department of High Talent and General Surgery and Center for Gut Microbiome Research and Med-X Institute, the First Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Linhua Lan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
241
|
Ito Y, Yamamoto T, Miyai K, Take J, Scherthan H, Rommel A, Eder S, Steinestel K, Rump A, Port M, Shinomiya N, Kinoshita M. Ascorbic acid-2 glucoside mitigates intestinal damage during pelvic radiotherapy in a rat bladder tumor model. Int J Radiat Biol 2021; 98:942-957. [PMID: 34871138 DOI: 10.1080/09553002.2021.2009145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE Ascorbic acid is a strong antioxidant and has potent radioprotective effects on radiation injuries. Ascorbic acid 2-glucoside (AA2G) is a stabilized derivative of ascorbic acid and rapidly hydrolyzed into ascorbic acid and glucose. Since there is the possibility that AA2G treatment interferes with the antitumor activity of radiotherapy, we investigated the effect of AA2G treatment during radiotherapy on acute radiation enteritis and antitumor activity of radiotherapy in rats. MATERIALS AND METHODS AY-27 rat bladder tumor cells were used to induce bladder tumors in rats. Two weeks after inoculation rats received fractionated pelvic radiotherapy in eight fractions for 4 weeks totaling 40 Gy. During radiotherapy, one group of rats received per os AA2G (ascorbic acid: 250 mg/kg/day) and its bolus engulfment (ascorbic acid: 250 mg/kg) 8 h before each X-irradiation fraction. Seven days after the last X-irradiation, we studied histology, DNA double strand break (DSB) damage (by 53BP1 foci staining), and the M1/M2 macrophage response by immunohistochemistry of paraffin-fixed bladder and intestinal tissues. RESULTS AA2G treatment reduced the intestinal damage (shortening of villi) but did not reduce antitumor effectiveness of radiotherapy against bladder tumors. Like the controls, AA2G-treated rats showed no residual tumor lesions in the bladder after X-irradiation. Both AA2G-treated and control groups showed similar persistent DSB damage (53BP1 foci) both in bladders and ilea seven days after radiotherapy. Radiotherapy tended to reduce CD163+ M2 macrophages, which are considered as an anti-inflammatory subtype favoring tissue repair, in the bladders. X-irradiation also reduced the occurrence of M2 macrophages in the ilea. AA2G treatment significantly increased CD163+/CD68+ macrophage ratio in the ilea of rats after pelvic irradiation in comparison to the sham irradiated control rats. AA2G treatment increased, albeit not significantly, the CD163+/CD68+ macrophage ratio in the irradiated bladders relative to the control irradiated rats. On the other hand, bladders and ilea of the irradiated rats with and without AA2G treatment showed similar frequencies of CD68+ macrophages. CONCLUSIONS AA2G treatment mitigated radiation-induced intestinal damage without reducing antitumor activity after fractionated pelvic radiotherapy against bladder tumors in rats. The beneficial effect of AA2G treatment seems to promote a restoration of the M2 answer as well as tissue remodeling and wound healing. Similar residual DNA damage in bladders and ilea seven days post-irradiation is consistent with tumor control in both groups.
Collapse
Affiliation(s)
- Yasutoshi Ito
- Military Medicine Research Unit, Test and Evaluation Command, Ground Self-Defense Force, Setagaya, Japan
| | - Tetsuo Yamamoto
- Military Medicine Research Unit, Test and Evaluation Command, Ground Self-Defense Force, Setagaya, Japan.,NBC Counter Medical Unit, Ground Self-Defense Force, Setagaya, Japan
| | - Kosuke Miyai
- Military Medicine Research Unit, Test and Evaluation Command, Ground Self-Defense Force, Setagaya, Japan.,Department of Pathology, Self-Defense Forces Central Hospital, Setagaya, Japan
| | - Junya Take
- Department of Pediatrics, National Defense Medical College, Tokorozawa, Japan
| | | | - Anna Rommel
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | - Stefan Eder
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | | | - Alexis Rump
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | - Matthias Port
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | | | - Manabu Kinoshita
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
242
|
Sharma D, Hamlet S, Vaquette C, Petcu EB, Ramamurthy P, Ivanovski S. Local delivery of hydrogel encapsulated vascular endothelial growth factor for the prevention of medication-related osteonecrosis of the jaw. Sci Rep 2021; 11:23371. [PMID: 34862395 PMCID: PMC8642483 DOI: 10.1038/s41598-021-02637-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022] Open
Abstract
The anti-angiogenic effects of bisphosphonates have been hypothesized as one of the major etiologic factors in the development of medication-related osteonecrosis of the jaw (MRONJ), a severe debilitating condition with limited treatment options. This study evaluated the potential of a gelatine-hyaluronic acid hydrogel loaded with the angiogenic growth factor, vascular endothelial growth factor (VEGF), as a local delivery system to aid in maintaining vascularization in a bisphosphonate-treated (Zoledronic Acid) rodent maxillary extraction defect. Healing was assessed four weeks after implantation of the VEGF-hydrogel into extraction sockets. Gross examination and histological assessment showed that total osteonecrosis and inflammatory infiltrate was significantly reduced in the presence of VEGF. Also, total vascularity and specifically neovascularization, was significantly improved in animals that received VEGF hydrogel. Gene expression of vascular, inflammatory and bone specific markers within the defect area were also significantly altered in the presence of VEGF. Furthermore, plasma cytokine levels were assessed to determine the systemic effect of locally delivered VEGF and showed similar outcomes. In conclusion, the use of locally delivered VEGF within healing extraction sockets assists bone healing and prevents MRONJ via a pro-angiogenic and immunomodulatory mechanism.
Collapse
Affiliation(s)
- Dileep Sharma
- College of Medicine and Dentistry, James Cook University, Cairns Campus, PO Box 6811, Cairns, 4870, Australia. .,Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia.
| | - Stephen Hamlet
- Menzies Health Institute Queensland, School of Medicine and Dentistry, Griffith University, Gold Coast Campus, Gold Coast, 4222, Australia
| | - Cedryck Vaquette
- School of Dentistry, Faculty of Health and Behavioral Sciences, The University of Queensland, Herston Campus, Brisbane, 4006, Australia
| | - Eugen Bogdan Petcu
- New York Institute of Technology College of Osteopathic Medicine (NYIT), Old Westbury, NY, 11545, USA.,School of Dentistry and Oral Health, Griffith University, Gold Coast, Australia
| | - Poornima Ramamurthy
- College of Medicine and Dentistry, James Cook University, Cairns Campus, PO Box 6811, Cairns, 4870, Australia
| | - Saso Ivanovski
- School of Dentistry, Faculty of Health and Behavioral Sciences, The University of Queensland, Herston Campus, Brisbane, 4006, Australia.
| |
Collapse
|
243
|
Kuroda H, Jamiyan T, Yamaguchi R, Kakumoto A, Abe A, Harada O, Masunaga A. Tumor microenvironment in triple-negative breast cancer: the correlation of tumor-associated macrophages and tumor-infiltrating lymphocytes. Clin Transl Oncol 2021; 23:2513-2525. [PMID: 34089486 PMCID: PMC8557183 DOI: 10.1007/s12094-021-02652-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/21/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE Immune cells such as cytotoxic T cells, helper T cells, B cells or tumor-associated macrophages (TAMs) contribute to the anti-tumor response or pro-tumorigenic effect in triple negative breast cancer (TNBC). The interrelation of TAMs, T and B tumor-infiltrating lymphocytes (TILs) in TNBC has not been fully elucidated. METHODS We evaluated the association of tumor-associated macrophages, T and B TILs in TNBC. RESULTS TNBCs with a high CD68+, CD163+ TAMs and low CD4+, CD8+, CD20+ TILs had a significantly shorter relapse-free survival (RFS) and overall survival (OS) than those with low CD68+, CD163+ TAMs and high CD4+, CD8+, CD20+ TILs. TNBCs with high CD68+ TAMs/low CD8+ TILs showed a significantly shorter RFS and OS and a significantly poorer prognosis than those with high CD68+ TAMs/high CD8+ TILs, low CD68+ TAMs/high CD8+ TILs, and low CD68+/low CD8+. TNBCs with high CD163+ TAMs/low CD8+, low CD20 + TILs showed a significantly shorter RFS and OS and a significantly poorer prognosis than those with high CD163+ TAMs/high CD8+ TILs and high CD163+ TAMs /high CD20+ TILs. CONCLUSIONS Our study suggests that TAMs further create an optimal tumor microenvironment (TME) for growth and invasion of cancer cells when evasion of immunoreactions due to T and B TILs occurs. In TNBCs, all these events combine to affect prognosis. The process of TME is highly complex in TNBCs and for an improved understanding, larger validation studies are necessary to confirm these findings.
Collapse
Affiliation(s)
- H Kuroda
- Department of Diagnostic Pathology, Tokyo Women's Medical University, Medical Center East, 2-1-10 Nishiogu, Arakawa-ku, Tokyo, 116-8567, Japan.
- Department of Diagnostic Pathology, Dokkyo Medical University, Mibu, Japan.
| | - T Jamiyan
- Department of Diagnostic Pathology, Dokkyo Medical University, Mibu, Japan
- Department of Pathology and Forensic Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - R Yamaguchi
- Department of Pathology & Laboratory Medicine, Kurume University Medical Center, Kurume, Japan
| | - A Kakumoto
- Department of Diagnostic Pathology, Tokyo Women's Medical University, Medical Center East, 2-1-10 Nishiogu, Arakawa-ku, Tokyo, 116-8567, Japan
- Department of Diagnostic Pathology, Nasu Red Cross Hospital, Otawara, Japan
| | - A Abe
- Breast Center, Dokkyo Medical University, Mibu, Japan
| | - O Harada
- Breast Center, Showa University, Tokyo, Japan
| | - A Masunaga
- Department of Diagnostic Pathology, Tokyo Women's Medical University, Medical Center East, 2-1-10 Nishiogu, Arakawa-ku, Tokyo, 116-8567, Japan
| |
Collapse
|
244
|
Meng F, Xiao Y, Xie L, Liu Q, Qian K. Diagnostic and prognostic value of ABC transporter family member ABCG1 gene in clear cell renal cell carcinoma. Channels (Austin) 2021; 15:375-385. [PMID: 33825659 PMCID: PMC8032227 DOI: 10.1080/19336950.2021.1909301] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/24/2022] Open
Abstract
As the most common histologic subtype of renal cancer, clear cell renal cell carcinoma (ccRCC) poses a serious threat to public health. However, there are no specific molecular-targeted drugs for ccRCC at present. Human ATP-binding cassette (ABC) transporter family plays an important role in homeostasis maintenance. This study aimed to evaluate the potential diagnostic value of ABC genes in ccRCC. A total of 952 samples of ccRCC patients (707) and controls (245) from three different datasets were included for analysis. Receiver operating characteristic analysis and t-test were used to analyze the differential expression of ABC genes in ccRCC patients and control samples at mRNA level during screening and validations. The Cancer Genome Atlas (TCGA-ccRCC) dataset was utilized to investigate the correlation between ABC genes expression and prognostic value in ccRCC. We then investigated the interactions between ABCG1 and proteins in the Comparative Toxicogenomics Database (CTD). Finally, we found that ATP-binding cassette transporter G member 1 (ABCG1) was over-expressed in ccRCC patients compared with healthy samples at mRNA level. Cox regression analysis and Kaplan-Meier analysis showed that ccRCC patients with high ABCG1 expression had better overall survival (OS) than those patients with low expression (hazard ratio (HR) = 0.662, p = 0.007). This study demonstrated that ABCG1 is a potential diagnostic and prognostic biomarker in ccRCC and discussed the molecular mechanisms underlying the relationship between ccRCC and ABCG1, which might provide guidance for better management and treatment of ccRCC in the future.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/diagnosis
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/diagnosis
- ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism
- Prognosis
- Male
- Female
- Middle Aged
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Gene Expression Regulation, Neoplastic
Collapse
Affiliation(s)
- Fucheng Meng
- Department of Infection Control, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yafei Xiao
- Department of Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Longxiang Xie
- Department of Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Qiao Liu
- Department of Pediatric Dentistry, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Keli Qian
- Department of Infection Control, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
245
|
Chen T, Wang Y, Nan Z, Wu J, Li A, Zhang T, Qu X, Li C. Interaction Between Macrophage Extracellular Traps and Colon Cancer Cells Promotes Colon Cancer Invasion and Correlates With Unfavorable Prognosis. Front Immunol 2021; 12:779325. [PMID: 34925357 PMCID: PMC8671452 DOI: 10.3389/fimmu.2021.779325] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 11/15/2021] [Indexed: 01/03/2023] Open
Abstract
BackgroundMacrophage extracellular traps (METs) and tumor-infiltrating macrophages contribute to the progression of several diseases. But the role of METs and tumor-infiltrating macrophages in colon cancer (CC) has not been illuminated. In this study, we aimed to clarify the prognostic value of METs for CC patients and to explore the interaction between CC cells and METs in vitro and in vivo.MethodsA training cohort consisting of 116 patients and a validation cohort of 94 patients were enrolled in this study. Immunofluorescence (IF) staining was conducted to determine METs formation in CC patients. Cox regression was used to perform prognostic analysis and screen out the best prognostic model. A nomogram was established to predict 5-year overall survival (OS). The correlation between METs with clinicopathological features and inflammatory markers was analyzed. The formation of METs in vitro was detected by SYTOX® green and IF staining, and the effect of METs on CC cells was detected by transwell assays. PAD2-IN-1, a selective inhibitor of peptidylarginine deiminase 2 (PAD2), was introduced to destroy the crosstalk between CC cells and METs in vitro and in vivo.ResultsMETs levels were higher in CC tissues and were an independent prognostic factor for CC patients. The prognostic model consisting of age, tumors local invasion, lymph node metastasis and METs were confirmed to be consistent and accurate for predicting the 5-year OS of CC patients. Besides, METs were correlated with distant metastasis and inflammation. Through in vitro experiments, we confirmed that there was a positive feedback loop between CC cells and METs, in that METs promoted the invasion of CC cells and CC cells enhanced the production of METs, in turn. This interaction could be blocked by PAD2-IN-1 inhibitors. More importantly, animal experiments revealed that PAD2-IN-1 inhibited METs formation and CC liver metastasis in vivo.ConclusionsMETs were the potential biomarker of CC patient prognosis. PAD2-IN-1 inhibited the crosstalk between CC cells and METs in vitro and in vivo, which should be emphasized in CC therapy.
Collapse
Affiliation(s)
- Tianli Chen
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Wang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhaodi Nan
- Institute of Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Wu
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ailu Li
- Department of Obstetrics and Gynecology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Tingguo Zhang
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xun Qu
- Institute of Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chen Li
- Department of Ultrasound, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Chen Li,
| |
Collapse
|
246
|
Huang Y, Chen L, Tang Z, Min Y, Yu W, Yang G, Zhang L. A Novel Immune and Stroma Related Prognostic Marker for Invasive Breast Cancer in Tumor Microenvironment: A TCGA Based Study. Front Endocrinol (Lausanne) 2021; 12:774244. [PMID: 34867821 PMCID: PMC8636929 DOI: 10.3389/fendo.2021.774244] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/01/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Breast cancer (BC) is the most frequent cancer in women. The tumor microenvironment (TME), consisting of blood vessels, immune cells, fibroblasts, and extracellular matrix, plays a pivotal role in tumorigenesis and progression. Increasing evidence has emphasized the importance of TME, especially the immune components, in patients with BC. Nevertheless, we still lack a deep understanding of the correlation between tumor invasion and TME status. METHODS Transcriptome and clinical data were retrieved from The Cancer Genome Atlas (TCGA) database. ESTIMATE algorithm was applied for quantifying stromal and immune scores. Then we screened out the differentially expressed genes (DEGs) through the intersection analysis. Furthermore, the establishment of protein-protein interaction (PPI) network and univariate COX regression analysis were utilized to determine the core genes in DEGs. In addition, we also performed Gene Set Enrichment Analysis (GSEA) and CIBERSORT analysis to distinguish the function of crucial gene expression and the proportion of tumor-infiltrating immune cells (TICs), respectively. RESULTS A total of 1178 samples (112 normal samples and 1066 tumor samples) were extracted from TCGA for calculation, and 226 DEGs were obtained from this assessment. Further intersection analysis revealed eight key genes, including ITK, CD3E, CCL19, CD2, SH2D1A, CD5, SLAMF6, SPN, which were proven to correlate with BC status. Moreover, ITK was picked out for further study. The results illustrated that high expression of BC patients had a more prolonged overall survival (OS) time than ITK low expression BC patients (p = 0.009), and ITK expression also presented the statistical significance in age, TNM staging, tumor size classification, and metastasis classification. Additionally, GSEA and CIBERSORT analysis indicated that ITK expression had an association with immune activity in TME. CONCLUSION ITK may be a potential indicator for prognosis prediction in patients with BC, and its biological behavior may promote our understanding of the molecular mechanism of tumor progression and targeted therapy.
Collapse
Affiliation(s)
- Yizhou Huang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lizhi Chen
- College of Pharmacy, Southwest Medical University, Luzhou, China
| | - Ziyi Tang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Min
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wanli Yu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lili Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
247
|
Duran CL, Borriello L, Karagiannis GS, Entenberg D, Oktay MH, Condeelis JS. Targeting Tie2 in the Tumor Microenvironment: From Angiogenesis to Dissemination. Cancers (Basel) 2021; 13:cancers13225730. [PMID: 34830883 PMCID: PMC8616247 DOI: 10.3390/cancers13225730] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary The dissemination of cancer cells from their original location to distant organs where they grow, a process called metastasis, causes more than 90% of cancer deaths. The identification of the molecular mechanisms of metastasis and the development of anti-metastatic therapies are essential to increase patient survival. In recent years, targeting the tumor microenvironment has become a promising avenue to prevent both tumor growth and metastasis. As the tumor microenvironment contains not only cancer cells but also blood vessels, immune cells, and other non-cancerous cells, it is naïve to think that therapy only affects a single cell type in this complex environment. Here we review the importance, and ways to inhibit the function, of one therapeutic target: the receptor Tie2. Tie2 is a receptor present on the cell surface of several cell types within the tumor microenvironment and regulates tumor angiogenesis, growth, and metastasis to distant organs. Abstract The Tie2 receptor tyrosine kinase is expressed in vascular endothelial cells, tumor-associated macrophages, and tumor cells and has been a major focus of research in therapies targeting the tumor microenvironment. The most extensively studied Tie2 ligands are Angiopoietin 1 and 2 (Ang1, Ang2). Ang1 plays a critical role in vessel maturation, endothelial cell migration, and survival. Ang2, depending on the context, may function to disrupt connections between the endothelial cells and perivascular cells, promoting vascular regression. However, in the presence of VEGF-A, Ang2 instead promotes angiogenesis. Tie2-expressing macrophages play a critical role in both tumor angiogenesis and the dissemination of tumor cells from the primary tumor to secondary sites. Therefore, Ang-Tie2 signaling functions as an angiogenic switch during tumor progression and metastasis. Here we review the recent advances and complexities of targeting Tie2 signaling in the tumor microenvironment as a possible anti-angiogenic, and anti-metastatic, therapy and describe its use in combination with chemotherapy.
Collapse
Affiliation(s)
- Camille L. Duran
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA; (C.L.D.); (L.B.); (D.E.); (M.H.O.)
- Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA;
| | - Lucia Borriello
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA; (C.L.D.); (L.B.); (D.E.); (M.H.O.)
- Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA;
| | - George S. Karagiannis
- Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA;
- Department of Microbiology and Immunology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
- Integrated Imaging Program, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
| | - David Entenberg
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA; (C.L.D.); (L.B.); (D.E.); (M.H.O.)
- Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA;
- Integrated Imaging Program, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
| | - Maja H. Oktay
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA; (C.L.D.); (L.B.); (D.E.); (M.H.O.)
- Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA;
- Integrated Imaging Program, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
- Department of Pathology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
| | - John S. Condeelis
- Department of Anatomy and Structural Biology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA; (C.L.D.); (L.B.); (D.E.); (M.H.O.)
- Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA;
- Integrated Imaging Program, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
- Department of Surgery, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
- Correspondence:
| |
Collapse
|
248
|
Seebacher NA, Krchniakova M, Stacy AE, Skoda J, Jansson PJ. Tumour Microenvironment Stress Promotes the Development of Drug Resistance. Antioxidants (Basel) 2021; 10:1801. [PMID: 34829672 PMCID: PMC8615091 DOI: 10.3390/antiox10111801] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 01/18/2023] Open
Abstract
Multi-drug resistance (MDR) is a leading cause of cancer-related death, and it continues to be a major barrier to cancer treatment. The tumour microenvironment (TME) has proven to play an essential role in not only cancer progression and metastasis, but also the development of resistance to chemotherapy. Despite the significant advances in the efficacy of anti-cancer therapies, the development of drug resistance remains a major impediment to therapeutic success. This review highlights the interplay between various factors within the TME that collectively initiate or propagate MDR. The key TME-mediated mechanisms of MDR regulation that will be discussed herein include (1) altered metabolic processing and the reactive oxygen species (ROS)-hypoxia inducible factor (HIF) axis; (2) changes in stromal cells; (3) increased cancer cell survival via autophagy and failure of apoptosis; (4) altered drug delivery, uptake, or efflux and (5) the induction of a cancer stem cell (CSC) phenotype. The review also discusses thought-provoking ideas that may assist in overcoming the TME-induced MDR. We conclude that stressors from the TME and exposure to chemotherapeutic agents are strongly linked to the development of MDR in cancer cells. Therefore, there remains a vast area for potential research to further elicit the interplay between factors existing both within and outside the TME. Elucidating the mechanisms within this network is essential for developing new therapeutic strategies that are less prone to failure due to the development of resistance in cancer cells.
Collapse
Affiliation(s)
| | - Maria Krchniakova
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Alexandra E. Stacy
- Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Patric J. Jansson
- Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St. Leonards, NSW 2065, Australia
| |
Collapse
|
249
|
Tumour microenvironment: a non-negligible driver for epithelial-mesenchymal transition in colorectal cancer. Expert Rev Mol Med 2021; 23:e16. [PMID: 34758892 DOI: 10.1017/erm.2021.13] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer remains the leading cause of death worldwide, and metastasis is still the major cause of treatment failure for cancer patients. Epithelial-mesenchymal transition (EMT) has been shown to play a critical role in the metastasis cascade of epithelium-derived carcinoma. Tumour microenvironment (TME) refers to the local tissue environment in which tumour cells produce and live, including not only tumour cells themselves, but also fibroblasts, immune and inflammatory cells, glial cells and other cells around them, as well as intercellular stroma, micro vessels and infiltrated biomolecules from the nearby areas, which has been proved to widely participate in the occurrence and progress of cancer. Emerging and accumulating studies indicate that, on one hand, mesenchymal cells in TME can establish 'crosstalk' with tumour cells to regulate their EMT programme; on the other, EMT-tumour cells can create a favourable environment for their own growth via educating stromal cells. Recently, our group has conducted a series of studies on the interaction between tumour-associated macrophages (TAMs) and colorectal cancer (CRC) cells in TME, confirming that the interaction between TAMs and CRC cells mediated by cytokines or exosomes can jointly promote the metastasis of CRC by regulating the EMT process of tumour cells and the M2-type polarisation process of TAMs. Herein, we present an overview to describe the current knowledge about EMT in cancer, summarise the important role of TME in EMT, and provide an update on the mechanisms of TME-induced EMT in CRC, aiming to provide new ideas for understanding and resisting tumour metastasis.
Collapse
|
250
|
Tada Y, Hojo M, Yuzawa K, Nagasawa A, Suzuki J, Inomata A, Moriyasu T, Nakae D. Iron oxide nanoparticles exert inhibitory effects on N-Bis(2-hydroxypropyl)nitrosamine (DHPN)-induced lung tumorigenesis in rats. Regul Toxicol Pharmacol 2021; 128:105072. [PMID: 34742869 DOI: 10.1016/j.yrtph.2021.105072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 10/19/2022]
Abstract
Iron oxide nanoparticles (magnetite) have been widely used in industry and medicine. However, the safety assessment of magnetite has not been fully completed. The present study was conducted to assess effects of magnetite on carcinogenic activity, using a medium-term bioassay protocol. A total of 100 male Fischer 344 rats, 6 weeks old, were randomly divided into 5 groups of 20 animals each, and given a basal diet and drinking water containing 0 or 0.1% of N-bis(2-hydroxypropyl)nitrosamine (DHPN) for 2 weeks. Two weeks later, the rats were intratracheally instilled magnetite 7 times at an interval of 4 weeks, at the doses of 0, 1.0 or 5.0 mg/kg body weight, and sacrificed at the end of the experimental period of 30 weeks. The multiplicities of macroscopic lung nodules and histopathologically diagnosed bronchiolo-alveolar hyperplasia, induced by DHPN, were both significantly decreased by the high dose of magnetite. The expression of minichromosome maintenance (MCM) protein 7 in non-tumoral alveolar epithelial cells, and the number of CD163-positive macrophages in tumor nodules were both significantly reduced by magnetite. It is suggested that magnetite exerts inhibitory effects against DHPN-induced lung tumorigenesis, by the reduction of alveolar epithelial proliferation and the M2 polarization of tumor-associated macrophages.
Collapse
Affiliation(s)
- Yukie Tada
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shin'juku, Tokyo, 169-0073, Japan.
| | - Motoki Hojo
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shin'juku, Tokyo, 169-0073, Japan
| | - Katsuhiro Yuzawa
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shin'juku, Tokyo, 169-0073, Japan
| | - Akemichi Nagasawa
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shin'juku, Tokyo, 169-0073, Japan
| | - Jin Suzuki
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shin'juku, Tokyo, 169-0073, Japan
| | - Akiko Inomata
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shin'juku, Tokyo, 169-0073, Japan
| | - Takako Moriyasu
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health, 3-24-1 Hyakunincho, Shin'juku, Tokyo, 169-0073, Japan
| | - Dai Nakae
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakura-ga-Oka, Setagaya, Tokyo, 156-8502, Japan.
| |
Collapse
|