1
|
Dai X, Cao B, Liu X, Meng W, Qiu Y, Sun Y, Zhang L, Li N, Liu Z, Li D, Xiao L, Li B, Zhang Q. Tumor vascular normalization by B7-H3 blockade augments T lymphocyte-mediated antitumor immunity. Eur J Pharmacol 2025; 993:177334. [PMID: 39892447 DOI: 10.1016/j.ejphar.2025.177334] [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: 09/28/2024] [Revised: 01/09/2025] [Accepted: 01/30/2025] [Indexed: 02/03/2025]
Abstract
Triple-negative breast cancer (TNBC), defined by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), presents unique clinical challenges and generally predicts a less favorable prognosis. Despite recent advancements in TNBC treatment, a subset of patients remains resistant to immunotherapy. B7-H3, a member of the B7 family of immune checkpoints, is correlated with poor outcomes in various cancers and is distinctively expressed in tumor vasculature, marking it as a potential biomarker for tumor-associated endothelial cells. We found high expression of B7-H3 in the endothelial cells of the postoperative tissue of TNBC patients. Elevated gene expression of CD276 (encoding B7-H3) and PECAM1 (encoding CD31) in TNBC is associated with poor prognosis. Anti-B7-H3 blockade reduces tumor burden and promotes lymphocyte infiltration in a TNBC mouse model. Additionally, anti-B7-H3 blockade promotes tumor vessel normalization and enhances programmed cell death ligand 1 (PD-L1) expression. Synergistic effects were observed when B7-H3 blockade was combined with programmed cell death protein 1 (PD-1) inhibition in the TNBC mouse model. Furthermore, anti-B7-H3 inhibits human umbilical vein endothelial cell (HUVEC) proliferation by suppression of the nuclear factor kappa-B (NF-κB) signaling pathway. Downregulation of B7-H3 expression in HUVECs promotes lymphocyte trans-endothelial migration. These findings suggest that B7-H3 represents a promising therapeutic target for TNBC, and the combination of anti-B7-H3 and anti-PD-1 therapies may have synergetic effects in treating TNBC.
Collapse
Affiliation(s)
- Xin Dai
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China; Department of Oncology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Boran Cao
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinnan Liu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wangyang Meng
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiran Qiu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital, Fudan University School of Medicine, Shanghai, China
| | - Yidan Sun
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lulu Zhang
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Nan Li
- Department of Pathology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenyu Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Dan Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lianbo Xiao
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Bin Li
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
2
|
Babič D, Jovčevska I, Zottel A. B7-H3 in glioblastoma and beyond: significance and therapeutic strategies. Front Immunol 2024; 15:1495283. [PMID: 39664380 PMCID: PMC11632391 DOI: 10.3389/fimmu.2024.1495283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024] Open
Abstract
Cancer has emerged as the second most prevalent disease and the leading cause of death, claiming the lives of 10 million individuals each year. The predominant varieties of cancer encompass breast, lung, colon, rectal, and prostate cancers. Among the more aggressive malignancies is glioblastoma, categorized as WHO stage 4 brain cancer. Following diagnosis, the typical life expectancy ranges from 12 to 15 months, as current established treatments like surgical intervention, radiotherapy, and chemotherapy using temozolomide exhibit limited effectiveness. Beyond conventional approaches, the exploration of immunotherapy for glioblastoma treatment is underway. A methodology involves CAR-T cells, monoclonal antibodies, ADCC and nanobodies sourced from camelids. Immunotherapy's recent focal point is the cellular ligand B7-H3, notably abundant in tumor cells while either scarce or absent in normal ones. Its expression elevates with cancer progression and serves as a promising prognostic marker. In this article, we delve into the essence of B7-H3, elucidating its function and involvement in signaling pathways. We delineate the receptors it binds to and its significance in glioblastoma and other cancer types. Lastly, we examine its role in immunotherapy and the utilization of nanobodies in this domain.
Collapse
|
3
|
Meeus F, Funeh CN, Awad RM, Zeven K, Autaers D, De Becker A, Van Riet I, Goyvaerts C, Tuyaerts S, Neyns B, Devoogdt N, De Vlaeminck Y, Breckpot K. Preclinical evaluation of antigen-sensitive B7-H3-targeting nanobody-based CAR-T cells in glioblastoma cautions for on-target, off-tumor toxicity. J Immunother Cancer 2024; 12:e009110. [PMID: 39562005 PMCID: PMC11575280 DOI: 10.1136/jitc-2024-009110] [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: 02/21/2024] [Accepted: 10/27/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Glioblastoma is the most common lethal primary brain tumor, urging evaluation of new treatment options. Chimeric antigen receptor (CAR)-T cells targeting B7 homolog 3 (B7-H3) are promising because of the overexpression of B7-H3 on glioblastoma cells but not on healthy brain tissue. Nanobody-based (nano)CARs are gaining increasing attention as promising alternatives to classical single-chain variable fragment-based (scFv)CARs, because of their single-domain nature and low immunogenicity. Still, B7-H3 nanoCAR-T cells have not been extensively studied in glioblastoma. METHODS B7-H3 nanoCAR- and scFvCAR-T cells were developed and evaluated in human glioblastoma models. NanoCAR-T cells targeting an irrelevant antigen served as control. T cell activation, cytokine secretion and killing capacity were evaluated in vitro using ELISA, live cell imaging and flow cytometry. Antigen-specific killing was assessed by generating B7-H3 knock-out cells using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9-genome editing. The tumor tracing capacity of the B7-H3 nanobody was first evaluated in vivo using nuclear imaging. Then, the therapeutic potential of the nanoCAR-T cells was evaluated in a xenograft glioblastoma model. RESULTS We showed that B7-H3 nanoCAR-T cells were most efficient in lysing B7-H3pos glioblastoma cells in vitro. Lack of glioblastoma killing by control nanoCAR-T cells and lack of B7-H3neg glioblastoma killing by B7-H3 nanoCAR-T cells showed antigen-specificity. We showed in vivo tumor targeting capacity of the B7-H3 nanobody-used for the nanoCAR design-in nuclear imaging experiments. Evaluation of the nanoCAR-T cells in vivo showed tumor control in mice treated with B7-H3 nanoCAR-T cells in contrast to progressive disease in mice treated with control nanoCAR-T cells. However, we observed limiting toxicity in mice treated with B7-H3 nanoCAR-T cells and showed that the B7-H3 nanoCAR-T cells are activated even by low levels of mouse B7-H3 expression. CONCLUSIONS B7-H3 nanoCAR-T cells showed promise for glioblastoma therapy following in vitro characterization, but limiting in vivo toxicity was observed. Off-tumor recognition of healthy mouse tissue by the cross-reactive B7-H3 nanoCAR-T cells was identified as a potential cause for this toxicity, warranting caution when using highly sensitive nanoCAR-T cells, recognizing the low-level expression of B7-H3 on healthy tissue.
Collapse
Affiliation(s)
- Fien Meeus
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cyprine Neba Funeh
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Robin Maximilian Awad
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Katty Zeven
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Dorien Autaers
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Ann De Becker
- Department of Hematology, Cellular Therapy Laboratory, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ivan Van Riet
- Department of Hematology, Cellular Therapy Laboratory, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Hematology and Immunology Research Team (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sandra Tuyaerts
- Department of Medical Oncology, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel, Brussels, Belgium
| | - Bart Neyns
- Department of Medical Oncology, University Hospital Brussels, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- Department of Medical Imaging (MIMA), Molecular Imaging and Therapy (MITH) research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Translational Oncology Research Center (TORC), Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
4
|
Qiu J, Fu Y, Tian T, Mao Y, Tian Q, Zhou L, Jin R, Zhuang L, Zhou G. Suppression of FOXC1 induces pyroptosis of the coronary artery through activation of JAK2. Atherosclerosis 2024; 396:118543. [PMID: 39182474 DOI: 10.1016/j.atherosclerosis.2024.118543] [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: 09/24/2023] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND AND AIMS Janus kinase 2 (JAK2) triggers endothelial pyroptosis and is associated with a multitude of pathological cardiovascular manifestations, including atherosclerosis. However, the associated transcriptional regulatory mechanisms remain unclear. In this study, we investigated a novel transcriptional regulator upstream of JAK2. METHODS We validated the binding and regulation of Forkhead box C1 (FOXC1) and JAK2 using chromatin immunoprecipitation and luciferase reporter assays. Immunofluorescence was used to detect protein localization in cells and tissues. Immunohistochemistry, hematoxylin-eosin (HE), Masson's trichrome, and Oil Red O staining were used to identify tissue lesions. Transcriptional functions were investigated using in vitro and in vivo coronary artery disease (CAD) atherosclerosis models. RESULTS The mRNA levels of JAK2 were considerably higher in both the cardiac tissues of mice and the peripheral blood of patients with CAD than in equivalent controls. JAK2 expression increased markedly in the coronary arteries of ApoeKO mice, whereas FOXC1 expression exhibited a decreasing trend. In vitro, FOXC1 bound to the JAK2 promoter region and inversely regulated the expression of JAK2. Mechanistic studies have revealed that the FOXC1-JAK2 pathway regulates pyroptosis and participates in the pathogenesis of human coronary artery endothelial cells (HCAECs). In vivo, the suppression of FOXC1 was confirmed to stimulate the levels of JAK2 and pyroptosis, contributing to the pathological progression of aortic and coronary artery damage. CONCLUSIONS We established the FOXC1-JAK2 regulatory pathway and verified its reverse-regulatory function in CAD pyroptosis. Our data emphasizes that FOXC1 is critical for the treatment of pyroptosis-induced injury in patients with CAD.
Collapse
Affiliation(s)
- Jiayun Qiu
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Yahong Fu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Ting Tian
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Yan Mao
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Qiang Tian
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Lanbo Zhou
- Department of Dermatology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Rui Jin
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Lili Zhuang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China.
| | - Guoping Zhou
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China.
| |
Collapse
|
5
|
Yang Z, Zhang X, Bai X, Xi X, Liu W, Zhong W. Anti-angiogenesis in colorectal cancer therapy. Cancer Sci 2024; 115:734-751. [PMID: 38233340 PMCID: PMC10921012 DOI: 10.1111/cas.16063] [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: 09/18/2023] [Revised: 11/16/2023] [Accepted: 12/16/2023] [Indexed: 01/19/2024] Open
Abstract
The morbidity of colorectal cancer (CRC) has risen to third place among malignant tumors worldwide. In addition, CRC is a common cancer in China whose incidence increases annually. Angiogenesis plays an important role in the development of tumors because it can bring the nutrients that cancer cells need and take away metabolic waste. Various mechanisms are involved in the formation of neovascularization, and vascular endothelial growth factor is a key mediator. Meanwhile, angiogenesis inhibitors and drug resistance (DR) are challenges to consider when formulating treatment strategies for patients with different conditions. Thus, this review will discuss the molecules, signaling pathways, microenvironment, treatment, and DR of angiogenesis in CRC.
Collapse
Affiliation(s)
- Zhenni Yang
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
- Department of Gastroenterology and HepatologyXing'an League People's HospitalXing'an LeagueChina
| | - Xuqian Zhang
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
- Department of Gastroenterology and HepatologyChina Aerospace Science and Industry CorporationBeijingChina
| | - Xiaozhe Bai
- Department of Gastroenterology and HepatologyXing'an League People's HospitalXing'an LeagueChina
| | - Xiaonan Xi
- State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjinChina
| | - Wentian Liu
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
| | - Weilong Zhong
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
| |
Collapse
|
6
|
Varghese E, Samuel SM, Brockmueller A, Shakibaei M, Kubatka P, Büsselberg D. B7-H3 at the crossroads between tumor plasticity and colorectal cancer progression: a potential target for therapeutic intervention. Cancer Metastasis Rev 2024; 43:115-133. [PMID: 37768439 PMCID: PMC11016009 DOI: 10.1007/s10555-023-10137-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
B7-H3 (B7 homology 3 protein) is an important transmembrane immunoregulatory protein expressed in immune cells, antigen-presenting cells, and tumor cells. Studies reveal a multifaceted role of B7-H3 in tumor progression by modulating various cancer hallmarks involving angiogenesis, immune evasion, and tumor microenvironment, and it is also a promising candidate for cancer immunotherapy. In colorectal cancer (CRC), B7-H3 has been associated with various aspects of disease progression, such as evasion of tumor immune surveillance, tumor-node metastasis, and poor prognosis. Strategies to block or interfere with B7-H3 in its immunological and non-immunological functions are under investigation. In this study, we explore the role of B7-H3 in tumor plasticity, emphasizing tumor glucose metabolism, angiogenesis, epithelial-mesenchymal transition, cancer stem cells, apoptosis, and changing immune signatures in the tumor immune landscape. We discuss how B7-H3-induced tumor plasticity contributes to immune evasion, metastasis, and therapy resistance. Furthermore, we delve into the most recent advancements in targeting B7-H3-based tumor immunotherapy as a potential approach to CRC treatment.
Collapse
Affiliation(s)
- Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar
| | - Aranka Brockmueller
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University Munich, Pettenkoferstr. 11, 80336, Munich, Germany
| | - Mehdi Shakibaei
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University Munich, Pettenkoferstr. 11, 80336, Munich, Germany
| | - Peter Kubatka
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar.
| |
Collapse
|
7
|
Fan X, Huang J, Hu B, Zhou J, Chen L. Tumor-expressed B7-H3 promotes vasculogenic mimicry formation rather than angiogenesis in non-small cell lung cancer. J Cancer Res Clin Oncol 2023; 149:8729-8741. [PMID: 37129607 DOI: 10.1007/s00432-023-04790-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/15/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Vasculogenic mimicry (VM), an alternative microvascular circulation independent of angiogenesis, is formed by aggressive cancer cells. Tumor-expressed B7-H3 has been reported to promote VM formation in hepatocellular carcinoma and modulate angiogenesis in breast cancer and colorectal cancer. However, its effects on VM generation and angiogenesis in non-small cell Lung cancer (NSCLC) remained to be elucidated. METHODS CRISPR/Cas9-mediated B7-H3 knockout (KO) was conducted in NSCLC A549 and H3255 cells. The expression of VM-related proteins, including vascular endothelial (VE)-cadherin and matrix metalloproteinase 14 (MMP14), and the secretion of vascular endothelial growth factor (VEGF) were measured by western blotting and chemiluminescence assay in both B7-H3 KO and mock-edited A549 and H3255 cells. To examine VM formation, a three-dimensional (3D) culture model was used for B7-H3 KO and mock A549 and H3255 cells. For in vivo analysis, xenograft mice models were established using B7-H3 KO and mock-edited A549 cells, and immunohistochemical (CD31) and histochemical (periodic acid-Schiff, PAS) double staining were performed to identify VM and endothelial vessels in tumor tissues. Finally, specific signaling inhibitors were used to analyze B7-H3-induced signaling pathway responsible for VE-cadherin and MMP14 expression and VM generation. RESULTS Higher expression of B7-H3 was associated with a worse prognosis and more advanced T-category in NSCLC. CRISPR/Cas9-mediated B7-H3 KO in A549 and H3255 cells led to decreased expression of VE-cadherin and MMP14; however, the secretion of VEGF by the two cell lines remained unchanged. In the 3D cell culture model, both B7-H3 KO A549 and H3255 cells showed a significant reduction in the formation of capillary-like tubular structures compared to mock-edited cells. In the in vivo xenograft model, mock-edited A549 cells formed excessive PAS+ CD31- VM channels, while B7-H3 KO restrained VM formation in the xenograft tumors. However, no significant differences were found in CD31+ endothelial vessels between xenografts formed by B7-H3 KO and mock-edited A549 cells. Finally, we analyzed the signaling pathway responsible for B7-H3-induced VM formation and found that selective inhibition of the phosphoinositide 3-kinase(PI3K)/protein kinase B (AKT) hyperactivation by LY294002 was associated with decreased expression of MMP14 and VE-cadherin, and in vitro VM formation by both A549 and H3255 cells. CONCLUSIONS Tumor-expressed B7-H3 acts via PI3K/AKT signaling pathway to promote VM formation by NSCLC cells while bears no effects on angiogenesis in NSCLC.
Collapse
Affiliation(s)
- Xingyu Fan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Junfeng Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bingqi Hu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Zhou
- Department of Laboratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liwen Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
8
|
Wang Q, Bode AM, Zhang T. Targeting CDK1 in cancer: mechanisms and implications. NPJ Precis Oncol 2023; 7:58. [PMID: 37311884 DOI: 10.1038/s41698-023-00407-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Cyclin dependent kinases (CDKs) are serine/threonine kinases that are proposed as promising candidate targets for cancer treatment. These proteins complexed with cyclins play a critical role in cell cycle progression. Most CDKs demonstrate substantially higher expression in cancer tissues compared with normal tissues and, according to the TCGA database, correlate with survival rate in multiple cancer types. Deregulation of CDK1 has been shown to be closely associated with tumorigenesis. CDK1 activation plays a critical role in a wide range of cancer types; and CDK1 phosphorylation of its many substrates greatly influences their function in tumorigenesis. Enrichment of CDK1 interacting proteins with Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was conducted to demonstrate that the associated proteins participate in multiple oncogenic pathways. This abundance of evidence clearly supports CDK1 as a promising target for cancer therapy. A number of small molecules targeting CDK1 or multiple CDKs have been developed and evaluated in preclinical studies. Notably, some of these small molecules have also been subjected to human clinical trials. This review evaluates the mechanisms and implications of targeting CDK1 in tumorigenesis and cancer therapy.
Collapse
Affiliation(s)
- Qiushi Wang
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA.
| | - Tianshun Zhang
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA.
| |
Collapse
|
9
|
Ng NS, Newbery M, Touffu A, Maksour S, Chung J, Carroll L, Zaw T, Wu Y, Ooi L. Edaravone and mitochondrial transfer as potential therapeutics for vanishing white matter disease astrocyte dysfunction. CNS Neurosci Ther 2023. [PMID: 36971196 PMCID: PMC10401142 DOI: 10.1111/cns.14190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 02/23/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
INTRODUCTION Previous research has suggested that vanishing white matter disease (VWMD) astrocytes fail to fully differentiate and respond differently to cellular stresses compared to healthy astrocytes. However, few studies have investigated potential VWMD therapeutics in monoculture patient-derived cell-based models. METHODS To investigate the impact of alterations in astrocyte expression and function in VWMD, astrocytes were differentiated from patient and control induced pluripotent stem cells and analyzed by proteomics, pathway analysis, and functional assays, in the absence and presence of stressors or potential therapeutics. RESULTS Vanishing white matter disease astrocytes demonstrated significantly reduced expression of astrocyte markers and markers of inflammatory activation or cellular stress relative to control astrocytes. These alterations were identified both in the presence and absence of polyinosinic:polycytidylic acid stimuli, which is used to simulate viral infections. Pathway analysis highlighted differential signaling in multiple pathways in VWMD astrocytes, including eukaryotic initiation factor 2 (EIF2) signaling, oxidative stress, oxidative phosphorylation (OXPHOS), mitochondrial function, the unfolded protein response (UPR), phagosome regulation, autophagy, ER stress, tricarboxylic acid cycle (TCA) cycle, glycolysis, tRNA signaling, and senescence pathways. Since oxidative stress and mitochondrial function were two of the key pathways affected, we investigated whether two independent therapeutic strategies could ameliorate astrocyte dysfunction: edaravone treatment and mitochondrial transfer. Edaravone treatment reduced differential VWMD protein expression of the UPR, phagosome regulation, ubiquitination, autophagy, ER stress, senescence, and TCA cycle pathways. Meanwhile, mitochondrial transfer decreased VWMD differential expression of the UPR, glycolysis, calcium transport, phagosome formation, and ER stress pathways, while further modulating EIF2 signaling, tRNA signaling, TCA cycle, and OXPHOS pathways. Mitochondrial transfer also increased the gene and protein expression of the astrocyte marker, glial fibrillary acidic protein (GFAP) in VWMD astrocytes. CONCLUSION This study provides further insight into the etiology of VWMD astrocytic failure and suggests edaravone and mitochondrial transfer as potential candidate VWMD therapeutics that can ameliorate disease pathways in astrocytes related to oxidative stress, mitochondrial dysfunction, and proteostasis.
Collapse
|
10
|
Gao Q, Cui L, Huang C, Chen Z, Wang X, Wen S, Zhao Y, Wang M, Shen B, Zhu W. Gastric cancer-derived mesenchymal stem cells promote gastric cancer cell lines migration by modulating CD276 expression. Exp Cell Res 2023; 422:113414. [PMID: 36368567 DOI: 10.1016/j.yexcr.2022.113414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/22/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
CD276 has been studied in a variety of cancers and diseases, but its regulatory mechanisms in gastric cancer is still unclear. Mesenchymal stem cells (MSCs), one of the important members of tumor microenvironment, play an important role in the occurrence, development and metastasis of tumor, but the relationship between gastric cancer mesenchymal stem cells (GCMSCs) and CD276 in gastric cancer needs to be further explored. The differential expression of CD276 was identified via UCLAN and GEPIA databases. Then, the impacts of CD276 were calculated on clinical prognosis using the Kaplan-Meier plotter and Cox analysis. GO, KEGG and GSEA analysis were used to explore potential mechanism under CD276. Next, the expression of CD276 in gastric cell lines were detected by Western blot. Immunocoprecipitation was used to explore the association between CD276 and COL1A1. And the effect of condition medium (CM) from GCMSCs on gastric cell lines migration analyzed. GC-MSCs activated the AKT/c-Myc/mTOR pathway of gastric cell lines and upregulated CD276 expression. Moreover, the upregulation of CD276 promoted the migration of gastric cancer cells. Taken together, this study shown that GCMSCs could up-regulate the expression of CD276 of gastric cell lines to promote tumor migration. Our results provide a new basis for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Qiuzhi Gao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Linjing Cui
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Chao Huang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Zhihong Chen
- Department of Gastrointestinal Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
| | - Xin Wang
- Department of Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Shaodi Wen
- Department of Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Yuanyuan Zhao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Mei Wang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Bo Shen
- Department of Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, 210009, China.
| | - Wei Zhu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
11
|
Zhao B, Li H, Xia Y, Wang Y, Wang Y, Shi Y, Xing H, Qu T, Wang Y, Ma W. Immune checkpoint of B7-H3 in cancer: from immunology to clinical immunotherapy. J Hematol Oncol 2022; 15:153. [PMID: 36284349 PMCID: PMC9597993 DOI: 10.1186/s13045-022-01364-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/30/2022] [Indexed: 11/28/2022] Open
Abstract
Immunotherapy for cancer is a rapidly developing treatment that modifies the immune system and enhances the antitumor immune response. B7-H3 (CD276), a member of the B7 family that plays an immunoregulatory role in the T cell response, has been highlighted as a novel potential target for cancer immunotherapy. B7-H3 has been shown to play an inhibitory role in T cell activation and proliferation, participate in tumor immune evasion and influence both the immune response and tumor behavior through different signaling pathways. B7-H3 expression has been found to be aberrantly upregulated in many different cancer types, and an association between B7-H3 expression and poor prognosis has been established. Immunotherapy targeting B7-H3 through different approaches has been developing rapidly, and many ongoing clinical trials are exploring the safety and efficacy profiles of these therapies in cancer. In this review, we summarize the emerging research on the function and underlying pathways of B7-H3, the expression and roles of B7-H3 in different cancer types, and the advances in B7-H3-targeted therapy. Considering different tumor microenvironment characteristics and results from preclinical models to clinical practice, the research indicates that B7-H3 is a promising target for future immunotherapy, which might eventually contribute to an improvement in cancer immunotherapy that will benefit patients.
Collapse
Affiliation(s)
- Binghao Zhao
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Huanzhang Li
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yu Xia
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yaning Wang
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yuekun Wang
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yixin Shi
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Hao Xing
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Tian Qu
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yu Wang
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Wenbin Ma
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
12
|
To kill a cancer: Targeting the immune inhibitory checkpoint molecule, B7-H3. Biochim Biophys Acta Rev Cancer 2022; 1877:188783. [PMID: 36028149 DOI: 10.1016/j.bbcan.2022.188783] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/30/2022] [Accepted: 08/19/2022] [Indexed: 12/26/2022]
Abstract
Targeting the anti-tumor immune response via the B7 family of immune-regulatory checkpoint proteins has revolutionized cancer treatment and resulted in punctuated responses in patients. B7-H3 has gained recent attention given its prominent deregulation and immunomodulatory role in a multitude of cancers. Numerous cancer studies have firmly established a strong link between deregulated B7-H3 expression and poorer outcomes. B7-H3 has been shown to augment cancer cell survival, proliferation, metastasis, and drug resistance by inducing an immune evasive phenotype through its effects on tumor-infiltrating immune cells, cancer cells, cancer-associated vasculature, and the stroma. Given the complex interplay between each of these components of the tumor microenvironment, a deeper understanding of B7-H3 signaling properties is inherently crucial to developing efficacious therapies that can target and inhibit these cancer-promoting interactions. This review delves into the various ways B7-H3 acts as an immunomodulator to facilitate immune evasion and promote tumor growth and spread. With post-transcriptional and post-translational modifications giving rise to different active isoforms coupled with recent discoveries of its putative receptors, B7-H3 can perform diverse functions. Here, we first discuss the dual co-stimulatory/co-inhibitory functions of B7-H3 in the context of normal physiology and cancer. We then discuss the crosstalk facilitated by B7-H3 between stromal components and tumor cells that promote tumor growth and metastasis in different populations of tumor cells, associated vasculature, and the stroma. Concurrently, we highlight therapeutic strategies that can exploit these interactions and their associated limitations, concluding with a special focus on the promise of next-gen in silico-based approaches to small molecule inhibitor drug discovery for B7-H3 that may mitigate these limitations.
Collapse
|
13
|
Picarda E, Galbo PM, Zong H, Rajan MR, Wallenius V, Zheng D, Börgeson E, Singh R, Pessin J, Zang X. The immune checkpoint B7-H3 (CD276) regulates adipocyte progenitor metabolism and obesity development. SCIENCE ADVANCES 2022; 8:eabm7012. [PMID: 35476450 PMCID: PMC9045715 DOI: 10.1126/sciadv.abm7012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/11/2022] [Indexed: 05/09/2023]
Abstract
The immune checkpoint B7-H3 (CD276) is a member of the B7 family that has been studied in the tumor microenvironment and immunotherapy, but its potential role in metabolism remains largely unknown. Here, we show that B7-H3 is highly expressed in mouse and human adipose tissue at steady state, with the highest levels in adipocyte progenitor cells. B7-H3 is rapidly down-regulated upon the initiation of adipocyte differentiation. Combined RNA sequencing and metabolic studies reveal that B7-H3 stimulates glycolytic and mitochondrial activity of adipocyte progenitors. Loss of B7-H3 in progenitors results in impaired oxidative metabolism program and increased lipid accumulation in derived adipocytes. Consistent with these observations, mice knocked out for B7-H3 develop spontaneous obesity, metabolic dysfunction, and adipose tissue inflammation. Our results reveal an unexpected metabolic role for B7-H3 in adipose tissue and open potential new avenues for the treatment of metabolic diseases by targeting the B7-H3 pathway.
Collapse
Affiliation(s)
- Elodie Picarda
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Phillip M. Galbo
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Haihong Zong
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Meenu Rohini Rajan
- Institute of Medicine, Department of Molecular and Clinical Medicine, The Wallenberg Laboratory and Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ville Wallenius
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Emma Börgeson
- Institute of Medicine, Department of Molecular and Clinical Medicine, The Wallenberg Laboratory and Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Vaestra Goetaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rajat Singh
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jeffrey Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
14
|
Harland N, Maurer FB, Abruzzese T, Bock C, Montes-Mojarro IA, Fend F, Aicher WK, Stenzl A, Amend B. Elevated Expression of the Immune Checkpoint Ligand CD276 (B7-H3) in Urothelial Carcinoma Cell Lines Correlates Negatively with the Cell Proliferation. Int J Mol Sci 2022; 23:4969. [PMID: 35563359 PMCID: PMC9100984 DOI: 10.3390/ijms23094969] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/15/2022] Open
Abstract
The cell surface molecule CD276 (B7-H3) is an immune checkpoint antigen. The elevated expression of CD276 on tumors contributes to the suppression of anti-tumor T-cell responses and correlates with poor prognosis. METHODS The expression of CD276 was explored in vitro on eight urothelial carcinoma cell lines (UM-UC) in comparison to eight normal urothelial cells (NUCs) by RT-qPCR, Western blotting, and flow cytometry. Cell proliferation was enumerated over consecutive passages. The expression of cancer stem cell markers CD24 and CD44, cytokeratins, and vimentin was investigated by immunofluorescence. The expression of CD276 in bladder tumor samples and metastases was explored by immunohistochemistry. RESULTS Expression of CD276 on cell surfaces was elevated on UM-UCs when compared to NUCs. In UM-UCs, CD276 transcripts correlated moderately positive with CD276 protein expression (ρ = 0.660) and strongly positive with CD276 surface-expression (ρ = 0.810). CD276 mRNA expression (ρ = -0.475) and CD276 protein expression (ρ = -0.417) had a significant negative correlation with proliferation, while a significant correlation between proliferation and cell surface expression was not observed in UM-UCs. CONCLUSION The expression of CD276 on UM-UC bladder tumor cell surfaces is elevated. Slow proliferating UM-UC cells express more CD276 mRNA and protein than fast proliferating cells. In patients, slow proliferating CD276high tumor (stem) cells may evade immune surveillance. However, cancer therapy targeting CD276 may be effective in the treatment of slow proliferating tumor cells.
Collapse
Affiliation(s)
- Niklas Harland
- Department of Urology, University of Tuebingen Hospital, Hoppe-Seyler-Str. 3, 72076 Tuebingen, Germany; (N.H.); (A.S.)
| | - Florian B. Maurer
- Center for Medical Research, University of Tuebingen Hospital, Waldhoernlestr. 22, 72072 Tuebingen, Germany; (F.B.M.); (T.A.); (C.B.); (W.K.A.)
| | - Tanja Abruzzese
- Center for Medical Research, University of Tuebingen Hospital, Waldhoernlestr. 22, 72072 Tuebingen, Germany; (F.B.M.); (T.A.); (C.B.); (W.K.A.)
| | - Cornelia Bock
- Center for Medical Research, University of Tuebingen Hospital, Waldhoernlestr. 22, 72072 Tuebingen, Germany; (F.B.M.); (T.A.); (C.B.); (W.K.A.)
| | - Ivonne A. Montes-Mojarro
- Institute for Pathology, Eberhard-Karls-University, 72072 Tuebingen, Germany; (I.A.M.-M.); (F.F.)
| | - Falko Fend
- Institute for Pathology, Eberhard-Karls-University, 72072 Tuebingen, Germany; (I.A.M.-M.); (F.F.)
| | - Wilhelm K. Aicher
- Center for Medical Research, University of Tuebingen Hospital, Waldhoernlestr. 22, 72072 Tuebingen, Germany; (F.B.M.); (T.A.); (C.B.); (W.K.A.)
| | - Arnulf Stenzl
- Department of Urology, University of Tuebingen Hospital, Hoppe-Seyler-Str. 3, 72076 Tuebingen, Germany; (N.H.); (A.S.)
| | - Bastian Amend
- Department of Urology, University of Tuebingen Hospital, Hoppe-Seyler-Str. 3, 72076 Tuebingen, Germany; (N.H.); (A.S.)
| |
Collapse
|
15
|
Ke X, Liao Z, Luo X, Chen JQ, Deng M, Huang Y, Wang Z, Wei M. Endothelial colony-forming cell-derived exosomal miR-21-5p regulates autophagic flux to promote vascular endothelial repair by inhibiting SIPL1A2 in atherosclerosis. Cell Commun Signal 2022; 20:30. [PMID: 35279183 PMCID: PMC8917727 DOI: 10.1186/s12964-022-00828-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/07/2022] [Indexed: 12/12/2022] Open
Abstract
Abstract
Background
Percutaneous transluminal coronary angioplasty (PTCA) represents an efficient therapeutic method for atherosclerosis but conveys a risk of causing restenosis. Endothelial colony-forming cell-derived exosomes (ECFC-exosomes) are important mediators during vascular repair. This study aimed to investigate the therapeutic effects of ECFC-exosomes in a rat model of atherosclerosis and to explore the molecular mechanisms underlying the ECFC-exosome-mediated effects on ox-LDL-induced endothelial injury.
Methods
The effect of ECFC-exosome-mediated autophagy on ox-LDL-induced human microvascular endothelial cell (HMEC) injury was examined by cell counting kit-8 assay, scratch wound assay, tube formation assay, western blot and the Ad-mCherry-GFP-LC3B system. RNA-sequencing assays, bioinformatic analysis and dual-luciferase reporter assays were performed to confirm the interaction between the miR-21-5p abundance of ECFC-exosomes and SIPA1L2 in HMECs. The role and underlying mechanism of ECFC-exosomes in endothelial repair were explored using a high-fat diet combined with balloon injury to establish an atherosclerotic rat model of vascular injury. Evans blue staining, haematoxylin and eosin staining and western blotting were used to evaluate vascular injury.
Results
ECFC-exosomes were incorporated into HMECs and promoted HMEC proliferation, migration and tube formation by repairing autophagic flux and enhancing autophagic activity. Subsequently, we demonstrated that miR-21-5p, which is abundant in ECFC-exosomes, binds to the 3’ untranslated region of SIPA1L2 to inhibit its expression, and knockout of miR-21-5p in ECFC-exosomes reversed ECFC-exosome-decreased SIPA1L2 expression in ox-LDL-induced HMEC injury. Knockdown of SIPA1L2 repaired autophagic flux and enhanced autophagic activity to promote cell proliferation in ox-LDL-treated HMECs. ECFC-exosome treatment attenuated vascular endothelial injury, regulated lipid balance and activated autophagy in an atherogenic rat model of vascular injury, whereas these effects were eliminated with ECFC-exosomes with knockdown of miR-21-5p.
Conclusions
Our study demonstrated that ECFC-exosomes protect against atherosclerosis- or PTCA-induced vascular injury by rescuing autophagic flux and inhibiting SIAP1L2 expression through delivery of miR-21-5p.
Collapse
|
16
|
Haydar D, Ibañez-Vega J, Krenciute G. T-Cell Immunotherapy for Pediatric High-Grade Gliomas: New Insights to Overcoming Therapeutic Challenges. Front Oncol 2021; 11:718030. [PMID: 34760690 PMCID: PMC8573171 DOI: 10.3389/fonc.2021.718030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 10/08/2021] [Indexed: 01/06/2023] Open
Abstract
Despite decades of research, pediatric central nervous system (CNS) tumors remain the most debilitating, difficult to treat, and deadliest cancers. Current therapies, including radiation, chemotherapy, and/or surgery, are unable to cure these diseases and are associated with serious adverse effects and long-term impairments. Immunotherapy using chimeric antigen receptor (CAR) T cells has the potential to elucidate therapeutic antitumor immune responses that improve survival without the devastating adverse effects associated with other therapies. Yet, despite the outstanding performance of CAR T cells against hematologic malignancies, they have shown little success targeting brain tumors. This lack of efficacy is due to a scarcity of targetable antigens, interactions with the immune microenvironment, and physical and biological barriers limiting the homing and trafficking of CAR T cells to brain tumors. In this review, we summarize experiences with CAR T-cell therapy for pediatric CNS tumors in preclinical and clinical settings and focus on the current roadblocks and novel strategies to potentially overcome those therapeutic challenges.
Collapse
Affiliation(s)
| | | | - Giedre Krenciute
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
17
|
Zhang M, Zhang H, Fu M, Zhang J, Zhang C, Lv Y, Fan F, Zhang J, Xu H, Ye D, Yang H, Hua W, Mao Y. The Inhibition of B7H3 by 2-HG Accumulation Is Associated With Downregulation of VEGFA in IDH Mutated Gliomas. Front Cell Dev Biol 2021; 9:670145. [PMID: 34079802 PMCID: PMC8165280 DOI: 10.3389/fcell.2021.670145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
B7H3 (also known as CD276) is a co-stimulator checkpoint protein of the cell surface B7 superfamily. Recently, the function beyond immune regulation of B7H3 has been widely studied. However, the expression preference and the regulation mechanism underlying B7H3 in different subtypes of gliomas is rarely understood. We show here that B7H3 expression is significantly decreased in IDH-mutated gliomas and in cultured IDH1-R132H glioma cells. Accumulation of 2-HG leads to a remarkable downregulation of B7H3 protein and the activity of IDH1-R132H mutant is responsible for B7H3 reduction in glioma cells. Inhibition of autophagy by inhibitors like leupeptin, chloroquine (CQ), and Bafilomycin A1 (Baf-A1) blocks the degradation of B7H3 in glioma cells. In the meantime, the autophagy flux is more active with higher LC3B-II and lower p62 in IDH1-R132H glioma cells than in IDH1-WT cells. Furthermore, sequence alignment analysis reveals potential LC3-interacting region (LIR) motifs “F-V-S/N-I/V” in B7H3. Moreover, B7H3 interacts with p62 and CQ treatment significantly enhances this interaction. Additionally, we find that B7H3 is positively correlated with VEGFA and MMP2 by bioinformatics analysis in gliomas. B7H3 and VEGFA are decreased in IDH-mutated gliomas and further reduced in 2-HGhigh gliomas compared to 2-HGlow glioma sections by IHC staining. Our study demonstrates that B7H3 is preferentially overexpressed in IDH wild-type gliomas and could serve as a potential theranostic target for the precise treatment of glioma patients with wild-type IDH.
Collapse
Affiliation(s)
- Mengli Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Huaichao Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Minjie Fu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingwen Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Cheng Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yingying Lv
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Fengfeng Fan
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinsen Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Hao Xu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Dan Ye
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, The Molecular and Cell Biology Lab, Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,Ministry of Education Frontiers Center for Brain Science, Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China.,The Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
18
|
The complexity of tumour angiogenesis based on recently described molecules. Contemp Oncol (Pozn) 2021; 25:33-44. [PMID: 33911980 PMCID: PMC8063899 DOI: 10.5114/wo.2021.105075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022] Open
Abstract
Tumour angiogenesis is a crucial factor associated with tumour growth, progression, and metastasis. The whole process is the result of an interaction between a wide range of different molecules, influencing each other. Herein we summarize novel discoveries related to the less known angiogenic molecules such as galectins, pentraxin-3, Ral-interacting protein of 76 kDa (RLIP76), long non-coding RNAs (lncRNAs), B7-H3, and delta-like ligand-4 (DLL-4) and their role in the process of tumour angiogenesis. These molecules influence the most important molecular pathways involved in the formation of blood vessels in cancer, including the vascular endothelial growth factor (VEGF)-vascular endothelial growth factor receptor interaction (VEGFR), HIF1-a activation, or PI3K/Akt/mTOR and JAK-STAT signalling pathways. Increased expression of galectins, RLIP76, and B7H3 has been proven in several malignancies. Pentraxin-3, which appears to inhibit tumour angiogenesis, shows reduced expression in tumour tissues. Anti-angiogenic treatment based mainly on VEGF inhibition has proved to be of limited effectiveness, leading to the development of drug resistance. The newly discovered molecules are of great interest as a potential source of new anti-cancer therapies. Their role as targets for new drugs and as prognostic markers in neoplasms is discussed in this review.
Collapse
|
19
|
Geng Q, Shen Z, Li L, Zhao J. COL1A1 is a prognostic biomarker and correlated with immune infiltrates in lung cancer. PeerJ 2021; 9:e11145. [PMID: 33850663 PMCID: PMC8018245 DOI: 10.7717/peerj.11145] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/03/2021] [Indexed: 12/23/2022] Open
Abstract
Objective Lung cancer (LC) is one of the top ten malignant tumors and the first leading cause of cancer-related death among both men and women worldwide. It is imperative to identify immune-related biomarkers for early LC diagnosis and treatment. Methods Three Gene Expression Omnibus (GEO) datasets were selected to acquire the differentially expressed genes(DEGs) between LC and normal lung samples through GEO2R tools of NCBI. To identify hub genes, the DEGs were performed functional enrichment analysis, the protein-protein interaction (PPI) network construction, and Lasso regression. Then, a nomogram was constructed to predict the prognosis of patients with carcinoma based on hub genes. We further evaluated the influence of COL1A1 on clinical prognosis using GSE3141, GSE31210, and TCGA database. Also, the correlations between COL1A1 and cancer immune infiltrates and the B7-CD28 family was investigated via TIMER and GEPIA. Further analysis of immunohistochemistry shown that the COL1A1 expression level is positively correlated with CD276 expression level. Results By difference analysis, there were 340 DEGs between LC and normal lung samples. Then, we picked out seven hub genes, which were identified as components of the risk signature to divide LC into low and high-risk groups. Among them, the expression of COL1A1 is highly correlated with overall survival(OS) and progression-free survival (PFS) (p < 0.05). Importantly, there is a moderate to strong positive relationships between COL1A1 expression level and infiltration level of CD4+ T cells, Macrophage, Neutrophil, and Dendritic cell, as well as CD276 expression level. Conclusion These findings suggest that COL1A1 is correlated with prognosis and immune infiltrating levels, including CD4+ T cells, Macrophage, Neutrophil, and Dendritic cell, as well as CD276 expression level, indicating COL1A1 can be a potential immunity-related biomarker and therapeutic target in LC.
Collapse
Affiliation(s)
- Qishun Geng
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhibo Shen
- Engineering Laboratory for Digital Telemedicine Service, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lifeng Li
- Engineering Laboratory for Digital Telemedicine Service, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Zhao
- Engineering Laboratory for Digital Telemedicine Service, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
20
|
Lu Z, Zhao ZX, Cheng P, Huang F, Guan X, Zhang MG, Chen HP, Liu Z, Jiang Z, Zheng ZX, Zou SM, Wang XS. B7-H3 immune checkpoint expression is a poor prognostic factor in colorectal carcinoma. Mod Pathol 2020; 33:2330-2340. [PMID: 32514163 DOI: 10.1038/s41379-020-0587-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 12/24/2022]
Abstract
Although PD-1/PD-L1 immunotherapy has been used successfully in treating many cancers, metastatic colorectal cancer (CRC) patients are not as responsive. B7-H3 is a promising target for immunotherapy and we found it to have the highest expression among B7-CD28 family members in CRC. Thus, the aim of the present study was to investigate B7-H3 expression in a large CRC cohort. B7-H3, B7-H4, and PD-L1 protein levels and differential lymphocyte infiltration were evaluated in tissue microarrays from 805 primary tumors and matched metastases. The relationships between immune markers, patient characteristics, and survival outcomes were determined. B7-H3 (50.9%) was detected in more primary tumors than B7-H4 (29.1%) or PD-L1 (29.2%), and elevated B7-H3 expression was associated with advanced overall stage. Co-expression of B7-H3 only with B7-H4 or PD-L1 was infrequent in primary tumors (6.3%, 5.7%, respectively). Moreover, B7-H3 in primary tumors was positively correlated with their respective expression at metastatic sites (ρ = 0.631; p < 0.001). No significant relationships between B7-H4 and PD-L1 and survival were observed; however, B7-H3 overexpression in primary tumors was significantly related to decreased disease-free survival. A positive relationship between B7-H3 expression and high density CD45RO T cell was observed in primary tumors, whereas B7-H4 and PD-L1 overexpression were related to CD3 T-cell infiltration. In conclusion, compared with B7-H4 and PD-L1, B7-H3 expression exhibited a higher prevalence and was significantly related to aggressiveness, worse prognosis and CD45RO T-cell infiltration in primary tumors. Further exploration of this potential target of immunotherapy in CRC patients is warranted.
Collapse
Affiliation(s)
- Zhao Lu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Xun Zhao
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pu Cheng
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Huang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Guan
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Guang Zhang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hai-Peng Chen
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Jiang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhao-Xu Zheng
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Shuang-Mei Zou
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xi-Shan Wang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
21
|
Lu H, Ma Y, Wang M, Shen J, Wu H, Li J, Gao N, Gu Y, Zhang X, Zhang G, Shi T, Chen W. B7-H3 confers resistance to Vγ9Vδ2 T cell-mediated cytotoxicity in human colon cancer cells via the STAT3/ULBP2 axis. Cancer Immunol Immunother 2020; 70:1213-1226. [PMID: 33119798 DOI: 10.1007/s00262-020-02771-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022]
Abstract
Immunotherapy based on γδT cells has limited efficiency in solid tumors, including colon cancer (CC). The immune evasion of tumor cells may be the main cause of the difficulties of γδT cell-based treatment. In the present study, we explored whether and how B7-H3 regulates the resistance of CC cells to the cytotoxicity of Vγ9Vδ2 (Vδ2) T cells. We observed that B7-H3 overexpression promoted, while B7-H3 knockdown inhibited, CC cell resistance to the killing effect of Vδ2 T cells in vitro and in vivo. Mechanistically, we showed that B7-H3-mediated CC cell resistance to the cytotoxicity of Vδ2 T cells involved a molecular pathway comprising STAT3 activation and decreased ULBP2 expression. ULBP2 blockade or knockdown abolished the B7-H3 silencing-induced increase in the cytotoxicity of Vδ2 T cells to CC cells. Furthermore, cryptotanshinone, a STAT3 phosphorylation inhibitor, reversed the B7-H3 overexpression-induced decrease in ULBP2 expression and attenuated the killing effect of Vδ2 T cells on CC cells. Moreover, there was a negative correlation between the expression of B7-H3 and ULBP2 in the tumor tissues of CC patients. Our results suggest that the B7-H3-mediated STAT3/ULBP2 axis may be a potential candidate target for improving the efficiency of γδT cell-based immunotherapy in CC.
Collapse
Affiliation(s)
- Huimin Lu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, 215100, Jiangsu, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China
| | - Yanchao Ma
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China
| | - Mingyuan Wang
- Suzhou Red Cross Blood Center, 355 Shizi Road, Suzhou, China
| | - Jin Shen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
| | - Hongya Wu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, 215100, Jiangsu, China
| | - Juntao Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
| | - Nan Gao
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
| | - Yanzheng Gu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, 215100, Jiangsu, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, 215100, Jiangsu, China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, 215100, Jiangsu, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, 215100, Jiangsu, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China.
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, China.
| | - Weichang Chen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 708 Renmin Road, Suzhou, 215100, Jiangsu, China.
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 708 Renmin Road, Suzhou, China.
- , 50 Donghuan Road, Suzhou, 215100, Jiangsu, China.
| |
Collapse
|
22
|
Chen G, Tu Y, Aladelusi TO, Zhao S, Chen J, Jin L, Zhu D. Knocking down B7H3 expression enhances cell proliferation of SHEDs via the SHP1/AKT signal axis. Biochem Biophys Res Commun 2020; 531:282-289. [PMID: 32800541 DOI: 10.1016/j.bbrc.2020.06.154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/26/2020] [Indexed: 01/12/2023]
Abstract
B7H3 is a member of B7 family of immunoregulatory transmembrane glycoproteins associated with maintaining immune tolerance, tumor cell proliferation, migration, invasion and metabolism, drug resistance, and stem cell differentiation. Neural crest-derived Multipotent Stem Cells (MSCs) from the dental pulp has become a good choice for tissue regeneration because it is easily obtainable and has strong regeneration potentials. Although there have been many studies investigating the role of B7H3 in cancer cells and immune cells, its role in the dental pulp stem cells regeneration is unknown. In this study, we chose SHEDs (stem cells from human exfoliated deciduous teeth) as a research model to analyze the expression and function of B7H3. The result showed that SHEDs were B7H3/CD90, B7H3/CD73, B7H3/CD105 double positive, and the expression of B7H3 is primarily located within the membrane. Downregulation of B7H3 expression significantly accelerated the expansion of SHEDs through the SHP1/AKT signal axis while upregulation of B7H3 expression decreased the proliferation of SHEDs. Hence, this study indicates that B7H3 is a stem cell surface molecule and might be used as a SHEDs marker whereby its downregulation enhances the proliferation of SHEDs via the activation of B7H3/SHP1/AKT signaling pathway.
Collapse
Affiliation(s)
- Gang Chen
- Department of Pediatric Dentistry, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology &Shanghai Key Research Institute of Stomatology, Shanghai, 200011, China
| | - Yaoyao Tu
- Department of Obstetrics and Gynecology, International Peace Marternity and Child Health Hospital, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Timothy O Aladelusi
- Department of Oral and Maxillofacial Surgery, College of Medicine, University of Ibadan, Nigeria
| | - Shiming Zhao
- Department of Pediatric Dentistry, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, China
| | - Jun Chen
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology &Shanghai Key Research Institute of Stomatology, Shanghai, 200011, China
| | - Li Jin
- Department of Obstetrics and Gynecology, International Peace Marternity and Child Health Hospital, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
| | - Dongwang Zhu
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology &Shanghai Key Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, College of Stomatoloy, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
23
|
Flem-Karlsen K, Fodstad Ø, Nunes-Xavier CE. B7-H3 Immune Checkpoint Protein in Human Cancer. Curr Med Chem 2020; 27:4062-4086. [PMID: 31099317 DOI: 10.2174/0929867326666190517115515] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/29/2019] [Accepted: 05/04/2019] [Indexed: 02/07/2023]
Abstract
B7-H3 belongs to the B7 family of immune checkpoint proteins, which are important regulators of the adaptive immune response and emerging key players in human cancer. B7-H3 is a transmembrane protein expressed on the surface of tumor cells, antigen presenting cells, natural killer cells, tumor endothelial cells, but can also be present in intra- and extracellular vesicles. Additionally, B7-H3 may be present as a circulating soluble isoform in serum and other body fluids. B7-H3 is overexpressed in a variety of tumor types, in correlation with poor prognosis. B7-H3 is a promising new immunotherapy target for anti-cancer immune response, as well as a potential biomarker. Besides its immunoregulatory role, B7-H3 has intrinsic pro-tumorigenic activities related to enhanced cell proliferation, migration, invasion, angiogenesis, metastatic capacity and anti-cancer drug resistance. B7-H3 has also been found to regulate key metabolic enzymes, promoting the high glycolytic capacity of cancer cells. B7-H3 receptors are still not identified, and little is known about the molecular mechanisms underlying B7-H3 functions. Here, we review the current knowledge on the involvement of B7-H3 in human cancer.
Collapse
Affiliation(s)
- Karine Flem-Karlsen
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Øystein Fodstad
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Caroline E Nunes-Xavier
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| |
Collapse
|
24
|
Testa U, Pelosi E, Castelli G. Endothelial Progenitors in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1263:85-115. [PMID: 32588325 DOI: 10.1007/978-3-030-44518-8_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor vascularization refers to the formation of new blood vessels within a tumor and is considered one of the hallmarks of cancer. Tumor vessels supply the tumor with oxygen and nutrients, required to sustain tumor growth and progression, and provide a gateway for tumor metastasis through the blood or lymphatic vasculature. Blood vessels display an angiocrine capacity of supporting the survival and proliferation of tumor cells through the production of growth factors and cytokines. Although tumor vasculature plays an essential role in sustaining tumor growth, it represents at the same time an essential way to deliver drugs and immune cells to the tumor. However, tumor vasculature exhibits many morphological and functional abnormalities, thus resulting in the formation of hypoxic areas within tumors, believed to represent a mechanism to maintain tumor cells in an invasive state.Tumors are vascularized through a variety of modalities, mainly represented by angiogenesis, where VEGF and other members of the VEGF family play a key role. This has represented the basis for the development of anti-VEGF blocking agents and their use in cancer therapy: however, these agents failed to induce significant therapeutic effects.Much less is known about the cellular origin of vessel network in tumors. Various cell types may contribute to tumor vasculature in different tumors or in the same tumor, such as mature endothelial cells, endothelial progenitor cells (EPCs), or the same tumor cells through a process of transdifferentiation. Early studies have suggested a role for bone marrow-derived EPCs; these cells do not are true EPCs but myeloid progenitors differentiating into monocytic cells, exerting a proangiogenic effect through a paracrine mechanism. More recent studies have shown the existence of tissue-resident endothelial vascular progenitors (EVPs) present at the level of vessel endothelium and their possible involvement as cells of origin of tumor vasculature.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy.
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
25
|
Lustgarten Guahmich N, Farber G, Shafiei S, McNally D, Redmond D, Kallinos E, Stuhlmann H, Dufort D, James D, Blobel CP. Endothelial deletion of ADAM10, a key regulator of Notch signaling, causes impaired decidualization and reduced fertility in female mice. Angiogenesis 2020; 23:443-458. [PMID: 32385775 DOI: 10.1007/s10456-020-09723-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/23/2020] [Indexed: 02/06/2023]
Abstract
During the initiation of pregnancy, the vasculature of the implantation site expands rapidly, yet little is known about this process or its role in fertility. Here, we report that endothelial-specific deletion of a disintegrin and metalloprotease 10 (ADAM10), an essential regulator of Notch signaling, results in severe subfertility in mice. We found that implantation sites develop until 5.5 days post conception (dpc) but are resorbed by 6.5 dpc in A10ΔEC mice. Analysis of the mutant implantation sites showed impaired decidualization and abnormal vascular patterning compared to controls. Moreover, RNA-seq analysis revealed changes in endothelial cell marker expression consistent with defective ADAM10/Notch signaling in samples from A10ΔEC mice, suggesting that this signaling pathways is essential for the physiological function of endometrial endothelial cells during early pregnancy. Our findings raise the possibility that impaired endothelial cell function could be a cause for repeated pregnancy loss (RPL) and infertility in humans.
Collapse
Affiliation(s)
- Nicole Lustgarten Guahmich
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA.,Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY, USA
| | - Gregory Farber
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Shiva Shafiei
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Dylan McNally
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY, USA
| | - Eleni Kallinos
- Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY, USA
| | - Heidi Stuhlmann
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Daniel Dufort
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Daylon James
- Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY, USA
| | - Carl P Blobel
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA. .,Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY, USA. .,Hospital for Special Surgery at Weill Cornell Medicine, 535 East 70th, New York, NY, 10021, USA.
| |
Collapse
|
26
|
Yan J, Bao H, Fan YJ, Jiang ZL, Qi YX, Han Y. Platelet-derived microvesicles promote endothelial progenitor cell proliferation in intimal injury by delivering TGF-β1. FEBS J 2020; 287:5196-5217. [PMID: 32167224 DOI: 10.1111/febs.15293] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/21/2020] [Accepted: 03/11/2020] [Indexed: 12/20/2022]
Abstract
Intimal injury is an early stage of several cardiovascular diseases. Endothelial progenitor cells (EPCs) play a significant role in endothelial repair following vascular injury. Once the intima is damaged, EPCs are mobilized from the bone marrow to the injury site. Meanwhile, the injury to the intimal surface triggers platelet degranulation, aggregation, and adhesion to the damaged endothelium, and exposed collagen stimulates platelet to secrete platelet-derived microvesicles (PMVs). However, the role of PMVs in EPC function during this process remains unknown. In an in vivo study, EPCs and platelets were found to adhere to the injury site in Sprague-Dawley (SD) rat vascular injury model. In vitro, collagen stimulation induced the release of PMVs, and collagen-activated PMVs (ac.PMVs) significantly promoted EPC proliferation. Transforming growth factor-β1 (TGF-β1) content was increased in ac.PMVs. Activated PMVs significantly upregulated Smad3 phosphorylation in EPCs and increased Smad3 nuclear translocation from the cytoplasm. TGF-β1 knockdown ac.PMVs downregulated EPC proliferation. Recombinant TGF-β1 enhanced EPC proliferation. The TGF-β1 inhibitor SB431542 significantly repressed the intracellular signal triggered by ac.PMVs. Furthermore, the Smad3-specific phosphorylation inhibitor SIS3 effectively reversed the cell proliferation induced by ac.PMVs. Smad3 translocated to the nucleus and enhanced EPC proliferation via its downstream genes tenascin C (TNC), CDKN1A, and CDKN2A. r-TGF-β1 promoted reendothelialization and EPC proliferation in vivo. Our data demonstrate that activated PMVs deliver TGF-β1 from collagen-activated platelets to EPCs, which in turn activates Smad3 phosphorylation and regulates TNC, CDKN1A, and CDKN2A expression to promote EPC proliferation, suggesting that PMVs act as a key transporter and a potential therapeutic target for vascular injury.
Collapse
Affiliation(s)
- Jing Yan
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, China
| | - Han Bao
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, China
| | - Yang-Jing Fan
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, China
| | - Zong-Lai Jiang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, China
| | - Ying-Xin Qi
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, China
| | - Yue Han
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, China
| |
Collapse
|