1
|
Singh AK, Dadey DY, Rau MJ, Fitzpatrick J, Shah HK, Saikia M, Townsend R, Thotala D, Hallahan DE, Kapoor V. Blocking the functional domain of TIP1 by antibodies sensitizes cancer to radiation therapy. Biomed Pharmacother 2023; 166:115341. [PMID: 37625322 DOI: 10.1016/j.biopha.2023.115341] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/11/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) and glioblastoma (GB) have poor prognoses. Discovery of new molecular targets is needed to improve therapy. Tax interacting protein 1 (TIP1), which plays a role in cancer progression, is overexpressed and radiation-inducible in NSCLC and GB. We evaluated the effect of an anti-TIP1 antibody alone and in combination with ionizing radiation (XRT) on NSCLC and GB in vitro and in vivo. NSCLC and GB cells were treated with anti-TIP1 antibodies and evaluated for proliferation, colony formation, endocytosis, and cell death. The efficacy of anti-TIP1 antibodies in combination with XRT on tumor growth was measured in mouse models of NSCLC and GB. mRNA sequencing was performed to understand the molecular mechanisms involved in the action of anti-TIP1 antibodies. We found that targeting the functional domain of TIP1 leads to endocytosis of the anti-TIP1 antibody followed by reduced proliferation and increased apoptosis-mediated cell death. Anti-TIP1 antibodies bound specifically (with high affinity) to cancer cells and synergized with XRT to significantly increase cytotoxicity in vitro and reduce tumor growth in mouse models of NSCLC and GB. Importantly, downregulation of cancer survival signaling pathways was found in vitro and in vivo following treatment with anti-TIP1 antibodies. TIP1 is a new therapeutic target for cancer treatment. Antibodies targeting the functional domain of TIP1 exhibited antitumor activity and enhanced the efficacy of radiation both in vitro and in vivo. Anti-TIP1 antibodies interrupt TIP1 function and are effective cancer therapy alone or in combination with XRT in mouse models of human cancer.
Collapse
Affiliation(s)
- Abhay K Singh
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - David Ya Dadey
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael J Rau
- Center for Cellular Imaging, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - James Fitzpatrick
- Center for Cellular Imaging, Washington University School of Medicine in St. Louis, St. Louis, MO, USA; Departments of Cell Biology & Physiology and Neuroscience, Washington University School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO,USA
| | - Harendra K Shah
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Minakshi Saikia
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Reid Townsend
- Department of Medicine, Washington University in St. Louis, St. Louis, MO,USA; Siteman Cancer Center, St. Louis, MO, USA
| | - Dinesh Thotala
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA; Siteman Cancer Center, St. Louis, MO, USA
| | - Dennis E Hallahan
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA; Siteman Cancer Center, St. Louis, MO, USA.
| | - Vaishali Kapoor
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA; Siteman Cancer Center, St. Louis, MO, USA.
| |
Collapse
|
2
|
Wang Y, Gan Y, Dong Y, Zhou J, Zhu E, Yuan H, Li X, Wang B. Tax1 binding protein 3 regulates osteogenic and adipogenic differentiation through inactivating Wnt/β-catenin signalling. J Cell Mol Med 2023; 27:950-961. [PMID: 36892460 PMCID: PMC10064035 DOI: 10.1111/jcmm.17702] [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: 09/21/2021] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 03/10/2023] Open
Abstract
Tax1 binding protein 3 (Tax1bp3) is a PDZ domain-containing protein that is overexpressed in cancer. Previous studies recognized Tax1bp3 as an inhibitor of β-catenin. Till now it is not known whether Tax1bp3 regulates osteogenic and adipogenic differentiation of mesenchymal progenitor cells. In the current study, the data showed that Tax1bp3 was expressed in bone and was increased in the progenitor cells when induced toward osteoblast and adipocyte differentiation. The overexpression of Tax1bp3 in the progenitor cells inhibited osteogenic differentiation and conversely stimulated adipogenic differentiation, and the knockdown of Tax1bp3 affected the differentiation of the progenitor cells oppositely. Ex vivo experiments using the primary calvarial osteoblasts from osteoblast-specific Tax1bp3 knock-in mice also demonstrated the anti-osteogenic and pro-adipogenic function of Tax1bp3. Mechanistic investigations revealed that Tax1bp3 inhibited the activation of canonical Wnt/β-catenin and bone morphogenetic proteins (BMPs)/Smads signalling pathways. Taken together, the current study has provided evidences demonstrating that Tax1bp3 inactivates Wnt/β-catenin and BMPs/Smads signalling pathways and reciprocally regulates osteogenic and adipogenic differentiation from mesenchymal progenitor cells. The inactivation of Wnt/β-catenin signalling may be involved in the reciprocal role of Tax1bp3.
Collapse
Affiliation(s)
- Yi Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien‐I Memorial Hospital & Institute of EndocrinologyTianjin Medical UniversityTianjinChina
| | - Ying Gan
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien‐I Memorial Hospital & Institute of EndocrinologyTianjin Medical UniversityTianjinChina
| | - Yuan Dong
- College of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Jie Zhou
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien‐I Memorial Hospital & Institute of EndocrinologyTianjin Medical UniversityTianjinChina
| | - Endong Zhu
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien‐I Memorial Hospital & Institute of EndocrinologyTianjin Medical UniversityTianjinChina
| | - Hairui Yuan
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien‐I Memorial Hospital & Institute of EndocrinologyTianjin Medical UniversityTianjinChina
| | - Xiaoxia Li
- College of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Baoli Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien‐I Memorial Hospital & Institute of EndocrinologyTianjin Medical UniversityTianjinChina
| |
Collapse
|
3
|
Chen B, Zhou X, Yang L, Zhou H, Meng M, Wu H, Liu Z, Zhang L, Li C. Glioma stem cell signature predicts the prognosis and the response to tumor treating fields treatment. CNS Neurosci Ther 2022; 28:2148-2162. [PMID: 36070228 PMCID: PMC9627385 DOI: 10.1111/cns.13956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Glioma stem cells (GSCs) play an important role in glioma recurrence and chemo-radiotherapy (CRT) resistance. Currently, there is a lack of efficient treatment approaches targeting GSCs. This study aimed to explore the potential personalized treatment of patients with GSC-enriched gliomas. METHODS Single-cell RNA sequencing (scRNA-seq) was used to identify the GSC-related genes. Then, machine learning methods were applied for clustering and validation. The least absolute shrinkage and selection operator (LASSO) and COX regression were used to construct the risk scores. Survival analysis was performed. Additionally, the incidence of chemo-radiotherapy resistance, immunotherapy status, and tumor treating field (TTF) therapy response were evaluated in high- and low-risk scores groups. RESULTS Two GSC clusters exhibited significantly different stemness indices, immune microenvironments, and genomic alterations. Based on GSC clusters, 11-gene GSC risk scores were constructed, which exhibited a high predictive value for prognosis. In terms of therapy, patients with high GSC risk scores had a higher risk of resistance to chemotherapy. TTF therapy can comprehensively inhibit the malignant biological characteristics of the high GSC-risk-score gliomas. CONCLUSION Our study constructed a GSC signature consisting of 11 GSC-specific genes and identified its prognostic value in gliomas. TTF is a promising therapeutic approach for patients with GSC-enriched glioma.
Collapse
Affiliation(s)
- Bo Chen
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina
| | - Xiaoxi Zhou
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina
| | - Liting Yang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina,Hypothalamic‐Pituitary Research Center, Xiangya HospitalCentral South UniversityChangshaChina,Clinical Diagnosis and Therapy Center for Glioma, Xiangya HospitalCentral South UniversityChangshaChina
| | - Hongshu Zhou
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina
| | - Ming Meng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina
| | - Hao Wu
- Department of Neurosurgery, The Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina,Hypothalamic‐Pituitary Research Center, Xiangya HospitalCentral South UniversityChangshaChina,Clinical Diagnosis and Therapy Center for Glioma, Xiangya HospitalCentral South UniversityChangshaChina
| | - Chuntao Li
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina,Hypothalamic‐Pituitary Research Center, Xiangya HospitalCentral South UniversityChangshaChina,Clinical Diagnosis and Therapy Center for Glioma, Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
4
|
Lewis CD, Singh AK, Hsu FF, Thotala D, Hallahan DE, Kapoor V. Targeting a Radiosensitizing Antibody-Drug Conjugate to a Radiation-Inducible Antigen. Clin Cancer Res 2021; 27:3224-3233. [PMID: 34074654 DOI: 10.1158/1078-0432.ccr-20-1725] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 02/14/2021] [Accepted: 04/07/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE We recently discovered that anti-TIP1 antibody activates endocytosis in cancer cells, which facilitates retention of antibody and dissociation of a conjugated drug. To improve the pharmacokinetics and cancer specificity of radiosensitizing drugs, we utilized antibody-drug conjugates (ADCs) that bind specifically to radiation-inducible antigen, TIP1, on non-small cell lung cancer (NSCLC). This approach exploits the long circulation time of antibodies to deliver a radiosensitizing drug to cancer each day during radiotherapy. EXPERIMENTAL DESIGN Antibodies to TIP1 were prioritized based on affinity, cancer-specific binding, and internalization. The lead antibody, 7H5, was conjugated with a cytotoxic drug MMAE because of its ability to radiosensitize cancer. Cytotoxicity, colony formation, and tumor growth studies were performed with 7H5-VcMMAE in combination with radiation. RESULTS 7H5 showed a high affinity to recombinant TIP1 protein and radiation-inducible TIP1 on the cancer cell surface. 7H5 undergoes endocytosis in NSCLC cells in vitro. We obtained an average drug-to-antibody ratio (DAR) of 4.25 for 7H5-VcMMAE. A 70% reduction in viable cells was observed following 7H5-VcMMAE treatment compared with 7H5 alone in both A549 and H1299 cells. 7H5-VcMMAE sensitized NSCLC cells to radiation, thereby significantly decreasing the surviving fraction. The ADC combined with radiation showed a prolonged delay in tumor growth and improved survival in A549 and H1299 tumor models. CONCLUSIONS Targeting radiation-inducible TIP1 with a radiosensitizing ADC is a promising strategy to enhance the therapeutic efficacy of NSCLC. This novel approach of targeting with ADCs to radiation-inducible antigens will lead to clinical trials in lung cancer patients treated with radiotherapy.
Collapse
Affiliation(s)
- Calvin D Lewis
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
- Department of Radiation Oncology, University of Iowa, Iowa City, Iowa
| | - Abhay K Singh
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Fong-Fu Hsu
- Division of Endocrinology, Metabolism and Lipid Research, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Dinesh Thotala
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri
| | - Dennis E Hallahan
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri
| | - Vaishali Kapoor
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri.
| |
Collapse
|
5
|
Quintero M, Liu S, Xia Y, Huang Y, Zou Y, Li G, Hu L, Singh N, Blumberg R, Cai Y, Xu H, Li H. Cdk5rap3 is essential for intestinal Paneth cell development and maintenance. Cell Death Dis 2021; 12:131. [PMID: 33504792 PMCID: PMC7841144 DOI: 10.1038/s41419-021-03401-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
Intestinal Paneth cells are professional exocrine cells that play crucial roles in maintenance of homeostatic microbiome, modulation of mucosal immunity, and support for stem cell self-renewal. Dysfunction of these cells may lead to the pathogenesis of human diseases such as inflammatory bowel disease (IBD). Cdk5 activator binding protein Cdk5rap3 (also known as C53 and LZAP) was originally identified as a binding protein of Cdk5 activator p35. Although previous studies have indicated its involvement in a wide range of signaling pathways, the physiological function of Cdk5rap3 remains largely undefined. In this study, we found that Cdk5rap3 deficiency resulted in very early embryonic lethality, indicating its indispensable role in embryogenesis. To further investigate its function in the adult tissues and organs, we generated intestinal epithelial cell (IEC)-specific knockout mouse model to examine its role in intestinal development and tissue homeostasis. IEC-specific deletion of Cdk5rap3 led to nearly complete loss of Paneth cells and increased susceptibility to experimentally induced colitis. Interestingly, Cdk5rap3 deficiency resulted in downregulation of key transcription factors Gfi1 and Sox9, indicating its crucial role in Paneth cell fate specification. Furthermore, Cdk5rap3 is highly expressed in mature Paneth cells. Paneth cell-specific knockout of Cdk5rap3 caused partial loss of Paneth cells, while inducible acute deletion of Cdk5rap3 resulted in disassembly of the rough endoplasmic reticulum (RER) and abnormal zymogen granules in the mature Paneth cells, as well as loss of Paneth cells. Together, our results provide definitive evidence for the essential role of Cdk5rap3 in Paneth cell development and maintenance.
Collapse
Affiliation(s)
- Michaela Quintero
- Department of Biochemistry & Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Siyang Liu
- Department of Biochemistry & Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Yanhua Xia
- Faculty of Basic Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Yonghong Huang
- Faculty of Basic Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Yi Zou
- Department of Metabolic Endocrinology, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ge Li
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Ling Hu
- Department of Metabolic Endocrinology, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Nagendra Singh
- Department of Biochemistry & Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Richard Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yafei Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Hong Xu
- Faculty of Basic Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Honglin Li
- Department of Biochemistry & Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
6
|
Köster F, Sauer L, Hoellen F, Ribbat-Idel J, Bräutigam K, Rody A, Banz-Jansen C. PSMD9 expression correlates with recurrence after radiotherapy in patients with cervical cancer. Oncol Lett 2020; 20:581-588. [PMID: 32565983 PMCID: PMC7285846 DOI: 10.3892/ol.2020.11622] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/31/2020] [Indexed: 11/25/2022] Open
Abstract
In the current retrospective cohort study, the expression of the Proteasome 26S non-ATPase Subunit 9 (PSMD9) was investigated in 102 patients with cervical cancer. The rat homologue of PSMD9, Bridge-1, was identified as a binding protein of the transcription factors PDX-1 and E-12 via its PDZ-domain. The aim of the current study was to evaluate the prognostic or predictive value of PSMD9 expression as a biomarker for patients with cervical cancer. Tissue microarrays were constructed from formalin-fixed paraffin-embedded tissue specimens of cervical cancer and peritumoral stroma after hysterectomy and a Bridge-1 antibody was used to perform immunohistochemistry. The immunoreactions were analyzed using an immunoreactive score, which evaluated the number of positive cells as well as their intensity of PSMD9 expression. A misinterpretation of statistically significant results after multiple testing was controlled by the false discovery rate correction using the algorithm of Benjamini and Hochberg. All tumor tissues and almost all peritumoral stroma tissues expressed PSMD9. The PSMD9 expression in tumor tissues was significantly higher compared with the peritumoral stroma. PSMD9 expression correlated significantly with the expression of the proliferation marker MIB-1. Patients with stronger PSMD9 expression tended to exhibit a higher odds ratio for the recurrence of the disease in all patients (n=102) as well as in the subgroup of 47 patients having received a combined chemoradiotherapy following hysterectomy. In the group of 62 patients having that received radiotherapy following hysterectomy, which included the chemoradiotherapy patients, a higher PSMD9 expression significantly increased the odds for a recurrence to 1.983-fold even after FDR correction (P=0.0304). In conclusion, PSMD9 was indicated to be overexpressed in tumor tissues and associated with tumor cell proliferation. Therefore, PSMD9 may be useful as a tumor marker. Furthermore, increased PSMD9 overexpression may be used to predict resistance against radiation.
Collapse
Affiliation(s)
- Frank Köster
- Department of Gynecology and Obstetrics, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany
| | - Lisa Sauer
- Department of Gynecology and Obstetrics, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany
| | - Friederike Hoellen
- Department of Gynecology and Obstetrics, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany
| | - Julika Ribbat-Idel
- Institute of Pathology, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany
| | - Karen Bräutigam
- Department of Gynecology and Obstetrics, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany
| | - Achim Rody
- Department of Gynecology and Obstetrics, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany
| | - Constanze Banz-Jansen
- Department of Gynecology and Obstetrics, Evangelisches Krankenhaus Bethel, D-33617 Bielefeld, Germany
| |
Collapse
|
7
|
Lim SH, Li CH, Jeong YI, Jang WY, Choi JM, Jung S. Enhancing Radiotherapeutic Effect With Nanoparticle-Mediated Radiosensitizer Delivery Guided By Focused Gamma Rays In Lewis Lung Carcinoma-Bearing Mouse Brain Tumor Models. Int J Nanomedicine 2019; 14:8861-8874. [PMID: 32009784 PMCID: PMC6859088 DOI: 10.2147/ijn.s227894] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/25/2019] [Indexed: 12/26/2022] Open
Abstract
Background Targeting radiosensitizer-incorporated nanoparticles to a tumor could allow for less normal tissue toxicity with more efficient drug release, thus improving the efficacy and safety of radiation treatment. The aim of this study was to improve tumor-specific delivery and bioavailability of a nanoparticle-mediated radiosensitizer in mouse brain tumor models. Methods A pH-sensitive nanoparticle, chitoPEGAcHIS, was conjugated to recombinant peptide HVGGSSV that could bind to tax-interaction protein 1 (TIP-1) as a radiation-inducible receptor. Then the c-Jun N-terminal kinase (JNK) inhibitor, SP600125 was incorporated into this copolymer to fabricate a HVGGSSV-chitoPEGAcHIS-SP600125 (HVSP-NP) nanoradiosensitizer. In vitro and in vivo radiation treatment were performed using a Gamma Knife unit. The tumor targetability of HVSP-NP was estimated by optical bioluminescence. Synergistic therapeutic effects of radiation treatment and HVSP-NP were investigated in Lewis lung carcinoma (LLC) cell-bearing mouse brain tumor models. Results The SP600125 JNK inhibitor effectively reduced DNA damage repair to irradiated LLC cells. A pH sensitivity assay indicated that HVSP-NP swelled at acidic pH and increased in diameter, and its release rate gradually increased. Optical bioluminescence assay showed that radiation induced TIP-1 expression in mouse brain tumor and that the nanoradiosensitizer selectively targeted irradiated tumors. Radiation treatment with HVSP-NP induced greater apoptosis and significantly inhibited tumor growth compared to radiation alone. Conclusion As a novel nanoradiosensitizer, HVSP-NP was found to be able to selectively target irradiated tumors and significantly increase tumor growth delay in LLC-bearing mouse brain tumor models. This research shows that delivering a pH-sensitive nanoradiosensitizer to a brain tumor in which TIP-1 is induced by radiation can result in improved radiosensitizer-release in an acidic microenvironment of tumor tissue and in created synergistic effects in radiation treatment.
Collapse
Affiliation(s)
- Sa-Hoe Lim
- Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Korea.,Brain Tumor Research Laboratory, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Chun-Hao Li
- Department of Neurosurgery, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, People's Republic of China
| | - Young-Il Jeong
- Biomedical Research Institute, Pusan National University Hospital, Pusan 602-739, Republic of Korea
| | - Woo-Youl Jang
- Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Korea.,Brain Tumor Research Laboratory, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Jin-Myung Choi
- Brain Tumor Research Laboratory, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Shin Jung
- Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Korea.,Brain Tumor Research Laboratory, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital, Hwasun, Korea
| |
Collapse
|
8
|
Yan H, Kapoor V, Nguyen K, Akers WJ, Li H, Scott J, Laforest R, Rogers B, Thotala D, Hallahan D. Anti-tax interacting protein-1 (TIP-1) monoclonal antibody targets human cancers. Oncotarget 2017; 7:43352-43362. [PMID: 27270318 PMCID: PMC5190028 DOI: 10.18632/oncotarget.9713] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/13/2016] [Indexed: 02/04/2023] Open
Abstract
Radiation-inducible neo-antigens are proteins expressed on cancer cell surface after exposure to ionizing radiation (IR). These neo-antigens provide opportunities to specifically target cancers while sparing normal tissues. Tax interacting protein-1 (TIP-1) is induced by irradiation and is translocated to the surface of cancer cells. We have developed a monoclonal antibody, 2C6F3, against TIP-1. Epitope mapping revealed that 2C6F3 binds to the QPVTAVVQRV epitope of the TIP-1 protein. 2C6F3 binds to the surface of lung cancer (A549, LLC) and glioma (D54, GL261) cell lines. 2C6F3 binds specifically to TIP-1 and ELISA analysis showed that unconjugated 2C6F3 efficiently blocked binding of radiolabeled 2C6F3 to purified TIP-1 protein. To study in vivo tumor binding, we injected near infrared (NIR) fluorochrome-conjugated 2C6F3 via tail vein in mice bearing subcutaneous LLC and GL261 heterotopic tumors. The NIR images indicated that 2C6F3 bound specifically to irradiated LLC and GL261 tumors, with little or no binding in un-irradiated tumors. We also determined the specificity of 2C6F3 to bind tumors in vivo using SPECT/CT imaging. 2C6F3 was conjugated with diethylene triamine penta acetic acid (DTPA) chelator and radiolabeled with 111Indium (111In). SPECT/CT imaging revealed that 111In-2C6F3 bound more to the irradiated LLC tumors compared to un-irradiated tumors. Furthermore, injection of DTPA-2C6F3 labeled with the therapeutic radioisotope, 90Y, (90Y-DTPA-2C6F3) significantly delayed LLC tumor growth. 2C6F3 mediated antibody dependent cell-mediated cytotoxicity (ADCC) and antibody dependent cell-mediated phagocytosis (ADCP) in vitro. In conclusion, the monoclonal antibody 2C6F3 binds specifically to TIP-1 on cancer and radio-immunoconjugated 2C6F3 improves tumor control.
Collapse
Affiliation(s)
- Heping Yan
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Vaishali Kapoor
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kim Nguyen
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Walter J Akers
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Hua Li
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jalen Scott
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Richard Laforest
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Buck Rogers
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Dinesh Thotala
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA.,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Dennis Hallahan
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri, USA.,Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA.,Siteman Cancer Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
9
|
Mohanty S, Ovee M, Banerjee M. PDZ Domain Recognition: Insight from Human Tax-Interacting Protein 1 (TIP-1) Interaction with Target Proteins. BIOLOGY 2015; 4:88-103. [PMID: 25665168 PMCID: PMC4381219 DOI: 10.3390/biology4010088] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/22/2015] [Accepted: 01/23/2015] [Indexed: 11/16/2022]
Abstract
Cellular signaling is primarily directed via protein-protein interactions. PDZ (PSD-95/Discs large/ZO-1 homologous) domains are well known protein-protein interaction modules involved in various key signaling pathways. Human Tax-interacting protein 1 (TIP-1), also known as glutaminase interaction protein (GIP), is a Class I PDZ domain protein that recognizes the consensus binding motif X-S/T-X-V/I/L-COOH of the C-terminus of its target proteins. We recently reported that TIP-1 not only interacts via the C-terminus of its target partner proteins but also recognizes an internal motif defined by the consensus sequence S/T-X-V/L-D in the target protein. Identification of new target partners containing either a C-terminal or internal recognition motif has rapidly expanded the TIP-1 protein interaction network. TIP-1 being composed solely of a single PDZ domain is unique among PDZ containing proteins. Since it is involved in many important signaling pathways, it is a possible target for drug design. In this mini review, we have discussed human TIP-1, its structure, mechanism of function, its interactions with target proteins containing different recognition motifs, and its involvement in human diseases. Understanding the molecular mechanisms of TIP-1 interactions with distinct target partners and their role in human diseases will be useful for designing novel therapeutics.
Collapse
Affiliation(s)
- Smita Mohanty
- Department of Chemistry, Oklahoma State University, Stillwater, OK 74078, USA.
| | - Mohiuddin Ovee
- Department of Chemistry, Oklahoma State University, Stillwater, OK 74078, USA.
| | - Monimoy Banerjee
- Department of Chemistry, Oklahoma State University, Stillwater, OK 74078, USA.
| |
Collapse
|
10
|
Wang H, Han M, Whetsell W, Wang J, Rich J, Hallahan D, Han Z. Tax-interacting protein 1 coordinates the spatiotemporal activation of Rho GTPases and regulates the infiltrative growth of human glioblastoma. Oncogene 2013; 33:1558-69. [PMID: 23563176 PMCID: PMC3965267 DOI: 10.1038/onc.2013.97] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 01/17/2013] [Accepted: 02/04/2013] [Indexed: 12/28/2022]
Abstract
PDZ domains represent one group of the major structural units that mediate protein interactions in intercellular contact, signal transduction and assembly of biological machineries. TIP-1 protein is composed of a single PDZ domain that distinguishes TIP-1 from other PDZ domain proteins that more often contain multiple protein domains and function as scaffolds for protein complex assembly. However, the biological functions of TIP-1, especially in cell transformation and tumor progression, are still controversial as observed in a variety of cell types. In this study, we have identified ARHGEF7, a guanine nucleotide exchange factor (GEF) for Rho GTPases, as one novel TIP-1 interacting protein in human glioblastoma cells. We found that the presence of TIP-1 protein is essential to the intracellular redistribution of ARHGEF7 and rhotekin, one Rho effector, and the spatiotemporally coordinated activation of Rho GTPases (RhoA, Cdc42 and Rac1) in migrating glioblastoma cells. TIP-1 knockdown resulted in both aberrant localization of ARHGEF7 and rhotekin, as well as abnormal activation of Rho GTPases that was accompanied with impaired motility of glioblastoma cells. Furthermore, TIP-1 knockdown suppressed tumor cell dispersal in orthotopic glioblastoma murine models. We also observed high levels of TIP-1 expression in human glioblastoma specimens, and the elevated TIP-1 levels are associated with advanced staging and poor prognosis in glioma patients. Although more studies are needed to further dissect the mechanism(s) by which TIP-1 modulates the intracellular redistribution and activation of Rho GTPases, this study suggests that TIP-1 holds potential as both a prognostic biomarker and a therapeutic target of malignant gliomas.
Collapse
Affiliation(s)
- H Wang
- 1] Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA [2] Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - M Han
- 1] Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA [2] Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Science, Kunming, China [3] Graduate School, Chinese Academy of Sciences, Beijing, China
| | - W Whetsell
- Department of Pathology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - J Wang
- 1] Department of Neurological Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA [2] Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - J Rich
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - D Hallahan
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Z Han
- 1] Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA [2] Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA [3] Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|