1
|
Hao H, Tian W, Gong J, Li J, Zhou S, Guo Z, Jiao Y, Xue D, Han S, Li P. Targeting the CXCR7 pathway with TC14012 to inhibit endothelial necroptosis and lung cancer metastasis. Biochem Pharmacol 2025; 236:116852. [PMID: 40049294 DOI: 10.1016/j.bcp.2025.116852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 04/20/2025]
Abstract
Endothelial necroptosis plays a crucial role in regulating cancer metastasis. Our previous research demonstrated that TC14012, which is an agonist of CXCR7, exhibits protective effects against endothelial injury. This study was designed to elucidate the effects of TC14012 on endothelial necroptosis and cancer lung metastasis, along with deciphering the underlying molecular mechanisms. The trans-well analysis system was used to evaluate the trans-endothelial migration ability of the tumor cells. Cell death was evaluated with Ethidium Homodimer 3 (EthD-3) staining and flow cytometry analysis. The expression and phosphorylation of MLKL or RIPK3 were evaluated using Western blot. The effects of TC14012 on cancer lung metastasis in vivo were determined using the mouse hematogenous metastasis model. The results showed that TC14012 treatment significantly suppressed trans-endothelial migration of lung cancer cells, through effectively counteracting endothelial cell death induced by the tumor cells in vitro. Upon inhibition of cell necroptosis with necrosulfonamide (NSA), an MLKL inhibitor, the suppressive effects of TC14012 on endothelial cell death were significantly alleviated. Further investigations unveiled that TC14012, via its interaction with CXCR7 receptor rather than CXCR4, impeded the phosphorylation and subsequent activation of the RIPK3/MLKL signaling cascade. Ultimately, in vivo experiments demonstrated that administration of TC14012 mitigated lung infiltration of pre-labeled tumor cells and reduced lung metastasis in mice subsequent to intravenous injection of tumor cells. In summary, TC14012 effectively retards lung cancer metastasis by inhibiting endothelial necroptosis and the consequential trans-endothelial migration of tumor cells, through modulating the CXCR7/RIPK3/MLKL signaling.
Collapse
Affiliation(s)
- Huifeng Hao
- Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Wenjia Tian
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
| | - Jingjing Gong
- Institute of Systems Biomedicine, Peking University Health Science Center, Beijing 100191, China
| | - Junfeng Li
- Departments of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shiyan Zhou
- Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhengwang Guo
- Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yanna Jiao
- Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Dong Xue
- Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Shuyan Han
- Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Pingping Li
- Department of Integration of Chinese and Western Medicine, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China.
| |
Collapse
|
2
|
Cuesta-Margolles G, Schlecht-Louf G, Bachelerie F. ACKR3 in Skin Homeostasis, an Overlooked Player in the CXCR4/CXCL12 Axis. J Invest Dermatol 2025; 145:1039-1049. [PMID: 39466217 DOI: 10.1016/j.jid.2024.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 10/29/2024]
Abstract
CXCL12 and its receptor CXCR4 emerge as critical regulators within the intricate network of processes ensuring skin homeostasis. In this review, we discuss their spatial distribution and function in steady-state skin; delve into their role in acute wound healing, with emphasis on fibrotic and regenerative responses; and explore their relevance in skin responses to commensals and pathogens. Given the lack of knowledge surrounding ACKR3, the atypical receptor of CXCL12, we speculate whether and how it might be involved in the processes mentioned earlier. Is ACKR3 the (a)typical friend who enjoys missing the party, or do we need to take a closer look?
Collapse
Affiliation(s)
| | - Géraldine Schlecht-Louf
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Françoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| |
Collapse
|
3
|
Dong Z, He L, Wu J, Xie C, Geng S, Wu J, Zhong C, Li X. Bisphenol A-induced cancer-associated adipocytes promotes breast carcinogenesis via CXCL12/AKT signaling. Mol Cell Endocrinol 2025; 599:112473. [PMID: 39863150 DOI: 10.1016/j.mce.2025.112473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Bisphenol A (BPA), a commonly used plastic additive, is believed to cause obesity. As an environmental endocrine disruptor, BPA is closely associated with the onset and progression of BC. However, the molecular mechanisms underlying the promotion of breast cancer by BPA remain unclear. As obesity is a significant risk factor for breast cancer, this study aimed at exploring whether BPA facilitates the progression of breast cancer by inducing obesity. Using the National Health and Nutrition Examination Survey data, a positive correlation was observed between BPA exposure and the risk of sex-specific cancers among US adults with body mass index ≥30, suggesting that obesity may be influenced by urinary BPA. 3T3-L1 cells differentiated into mature adipocytes following treatment with 10-8 M BPA, and subsequent treatment with 4T1-conditioned medium acquired properties associated with cancer-associated adipocytes (CAAs). Network pharmacology suggested that CXCL12 may serve as a key target gene in breast cancer progression. Follow-up PCR analysis revealed high CXCL12 expression in BPA-induced CAAs. Overexpression of CXCL12 promoted epithelial-mesenchymal transition (EMT) and 4T1 cell migration by activating the AKT pathway. In vivo, BPA-induced CAAs accelerated tumor growth compared to a controls xenografted with only 4T1 cells. In tissues from the BPA-CAAs group, the expression of CXCL12, markers associated with CAAs, phosphorylated AKT, N-cadherin, and vimentin was markedly elevated, whereas the expression of E-cadherin was reduced. In conclusion, BPA may induce adipose cells to differentiate into CAA-like cells and subsequently advance breast cancer EMT through the CXCL12/AKT pathway.
Collapse
Affiliation(s)
- Zhiyuan Dong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Liping He
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jinyi Wu
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Shanshan Geng
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jieshu Wu
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Xiaoting Li
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
4
|
Liu A, Liu Y, Ye RD. Structural basis of CXCR4 assembly and regulation. Cell Rep 2025; 44:115255. [PMID: 39891908 DOI: 10.1016/j.celrep.2025.115255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/17/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025] Open
Abstract
CXC chemokine receptor 4 (CXCR4) is a well-established drug target and a key representative of the chemokine receptor family. Chemokine receptors tend to assemble, and this assembly plays a critical role in regulating their functions. However, structural information regarding the organization of these receptors remains limited. Here, we present the cryoelectron microscopy (cryo-EM) structure of a CXCR4 homo-tetramer. In this tetramer, each protomer interfaces with adjacent protomers via TM1/2 and TM5/6/7, aligning at a 90° angle to assemble into a C4 rotationally symmetric arrangement. Each protomer allosterically regulates the others, with Q272 in the ECL3 loop interacting with K38 (TM1) and V99 (TM2) of the adjacent protomer, resulting in a mutually inhibitory configuration. These findings reveal an allosteric and antagonistic mechanism that prevents excessive activation, providing a structural framework for understanding the molecular mechanisms driving CXCR4 self-assembly and offering insights that could inspire further therapeutic strategies.
Collapse
Affiliation(s)
- Aijun Liu
- Dongguan Songshan Lake Central Hospital, Dongguan Third People's Hospital, The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong 523326, China; Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China.
| | - Yezhou Liu
- Dongguan Songshan Lake Central Hospital, Dongguan Third People's Hospital, The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong 523326, China; Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Richard D Ye
- Dongguan Songshan Lake Central Hospital, Dongguan Third People's Hospital, The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong 523326, China; Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China; The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Guangdong 518000, China.
| |
Collapse
|
5
|
Pan KS, Wang Z, Pfeil C, Bergkamp ND, Mobach S, Roth S, Rizk A, Lohse MJ, Annibale P, Siderius M, Zimmermann M, Smit MJ, Bosma R. Pharmacological characterization of a clinical candidate, TG-0054, a small molecule inverse agonist targeting CXCR4. Mol Pharmacol 2025; 107:100015. [PMID: 40156952 DOI: 10.1016/j.molpha.2025.100015] [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: 07/08/2024] [Accepted: 01/08/2025] [Indexed: 04/01/2025] Open
Abstract
CXCR4 is an important therapeutic target for hematopoietic stem cell mobilization, which enhances the success of autologous stem cell transplantation for treating blood cancers such as lymphomas and myeloma. As CXCR4 has been shown to be involved in various inflammatory diseases, cancer progression, and cell entry by the human immunodeficiency virus, understanding the molecular mechanism of CXCR4 inhibitors has potential implications in a wide area of diseases. Here, we present an exploratory study which involves the molecular pharmacological characterization of TG-0054 (burixafor, GPC-100), a clinical candidate for hematopoietic stem cell mobilization. TG-0054 inhibited CXCL12 binding at CXCR4, and antagonized both Gαi and β-arrestin2 recruitment as well as the downstream Gαi-attenuation of cAMP signaling pathway, with pIC50 of 7.7, 8.0, and 7.9, respectively. Compared with the clinically used antagonist AMD3100 and the prototypical inverse agonist Isothiourea-1t (IT1t), TG-0054 displayed a unique pharmacological profile. Like IT1t, TG-0054 inhibited the constitutive Gαi signaling of CXCR4. However, in contrast to IT1t and other reported inverse agonists, TG-0054 was not able to induce monomerization of CXCR4 oligomeric complexes. Considering the unique properties of TG-0054 on CXCR4, TG-0054 is an interesting tool compound for studying the relevance of inverse agonism as well as CXCR4 monomerization in various pathologies. SIGNIFICANCE STATEMENT: CXCR4-targeted therapeutics hold important potential for the treatment of blood cancers. TG-0054 has inverse agonistic properties and is a non-CXCR4-monomerizing small molecule antagonist, unlike other well studied CXCR4 small molecule antagonists.
Collapse
Affiliation(s)
- Kylie S Pan
- InterAx Biotech AG, Villigen, Switzerland; Division of Medicinal Chemistry, Faculty of Sciences, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ziming Wang
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Cy Pfeil
- Division of Medicinal Chemistry, Faculty of Sciences, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Nick D Bergkamp
- Division of Medicinal Chemistry, Faculty of Sciences, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Simon Mobach
- Division of Medicinal Chemistry, Faculty of Sciences, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | | | - Martin J Lohse
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Paolo Annibale
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; School of Physics and Astronomy, University of St Andrews, St Andrews, United Kingdom
| | - Marco Siderius
- Division of Medicinal Chemistry, Faculty of Sciences, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Martine J Smit
- Division of Medicinal Chemistry, Faculty of Sciences, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Reggie Bosma
- Division of Medicinal Chemistry, Faculty of Sciences, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Eigenbrood J, Wong N, Mallory P, Pereira J, Morris-II DW, Beck JA, Cronk JC, Sayers CM, Mendez M, Kaiser L, Galindo J, Singh J, Cardamone A, Pore M, Kelly M, LeBlanc AK, Cotter J, Kaplan RN, McEachron TA. Spatial profiling identifies regionally distinct microenvironments and targetable immunosuppressive mechanisms in pediatric osteosarcoma pulmonary metastases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.631350. [PMID: 39896512 PMCID: PMC11785069 DOI: 10.1101/2025.01.22.631350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Osteosarcoma is the most common malignant bone tumor in young patients and remains a significant clinical challenge, particularly in the context of metastatic disease. Despite extensive documentation of genomic alterations in osteosarcoma, studies detailing the immunosuppressive mechanisms within the metastatic osteosarcoma microenvironment are lacking. Our objective was to characterize the spatial transcriptional landscape of metastatic osteosarcoma to reveal these immunosuppressive mechanisms and identify promising therapeutic targets. Here, we performed spatial transcriptional profiling on a cohort of osteosarcoma pulmonary metastases from pediatric patients. We reveal a conserved spatial gene expression pattern resembling a foreign body granuloma, characterized by peripheral inflammatory signaling, fibrocollagenous encapsulation, lymphocyte exclusion, and peritumoral macrophage accumulation. We also show that the intratumoral microenvironment of these lesions lack inflammatory signaling. Additionally, we identified CXCR4 as an actionable immunomodulatory target that bridges both the intratumoral and extratumoral microenvironments and highlights the spatial heterogeneity and complexity of this pathway. Collectively, this study reveals that metastatic osteosarcoma specimens are comprised of multiple regionally distinct immunosuppressive microenvironments.
Collapse
Affiliation(s)
- Jason Eigenbrood
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Current Address: University of Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK
- These authors contributed equally to this manuscript
| | - Nathan Wong
- Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- These authors contributed equally to this manuscript
| | - Paul Mallory
- Imaging Mass Cytometry Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Janice Pereira
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Douglass W Morris-II
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jessica A Beck
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - James C Cronk
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Carly M Sayers
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Monica Mendez
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Linus Kaiser
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Julie Galindo
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Jatinder Singh
- Center for Cancer Research Single Cell Analysis Facility, Cancer Research Technology Program, Frederick National Laboratory, Bethesda, MD, USA
| | - Ashley Cardamone
- Imaging Mass Cytometry Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Milind Pore
- Imaging Mass Cytometry Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Michael Kelly
- Center for Cancer Research Single Cell Analysis Facility, Cancer Research Technology Program, Frederick National Laboratory, Bethesda, MD, USA
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jennifer Cotter
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rosandra N Kaplan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Troy A McEachron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
7
|
Xia S, Chen L, Yu M, Li J, Chen J, Xu F, Ni M, Liu C, Wu X, Chen X, Li J. Genetic and therapeutic heterogeneity shape the baseline and longitudinal immune ecosystem of ovarian clear cell carcinoma. J Immunother Cancer 2024; 12:e010069. [PMID: 39608974 PMCID: PMC11603735 DOI: 10.1136/jitc-2024-010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/06/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Ovarian clear cell carcinoma (OCCC) is a rare and chemo-resistant subtype of ovarian cancer. While immunotherapy has demonstrated effectiveness in some OCCC cases, the mechanisms for heterogeneous immunoreactivity and potential combinatory strategies remain unclear. METHODS Tumor samples from 13 patients with OCCC underwent single-cell mRNA-seq and TCR-seq to generate 1 40 683 cells transcriptome, while additionally 31 formalin-fixed paraffin-embedded samples were used for immunohistochemistry. Spatial transcriptomics of two OCCC samples and bulk RNA-seq of 58 patients were incorporated for spatial and interpatient level explorations. Serum tumor markers and radiologic images of three patients with OCCC who received combinatory VEGF and PD-1 inhibition were retrospectively analyzed. RESULTS OCCC exhibited a dynamic immune architecture shaped by genetic and therapeutic pressure. ARID1A mutation linked to baseline immune activation, correlated with an enrichment of neoantigen-reactive CXCL13+ CTLA4+ CD8+ T cells (p<0.001) and enhanced FASLG-FAS interactions. Recurrent OCCC was fibrotic, angiogenic, and immunosuppressive, exhibiting metabolic reprogramming towards activated activity in fatty acid metabolism. High CD36 (log-rank p=0.012, HR: 4.515) and CD47 expression (log-rank p=0.037, HR: 3.246) indicated worse progression-free survival. Treatment with bevacizumab increased intratumoral T cell infiltration and activated T cell interferon-γ signaling. Retrospective analysis of clinical cases revealed that combination therapy with anti-VEGF (vascular endothelial growth factor) and anti-PD-1 agents exerted clinical benefits in patients with OCCC with persistent, recurrent, and metastatic disease. CONCLUSIONS ARID1A mutation correlated with OCCC baseline immune activation. Stromal reconstruction and tumor metabolic reprogramming functioned as key processes of OCCC dynamic progression. VEGF inhibition remodeled OCCC stroma, restored T cell function and potentiated immunotherapy. CD36 and CD47 might be potential therapeutic targets for recurrent OCCC.
Collapse
Affiliation(s)
- Siyu Xia
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lihua Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Min Yu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Jiana Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Jiaxin Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Fei Xu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Mengdong Ni
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Chaohua Liu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaojun Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Jiajia Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| |
Collapse
|
8
|
Song D, Wu Y, Li J, Liu J, Yi Z, Wang X, Sun J, Li L, Wu Q, Chen Y, Fang H, Luan T, Du H, Huang J, Peng W, Wei Y, Li F, Li Q, Zhang L, Zhu Y, Wan J, Ren G, Li H. Insulin-like growth factor 2 drives fibroblast-mediated tumor immunoevasion and confers resistance to immunotherapy. J Clin Invest 2024; 134:e183366. [PMID: 39545420 PMCID: PMC11563680 DOI: 10.1172/jci183366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/20/2024] [Indexed: 11/17/2024] Open
Abstract
T cell exclusion is crucial in enabling tumor immune evasion and immunotherapy resistance. However, the key genes driving this process remain unclear. We uncovered a notable increase of insulin-like growth factor 2 (IGF2) in immune-excluded tumors, predominantly secreted by cancer-associated fibroblasts (CAFs). Using mice with systemic or fibroblast-specific deletion of IGF2, we demonstrated that IGF2 deficiency enhanced the infiltration and cytotoxic activity of CD8+ T cells, leading to a reduction in tumor burden. Integration of spatial and single-cell transcriptomics revealed that IGF2 promoted interaction between CAFs and T cells via CXCL12 and programmed death ligand 1 (PD-L1). Mechanistically, autocrine IGF2 activated PI3K/AKT signaling by binding to the IGF1 receptor (IGF1R) on CAFs, which was required for the immunosuppressive functions of CAFs. Furthermore, genetic ablation of IGF2 or targeted inhibition of the IGF2/IGF1R axis with the inhibitor linsitinib markedly boosted the response to immune checkpoint blockade. Clinically, elevated levels of IGF2 in tumors or plasma correlated with an adverse prognosis and reduced efficacy of anti-programmed death 1 treatment. Together, these results highlight the pivotal role of IGF2 in promoting CAF-mediated immunoevasion, indicating its potential as a biomarker and therapeutic target in immunotherapy.
Collapse
Affiliation(s)
- Daqiang Song
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yushen Wu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Oncology
| | - Jie Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiazhou Liu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast and Thyroid Surgery, and
| | - Ziying Yi
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyu Wang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiazheng Sun
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liuying Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qianxue Wu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuru Chen
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huiying Fang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tiankuo Luan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | - Jing Huang
- Department of Respiratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyan Peng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxian Wei
- Department of Breast and Thyroid Surgery, and
| | - Fan Li
- Department of Breast and Thyroid Surgery, and
| | - Qin Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Li Zhang
- Department of Pathophysiology and
| | - Yong Zhu
- Research Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jingyuan Wan
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast and Thyroid Surgery, and
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Perez Almeria CV, Otun O, Schlimgen R, Lamme TD, Crudden C, Youssef N, Musli L, Jenjak S, Bobkov V, Drube J, Hoffmann C, Volkman BF, Granier S, Bechara C, Siderius M, Heukers R, Schafer CT, Smit MJ. Constitutive activity of an atypical chemokine receptor revealed by inverse agonistic nanobodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621790. [PMID: 39574661 PMCID: PMC11580867 DOI: 10.1101/2024.11.04.621790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Chemokine stimulation of atypical chemokine receptor 3 (ACKR3) does not activate G proteins but recruits arrestins. It is a chemokine scavenger that indirectly influences responses by restricting the availability of CXCL12, an agonist shared with the canonical receptor CXCR4. ACKR3 is upregulated in numerous disorders. Due to limited insights in chemokine-activated ACKR3 signaling, it is unclear how ACKR3 contributes to pathological phenotypes. One explanation may be that high constitutive activity of ACKR3 drives non-canonical signaling through a basal receptor state. Here we characterize the constitutive action of ACKR3 using novel inverse agonistic nanobodies to suppress basal activity. These new tools promote an inactive receptor conformation which decreased arrestin engagement and inhibited constitutive internalization. Basal, non-chemotactic, breast cancer cell motility was also suppressed, suggesting a role for ACKR3 in this process. The basal receptor activity in pathophysiology may provide a new therapeutic approach for targeting ACKR3.
Collapse
Affiliation(s)
- Claudia V Perez Almeria
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Department of Chemistry & Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands
| | - Omolade Otun
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Roman Schlimgen
- Department of Biochemistry, Medical College of Wisconsin, USA
| | - Thomas D Lamme
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Department of Chemistry & Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands
| | - Caitrin Crudden
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Department of Chemistry & Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands
| | - Noureldine Youssef
- Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Lejla Musli
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Department of Chemistry & Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands
| | - Shawn Jenjak
- Department of Biochemistry, Medical College of Wisconsin, USA
| | - Vladimir Bobkov
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Department of Chemistry & Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands
| | - Julia Drube
- Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Carsten Hoffmann
- Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, USA
| | - Sébastien Granier
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Cherine Bechara
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
- Institut Universitaire de France, Paris, France
| | - Marco Siderius
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Department of Chemistry & Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands
| | - Raimond Heukers
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Department of Chemistry & Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands
- QVQ Holding BV, Utrecht, the Netherlands
| | - Christopher T Schafer
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Department of Chemistry & Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands
| | - Martine J Smit
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Department of Chemistry & Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit, Amsterdam, the Netherlands
| |
Collapse
|
10
|
Mu Y, Zhang Z, Zhou H, Jin M, Ma L, Liu B, Ma C, Hu X, Zhang Y, Wang DA. A biomimetic targeted nanosystem delivering synergistic inhibitors for glioblastoma immune microenvironment reprogramming and treatment. Mater Today Bio 2024; 28:101222. [PMID: 39296357 PMCID: PMC11407969 DOI: 10.1016/j.mtbio.2024.101222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/21/2024] Open
Abstract
Efficient drug delivery across the blood-brain barrier is imperative for treating glioblastoma (GBM). This study utilized the GBM cell membrane to construct a biomimetic targeted nanosystem (GMNPs@AMD/RAPA) that hierarchically releases the CXCR4 antagonist AMD3100 and the mTOR pathway inhibitor rapamycin (RAPA) for reprogramming the tumor immune microenvironment and suppressing the progression of GBM. By initially inhibiting the CXCL12/CXCR4 axis, the tumor microenvironment (TME) was reprogrammed to enhance the infiltration of cytotoxic T lymphocytes (CTLs) into the TME while suppressing tumor cell survival, proliferation, and angiogenesis. Subsequently, through further cellular uptake and degradation of the nanoparticles, the mTOR pathway inhibitor RAPA was released, further suppressing the tumor progression. This study successfully combined chemotherapy and immunotherapy, achieving effective synergistic therapeutic effects, and suppressing the progression of GBM.
Collapse
Affiliation(s)
- Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Min Jin
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Bangheng Liu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Cheng Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Xu Hu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Yi Zhang
- School of Integrated Circuit Science and Engineering, University of Electronic Science and Engineering of China, Chengdu, Sichuan, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| |
Collapse
|
11
|
Isci D, Kuppens A, Scalisi J, Cokaiko J, D'Uonnolo G, Wantz M, Szpakowska M, Chevigné A, Rogister B, Neirinckx V. Heterogeneous expression of the atypical chemokine receptor ACKR3 in glioblastoma patient-derived tissue samples and cell cultures. Sci Rep 2024; 14:21925. [PMID: 39300240 PMCID: PMC11412975 DOI: 10.1038/s41598-024-73064-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive glial tumor of the adult brain, associated with invariably fatal outcome, and a deeper understanding of the underlying malignant mechanisms is necessary to address the current therapeutic failure. We previously demonstrated the role of the CXCL12/CXCR4 axis in GBM cell migration and resistance to ionizing radiation. The atypical chemokine receptor ACKR3, responsible for CXCL12 scavenging, was previously suggested as additional important player in the context of GBM. Following validation of the detection tools, we observed that ACKR3 is expressed within GBM patient tumor tissue, distributed in diverse cell types. In contrast to CXCR4, ACKR3 expression in patient-derived stem-like cells (GSCs) remains however low, while ACKR3 gene expression by tumor cells appears to be modulated by the in-vivo environment. Using overexpression models, we also showed that in vitro ACKR3 had no significant direct effect on cell proliferation or invasion. Altogether, these results suggest that in vitro ACKR3 plays a minor role in malignant GBM cell biology and that its expression is possibly regulated by in-vivo influences. The subtle and multifaceted functions ACKR3 could exert in GBM should therefore only be tackled within a comprehensive tumor microenvironment considering tumoral but also non-tumoral cells.
Collapse
Affiliation(s)
- Damla Isci
- Laboratory of Nervous System Diseases and Therapy, GIGA Neuroscience, GIGA Institute, University of Liège, Liège, Belgium
| | - Amandine Kuppens
- Laboratory of Nervous System Diseases and Therapy, GIGA Neuroscience, GIGA Institute, University of Liège, Liège, Belgium
| | - Joshua Scalisi
- Laboratory of Nervous System Diseases and Therapy, GIGA Neuroscience, GIGA Institute, University of Liège, Liège, Belgium
| | - Julie Cokaiko
- Laboratory of Nervous System Diseases and Therapy, GIGA Neuroscience, GIGA Institute, University of Liège, Liège, Belgium
| | - Giulia D'Uonnolo
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Strassen, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - May Wantz
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Martyna Szpakowska
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Andy Chevigné
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Bernard Rogister
- Laboratory of Nervous System Diseases and Therapy, GIGA Neuroscience, GIGA Institute, University of Liège, Liège, Belgium
- Neurology Department, University Hospital, University of Liège, Liège, Belgium
| | - Virginie Neirinckx
- Laboratory of Nervous System Diseases and Therapy, GIGA Neuroscience, GIGA Institute, University of Liège, Liège, Belgium.
| |
Collapse
|
12
|
Yang J, Tian E, Chen L, Liu Z, Ren Y, Mao W, Zhang Y, Zhang J. Development and therapeutic perspectives of CXCR4 antagonists for disease therapy. Eur J Med Chem 2024; 275:116594. [PMID: 38879970 DOI: 10.1016/j.ejmech.2024.116594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Chemokine receptor 4 (CXCR4) is a subtype receptor protein of the GPCR family with a seven-transmembrane structure widely distributed in human tissues. CXCR4 is involved in diseases (e.g., HIV-1 infection), cancer proliferation and metastasis, inflammation signaling pathways, and leukemia, making it a promising drug target. Clinical trials on CXCR4 antagonists mainly focused on peptides and antibodies, with a few small molecule compounds, such as AMD11070 (2) and MSX-122 (3), showing promise in cancer treatment. This perspective discusses the structure-activity relationship (SAR) of CXCR4 and its role in diseases, mainly focusing on the SAR of CXCR4 antagonists. It also explores the standard structural features and target interactions of CXCR4 binding in different disease categories. Furthermore, it investigates various modification strategies to propose potential improvements in the effectiveness of CXCR4 drugs.
Collapse
Affiliation(s)
- Jun Yang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Erkang Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Li Chen
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zihang Liu
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Wuyu Mao
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yiwen Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
13
|
Jiang H, Xia W, Xia T, Jiang L, Yu J, Zhu X, Lin C, Lou C, Wang W, Chai Y, Wan R, Wang J, Xue X, Pan X. Chemotactic recruitment of genetically engineered cell membrane-camouflaged metal-organic framework nanoparticles for ischemic osteonecrosis treatment. Acta Biomater 2024; 185:410-428. [PMID: 39029641 DOI: 10.1016/j.actbio.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/26/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024]
Abstract
Ischemic osteonecrosis, particularly glucocorticoid-induced osteonecrosis of the femoral head (GIONFH), is primarily due to the dysfunction of osteogenesis and angiogenesis. miRNA, as a therapeutic system with immense potential, plays a vital role in the treatment of various diseases. However, due to the unique microenvironmental structure of bone tissue, especially in the case of GIONFH, where there is a deficiency in the vascular system, it is challenging to effectively target and deliver to the ischemic osteonecrosis area. A drug delivery system assisted by genetically engineered cell membranes holds promise in addressing the challenge of targeted miRNA delivery. Herein, we leverage the potential of miR-21 in modulating osteogenesis and angiogenesis to design an innovative biomimetic nanoplatform system. First, we employed metal-organic frameworks (MOFs) as the core structure to load miR-21-m (miR-21-m@MOF). The nanoparticles were further coated with the membrane of bone marrow mesenchymal stem cells overexpressing CXCR4 (CM-miR-21-m@MOF), enhancing their ability to target ischemic bone areas via the CXCR4-SDF1 axis. These biomimetic nanocomposites possess both bone-targeting and ischemia-guiding capabilities, actively targeting GIONFH lesions to release miR-21-m into target cells, thereby silencing PTEN gene and activating the PI3K-AKT signaling pathway to regulate osteogenesis and angiogenesis. This innovative miRNA delivery system provides a promising therapeutic avenue for GIONFH and potentially other related ischemic bone diseases. STATEMENT OF SIGNIFICANCE.
Collapse
Affiliation(s)
- Hongyi Jiang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Weijie Xia
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tian Xia
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, People's Republic of China
| | - Liting Jiang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiachen Yu
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xinyi Zhu
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Chihao Lin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Chao Lou
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Weidan Wang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yingqian Chai
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, People's Republic of China
| | - Renwen Wan
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jilong Wang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, People's Republic of China.
| | - Xinghe Xue
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Xiaoyun Pan
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second Clinical School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
14
|
Anbuhl SM, Dervillez X, Neubacher S, Schriek AI, Bobkov V, de Taeye SW, Szpakowska M, Siderius M, Grossmann TN, Chevigné A, Smit MJ, Heukers R. Multivalent CXCR4-targeting nanobody formats differently affect affinity, receptor clustering, and antagonism. Biochem Pharmacol 2024; 227:116457. [PMID: 39098732 DOI: 10.1016/j.bcp.2024.116457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/12/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
The chemokine receptor CXCR4 is involved in the development and migration of stem and immune cells but is also implicated in tumor progression and metastasis for a variety of cancers. Antagonizing ligand (CXCL12)-induced CXCR4 signaling is, therefore, of therapeutic interest. Currently, there are two small-molecule CXCR4 antagonists on the market for the mobilization of hematopoietic stem cells. Other molecules with improved potencies and safety profiles are being developed for different indications, including cancer. Moreover, multiple antagonistic nanobodies targeting CXCR4 displayed similar or better potencies as compared to the CXCR4-targeting molecule AMD3100 (Plerixafor), which was further enhanced through avid binding of bivalent derivatives. In this study, we aimed to compare the affinities of various multivalent nanobody formats which might be differently impacted by avidity. By fusion to a flexible GS-linker, Fc-region of human IgG1, different C4bp/CLR multimerization domains, or via site-directed conjugation to a trivalent linker scaffold, we generated different types of multivalent nanobodies with varying valencies ranging from bivalent to decavalent. Of these, C-terminal fusion, especially to human Fc, was most advantageous with a 2-log-fold and 3-log-fold increased potency in inhibiting CXCL12-mediated Gαi- or β-arrestin recruitment, respectively. Overall, we describe strategies for generating multivalent and high-potency CXCR4 antagonistic nanobodies able to induce receptor clustering and conclude that fusion to an Fc-tail results in the highest avidity effect irrespective of the hinge linker.
Collapse
Affiliation(s)
- Stephanie M Anbuhl
- QVQ Holding BV, 3584 CL Utrecht, The Netherlands; Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HV, The Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), 1081 HV, Amsterdam, The Netherlands
| | - Xavier Dervillez
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Saskia Neubacher
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), 1081 HV, Amsterdam, The Netherlands; Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, The Netherlands; Incircular BV, 1081 HZ Amsterdam, The Netherlands
| | - Angela I Schriek
- Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam UMC, Location University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Infectious diseases, Amsterdam, The Netherlands
| | - Vladimir Bobkov
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HV, The Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), 1081 HV, Amsterdam, The Netherlands; Argenx, 9052 Ghent, Belgium
| | - Steven W de Taeye
- Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Amsterdam UMC, Location University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Infectious diseases, Amsterdam, The Netherlands
| | - Martyna Szpakowska
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Marco Siderius
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HV, The Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), 1081 HV, Amsterdam, The Netherlands
| | - Tom N Grossmann
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), 1081 HV, Amsterdam, The Netherlands; Department of Chemistry & Pharmaceutical Sciences, Vrije Universiteit Amsterdam, The Netherlands; Incircular BV, 1081 HZ Amsterdam, The Netherlands
| | - Andy Chevigné
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Martine J Smit
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HV, The Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), 1081 HV, Amsterdam, The Netherlands
| | - Raimond Heukers
- QVQ Holding BV, 3584 CL Utrecht, The Netherlands; Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HV, The Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
15
|
Liu Y, Liu A, Li X, Liao Q, Zhang W, Zhu L, Ye RD. Cryo-EM structure of monomeric CXCL12-bound CXCR4 in the active state. Cell Rep 2024; 43:114578. [PMID: 39093700 DOI: 10.1016/j.celrep.2024.114578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/17/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024] Open
Abstract
CXCR4 binding of its endogenous agonist CXCL12 leads to diverse functions, including bone marrow retention of hematopoietic progenitors and cancer metastasis. However, the structure of the CXCL12-bound CXCR4 remains unresolved despite available structures of CXCR4 in complex with antagonists. Here, we present the cryoelectron microscopy (cryo-EM) structure of the CXCL12-CXCR4-Gi complex at an overall resolution of 2.65 Å. CXCL12 forms a 1:1 stoichiometry complex with CXCR4, following the two-site model. The first 8 amino acids of mature CXCL12 are crucial for CXCR4 activation by forming polar interactions with minor sub-pocket residues in the transmembrane binding pocket. The 3.2-Å distance between V3 of CXCL12 and the "toggle switch" W6.48 marks the deepest insertion among all chemokine-receptor pairs, leading to conformational changes of CXCR4 for G protein activation. These results, combined with functional assays and computational analysis, provide the structural basis for CXCR4 activation by CXCL12.
Collapse
Affiliation(s)
- Yezhou Liu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Aijun Liu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China; Dongguan Songshan Lake Central Hospital, Dongguan Third People's Hospital, The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong 523326, China
| | - Xinyu Li
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Qiwen Liao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Weijia Zhang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Lizhe Zhu
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China.
| | - Richard D Ye
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China; The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Guangdong 518048, China.
| |
Collapse
|
16
|
Mu Y, Zhang Z, Zhou H, Ma L, Wang DA. Applications of nanotechnology in remodeling the tumour microenvironment for glioblastoma treatment. Biomater Sci 2024; 12:4045-4064. [PMID: 38993162 DOI: 10.1039/d4bm00665h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
With the increasing research and deepening understanding of the glioblastoma (GBM) tumour microenvironment (TME), novel and more effective therapeutic strategies have been proposed. The GBM TME involves intricate interactions between tumour and non-tumour cells, promoting tumour progression. Key therapeutic goals for GBM treatment include improving the immunosuppressive microenvironment, enhancing the cytotoxicity of immune cells against tumours, and inhibiting tumour growth and proliferation. Consequently, remodeling the GBM TME using nanotechnology has emerged as a promising approach. Nanoparticle-based drug delivery enables targeted delivery, thereby improving treatment specificity, facilitating combination therapies, and optimizing drug metabolism. This review provides an overview of the GBM TME and discusses the methods of remodeling the GBM TME using nanotechnology. Specifically, it explores the application of nanotechnology in ameliorating immune cell immunosuppression, inducing immunogenic cell death, stimulating, and recruiting immune cells, regulating tumour metabolism, and modulating the crosstalk between tumours and other cells.
Collapse
Affiliation(s)
- Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
| | - Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
- Centre for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, Hong Kong SAR 999077, China
| |
Collapse
|
17
|
Liu J, Zhao W, Luan Y, Tian Z. FAM189A2 plays a tumour suppressor role in lung adenocarcinoma by influencing cell apoptosis, CXCR4 expression and tight junction proteins. Tissue Cell 2024; 89:102441. [PMID: 38878656 DOI: 10.1016/j.tice.2024.102441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 07/28/2024]
Abstract
Transmembrane proteins play key roles in the development of lung cancer. The family with sequence similarity 189 member A2 (FAM189A2) gene encodes a transmembrane structural protein, yet its involvement in lung adenocarcinoma remains largely unexplored. This study elucidated its role in lung adenocarcinoma and its possible molecular mechanism. Our findings revealed diminished expression levels of FAM189A2 in LUAD tissues. Additionally, the activity of LUAD cells was significantly inhibited by overexpression of FAM189A2. Following FAM189A2 overexpression, the expression of OCLN and TJP2 was upregulated in LUAD cells, while CXCR4 expression experiences a notable decrease. Moreover, the coimmunoprecipitation experiment confirmed the direct interaction between FAM189A2 and CXCR4. The infiltration levels of T cells (CD4+ memory resting, CD8+, regulatory), NK cells, B memory cells, endothelial cells and cancer-associated fibroblasts were significantly correlated with FAM189A2 expression. These results indicate FAM189A2 may act as a tumour suppressor in LUAD through tight junction protein (TJP) and CXCR4 regulation. Moreover, FAM189A2 is significantly correlated with the immune microenvironment of LUAD, which may be involved in prognosis and immunotherapeutic efficacy.
Collapse
Affiliation(s)
- Jiakun Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China; Department of Thoracic Surgery, Hebei Chest Hospital, Shijiazhuang, PR China
| | - Wei Zhao
- Department of Prevention and Health Care, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Yanchao Luan
- Department of Thoracic Surgery, Hebei Chest Hospital, Shijiazhuang, PR China
| | - Ziqiang Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China.
| |
Collapse
|
18
|
Hu Y, Xu Z, Zhou D, Hou H, Liu B, Long H, Hu W, Tang Y, Wang J, Wei D, Zhao Q. CXCR4 promotes migration, invasion, and epithelial-mesenchymal transition of papillary thyroid carcinoma by activating STAT3 signaling pathway. J Cancer Res Ther 2024; 20:1241-1250. [PMID: 39206986 DOI: 10.4103/jcrt.jcrt_2395_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/02/2024] [Indexed: 09/04/2024]
Abstract
AIMS Papillary thyroid cancer (PTC) is a serious threat to human health worldwide, while metastasis in the early phase limits therapeutic success and leads to poor survival outcomes. The CXC chemokine receptor type 4 (CXCR4) plays an important role in many cellular movements such as transcriptional modulation, cell skeleton rearrangement, and cell migration, and the change in CXCR4 levels are crucial in various diseases including cancer. In this study, we explored the role of CXCR4 in the migration and invasion of PTC and investigated the potential mechanisms underlying its effects. SUBJECTS AND METHODS We analyzed the expression levels of CXCR4 in PTC tissues and cell lines. Would healing migration, Transwell invasion assay in vitro, and tail-vein lung metastasis assay In vivo were performed to evaluated the migration and invasion abilities of PTC cells with stable CXCR4 knockdown or overexpression. Signal transducers and activators of transcription (STAT3) signaling pathway-related protein expressions were examined by Western blotting assays. RESULTS The results showed that CXCR4 was highly expressed in PTC cell lines and PTC tissues. CXCR4 knockdown in PTC cells dampened the migration, invasion, and epithelial-mesenchymal transition (EMT), whereas CXCR4 overexpression enhanced these properties. In vivo, we also found that CXCR4 promoted the metastasis of PTC. Mechanistic studies showed that CXCR4 played these vital roles through the STAT3 signaling pathway. Furthermore, PTC patients with high CXCR4 or p-STAT3 expression correlated with aggressive clinical characteristics such as extrathyroidal extension (ETE), and lymph node metastasis (LNM). CONCLUSIONS We provided evidence that CXCR4 might activate the STAT3 signaling pathway and further promote PTC development. Thus, CXCR4 might be a novel therapeutic target for PTC.
Collapse
Affiliation(s)
- Yajie Hu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhipeng Xu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Dongsheng Zhou
- Department of Thyroid Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Haitao Hou
- Department of Breast and Thyroid Surgery, Tengzhou Central People's Hospital, Zaozhuang, Shandong, China
| | - Bin Liu
- Department of Breast and Thyroid Surgery, Tengzhou Central People's Hospital, Zaozhuang, Shandong, China
| | - Houlong Long
- Department of Breast and Thyroid Surgery, Tengzhou Central People's Hospital, Zaozhuang, Shandong, China
| | - Wenxin Hu
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Yuanqi Tang
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Jianning Wang
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Dan Wei
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Quanlin Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
19
|
Otun O, Aljamous C, Del Nero E, Arimont-Segura M, Bosma R, Zarzycka B, Girbau T, Leyrat C, de Graaf C, Leurs R, Durroux T, Granier S, Cong X, Bechara C. Conformational dynamics underlying atypical chemokine receptor 3 activation. Proc Natl Acad Sci U S A 2024; 121:e2404000121. [PMID: 39008676 PMCID: PMC11287255 DOI: 10.1073/pnas.2404000121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/28/2024] [Indexed: 07/17/2024] Open
Abstract
Atypical Chemokine Receptor 3 (ACKR3) belongs to the G protein-coupled receptor family but it does not signal through G proteins. The structural properties that govern the functional selectivity and the conformational dynamics of ACKR3 activation are poorly understood. Here, we combined hydrogen/deuterium exchange mass spectrometry, site-directed mutagenesis, and molecular dynamics simulations to examine the binding mode and mechanism of action of ACKR3 ligands of different efficacies. Our results show that activation or inhibition of ACKR3 is governed by intracellular conformational changes of helix 6, intracellular loop 2, and helix 7, while the DRY motif becomes protected during both processes. Moreover, we identified the binding sites and the allosteric modulation of ACKR3 upon β-arrestin 1 binding. In summary, this study highlights the structure-function relationship of small ligands, the binding mode of β-arrestin 1, the activation dynamics, and the atypical dynamic features in ACKR3 that may contribute to its inability to activate G proteins.
Collapse
Affiliation(s)
- Omolade Otun
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Christelle Aljamous
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Elise Del Nero
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Marta Arimont-Segura
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Reggie Bosma
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Barbara Zarzycka
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Tristan Girbau
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Cédric Leyrat
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Chris de Graaf
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Rob Leurs
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Thierry Durroux
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Sébastien Granier
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Xiaojing Cong
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Cherine Bechara
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
- Institut Universitaire de France, Paris75005, France
| |
Collapse
|
20
|
Roy S, Sinha S, Silas AJ, Ghassemian M, Kufareva I, Ghosh P. Growth factor-dependent phosphorylation of Gα i shapes canonical signaling by G protein-coupled receptors. Sci Signal 2024; 17:eade8041. [PMID: 38833528 PMCID: PMC11328959 DOI: 10.1126/scisignal.ade8041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 05/17/2024] [Indexed: 06/06/2024]
Abstract
A long-standing question in the field of signal transduction is how distinct signaling pathways interact with each other to control cell behavior. Growth factor receptors and G protein-coupled receptors (GPCRs) are the two major signaling hubs in eukaryotes. Given that the mechanisms by which they signal independently have been extensively characterized, we investigated how they may cross-talk with each other. Using linear ion trap mass spectrometry and cell-based biophysical, biochemical, and phenotypic assays, we found at least three distinct ways in which epidermal growth factor affected canonical G protein signaling by the Gi-coupled GPCR CXCR4 through the phosphorylation of Gαi. Phosphomimicking mutations in two residues in the αE helix of Gαi (tyrosine-154/tyrosine-155) suppressed agonist-induced Gαi activation while promoting constitutive Gβγ signaling. Phosphomimicking mutations in the P loop (serine-44, serine-47, and threonine-48) suppressed Gi activation entirely, thus completely segregating growth factor and GPCR pathways. As expected, most of the phosphorylation events appeared to affect intrinsic properties of Gαi proteins, including conformational stability, nucleotide binding, and the ability to associate with and to release Gβγ. However, one phosphomimicking mutation, targeting the carboxyl-terminal residue tyrosine-320, promoted mislocalization of Gαi from the plasma membrane, a previously uncharacterized mechanism of suppressing GPCR signaling through G protein subcellular compartmentalization. Together, these findings elucidate not only how growth factor and chemokine signals cross-talk through the phosphorylation-dependent modulation of Gαi but also how such cross-talk may generate signal diversity.
Collapse
Affiliation(s)
- Suchismita Roy
- Department of Cellular and Molecular Medicine, University of California San Diego, CA 92093, USA
| | - Saptarshi Sinha
- Department of Cellular and Molecular Medicine, University of California San Diego, CA 92093, USA
| | - Ananta James Silas
- Department of Cellular and Molecular Medicine, University of California San Diego, CA 92093, USA
| | - Majid Ghassemian
- Department of Chemistry and Biochemistry, Biomolecular and Proteomics Mass Spectrometry Facility, University of California San Diego, San Diego, CA 92093, USA
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, CA 92093, USA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, CA 92093, USA
- Department of Medicine, University of California San Diego, CA 92093, USA
- Moore’s Comprehensive Cancer Center, University of California San Diego, CA 92093, USA
| |
Collapse
|
21
|
Jia F, Sun S, Li J, Wang W, Huang H, Hu X, Pan S, Chen W, Shen L, Yao Y, Zheng S, Chen H, Xia W, Yuan H, Zhou J, Yu X, Zhang T, Zhang B, Huang J, Ni C. Neoadjuvant chemotherapy-induced remodeling of human hormonal receptor-positive breast cancer revealed by single-cell RNA sequencing. Cancer Lett 2024; 585:216656. [PMID: 38266804 DOI: 10.1016/j.canlet.2024.216656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 01/26/2024]
Abstract
Hormone receptor-positive breast cancer (HR+ BC) is known to be relatively insensitive to chemotherapy, and since chemotherapy has remained the major neoadjuvant therapy for HR+ BC, the undetermined mechanism of chemoresistance and how chemotherapy reshapes the immune microenvironment need to be explored by high-throughput technology. By using single-cell RNA sequencing and multiplexed immunofluorescence staining analysis of HR+ BC samples (paired pre- and post-neoadjuvant chemotherapy (NAC)), the levels of previously unrecognized immune cell subsets, including CD8+ T cells with pronounced expression of T-cell development (LMNA) and cytotoxicity (FGFBP2) markers, CD4+ T cells characterized by proliferation marker (ATP1B3) expression and macrophages characterized by CD52 expression, were found to be increased post-NAC, which were predictive of chemosensitivity and their antitumor function was also validated with in vitro experiments. In terms of immune checkpoint expression of CD8+ T cells, we found their changes were inconsistent post-NAC, that LAG3, VSIR were decreased, and PDCD1, HAVCR2, CTLA4, KLRC1 and BTLA were increased. In addition, we have identified novel genomic and transcriptional patterns of chemoresistant cancer cells, both innate and acquired, and have confirmed their prognostic value with TCGA cohorts. By shedding light on the ecosystem of HR+ BC reshaped by chemotherapy, our results uncover valuable candidates for predicting chemosensitivity and overcoming chemoresistance in HR+ BC.
Collapse
Affiliation(s)
- Fang Jia
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Shanshan Sun
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China
| | - Jiaxin Li
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Wenwen Wang
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huanhuan Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Xiaoxiao Hu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng Pan
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wuzhen Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lesang Shen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Yao
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siwei Zheng
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hailong Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjie Xia
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Hongjun Yuan
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jun Zhou
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuyan Yu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Zhang
- Department of Radiotherapy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing Zhang
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Jian Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| | - Chao Ni
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China.
| |
Collapse
|
22
|
Liu Z, Zhang Z, Zhang Y, Zhou W, Zhang X, Peng C, Ji T, Zou X, Zhang Z, Ren Z. Spatial transcriptomics reveals that metabolic characteristics define the tumor immunosuppression microenvironment via iCAF transformation in oral squamous cell carcinoma. Int J Oral Sci 2024; 16:9. [PMID: 38287007 PMCID: PMC10824761 DOI: 10.1038/s41368-023-00267-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 01/31/2024] Open
Abstract
Tumor progression is closely related to tumor tissue metabolism and reshaping of the microenvironment. Oral squamous cell carcinoma (OSCC), a representative hypoxic tumor, has a heterogeneous internal metabolic environment. To clarify the relationship between different metabolic regions and the tumor immune microenvironment (TME) in OSCC, Single cell (SC) and spatial transcriptomics (ST) sequencing of OSCC tissues were performed. The proportion of TME in the ST data was obtained through SPOTlight deconvolution using SC and GSE103322 data. The metabolic activity of each spot was calculated using scMetabolism, and k-means clustering was used to classify all spots into hyper-, normal-, or hypometabolic regions. CD4T cell infiltration and TGF-β expression is higher in the hypermetabolic regions than in the others. Through CellPhoneDB and NicheNet cell-cell communication analysis, it was found that in the hypermetabolic region, fibroblasts can utilize the lactate produced by glycolysis of epithelial cells to transform into inflammatory cancer-associated fibroblasts (iCAFs), and the increased expression of HIF1A in iCAFs promotes the transcriptional expression of CXCL12. The secretion of CXCL12 recruits regulatory T cells (Tregs), leading to Treg infiltration and increased TGF-β secretion in the microenvironment and promotes the formation of a tumor immunosuppressive microenvironment. This study delineates the coordinate work axis of epithelial cells-iCAFs-Tregs in OSCC using SC, ST and TCGA bulk data, and highlights potential targets for therapy.
Collapse
Affiliation(s)
- Zheqi Liu
- Department of Oral and Maxillofacial - Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Oral and Maxillofacial Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhen Zhang
- Department of Oral and Maxillofacial - Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yu Zhang
- Department of Oral and Maxillofacial - Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Wenkai Zhou
- Department of Oral and Maxillofacial - Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xu Zhang
- Department of Oral and Maxillofacial - Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Canbang Peng
- School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Tong Ji
- Department of Oral and Maxillofacial Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Zou
- Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Pathology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial - Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.
- Hainan Western Central Hospital, Academician Zhang Zhiyuan Team Innovation Center, Danzhou, China.
| | - Zhenhu Ren
- Department of Oral and Maxillofacial - Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.
- Hainan Western Central Hospital, Academician Zhang Zhiyuan Team Innovation Center, Danzhou, China.
| |
Collapse
|
23
|
Fang F, Yang J, Wang J, Li T, Wang E, Zhang D, Liu X, Zhou C. The role and applications of extracellular vesicles in osteoporosis. Bone Res 2024; 12:4. [PMID: 38263267 PMCID: PMC10806231 DOI: 10.1038/s41413-023-00313-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/13/2023] [Accepted: 11/28/2023] [Indexed: 01/25/2024] Open
Abstract
Osteoporosis is a widely observed condition characterized by the systemic deterioration of bone mass and microarchitecture, which increases patient susceptibility to fragile fractures. The intricate mechanisms governing bone homeostasis are substantially impacted by extracellular vesicles (EVs), which play crucial roles in both pathological and physiological contexts. EVs derived from various sources exert distinct effects on osteoporosis. Specifically, EVs released by osteoblasts, endothelial cells, myocytes, and mesenchymal stem cells contribute to bone formation due to their unique cargo of proteins, miRNAs, and cytokines. Conversely, EVs secreted by osteoclasts and immune cells promote bone resorption and inhibit bone formation. Furthermore, the use of EVs as therapeutic modalities or biomaterials for diagnosing and managing osteoporosis is promising. Here, we review the current understanding of the impact of EVs on bone homeostasis, including the classification and biogenesis of EVs and the intricate regulatory mechanisms of EVs in osteoporosis. Furthermore, we present an overview of the latest research progress on diagnosing and treating osteoporosis by using EVs. Finally, we discuss the challenges and prospects of translational research on the use of EVs in osteoporosis.
Collapse
Affiliation(s)
- Fei Fang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jie Yang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jiahe Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Tiantian Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Erxiang Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Demao Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China.
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
24
|
Abstract
For our immune system to contain or eliminate malignant solid tumours, both myeloid and lymphoid haematopoietic cells must not only extravasate from the bloodstream into the tumour tissue but also further migrate to various specialized niches of the tumour microenvironment to functionally interact with each other, with non-haematopoietic stromal cells and, ultimately, with cancer cells. These interactions regulate local immune cell survival, proliferative expansion, differentiation and their execution of pro-tumour or antitumour effector functions, which collectively determine the outcome of spontaneous or therapeutically induced antitumour immune responses. None of these interactions occur randomly but are orchestrated and critically depend on migratory guidance cues provided by chemokines, a large family of chemotactic cytokines, and their receptors. Understanding the functional organization of the tumour immune microenvironment inevitably requires knowledge of the multifaceted roles of chemokines in the recruitment and positioning of its cellular constituents. Gaining such knowledge will not only generate new insights into the mechanisms underlying antitumour immunity or immune tolerance but also inform the development of biomarkers (or 'biopatterns') based on spatial tumour tissue analyses, as well as novel strategies to therapeutically engineer immune responses in patients with cancer. Here we will discuss recent observations on the role of chemokines in the tumour microenvironment in the context of our knowledge of their physiological functions in development, homeostasis and antimicrobial responses.
Collapse
Affiliation(s)
- Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Julia K Lill
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lukas M Altenburger
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Sukhtankar DD, Fung JJ, Kim MN, Cayton T, Chiou V, Caculitan NG, Zalicki P, Kim S, Jo Y, Kim S, Lee JM, Choi J, Mun S, Chin A, Jang Y, Lee JY, Kim G, Kim EH, Huh WK, Jeong JY, Seen DS, Cardarelli PM. GPC-100, a novel CXCR4 antagonist, improves in vivo hematopoietic cell mobilization when combined with propranolol. PLoS One 2023; 18:e0287863. [PMID: 37878624 PMCID: PMC10599528 DOI: 10.1371/journal.pone.0287863] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/27/2023] [Indexed: 10/27/2023] Open
Abstract
Autologous Stem Cell Transplant (ASCT) is increasingly used to treat hematological malignancies. A key requisite for ASCT is mobilization of hematopoietic stem cells into peripheral blood, where they are collected by apheresis and stored for later transplantation. However, success is often hindered by poor mobilization due to factors including prior treatments. The combination of G-CSF and GPC-100, a small molecule antagonist of CXCR4, showed potential in a multiple myeloma clinical trial for sufficient and rapid collection of CD34+ stem cells, compared to the historical results from the standards of care, G-CSF alone or G-CSF with plerixafor, also a CXCR4 antagonist. In the present study, we show that GPC-100 has high affinity towards the chemokine receptor CXCR4, and it potently inhibits β-arrestin recruitment, calcium flux and cell migration mediated by its ligand CXCL12. Proximity Ligation Assay revealed that in native cell systems with endogenous receptor expression, CXCR4 co-localizes with the beta-2 adrenergic receptor (β2AR). Co-treatment with CXCL12 and the β2AR agonist epinephrine synergistically increases β-arrestin recruitment to CXCR4 and calcium flux. This increase is blocked by the co-treatment with GPC-100 and propranolol, a non-selective beta-adrenergic blocker, indicating a functional synergy. In mice, GPC-100 mobilized more white blood cells into peripheral blood compared to plerixafor. GPC-100 induced mobilization was further amplified by propranolol pretreatment and was comparable to mobilization by G-CSF. Addition of propranolol to the G-CSF and GPC-100 combination resulted in greater stem cell mobilization than the G-CSF and plerixafor combination. Together, our studies suggest that the combination of GPC-100 and propranolol is a novel strategy for stem cell mobilization and support the current clinical trial in multiple myeloma registered as NCT05561751 at www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Devki D. Sukhtankar
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Juan José Fung
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Mi-na Kim
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Thomas Cayton
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Valerie Chiou
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Niña G. Caculitan
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Piotr Zalicki
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Sujeong Kim
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Yoonjung Jo
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - SoHui Kim
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Jae Min Lee
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Junhee Choi
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | | | - Ashley Chin
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Yongdae Jang
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Ji Yeong Lee
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Gowoon Kim
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Eun Hee Kim
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Won-Ki Huh
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- Institute of Microbiology, Seoul National University, Seoul, Republic of Korea
| | - Jae-Yeon Jeong
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | | | - Pina M. Cardarelli
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| |
Collapse
|
26
|
Yang L, Zhang S, Pu P. Comprehensive analysis of ACKR family members in breast cancer using prognostic values. Oncol Lett 2023; 26:425. [PMID: 37664667 PMCID: PMC10472033 DOI: 10.3892/ol.2023.14011] [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: 05/12/2023] [Accepted: 07/25/2023] [Indexed: 09/05/2023] Open
Abstract
Breast cancer (BC) is by far the most prevalent malignancy found in the female population. Atypical chemokine receptors (ACKRs) are a subclass of G-protein-coupled receptors, which are characterized by disrupted ligand binding and a breakdown of signaling following ligand binding. The evolution and function of multiple ACKRs in BC have yet to be fully elucidated, although certain findings on this family have been reported in several studies in Homo sapiens and other species. The present study identified that the expression level of ACKRs was significantly lower in breast carcinoma (BRCA) tissues compared with normal breast tissues through searches of the Tumor Immune Estimation Resource, UALCAN and Gene Expression Profiling Interactive Analysis databases. Additionally, when comparing BRCA tissues with normal breast tissues, it was found that there was obvious hypomethylation in the promoters of ACKR1, ACKR3 and ACKR5, as well as a marked hypermethylation in the promoters of ACKR2 and ACKR6. In determining the prognosis of patients with BRCA, the expression levels of ACKR1, ACKR2, ACKR3, ACKR4 and ACKR6 were all found to be important factors. The values for distant metastasis-free survival (DMFS), overall survival (OS) and recurrence-free survival (RFS) were all found to be lower in patients with BRCA who had a low expression level of ACKR1. In addition, the RFS rates for patients with BRCA were lower when the expression of ACKR2 was low, and worse values for DMFS, OS and RFS were found to be highly correlated with higher expression levels of ACKR3. Moreover, the DMFS, OS, RFS and predictive power score values were worse in those patients with low ACKR4 expression, and the RFS values for patients with BRCA were also found to be lower when the expression level of ACKR6 was low. Additionally, dendritic cells, macrophages, neutrophils, T cells with CD4+ status, T cells with CD8+ status and B cells were all substantially linked with ACKR expression, as well as immune cell infiltration. Taken together, the findings of the present study may offer a theoretical foundation for the creation of novel targets and prognostic indicators for BRCA therapy.
Collapse
Affiliation(s)
- Lixian Yang
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei 054000, P.R. China
| | - Shiyu Zhang
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei 054000, P.R. China
| | - Pengpeng Pu
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei 054000, P.R. China
| |
Collapse
|
27
|
Xu H, Lin S, Zhou Z, Li D, Zhang X, Yu M, Zhao R, Wang Y, Qian J, Li X, Li B, Wei C, Chen K, Yoshimura T, Wang JM, Huang J. New genetic and epigenetic insights into the chemokine system: the latest discoveries aiding progression toward precision medicine. Cell Mol Immunol 2023; 20:739-776. [PMID: 37198402 PMCID: PMC10189238 DOI: 10.1038/s41423-023-01032-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/14/2023] [Indexed: 05/19/2023] Open
Abstract
Over the past thirty years, the importance of chemokines and their seven-transmembrane G protein-coupled receptors (GPCRs) has been increasingly recognized. Chemokine interactions with receptors trigger signaling pathway activity to form a network fundamental to diverse immune processes, including host homeostasis and responses to disease. Genetic and nongenetic regulation of both the expression and structure of chemokines and receptors conveys chemokine functional heterogeneity. Imbalances and defects in the system contribute to the pathogenesis of a variety of diseases, including cancer, immune and inflammatory diseases, and metabolic and neurological disorders, which render the system a focus of studies aiming to discover therapies and important biomarkers. The integrated view of chemokine biology underpinning divergence and plasticity has provided insights into immune dysfunction in disease states, including, among others, coronavirus disease 2019 (COVID-19). In this review, by reporting the latest advances in chemokine biology and results from analyses of a plethora of sequencing-based datasets, we outline recent advances in the understanding of the genetic variations and nongenetic heterogeneity of chemokines and receptors and provide an updated view of their contribution to the pathophysiological network, focusing on chemokine-mediated inflammation and cancer. Clarification of the molecular basis of dynamic chemokine-receptor interactions will help advance the understanding of chemokine biology to achieve precision medicine application in the clinic.
Collapse
Affiliation(s)
- Hanli Xu
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China
| | - Shuye Lin
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, 101149, Beijing, China
| | - Ziyun Zhou
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China
| | - Duoduo Li
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China
| | - Xiting Zhang
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China
| | - Muhan Yu
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China
| | - Ruoyi Zhao
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China
| | - Yiheng Wang
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China
| | - Junru Qian
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China
| | - Xinyi Li
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China
| | - Bohan Li
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China
| | - Chuhan Wei
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China
| | - Keqiang Chen
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Teizo Yoshimura
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Ji Ming Wang
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Jiaqiang Huang
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, 3 ShangyuanCun, Haidian District, 100044, Beijing, P.R. China.
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, 101149, Beijing, China.
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA.
| |
Collapse
|
28
|
Mayor F, Murga C. G Protein-Coupled Receptor Kinases Take Central Stage. Cells 2022; 12:cells12010023. [PMID: 36611817 PMCID: PMC9818062 DOI: 10.3390/cells12010023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The relevance of the family of G protein-coupled receptor kinases (GRKs) is based on its key participation in the regulation and intracellular dynamics of the largest family of membrane receptors, namely G protein-coupled receptors (GPCRs) [...].
Collapse
Affiliation(s)
- Federico Mayor
- Departamento de Biología Molecular, Instituto Universitario de Biología Molecular (IUBM) and Centro de Biología Molecular Severo Ochoa (CBMSO), Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Princesa, 28006 Madrid, Spain
- CIBER Cardiovascular (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28035 Madrid, Spain
| | - Cristina Murga
- Departamento de Biología Molecular, Instituto Universitario de Biología Molecular (IUBM) and Centro de Biología Molecular Severo Ochoa (CBMSO), Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Princesa, 28006 Madrid, Spain
- CIBER Cardiovascular (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28035 Madrid, Spain
- Correspondence:
| |
Collapse
|
29
|
Koch C, Fischer NC, Puchert M, Engele J. Interactions of the chemokines CXCL11 and CXCL12 in human tumor cells. BMC Cancer 2022; 22:1335. [PMID: 36539774 PMCID: PMC9768901 DOI: 10.1186/s12885-022-10451-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The chemokines, CXCL12 and CXCL11, are upregulated in tumors from many organs and control their progression. CXCL12 and CXCL11 affect tumor cell functions by either binding their prime receptors, CXCR4 and CXCR3, respectively, and/or CXCR7 as a common second chemokine receptor. In humans, CXCR3 exists in the functional splice variants, CXCR3A and CXCR3B, which either have pro- or anti-tumor activity, respectively. Despite the intimate crosstalk between the CXCL12- and CXCL11-system, the impact of a combination of CXCL12 and CXCL11 on tumor progression remains vague. METHODS In the present work, we have analyzed CXCL12 and CXCL11 for combined effects on migration, invasion, proliferation, and cytostatic-induced apoptosis of the human tumor cells, A549, A767, A772, DLD-1, and MDA-MB-231. RESULTS We demonstrate that the mode of interaction differs with respect to cell type and function and allows for either potentiation, attenuation or no changes of cellular responses. The divergent responses are not the result of the distinct use of different CXCL12- and CXCL11-receptors by the respective tumor cells, but in case of cell migration seem to be associated with the activation of p38 signaling pathways. CONCLUSIONS Our findings point to therapeutic limitations of ongoing efforts to selectively target CXCR3, CXCR4, or CXCR7 in cancer patients, and rather favor individualized targeting strategies.
Collapse
Affiliation(s)
- Christian Koch
- Institute of Anatomy, University of Leipzig, Medical Faculty, Liebigstr. 13, 04103, Leipzig, Germany
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Nina Charlotte Fischer
- Institute of Anatomy, University of Leipzig, Medical Faculty, Liebigstr. 13, 04103, Leipzig, Germany
| | - Malte Puchert
- Institute of Anatomy, University of Leipzig, Medical Faculty, Liebigstr. 13, 04103, Leipzig, Germany
| | - Jürgen Engele
- Institute of Anatomy, University of Leipzig, Medical Faculty, Liebigstr. 13, 04103, Leipzig, Germany.
| |
Collapse
|
30
|
Yang Y, Yuan L, Cao H, Guo J, Zhou X, Zeng Z. Application and Molecular Mechanisms of Extracellular Vesicles Derived from Mesenchymal Stem Cells in Osteoporosis. Curr Issues Mol Biol 2022; 44:6346-6367. [PMID: 36547094 PMCID: PMC9776574 DOI: 10.3390/cimb44120433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Osteoporosis (OP) is a chronic bone disease characterized by decreased bone mass, destroyed bone microstructure, and increased bone fragility. Accumulative evidence shows that extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) (MSC-EVs), especially exosomes (Exos), exhibit great potential in the treatment of OP. However, the research on MSC-EVs in the treatment of OP is still in the initial stage. The potential mechanism has not been fully clarified. Therefore, by reviewing the relevant literature of MSC-EVs and OP in recent years, we summarized the latest application of bone targeted MSC-EVs in the treatment of OP and further elaborated the potential mechanism of MSC-EVs in regulating bone formation, bone resorption, bone angiogenesis, and immune regulation through internal bioactive molecules to alleviate OP, providing a theoretical basis for the related research of MSC-EVs in the treatment of OP.
Collapse
Affiliation(s)
- Yajing Yang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
- Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Lei Yuan
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Hong Cao
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Jianmin Guo
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
- Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Zhipeng Zeng
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| |
Collapse
|
31
|
Chen F, Yang L, Peng X, Mao M, Liu X, Song J, Hu J. Histone deacetylase 2 regulates STAT1-dependent upregulation of atypical chemokine receptor 3 to induce M2 macrophage migration and immune escape in hepatocellular carcinoma. Mol Immunol 2022; 151:204-217. [PMID: 36179603 DOI: 10.1016/j.molimm.2022.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 10/31/2022]
Abstract
Atypical chemokine receptor 3 (ACKR3) has been linked to the tumor microenvironment. This work investigates the effects of ACKR3 and its regulatory molecules on the chemotactic migration of tumor-associated macrophages (TAMs) in hepatocellular carcinoma (HCC). RT-qPCR and western blot assays identified elevated ACKR3 and HDAC2 levels in HCC tissues and cells. Knockdown or overexpression of ACKR3 was induced in HCC cells through vectors of lentivirus plasmids, and then the conditioned medium of the HCC cells was collected to stimulate TAMs. The stimulated TAMs were co-cultured with CD3+ T cells. ACKR3 knockdown in HCC cells inhibited migration of TAMs and promoted M1 polarization, which restored the activity of T cells. Histone deacetylase 2 (HDAC2) recruited signal transducer and activator of transcription 1 (STAT1) to the ACKR3 promoter to activate ACKR3 transcription. HDAC2 silencing suppressed nuclear translocation of STAT1 and decreased ACKR3 expression. HDAC2 knockdown in HCC cells similarly suppressed TAM migration, promoted M1 polarization, and restored T cell function, but these changes were inversed by ACKR3 upregulation. HDAC2 or ACKR3 silencing weakened tumor growth and immune escape in mice. In conclusion, this study demonstrates that HDAC2 upregulates ACKR3 via STAT1 to induce migration of M2 macrophages and immune escape in HCC.
Collapse
Affiliation(s)
- Fu Chen
- The Seventh Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Liang Yang
- The Third Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Xueqiang Peng
- The Third Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Minghuan Mao
- Department of Urology Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Xiaodan Liu
- The Fifth Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Jianbo Song
- Interventional Department, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Jingyao Hu
- The Seventh Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China.
| |
Collapse
|
32
|
Ghasemi K, Ghasemi K. MSX-122: Is an effective small molecule CXCR4 antagonist in cancer therapy? Int Immunopharmacol 2022; 108:108863. [PMID: 35623288 DOI: 10.1016/j.intimp.2022.108863] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 11/05/2022]
Abstract
Chemokines, a subgroup of cytokines along with their receptors, are involved in various biologic processes and regulation of a wide range of immune responses in different physiologic and pathologic states such as tissue repair, infection, and inflammation. C-X-C motif chemokine receptor 4 (CXCR4), a G-protein-coupled receptor (GPCR), has one identified natural ligand termed stromal-derived factor-1(SDF-1 or CXCL12). Evidence demonstrated that the ligation of SDF-1 to CXCR4 initiates several intracellular signaling pathways, regulating cell proliferation, survival, chemotaxis, migration, angiogenesis, adhesion, as well as bone marrow (BM)-resident cells homing and mobilization. Additionally, CXCR4 is expressed by tumor cells in blood malignancies and solid tumors. Therefore, CXCR4 is considered a potential therapeutic target in cancer therapy, and CXCR4 antagonists, including AMD3100, MSX-122, BPRCX807, WZ811, Motixafortide, TN14003, AMD3465, and AMD1170, have been employed in experimental and clinical studies to enhance cancer therapy. MSX-122 is a specific small-molecule antagonist of CXCR4/CXCL12 and the only orally available non-peptide CXCR4 antagonist with promising anti-cancer properties. Studies have shown that MSX-122 is particularly important in treating metastatic cancers and has great therapeutic potential. Accordingly, this review summarized the characteristics of MSX-122 and its effects on the CXCL12/CXCR4 axis as well as cancer therapy.
Collapse
Affiliation(s)
- Kimia Ghasemi
- Department of Pharmacology and Toxicology, School of Pharmacy, Fertility and Infertility Research Center, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kosar Ghasemi
- Department of Pharmacology and Toxicology, School of Pharmacy, Cellular and Molecular Research Center, Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
33
|
The Chemokine System in Oncogenic Pathways Driven by Viruses: Perspectives for Cancer Immunotherapy. Cancers (Basel) 2022; 14:cancers14030848. [PMID: 35159113 PMCID: PMC8834488 DOI: 10.3390/cancers14030848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Oncoviruses are viruses with oncogenic potential, responsible for almost 20% of human cancers worldwide. They are from various families, some of which belong to the microbial communities that inhabit several sites in the body of healthy humans. As a result, they most often establish latent infections controlled by the arsenal of human host responses that include the chemokine system playing key roles at the interface between tissue homeostasis and immune surveillance. Yet, chemokines and their receptors also contribute to oncogenic processes as they are targeted by the virus-induced deregulations of host responses and/or directly encoded by viruses. Thus, the chemokine system offers a strong rationale for therapeutic options, some few already approved or in trials, and future ones that we are discussing in view of the pharmacological approaches targeting the different functions of chemokines operating in both cancer cells and the tumor microenvironment. Abstract Chemokines interact with glycosaminoglycans of the extracellular matrix and activate heptahelical cellular receptors that mainly consist of G Protein-Coupled Receptors and a few atypical receptors also with decoy activity. They are well-described targets of oncogenic pathways and key players in cancer development, invasiveness, and metastasis acting both at the level of cancer cells and cells of the tumor microenvironment. Hence, they can regulate cancer cell proliferation and survival and promote immune or endothelial cell migration into the tumor microenvironment. Additionally, oncogenic viruses display the potential of jeopardizing the chemokine system by encoding mimics of chemokines and receptors as well as several products such as oncogenic proteins or microRNAs that deregulate their human host transcriptome. Conversely, the chemokine system participates in the host responses that control the virus life cycle, knowing that most oncoviruses establish asymptomatic latent infections. Therefore, the deregulated expression and function of chemokines and receptors as a consequence of acquired or inherited mutations could bias oncovirus infection toward pro-oncogenic pathways. We here review these different processes and discuss the anticancer therapeutic potential of targeting chemokine availability or receptor activation, from signaling to decoy-associated functions, in combination with immunotherapies.
Collapse
|
34
|
Flatmark K, Torgunrud A, Fleten KG, Davidson B, Juul HV, Mensali N, Lund-Andersen C, Inderberg EM. Peptide vaccine targeting mutated GNAS: a potential novel treatment for pseudomyxoma peritonei. J Immunother Cancer 2021; 9:jitc-2021-003109. [PMID: 34711663 PMCID: PMC8557294 DOI: 10.1136/jitc-2021-003109] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
Background Pseudomyxoma peritonei (PMP) is a rare, slow-growing abdominal cancer with no efficacious treatment options in non-resectable and recurrent cases. Otherwise, rare activating mutations in the GNAS oncogene are remarkably frequent in PMP and the mutated gene product, guanine nucleotide-binding protein α subunit (Gsα), is a potential tumor neoantigen, presenting an opportunity for targeting by a therapeutic cancer vaccine. Methods Tumor and blood samples were collected from 25 patients undergoing surgery for PMP (NCT02073500). GNAS mutation analysis was performed by next-generation targeted sequencing or digital droplet PCR. Responses to stimulation with Gsα mutated (point mutations R201H and R201C) 30 mer peptides were analyzed in peripheral blood T cells derived from patients with PMP and healthy donors. Fresh PMP tumor samples were analyzed by mass cytometry using a panel of 35 extracellular markers, and cellular subpopulations were clustered and visualized using the visual stochastic network embedding analysis tool. Results GNAS mutations were detected in 22/25 tumor samples (88%; R201H and R201C mutations detected in 16 and 6 cases, respectively). Strong T cell proliferation against Gsα mutated peptides was observed in 18/24 patients with PMP. Mass cytometry analysis of tumor revealed infiltration of CD3 +T cells in most samples, with variable CD4+:CD8 + ratios. A large proportion of T cells expressed immune checkpoint molecules, in particular programmed death receptor-1 and T cell immunoreceptor with Ig and ITIM, indicating that these T cells were antigen experienced. Conclusion These findings point to the existence of a pre-existing immunity in patients with PMP towards mutated Gsα, which has been insufficient to control tumor growth, possibly because of inhibition of antitumor T cells by upregulation of immune checkpoint molecules. The results form a rationale for exploring peptide vaccination with Gsα peptides in combination with immune checkpoint inhibiton as a possible curative treatment for PMP and other GNAS mutated cancers.
Collapse
Affiliation(s)
- Kjersti Flatmark
- Department of Gastroenterological Surgery, Oslo University Hospital, Oslo, Norway .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Annette Torgunrud
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Karianne G Fleten
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Ben Davidson
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Hedvig V Juul
- Department of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Nadia Mensali
- Department of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Christin Lund-Andersen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Else Marit Inderberg
- Department of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
35
|
Palmer CB, Meyrath M, Canals M, Kostenis E, Chevigné A, Szpakowska M. Atypical opioid receptors: unconventional biology and therapeutic opportunities. Pharmacol Ther 2021; 233:108014. [PMID: 34624426 DOI: 10.1016/j.pharmthera.2021.108014] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/13/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022]
Abstract
Endogenous opioid peptides and prescription opioid drugs modulate pain, anxiety and stress by activating four opioid receptors, namely μ (mu, MOP), δ (delta, DOP), κ (kappa, KOP) and the nociceptin/orphanin FQ receptor (NOP). Interestingly, several other receptors are also activated by endogenous opioid peptides and influence opioid-driven signaling and biology. However, they do not meet the criteria to be recognized as classical opioid receptors, as they are phylogenetically distant from them and are insensitive to classical non-selective opioid receptor antagonists (e.g. naloxone). Nevertheless, accumulating reports suggest that these receptors may be interesting alternative targets, especially for the development of safer analgesics. Five of these opioid peptide-binding receptors belong to the family of G protein-coupled receptors (GPCRs)-two are members of the Mas-related G protein-coupled receptor X family (MrgX1, MrgX2), two of the bradykinin receptor family (B1, B2), and one is an atypical chemokine receptor (ACKR3). Additionally, the ion channel N-methyl-d-aspartate receptors (NMDARs) are also activated by opioid peptides. In this review, we recapitulate the implication of these alternative receptors in opioid-related disorders and discuss their unconventional biology, with members displaying signaling to scavenging properties. We provide an overview of their established and emerging roles and pharmacology in the context of pain management, as well as their clinical relevance as alternative targets to overcome the hurdles of chronic opioid use. Given the involvement of these receptors in a wide variety of functions, including inflammation, chemotaxis, anaphylaxis or synaptic transmission and plasticity, we also discuss the challenges associated with the modulation of both their canonical and opioid-driven signaling.
Collapse
Affiliation(s)
- Christie B Palmer
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Max Meyrath
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Meritxell Canals
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, UK
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Andy Chevigné
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
| | - Martyna Szpakowska
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| |
Collapse
|
36
|
Tao Y, Li Y, Zhang D, Xu L, Chen J, Sang Y, Piao H, Jing X, Yu M, Fu Q, Zhou S, Li D, Du M. Decidual CXCR4 + CD56 bright NK cells as a novel NK subset in maternal-foetal immune tolerance to alleviate early pregnancy failure. Clin Transl Med 2021; 11:e540. [PMID: 34709764 PMCID: PMC8516340 DOI: 10.1002/ctm2.540] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/12/2021] [Accepted: 08/06/2021] [Indexed: 01/15/2023] Open
Abstract
Natural killer (NK) cells preferentially accumulate at maternal-foetal interface and are believed to play vital immune-modulatory roles during early pregnancy and related immunological dysfunction may result in pregnant failure such as recurrent miscarriage (RM). However, the mechanisms underlying the establishment of maternal-foetal immunotolerance are complex but clarifying the roles of decidual NK (dNK) cells offers the potential to design immunotherapeutic strategies to assist RM patients. In this report, we analysed RNA sequencing on peripheral NK (pNK) and decidual NK cells during early pregnancy; we identified an immunomodulatory dNK subset CXCR4+ CD56bright dNK and investigated its origin and phenotypic and functional characteristics. CXCR4+ CD56bright dNK displayed a less activated and cytotoxic phenotype but an enhanced immunomodulatory potential relative to the CXCR4 negative subset. CXCR4+ CD56bright dNK promote Th2 shift in an IL-4-dependent manner and can be recruited from peripheral blood and reprogramed by trophoblasts, as an active participant in the establishment of immune-tolerance during early pregnancy. Diminished CXCR4+ dNK cells and their impaired ability to induce Th2 differentiation were found in RM patients and mouse models of spontaneous abortion. Moreover, adoptive transfer of CXCR4+ dNK cells to NK-deficient (Nfil3-/-) mice showed great therapeutic potential of CXCR4+ dNK via recovering the Th2/Th1 bias and reducing embryo resorption rates. The identification of this new dNK cell subset may lay the foundation for understanding NK cell mechanisms in early pregnancy and provide potential prognostic factors for the diagnosis and therapy of RM.
Collapse
MESH Headings
- Abortion, Habitual/blood
- Abortion, Habitual/immunology
- Abortion, Habitual/prevention & control
- Animals
- Decidua/immunology
- Disease Models, Animal
- Female
- Humans
- Immune Tolerance/immunology
- Killer Cells, Natural/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Neural Cell Adhesion Molecules/blood
- Neural Cell Adhesion Molecules/genetics
- Neural Cell Adhesion Molecules/immunology
- Pregnancy
- Pregnancy Trimester, First
- Receptors, CXCR4/blood
- Receptors, CXCR4/genetics
- Receptors, CXCR4/immunology
Collapse
Affiliation(s)
- Yu Tao
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghaiChina
- Department of Assisted ReproductionShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Yan‐Hong Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghaiChina
| | - Di Zhang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghaiChina
| | - Ling Xu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghaiChina
| | - Jia‐Jia Chen
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghaiChina
| | - Yi‐Fei Sang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghaiChina
| | - Hai‐Lan Piao
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghaiChina
| | - Xue‐Ling Jing
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghaiChina
| | - Min Yu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghaiChina
| | - Qiang Fu
- Department of ImmunologyBinzhou Medical CollegeYantaiPeople's Republic of China
| | - Sheng‐Tao Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, Department of Obstetrics and Gynecology, West China Second University HospitalSichuan University and Collaborative Innovation CenterChengduPeople's Republic of China
| | - Da‐Jin Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghaiChina
| | - Mei‐Rong Du
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghaiChina
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
- State Key Laboratory of Quality Research in Chinese Medicine and School of PharmacyMacau University of Science and TechnologyMacauChina
| |
Collapse
|
37
|
Tuo L, Song H, Jiang D, Bai X, Song G. Mesenchymal stem cells transfected with anti-miRNA-204-3p inhibit acute rejection after heart transplantation by targeting C-X-C motif chemokine receptor 4 (CXCR4) in vitro. J Thorac Dis 2021; 13:5077-5092. [PMID: 34527345 PMCID: PMC8411131 DOI: 10.21037/jtd-21-1293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 08/18/2021] [Indexed: 11/28/2022]
Abstract
Background Mesenchymal stem cells (MSCs) are a promising treatment for acute rejection (AR) after heart transplantation (HTx) owing to their immunomodulatory functions by promoting the transformation of macrophages from the M0 to M2 phenotype. However, it is undetermined whether surface expression of C-X-C motif chemokine receptor 4 (CXCR4) by MSCs influences macrophage polarization. In this study, we investigated the effects of MSCs on macrophages caused by CXCR4, and detected the underlying mechanism, which may contribute to improving HTx outcomes. Methods The MSCs were extracted from rat bone marrow and identified using flow cytometry. We subsequently observed the effects of CXCR4 and anti-miRNA-204-3p on cell proliferation and migration, and the effects on macrophage polarization. Dual luciferase reporter assay was used to explore whether miRNA-204-3p was an upstream microRNA (miRNA) of CXCR4. A series of rescue experiments were performed to further confirm the inhibitory effect of miRNA-204-3p on CXCR4. Results The results showed that CXCR4 could promote the proliferation and migration of MSCs. Furthermore, it facilitated MSC-mediated macrophage transformation from the M0 to M2 phenotype. In addition, miRNA-204-3p inhibited the function of CXCR4 of MSCs. Conclusions Regulated by miRNA-204-3p, CXCR4 could inhibit the progression of AR after HTx. This study provides a new insight of the treatment of AR after HTx.
Collapse
Affiliation(s)
- Lei Tuo
- Department of Cardiovascular Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Cardiovascular Surgery, Weifang Yidu Central Hospital, Qingzhou, China
| | - Hao Song
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Detian Jiang
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao Bai
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Guangmin Song
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
38
|
Ortiz Zacarías NV, Bemelmans MP, Handel TM, de Visser KE, Heitman LH. Anticancer opportunities at every stage of chemokine function. Trends Pharmacol Sci 2021; 42:912-928. [PMID: 34521537 DOI: 10.1016/j.tips.2021.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 02/01/2023]
Abstract
The chemokine system, comprising 48 chemokines and 23 receptors, is critically involved in several hallmarks of cancer. Yet, despite extensive efforts from the pharmaceutical sector, only two drugs aimed at this system are currently approved for clinical use against cancer. To date, numerous pharmacological approaches have been developed to successfully intervene at different stages of chemokine function: (i) chemokine availability; (ii) chemokine-glycosaminoglycan binding; and (iii) chemokine receptor binding. Many of these strategies have been tested in preclinical cancer models, and some have advanced to clinical trials as potential anticancer therapies. Here we will review the strategies and growing pharmacological toolbox for manipulating the chemokine system in cancer, and address novel methods poised for future (pre)clinical testing.
Collapse
Affiliation(s)
- Natalia V Ortiz Zacarías
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands; Oncode Institute, Leiden University, Leiden, The Netherlands
| | - Martijn P Bemelmans
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Tracy M Handel
- University of California San Diego, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, USA
| | - Karin E de Visser
- Oncode Institute, Leiden University, Leiden, The Netherlands; Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Immunology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands; Oncode Institute, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
39
|
GNAi2/gip2-Regulated Transcriptome and Its Therapeutic Significance in Ovarian Cancer. Biomolecules 2021; 11:biom11081211. [PMID: 34439877 PMCID: PMC8393559 DOI: 10.3390/biom11081211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/20/2022] Open
Abstract
Increased expression of GNAi2, which encodes the α-subunit of G-protein i2, has been correlated with the late-stage progression of ovarian cancer. GNAi2, also referred to as the proto-oncogene gip2, transduces signals from lysophosphatidic acid (LPA)-activated LPA-receptors to oncogenic cellular responses in ovarian cancer cells. To identify the oncogenic program activated by gip2, we carried out micro-array-based transcriptomic and bioinformatic analyses using the ovarian cancer cell-line SKOV3, in which the expression of GNAi2/gip2 was silenced by specific shRNA. A cut-off value of 5-fold change in gene expression (p < 0.05) indicated that a total of 264 genes were dependent upon gip2-expression with 136 genes coding for functional proteins. Functional annotation of the transcriptome indicated the hitherto unknown role of gip2 in stimulating the expression of oncogenic/growth-promoting genes such as KDR/VEGFR2, CCL20, and VIP. The array results were further validated in a panel of High-Grade Serous Ovarian Carcinoma (HGSOC) cell lines that included Kuramochi, OVCAR3, and OVCAR8 cells. Gene set enrichment analyses using DAVID, STRING, and Cytoscape applications indicated the potential role of the gip2-stimulated transcriptomic network involved in the upregulation of cell proliferation, adhesion, migration, cellular metabolism, and therapy resistance. The results unravel a multi-modular network in which the hub and bottleneck nodes are defined by ACKR3/CXCR7, IL6, VEGFA, CYCS, COX5B, UQCRC1, UQCRFS1, and FYN. The identification of these genes as the critical nodes in GNAi2/gip2 orchestrated onco-transcriptome establishes their role in ovarian cancer pathophysiology. In addition, these results also point to these nodes as potential targets for novel therapeutic strategies.
Collapse
|
40
|
Differential Involvement of ACKR3 C-Tail in β-Arrestin Recruitment, Trafficking and Internalization. Cells 2021; 10:cells10030618. [PMID: 33799570 PMCID: PMC8002179 DOI: 10.3390/cells10030618] [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: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Background: The atypical chemokine receptor 3 (ACKR3) belongs to the superfamily of G protein-coupled receptors (GPCRs). Unlike classical GPCRs, this receptor does not activate G proteins in most cell types but recruits β-arrestins upon activation. ACKR3 plays an important role in cancer and vascular diseases. As recruitment of β-arrestins is triggered by phosphorylation of the C-terminal tail of GPCRs, we studied the role of different potential phosphorylation sites within the ACKR3 C-tail to further delineate the molecular mechanism of internalization and trafficking of this GPCR. Methods: We used various bioluminescence and fluorescence resonance energy transfer-based sensors and techniques in Human Embryonic Kidney (HEK) 293T cells expressing WT or phosphorylation site mutants of ACKR3 to measure CXCL12-induced recruitment of β-arrestins and G-protein-coupled receptor kinases (GRKs), receptor internalization and trafficking. Results: Upon CXCL12 stimulation, ACKR3 recruits both β-arrestin 1 and 2 with equivalent kinetic profiles. We identified interactions with GRK2, 3 and 5, with GRK2 and 3 being important for β-arrestin recruitment. Upon activation, ACKR3 internalizes and recycles back to the cell membrane. We demonstrate that β-arrestin recruitment to the receptor is mainly determined by a single cluster of phosphorylated residues on the C-tail of ACKR3, and that residue T352 and in part S355 are important residues for β-arrestin1 recruitment. Phosphorylation of the C-tail appears essential for ligand-induced internalization and important for differential β-arrestin recruitment. GRK2 and 3 play a key role in receptor internalization. Moreover, ACKR3 can still internalize when β-arrestin recruitment is impaired or in the absence of β-arrestins, using alternative internalization pathways. Our data indicate that distinct residues within the C-tail of ACKR3 differentially regulate CXCL12-induced β-arrestin recruitment, ACKR3 trafficking and internalization.
Collapse
|
41
|
Hu Y, Li X, Zhang Q, Gu Z, Luo Y, Guo J, Wang X, Jing Y, Chen X, Su J. Exosome-guided bone targeted delivery of Antagomir-188 as an anabolic therapy for bone loss. Bioact Mater 2021; 6:2905-2913. [PMID: 33718671 PMCID: PMC7917458 DOI: 10.1016/j.bioactmat.2021.02.014] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/09/2021] [Accepted: 02/13/2021] [Indexed: 12/14/2022] Open
Abstract
The differentiation shift from osteogenesis to adipogenesis of bone marrow mesenchymal stem cells (BMSCs) characterizes many pathological bone loss conditions. Stromal cell-derived factor-1 (SDF1) is highly enriched in the bone marrow for C-X-C motif chemokine receptor 4 (CXCR4)-positive hematopoietic stem cell (HSC) homing and tumor bone metastasis. In this study, we displayed CXCR4 on the surface of exosomes derived from genetically engineered NIH-3T3 cells. CXCR4+ exosomes selectively accumulated in the bone marrow. Then, we fused CXCR4+ exosomes with liposomes carrying antagomir-188 to produce hybrid nanoparticles (NPs). The hybrid NPs specifically gathered in the bone marrow and released antagomir-188, which promoted osteogenesis and inhibited adipogenesis of BMSCs and thereby reversed age-related trabecular bone loss and decreased cortical bone porosity in mice. Taken together, this study presents a novel way to obtain bone-targeted exosomes via surface display of CXCR4 and a promising anabolic therapeutic approach for age-related bone loss. Surface display of CXCR4 grants exosomes bone targeting properties. Exosome-liposome hybrid nanoparticles carrying nucleic acid target bone. Antagomir-188 loaded hybrid nanoparticles regulate MSC differentiation in aged mice.
Collapse
Affiliation(s)
- Yan Hu
- Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xiaoqun Li
- Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Qin Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhengrong Gu
- Department of Orthopedics, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China
| | - Ying Luo
- Centre Laboratory, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jiawei Guo
- Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xiuhui Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Xiao Chen
- Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.,Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Jiacan Su
- Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
42
|
The G Protein-Coupled Receptor Kinases (GRKs) in Chemokine Receptor-Mediated Immune Cell Migration: From Molecular Cues to Physiopathology. Cells 2021; 10:cells10010075. [PMID: 33466410 PMCID: PMC7824814 DOI: 10.3390/cells10010075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/18/2020] [Accepted: 12/30/2020] [Indexed: 02/07/2023] Open
Abstract
Although G protein-coupled receptor kinases (GRKs) have long been known to regulate G protein-coupled receptor (GPCR) desensitization, their more recently characterized functions as scaffolds and signalling adapters underscore that this small family of proteins governs a larger array of physiological functions than originally suspected. This review explores how GRKs contribute to the complex signalling networks involved in the migration of immune cells along chemokine gradients sensed by cell surface GPCRs. We outline emerging evidence indicating that the coordinated docking of several GRKs on an active chemokine receptor determines a specific receptor phosphorylation barcode that will translate into distinct signalling and migration outcomes. The guidance cues for neutrophil migration are emphasized based on several alterations affecting GRKs or GPCRs reported to be involved in pathological conditions.
Collapse
|