1
|
Dabour MS, George MY, Grant MKO, Zordoky BN. Canagliflozin differentially modulates carfilzomib-induced endoplasmic reticulum stress in multiple myeloma and endothelial cells. Arch Toxicol 2024:10.1007/s00204-024-03913-0. [PMID: 39645617 DOI: 10.1007/s00204-024-03913-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/20/2024] [Indexed: 12/09/2024]
Abstract
Carfilzomib (CFZ), a second-generation proteasome inhibitor, is a key treatment for multiple myeloma (MM), but its use is associated with significant cardiovascular adverse events (CVAEs), including heart failure and hypertension. Endothelial dysfunction is believed to contribute to these CVAEs. Building on our previous findings that CFZ induces endothelial toxicity and that canagliflozin protects against CFZ-induced endothelial apoptosis, this study aimed to evaluate CFZ-induced endoplasmic reticulum (ER) stress and autophagy in endothelial and MM cells, as well as the impact of canagliflozin on these processes and its impact on the anticancer effects of CFZ in MM cells. Endothelial cells (HUVECs and EA.hy926) and multiple myeloma cells (RPMI8226) were treated with 0.5 µM CFZ, either alone or in combination with canagliflozin (5-20 µM), to assess the effects on ER stress and autophagy in both cell types. CFZ induced ER stress in endothelial and MM cells. In endothelial cells, canagliflozin mitigated CFZ-induced markers of ER stress, while unexpectedly upregulating CFZ-induced CHOP. Whereas, in MM cells, canagliflozin did not alter CFZ-induced ER stress, but instead further upregulated CFZ-induced ATF-4. In addition, CFZ induced autophagy in endothelial cells while inhibiting it in MM cells. Canagliflozin abrogated CFZ-induced autophagy in endothelial cells. In striking contrast to its effects in endothelial cells, canagliflozin enhanced the cytotoxic effects of CFZ in MM cells. Intriguingly, in an innovative co-culture system, canagliflozin enhanced CFZ-induced apoptosis in MM cells while protecting endothelial cells. These findings underscore the dual role of canagliflozin in reducing CFZ-induced endothelial toxicity, while enhancing its cytotoxic effect in MM.
Collapse
Affiliation(s)
- Mohamed S Dabour
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, 55455, USA
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Mina Y George
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, 55455, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Marianne K O Grant
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, 55455, USA
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, 55455, USA.
| |
Collapse
|
2
|
Lin HT, Takagi M, Kubara K, Yamazaki K, Michikawa F, Okumura T, Naruto T, Morio T, Miyazaki K, Taniguchi H, Otsu M. Monoallelic KRAS (G13C) mutation triggers dysregulated expansion in induced pluripotent stem cell-derived hematopoietic progenitor cells. Stem Cell Res Ther 2024; 15:106. [PMID: 38627844 PMCID: PMC11021011 DOI: 10.1186/s13287-024-03723-2] [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: 01/25/2023] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Although oncogenic RAS mutants are thought to exert mutagenic effects upon blood cells, it remains uncertain how a single oncogenic RAS impacts non-transformed multipotent hematopoietic stem or progenitor cells (HPCs). Such potential pre-malignant status may characterize HPCs in patients with RAS-associated autoimmune lymphoproliferative syndrome-like disease (RALD). This study sought to elucidate the biological and molecular alterations in human HPCs carrying monoallelic mutant KRAS (G13C) with no other oncogene mutations. METHODS We utilized induced pluripotent stem cells (iPSCs) derived from two unrelated RALD patients. Isogenic HPC pairs harboring either wild-type KRAS or monoallelic KRAS (G13C) alone obtained following differentiation enabled reliable comparative analyses. The compound screening was conducted with an established platform using KRAS (G13C) iPSCs and differentiated HPCs. RESULTS Cell culture assays revealed that monoallelic KRAS (G13C) impacted both myeloid differentiation and expansion characteristics of iPSC-derived HPCs. Comprehensive RNA-sequencing analysis depicted close clustering of HPC samples within the isogenic group, warranting that comparative studies should be performed within the same genetic background. When compared with no stimulation, iPSC-derived KRAS (G13C)-HPCs showed marked similarity with the wild-type isogenic control in transcriptomic profiles. After stimulation with cytokines, however, KRAS (G13C)-HPCs exhibited obvious aberrant cell-cycle and apoptosis responses, compatible with "dysregulated expansion," demonstrated by molecular and biological assessment. Increased BCL-xL expression was identified amongst other molecular changes unique to mutant HPCs. With screening platforms established for therapeutic intervention, we observed selective activity against KRAS (G13C)-HPC expansion in several candidate compounds, most notably in a MEK- and a BCL-2/BCL-xL-inhibitor. These two compounds demonstrated selective inhibitory effects on KRAS (G13C)-HPCs even with primary patient samples when combined. CONCLUSIONS Our findings indicate that a monoallelic oncogenic KRAS can confer dysregulated expansion characteristics to non-transformed HPCs, which may constitute a pathological condition in RALD hematopoiesis. The use of iPSC-based screening platforms will lead to discovering treatments that enable selective inhibition of RAS-mutated HPC clones.
Collapse
Affiliation(s)
- Huan-Ting Lin
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan.
| | - Masatoshi Takagi
- Department of Pediatrics and Developmental Biology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan
| | - Kenji Kubara
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, 300-2635, Japan
| | - Kazuto Yamazaki
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, 300-2635, Japan
| | - Fumiko Michikawa
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, 300-2635, Japan
| | - Takashi Okumura
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Takuya Naruto
- Department of Pediatrics and Developmental Biology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan
| | - Koji Miyazaki
- Department of Transfusion and Cell Transplantation, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hideki Taniguchi
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
- Department of Regenerative Medicine, Graduate School of Medicine, Yokohama City University, Yokohama, Kanagawa, 236-0004, Japan
| | - Makoto Otsu
- Department of Transfusion and Cell Transplantation, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan.
- Division of Hematology, Department of Medical Laboratory Sciences, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0373, Japan.
| |
Collapse
|
3
|
Shevchenko JA, Nazarov KV, Alshevskaya AA, Sennikov SV. Erythroid Cells as Full Participants in the Tumor Microenvironment. Int J Mol Sci 2023; 24:15141. [PMID: 37894821 PMCID: PMC10606658 DOI: 10.3390/ijms242015141] [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: 09/12/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
The tumor microenvironment is an important factor that can determine the success or failure of antitumor therapy. Cells of hematopoietic origin are one of the most important mediators of the tumor-host interaction and, depending on the cell type and functional state, exert pro- or antitumor effects in the tumor microenvironment or in adjacent tissues. Erythroid cells can be full members of the tumor microenvironment and exhibit immunoregulatory properties. Tumor growth is accompanied by the need to obtain growth factors and oxygen, which stimulates the appearance of the foci of extramedullary erythropoiesis. Tumor cells create conditions to maintain the long-term proliferation and viability of erythroid cells. In turn, tumor erythroid cells have a number of mechanisms to suppress the antitumor immune response. This review considers current data on the existence of erythroid cells in the tumor microenvironment, formation of angiogenic clusters, and creation of optimal conditions for tumor growth. Despite being the most important life-support function of the body, erythroid cells support tumor growth and do not work against it. The study of various signaling mechanisms linking tumor growth with the mobilization of erythroid cells and the phenotypic and functional differences between erythroid cells of different origin allows us to identify potential targets for immunotherapy.
Collapse
Affiliation(s)
- Julia A. Shevchenko
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (J.A.S.); (K.V.N.)
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution, Ministry of Health of the Russian Federation, Higher Education I.M. Sechenov First Moscow State Medical University, Sechenov University, 119048 Moscow, Russia;
| | - Kirill V. Nazarov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (J.A.S.); (K.V.N.)
| | - Alina A. Alshevskaya
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution, Ministry of Health of the Russian Federation, Higher Education I.M. Sechenov First Moscow State Medical University, Sechenov University, 119048 Moscow, Russia;
| | - Sergey V. Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (J.A.S.); (K.V.N.)
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution, Ministry of Health of the Russian Federation, Higher Education I.M. Sechenov First Moscow State Medical University, Sechenov University, 119048 Moscow, Russia;
| |
Collapse
|
4
|
Bozorgmehr N, Okoye I, Mashhouri S, Lu J, Koleva P, Walker J, Elahi S. CD71 + erythroid cells suppress T-cell effector functions and predict immunotherapy outcomes in patients with virus-associated solid tumors. J Immunother Cancer 2023; 11:jitc-2022-006595. [PMID: 37236637 DOI: 10.1136/jitc-2022-006595] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer. However, only a portion of patients respond to such treatments. Therefore, it remains a prevailing clinical need to identify factors associated with acquired resistance or lack of response to ICIs. We hypothesized that the immunosuppressive CD71+ erythroid cells (CECs) within the tumor and/or distant 'out-of-field' may impair antitumor response. METHODS We studied 38 patients with cancer through a phase II clinical trial investigating the effects of oral valproate combined with avelumab (anti-programmed death-ligand 1 (PD-L1)) in virus-associated solid tumors (VASTs). We quantified the frequency/functionality of CECs in blood and biopsies of patients. Also, we established an animal model of melanoma (B16-F10) to investigate the possible effects of erythropoietin (EPO) treatment on anti-PD-L1 therapy. RESULTS We found a substantial expansion of CECs in the blood of patients with VAST compared with healthy controls. We noted that the frequency of CECs in circulation was significantly higher at the baseline and throughout the study in non-responders versus responders to PD-L1 therapy. Moreover, we observed that CECs in a dose-dependent manner suppress effector functions of autologous T cells in vitro. The subpopulation of CD45+CECs appears to have a more robust immunosuppressive property compared with their CD45- counterparts. This was illustrated by a stronger expression of reactive oxygen species, PD-L1/PD-L2, and V-domain Ig suppressor of T-cell activation in this subpopulation. Lastly, we found a higher frequency of CECs in the blood circulation at the later cancer stage and their abundance was associated with anemia, and a poor response to immunotherapy. Finally, we report the expansion of CECs in the spleen and tumor microenvironment of mice with melanoma. We found that although CECs in tumor-bearing mice secret artemin, this was not the case for VAST-derived CECs in humans. Notably, our results imply that EPO, a frequently used drug for anemia treatment in patients with cancer, may promote the generation of CECs and subsequently abrogates the therapeutic effects of ICIs (eg, anti-PD-L1). CONCLUSIONS Our results demonstrate that anemia by the expansion of CECs may enhance cancer progression. Notably, measuring the frequency of CECs may serve as a valuable biomarker to predict immunotherapy outcomes.
Collapse
Affiliation(s)
- Najmeh Bozorgmehr
- Department of Dentistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Isobel Okoye
- Department of Dentistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Siavash Mashhouri
- Department of Dentistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Julia Lu
- Department of Dentistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Petya Koleva
- Department of Dentistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - John Walker
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shokrollah Elahi
- Department of Dentistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Pecar G, Liu S, Hooda J, Atkinson JM, Oesterreich S, Lee AV. RET signaling in breast cancer therapeutic resistance and metastasis. Breast Cancer Res 2023; 25:26. [PMID: 36918928 PMCID: PMC10015789 DOI: 10.1186/s13058-023-01622-7] [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: 05/23/2022] [Accepted: 02/16/2023] [Indexed: 03/15/2023] Open
Abstract
RET, a single-pass receptor tyrosine kinase encoded on human chromosome 10, is well known to the field of developmental biology for its role in the ontogenesis of the central and enteric nervous systems and the kidney. In adults, RET alterations have been characterized as drivers of non-small cell lung cancer and multiple neuroendocrine neoplasms. In breast cancer, RET signaling networks have been shown to influence diverse functions including tumor development, metastasis, and therapeutic resistance. While RET is known to drive the development and progression of multiple solid tumors, therapeutic agents selectively targeting RET are relatively new, though multiple multi-kinase inhibitors have shown promise as RET inhibitors in the past; further, RET has been historically neglected as a potential therapeutic co-target in endocrine-refractory breast cancers despite mounting evidence for a key pathologic role and repeated description of a bi-directional relationship with the estrogen receptor, the principal driver of most breast tumors. Additionally, the recent discovery of RET enrichment in breast cancer brain metastases suggests a role for RET inhibition specific to advanced disease. This review assesses the status of research on RET in breast cancer and evaluates the therapeutic potential of RET-selective kinase inhibitors across major breast cancer subtypes.
Collapse
Affiliation(s)
- Geoffrey Pecar
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, The Assembly, Room 2051, 5051 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Simeng Liu
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA
- School of Medicine, Tsinghua University, Beijing, China
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jagmohan Hooda
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, The Assembly, Room 2051, 5051 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Jennifer M Atkinson
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, The Assembly, Room 2051, 5051 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, The Assembly, Room 2051, 5051 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Adrian V Lee
- Women's Cancer Research Center, UPMC Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, The Assembly, Room 2051, 5051 Centre Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
6
|
JHDM1D-AS1-driven inhibition of miR-940 releases ARTN expression to induce breast carcinogenesis. Clin Transl Oncol 2023:10.1007/s12094-023-03102-y. [PMID: 36862282 DOI: 10.1007/s12094-023-03102-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/23/2023] [Indexed: 03/03/2023]
Abstract
INTRODUCTION As ceRNA network of long non-coding RNA (lncRNA)-microRNA (miR)-messenger RNAs (mRNA) can be predicted on the basis of bioinformatics tools, we are now one step closer to deeper understanding carcinogenic mechanisms. In this study, we clarified the mechanistic understanding of JHDM1D-AS1-miR-940-ARTN ceRNA network in the development of breast cancer (BC). MATERIALS AND METHODS The lncRNA-miRNA-mRNA interaction of interest was predicted by in silico analysis and identified by conducting RNA immunoprecipitation, RNA pull-down and luciferase assays. The expression patterns of JHDM1D-AS1, miR-940 and ARTN in BC cells were altered by lentivirus infection and plasmid transfection for functional assays on the biological properties of BC cells. Finally, the tumorigenic and metastatic abilities of BC cells were assessed in vivo. RESULTS JHDM1D-AS1 was highly expressed, while miR-940 was poorly expressed in BC tissues and cells. JHDM1D-AS1 could competitively bind to miR-940, whereby promoting the malignant behaviors of BC cells. Furthermore, ARTN was identified as a target gene of miR-940. Through targeting ARTN, miR-940 exerted a tumor-suppressive role. In vivo experiments further confirmed that JHDM1D-AS1 enhanced the tumorigenesis and metastasis through up-regulation of ARTN. CONCLUSIONS Taken together, our study demonstrated the involvement of ceRNA network JHDM1D-AS1-miR-940-ARTN in the progression of BC, which highlighted promising therapeutic targets for BC treatment.
Collapse
|
7
|
Zhu M, Zhou L, Fu J, Wang Y, Xu X, Wu J, Kong X, Li J, Zhou Z, Zhou H. Artemin Promotes the Migration and Invasion of Cervical Cancer Cells through AKT/mTORC1 Signaling. JOURNAL OF ONCOLOGY 2022; 2022:3332485. [PMID: 36471885 PMCID: PMC9719425 DOI: 10.1155/2022/3332485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/17/2022] [Accepted: 11/10/2022] [Indexed: 08/25/2024]
Abstract
BACKGROUND The neurotrophic factor Artemin (ARTN) is involved in tumor proliferation and metastasis. Nonetheless, ARTN's significance in cervical cancer (CC) has not been studied. In our study, we propose to investigate the biological function of ARTN in CC as well as its particular regulatory mechanism. METHODS Immunohistochemistry (IHC) was used to examine the degree of ARTN protein expression in CC patient tissue. Real-time PCR and Western blotting were performed to reveal related genes' levels in CC cells. The CCK-8 test, the colony formation assay, the wound-healing assay, and the transwell assay were utilized to determine the proliferation, migration, and invasion capabilities, respectively. To generate lung metastasis models, stable ARTN-expressing SiHa cells were injected into the caudal tail vein of mice. IHC was used to examine the protein levels in CC mice model tissues. RESULTS ARTN was overexpressed in CC tissues relative to normal cervical tissues and linked positively with lymph node metastases (P=0.012) and recurrence (P=0.015) in CC patients. In vitro, ARTN overexpression promoted the proliferation, invasion, and migration of CC cells. In contrast, the consequences of depleting endogenous ARTN were the opposite. Moreover, overexpression of ARTN increased lung metastasis of CC cells in vivo and shortened the lifespan of mice models. In addition, ARTN overexpression significantly enhanced AKT phosphorylation on Ser473 and mTOR phosphorylation on Ser2448 and promoted the epithelial-mesenchymal transition (EMT) cascade. In addition, rapamycin, a selective inhibitor of mTORC1, might rescue the EMT phenotype caused by ARTN. CONCLUSION Our findings suggested that ARTN may enhance CC metastasis through the AKT/mTORC1 pathway. ARTN is anticipated to be a novel potential therapeutic target for the treatment of CC metastases.
Collapse
Affiliation(s)
- Mengjing Zhu
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Ling Zhou
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jian Fu
- Department of Gynecology, Suqian People's Hospital of Nanjing Drum Tower Hospital Group, Suqian 223800, China
| | - Yijin Wang
- Department of Gynecology, Medical School of Southeast University Nanjing Drum Tower Hospital, Nanjing 210008, China
| | - Xiaofeng Xu
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jun Wu
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xiangyi Kong
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jian Li
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Zhe Zhou
- Department of Gynecology, Lianyungang Maternal and Child Health Hospital, Lianyungang 222006, China
| | - Huaijun Zhou
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
8
|
Srivastava A, Tommasi C, Sessions D, Mah A, Bencomo T, Garcia JM, Jiang T, Lee M, Shen JY, Seow LW, Nguyen A, Rajapakshe K, Coarfa C, Tsai KY, Lopez-Pajares V, Lee CS. MAB21L4 Deficiency Drives Squamous Cell Carcinoma via Activation of RET. Cancer Res 2022; 82:3143-3157. [PMID: 35705526 PMCID: PMC9444977 DOI: 10.1158/0008-5472.can-22-0047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/02/2022] [Accepted: 06/10/2022] [Indexed: 02/04/2023]
Abstract
Epithelial squamous cell carcinomas (SCC) most commonly originate in the skin, where they display disruptions in the normally tightly regulated homeostatic balance between keratinocyte proliferation and terminal differentiation. We performed a transcriptome-wide screen for genes of unknown function that possess inverse expression patterns in differentiating keratinocytes compared with cutaneous SCC (cSCC), leading to the identification of MAB21L4 (C2ORF54) as an enforcer of terminal differentiation that suppresses carcinogenesis. Loss of MAB21L4 in human cSCC organoids increased expression of RET to enable malignant progression. In addition to transcriptional upregulation of RET, deletion of MAB21L4 preempted recruitment of the CacyBP-Siah1 E3 ligase complex to RET and reduced its ubiquitylation. In SCC organoids and in vivo tumor models, genetic disruption of RET or selective inhibition of RET with BLU-667 (pralsetinib) suppressed SCC growth while inducing concomitant differentiation. Overall, loss of MAB21L4 early during SCC development blocks differentiation by increasing RET expression. These results suggest that targeting RET activation is a potential therapeutic strategy for treating SCC. SIGNIFICANCE Downregulation of RET mediated by MAB21L4-CacyBP interaction is required to induce epidermal differentiation and suppress carcinogenesis, suggesting RET inhibition as a potential therapeutic approach in squamous cell carcinoma.
Collapse
Affiliation(s)
- Ankit Srivastava
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA.,Department of Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institute, Stockholm 17177, Sweden
| | - Cristina Tommasi
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA
| | - Dane Sessions
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA
| | - Angela Mah
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA
| | - Tomas Bencomo
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA
| | - Jasmine M. Garcia
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA
| | - Tiffany Jiang
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA
| | - Michael Lee
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA
| | - Joseph Y. Shen
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA
| | - Lek Wei Seow
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA
| | - Audrey Nguyen
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA
| | - Kimal Rajapakshe
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristian Coarfa
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kenneth Y. Tsai
- Departments of Anatomic Pathology & Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute; Tampa, FL 33612, USA
| | | | - Carolyn S. Lee
- Stanford Program in Epithelial Biology, Stanford University, Stanford, CA 94305 USA.,Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA 94304 USA
| |
Collapse
|
9
|
Sheldon H, Zhang W, Bridges E, Ang KH, Lin S, Masiero M, Li D, Handford PA, Whiteman P, Fischer R, Buffa F, Vatish M, Banham AH, Harris AL. ELTD1 is present in extracellular vesicles derived from endothelial cells as a cleaved extracellular domain which induces in vivo angiogenesis. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e52. [PMID: 38939053 PMCID: PMC11080856 DOI: 10.1002/jex2.52] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 06/29/2024]
Abstract
ELTD1/ADGRL4 is an adhesion GPCR with an important role in angiogenesis. We recently identified a role for ELTD1 in wound repair and inflammation. Activation of ELTD1 in endothelial cells results in a type II EMT to myofibroblast-like cells that have enhanced angiogenic ability. Furthermore, expression of Eltd1 in murine breast cancer cells increases tumour growth by increasing blood vessel size and perfusion and by creating an immunosuppressive microenvironment. As extracellular vesicles (EVs) are known to be involved in vascular development, growth and maturation we investigated the composition and functional effects of the EVs isolated from ELTD1 expressing cells to elucidate their role in these processes. A highly glycosylated form of the extracellular domain (ECD) of ELTD1 is readily incorporated into EVs. Using mass spectrometry-based proteomics we identified proteins that are enriched in ELTD1-EVs and are involved in haemostasis and immune responses. ELTD1 enriched EVs were pro-angiogenic in vivo and in vitro and the presence of the ECD alone induced endothelial sprouting. In endothelial cells experiencing laminar flow, ELTD1 levels were reduced in the EVs when they are quiescent, showing a relationship between ELTD1 and the activation state of the endothelium. Using FACS, we detected a significant increase in vesicular ELTD1 in the plasma of patients with preeclampsia, a condition characterized by endothelial dysfunction. These data confirm a role for ELTD1 in wound repair and inflammation and reveal its potential as a biomarker of vessel dysfunction.
Collapse
Affiliation(s)
- Helen Sheldon
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Wei Zhang
- Nuffield Department of Women's & Reproductive Health, Women's CentreUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Esther Bridges
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Koon Hwee Ang
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Salwa Lin
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Massimo Masiero
- Nuffield Division of Clinical Laboratory SciencesRadcliffe Department of MedicineJohn Radcliffe HospitalOxfordUK
| | - Demin Li
- Nuffield Division of Clinical Laboratory SciencesRadcliffe Department of MedicineJohn Radcliffe HospitalOxfordUK
| | | | - Pat Whiteman
- Department of BiochemistryUniversity of OxfordOxfordUK
| | - Roman Fischer
- Nuffield Department of MedicineTarget Discovery InstituteOxford University, NDM Research BuildingOxfordUK
| | - Francesca Buffa
- Department of OncologyUniversity of OxfordChurchill HospitalOxfordUK
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, Women's CentreUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| | - Alison H. Banham
- Nuffield Division of Clinical Laboratory SciencesRadcliffe Department of MedicineJohn Radcliffe HospitalOxfordUK
| | - Adrian L. Harris
- Cancer Research UK Molecular Oncology LaboratoriesWeatherall Institute of Molecular MedicineUniversity of OxfordJohn Radcliffe HospitalOxfordUK
| |
Collapse
|
10
|
Wang X, Du C, Xu Q, Zhang Y, Zhou X, Wang X. Artemin affects the survival and prognosis of endometrial cancer patients via regulating tumor cell proliferation. Cancer Treat Res Commun 2022; 32:100597. [PMID: 35772316 DOI: 10.1016/j.ctarc.2022.100597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 06/15/2023]
Abstract
The main aim of the study is to analyze the impact of Artemin on survival and prognosis in endometrial cancer (EC) patients by bioinformatics methods. As a member of the glial-derived neurotrophic factor (GDNF) family, Artemin is not only important in the repair process of nerve damage, but also involved in tumorigenesis and metastasis. In this study, we demonstrated that Artemin mRNA was overexpressed in EC tissues. Artemin expression was closely related to the FIGO stage, pathologic differentiation, deep myometrial infiltration, lymphatic metastasis, and survival status. Univariate and multivariate Cox regression analysis showed that ectopic overexpression of Artemin predicted poor survival prognosis. Artemin expression could be used as an independent risk factor for the prognosis of EC patients. The proliferation of EC cells was significantly downregulated by silencing of Artemin. Artemin promotes tumor progression by regulating the proliferation of EC cells, thereby affecting the prognosis of EC patients.
Collapse
Affiliation(s)
- Xiaohua Wang
- Department of Gynecology, Affiliated Hospital of Chengde Medical University, Chengde, China.
| | - Chao Du
- Department of Biochemistry, Chengde Medical University, Chengde, China
| | - Qian Xu
- Department of Biochemistry, Chengde Medical University, Chengde, China
| | - Yujuan Zhang
- Department of Gynecology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Xiaohui Zhou
- Department of Biochemistry, Chengde Medical University, Chengde, China
| | - Xingcha Wang
- Department of Gynecology, Affiliated Hospital of Chengde Medical University, Chengde, China
| |
Collapse
|
11
|
Shi Y, Huang X, Du Z, Tan J. Analysis of single-cell RNA-sequencing data identifies a hypoxic tumor subpopulation associated with poor prognosis in triple-negative breast cancer. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:5793-5812. [PMID: 35603379 DOI: 10.3934/mbe.2022271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of mammary carcinoma characterized by low expression levels of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Along with the rapid development of the single-cell RNA-sequencing (scRNA-seq) technology, the heterogeneity within the tumor microenvironment (TME) could be studied at a higher resolution level, facilitating an exploration of the mechanisms leading to poor prognosis during tumor progression. In previous studies, hypoxia was considered as an intrinsic characteristic of TME in solid tumors, which would activate downstream signaling pathways associated with angiogenesis and metastasis. Moreover, hypoxia-related genes (HRGs) based risk score models demonstrated nice performance in predicting the prognosis of TNBC patients. However, it is essential to further investigate the heterogeneity within hypoxic TME, such as intercellular communications. In the present study, utilizing single-sample Gene Set Enrichment Analysis (ssGSEA) and cell-cell communication analysis on the scRNA-seq data retrieved from Gene Expression Omnibus (GEO) database with accession number GSM4476488, we identified four tumor subpopulations with diverse functions, particularly a hypoxia-related one. Furthermore, results of cell-cell communication analysis revealed the dominant role of the hypoxic tumor subpopulation in angiogenesis- and metastasis-related signaling pathways as a signal sender. Consequently, regard the TNBC cohorts acquired from The Cancer Genome Atlas (TCGA) and GEO as train set and test set respectively, we constructed a risk score model with reliable capacity for the prediction of overall survival (OS), where ARTN and L1CAM were identified as risk factors promoting angiogenesis and metastasis of tumors. The expression of ARTN and L1CAM were further analyzed through tumor immune estimation resource (TIMER) platform. In conclusion, these two marker genes of the hypoxic tumor subpopulation played vital roles in tumor development, indicating poor prognosis in TNBC patients.
Collapse
Affiliation(s)
- Yi Shi
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China
| | - Xiaoqian Huang
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China
| | - Zhaolan Du
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China
| | - Jianjun Tan
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China
| |
Collapse
|
12
|
Relevance of miR-223 as Potential Diagnostic and Prognostic Markers in Cancer. BIOLOGY 2022; 11:biology11020249. [PMID: 35205115 PMCID: PMC8869096 DOI: 10.3390/biology11020249] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 12/21/2022]
Abstract
In 1993, the discovery of microRNAs in Caenorhabditis elegans (C. elegans) altered the paradigmatic view of RNA biology and post-transcriptional gene regulation. Further study revealed the role of microRNAs in disease development and progression. In particular, this review highlights microRNA-223 (miR-223 or miRNA-223) expression in malignant neoplastic disorders. miR-223 expression controls aspects of hematopoiesis and apoptosis, and cell proliferation, migration, and invasion. miR-223 regulates a number of gene targets, including cytoplasmic activation/proliferation-associated protein-1 (Caprin-1), insulin-like growth factor-1 receptor (IGF-1R), and other cell proliferation- and cell cycle-associated genes. Several studies have proposed miR-223 as a novel biomarker for early cancer diagnosis. Here, we emphasize miR-223′s role in the development and progression of cancer.
Collapse
|
13
|
Zhu H, Huang M, Luo J, Ji X, Liu Q. Deficiency of GFRα1 promotes hepatocellular carcinoma progression but enhances oxaliplatin-mediated anti-tumor efficacy. Pharmacol Res 2021; 172:105815. [PMID: 34391932 DOI: 10.1016/j.phrs.2021.105815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 11/18/2022]
Abstract
Neurotrophic factors and their receptors have been identified to promote tumor progression. GFRα1, the receptor for glial cell line-derived neurotrophic factor (GDNF), has been demonstrated to be predominantly expressed in adult liver tissue. Our preliminary data showed that GFRα1 is significantly downregulated in hepatocellular carcinoma (HCC) tissue, compared to the matched non-neoplastic tissue. However, the role of GFRα1 in HCC progression remains unknown. Here we found that the expression of GFRα1 in HCC tissue is inversely correlated with the poorer prognosis of HCC patients. Silencing of GFRα1 expression markedly enhances HCC cell growth, tumor metastasis, as well as shortens the survival of HCC tumor-bearing mice. Forced expression of GFRα1 in HCC cells significantly reverses the tumor-promoting effects of GFRα1 silencing, and AAV8-mediated GFRα1 transfection in HCC tumor tissues significantly impedes tumor growth and prolongs the survival of HCC tumor-bearing mice. These results are also verified in vivo in GFRα1 knock-out mice model, with increased DEN-induced HCC carcinogenesis. Mechanistically, GFRα1 could inhibit epithelial-to-mesenchymal transition (EMT) of HCC cells, by upregulating expression of Claudin-1 and ZO-1. Of note, silencing of GFRα1 expression promotes oxaliplatin-mediated HCC cell apoptosis resulting in prolonged survival of HCC-bearing mice, and forced expression of GFRα1 markedly increased oxaliplatin resistance of HCC cells. These results demonstrate that deficiency of GFRα1 promotes HCC progression but enhances chemotherapeutic anti-tumor efficacy, suggesting that GFRα1 may be a candidate prognostic biomarker and a potential therapeutic target in HCC.
Collapse
Affiliation(s)
- Ha Zhu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Mingyan Huang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Jianhua Luo
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Xinpei Ji
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China; School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Qiuyan Liu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
14
|
Saha D, Ryan KR, Lakkaniga NR, Acharya B, Garcia NG, Smith EL, Frett B. Targeting Rearranged during Transfection in Cancer: A Perspective on Small-Molecule Inhibitors and Their Clinical Development. J Med Chem 2021; 64:11747-11773. [PMID: 34402300 DOI: 10.1021/acs.jmedchem.0c02167] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Rearranged during transfection (RET) is a receptor tyrosine kinase essential for the normal development and maturation of a diverse range of tissues. Aberrant RET signaling in cancers, due to RET mutations, gene fusions, and overexpression, results in the activation of downstream pathways promoting survival, growth, and metastasis. Pharmacological manipulation of RET is effective in treating RET-driven cancers, and efforts toward developing RET-specific therapies have increased over the last 5 years. In 2020, RET-selective inhibitors pralsetinib and selpercatinib achieved clinical approval, which marked the first approvals for kinase inhibitors specifically developed to target the RET oncoprotein. This Perspective discusses current development and clinical applications for RET precision medicine by providing an overview of the incremental improvement of kinase inhibitors for use in RET-driven malignancies.
Collapse
Affiliation(s)
- Debasmita Saha
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Katie Rose Ryan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Naga Rajiv Lakkaniga
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Baku Acharya
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Noemi Garcia Garcia
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Erica Lane Smith
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| |
Collapse
|
15
|
Zhuang QS, Sun XB, Chong QY, Banerjee A, Zhang M, Wu ZS, Zhu T, Pandey V, Lobie PE. ARTEMIN Promotes Oncogenicity and Resistance to 5-Fluorouracil in Colorectal Carcinoma by p44/42 MAPK Dependent Expression of CDH2. Front Oncol 2021; 11:712348. [PMID: 34422665 PMCID: PMC8377398 DOI: 10.3389/fonc.2021.712348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
ARTEMIN (ARTN), one of the glial-cell derived neurotrophic factor family of ligands, has been reported to be associated with a number of human malignancies. In this study, the enhanced expression of ARTN in colorectal carcinoma (CRC) was observed; the expression of ARTN positively correlated with lymph node metastases and advanced tumor stages and predicted poor prognosis. Forced expression of ARTN in CRC cells enhanced oncogenic behavior, mesenchymal phenotype, stem cell-like properties and tumor growth and metastasis in a xenograft model. These functions were conversely inhibited by depletion of endogenous ARTN. Forced expression of ARTN reduced the sensitivity of CRC cells to 5-FU treatment; and 5-FU resistant CRC cells harbored enhanced expression of ARTN. The oncogenic functions of ARTN were demonstrated to be mediated by p44/42 MAP kinase dependent expression of CDH2 (CADHERIN 2, also known as N-CADHERIN). Inhibition of p44/42 MAP kinase activity or siRNA mediated depletion of endogenous CDH2 reduced the enhanced oncogenicity and chemoresistance consequent to forced expression of ARTN induced cell functions; and forced expression of CDH2 rescued the reduced mesenchymal properties and resistance to 5-FU after ARTN depletion. In conclusion, ARTN may be of prognostic and theranostic utility in CRC.
Collapse
Affiliation(s)
- Qiu-Shi Zhuang
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore.,Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Xin-Bao Sun
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Qing-Yun Chong
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore
| | - Arindam Banerjee
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore.,Department of Chemical Engineering, Indian Institute of Technology, Kharagpur, India
| | - Min Zhang
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zheng-Sheng Wu
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Tao Zhu
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Vijay Pandey
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore.,Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|
16
|
Hou Y, Liang HL, Yu X, Liu Z, Cao X, Rao E, Huang X, Wang L, Li L, Bugno J, Fu Y, Chmura SJ, Wu W, Luo SZ, Zheng W, Arina A, Jutzy J, McCall AR, Vokes EE, Pitroda SP, Fu YX, Weichselbaum RR. Radiotherapy and immunotherapy converge on elimination of tumor-promoting erythroid progenitor cells through adaptive immunity. Sci Transl Med 2021; 13:13/582/eabb0130. [PMID: 33627484 DOI: 10.1126/scitranslmed.abb0130] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/20/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
Tumor-induced CD45-Ter119+CD71+ erythroid progenitor cells, termed "Ter cells," promote tumor progression by secreting artemin (ARTN), a neurotrophic peptide that activates REarranged during Transfection (RET) signaling. We demonstrate that both local tumor ionizing radiation (IR) and anti-programmed death ligand 1 (PD-L1) treatment decreased tumor-induced Ter cell abundance in the mouse spleen and ARTN secretion outside the irradiation field in an interferon- and CD8+ T cell-dependent manner. Recombinant erythropoietin promoted resistance to radiotherapy or anti-PD-L1 therapies by restoring Ter cell numbers and serum ARTN concentration. Blockade of ARTN or potential ARTN signaling partners, or depletion of Ter cells augmented the antitumor effects of both IR and anti-PD-L1 therapies in mice. Analysis of samples from patients who received radioimmunotherapy demonstrated that IR-mediated reduction of Ter cells, ARTN, and GFRα3, an ARTN signaling partner, were each associated with tumor regression. Patients with melanoma who received immunotherapy exhibited favorable outcomes associated with decreased expression of GFRα3. These findings demonstrate an out-of-field, or "abscopal," effect mediated by adaptive immunity, which is induced during local tumor irradiation. This effect, in turn, governs the therapeutic effects of radiation and immunotherapy. Therefore, our results identify multiple targets to potentially improve outcomes after radiotherapy and immunotherapy.
Collapse
Affiliation(s)
- Yuzhu Hou
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, ShaanXi 710061, China. .,Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA
| | - Hua L Liang
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA
| | - Xinshuang Yu
- Department of Oncology, First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Zhida Liu
- Department of Pathology, University of Texas Southwest Medical Center, Dallas, TX 75235, USA
| | - Xuezhi Cao
- Department of Pathology, University of Texas Southwest Medical Center, Dallas, TX 75235, USA
| | - Enyu Rao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaona Huang
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA
| | - Liangliang Wang
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA
| | - Lei Li
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA
| | - Jason Bugno
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, IL 60637, USA
| | - Yanbin Fu
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA
| | - Steven J Chmura
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA
| | - Wenjun Wu
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Sean Z Luo
- Whitney Young High School, Chicago, IL 60607, USA
| | - Wenxin Zheng
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA
| | - Ainhoa Arina
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA
| | - Jessica Jutzy
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA
| | - Anne R McCall
- Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Everett E Vokes
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Sean P Pitroda
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwest Medical Center, Dallas, TX 75235, USA.
| | - Ralph R Weichselbaum
- Ludwig Center for Metastasis Research, Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637 USA.
| |
Collapse
|
17
|
Romero-Trejo D, Mejía-Rodríguez R, Sierra-Mondragón E, Navarrete A, Pérez-Tapia M, González RO, Segovia J. The systemic administration of neural stem cells expressing an inducible and soluble form of growth arrest specific 1 inhibits mammary gland tumor growth and the formation of metastases. Cytotherapy 2020; 23:223-235. [PMID: 33168454 DOI: 10.1016/j.jcyt.2020.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/21/2020] [Accepted: 09/30/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND AIMS Metastasis to different organs is the major cause of death in breast cancer patients. The poor clinical prognosis and lack of successful treatments for metastatic breast cancer patients demand the development of new tumor-selective therapies. Thus, it is necessary to develop treatments capable of releasing therapeutic agents to both primary tumors and metastases that avoid toxic side effects in normal tissue, and neural stem cells are an attractive vehicle for tracking tumor cells and delivering anti-cancer agents. The authorspreviously demonstrated that a soluble form of growth arrest specific 1 (GAS1) inhibits the growth of triple-negative breast tumors and glioblastoma. METHODS In this study, the authors engineered ReNcell CX (EMD Millipore, Temecula, CA, USA) neural progenitor cells to express truncated GAS1 (tGAS1) under a tetracycline/on inducible system using lentiviral vectors. RESULTS Here the authors show that treatment with ReNcell-tGAS1 in combination with tetracycline decreased primary tumor growth and inhibited the formation of metastases in tumor-bearing mice by diminishing the phosphorylation of AKT and ERK1/2 in orthotopic mammary gland tumors. Moreover, the authors observed that ReNcell-tGAS1 prolonged the survival of 4T1 tumor-bearing mice. CONCLUSIONS These data suggest that the delivery of tGAS1 by ReNcell cells could be an effective adjuvant for the treatment of triple-negative breast cancer.
Collapse
Affiliation(s)
- Daniel Romero-Trejo
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, México
| | - Rosalinda Mejía-Rodríguez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, México
| | - Edith Sierra-Mondragón
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, México
| | - Araceli Navarrete
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, México
| | - Mayra Pérez-Tapia
- Departamento de Inmunología Escuela Nacional de Ciencias Biológicas, del Instituto Politécnico Nacional, México
| | - Rosa O González
- Departamento de Matemáticas, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), México
| | - José Segovia
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, México.
| |
Collapse
|
18
|
Engelsen AST, Wnuk-Lipinska K, Bougnaud S, Pelissier Vatter FA, Tiron C, Villadsen R, Miyano M, Lotsberg ML, Madeleine N, Panahandeh P, Dhakal S, Tan TZ, Peters SD, Grøndal S, Aziz SM, Nord S, Herfindal L, Stampfer MR, Sørlie T, Brekken RA, Straume O, Halberg N, Gausdal G, Thiery JP, Akslen LA, Petersen OW, LaBarge MA, Lorens JB. AXL Is a Driver of Stemness in Normal Mammary Gland and Breast Cancer. iScience 2020; 23:101649. [PMID: 33103086 PMCID: PMC7578759 DOI: 10.1016/j.isci.2020.101649] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 08/03/2020] [Accepted: 10/02/2020] [Indexed: 12/17/2022] Open
Abstract
The receptor tyrosine kinase AXL is associated with epithelial plasticity in several solid tumors including breast cancer and AXL-targeting agents are currently in clinical trials. We hypothesized that AXL is a driver of stemness traits in cancer by co-option of a regulatory function normally reserved for stem cells. AXL-expressing cells in human mammary epithelial ducts co-expressed markers associated with multipotency, and AXL inhibition abolished colony formation and self-maintenance activities while promoting terminal differentiation in vitro. Axl-null mice did not exhibit a strong developmental phenotype, but enrichment of Axl + cells was required for mouse mammary gland reconstitution upon transplantation, and Axl-null mice had reduced incidence of Wnt1-driven mammary tumors. An AXL-dependent gene signature is a feature of transcriptomes in basal breast cancers and reduced patient survival irrespective of subtype. Our interpretation is that AXL regulates access to epithelial plasticity programs in MaSCs and, when co-opted, maintains acquired stemness in breast cancer cells.
Collapse
Affiliation(s)
- Agnete S T Engelsen
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy Cancer Campus Grand Paris, 94800 Villejuif, France
| | | | - Sebastien Bougnaud
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
| | - Fanny A Pelissier Vatter
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
| | - Crina Tiron
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - René Villadsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Copenhagen N 2200, Denmark
| | - Masaru Miyano
- Biolgical Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.,Department of Population Sciences, Beckman Research Institute at City of Hope, Duarte, CA 91910, USA
| | - Maria L Lotsberg
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
| | - Noëlly Madeleine
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Pouda Panahandeh
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Sushil Dhakal
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Tuan Zea Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | | | - Sturla Grøndal
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Sura M Aziz
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,Department of Pathology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Silje Nord
- Department of Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Lars Herfindal
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Martha R Stampfer
- Biolgical Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Therese Sørlie
- Department of Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Oddbjørn Straume
- Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,Department of Oncology and Medical Physics, Haukeland University Hospital, 5021 Bergen, Norway
| | - Nils Halberg
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Gro Gausdal
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway
| | - Jean Paul Thiery
- Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy Cancer Campus Grand Paris, 94800 Villejuif, France.,Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, A-STAR, Singapore 138673, Singapore.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health, Bio-island, Guangzhou, 510320, China
| | - Lars A Akslen
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,Department of Pathology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Ole W Petersen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Copenhagen N 2200, Denmark.,Novo Nordisk Foundation Center for Stem Cell Biology, University of Copenhagen, Copenhagen, Copenhagen N 2200, Denmark
| | - Mark A LaBarge
- Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway.,Biolgical Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.,Department of Population Sciences, Beckman Research Institute at City of Hope, Duarte, CA 91910, USA
| | - James B Lorens
- Department of Biomedicine, University of Bergen, 5021 Bergen, Norway.,Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
| |
Collapse
|
19
|
Connolly E, Morgan DJ, Franklin M, Simpson A, Shah R, Brand OJ, Jagger CP, Casulli J, Mohamed K, Grabiec AM, Hussell T. Neurturin regulates the lung-resident macrophage inflammatory response to viral infection. Life Sci Alliance 2020; 3:3/12/e202000780. [PMID: 33020210 PMCID: PMC7556752 DOI: 10.26508/lsa.202000780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 11/24/2022] Open
Abstract
The neurotrophic factor RET is induced in lung macrophages by viral triggers and activation via its ligand neurturin regulates matrix proteins and cytokines that shape the inflammatory response. Lung-resident macrophages are crucial to the maintenance of health and in the defence against lower respiratory tract infections. Macrophages adapt to local environmental cues that drive their appropriate function; however, this is often dysregulated in many inflammatory lung pathologies. In mucosal tissues, neuro-immune interactions enable quick and efficient inflammatory responses to pathogenic threats. Although a number of factors that influence the antimicrobial response of lung macrophages are known, the role of neuronal factors is less well understood. Here, we show an intricate circuit involving the neurotrophic factor, neurturin (NRTN) on human lung macrophages that dampens pro-inflammatory cytokine release and modulates the type of matrix metalloproteinases produced in response to viral stimuli. This circuit involves type 1 interferon–induced up-regulation of RET that when combined with the glial cell line-derived neurotrophic factor (GDNF) receptor α2 (GFRα2) allows binding to epithelial-derived NRTN. Our research highlights a non-neuronal immunomodulatory role for NRTN and a novel process leading to a specific antimicrobial immune response by human lung-resident macrophages.
Collapse
Affiliation(s)
- Emma Connolly
- The Lydia Becker Institute for Immunology and Inflammation, The University of Manchester, Manchester, UK.,Manchester Collaborative Centre for Inflammation Research, The University of Manchester, Manchester, UK
| | - David J Morgan
- The Lydia Becker Institute for Immunology and Inflammation, The University of Manchester, Manchester, UK.,Manchester Collaborative Centre for Inflammation Research, The University of Manchester, Manchester, UK
| | - Miriam Franklin
- The Lydia Becker Institute for Immunology and Inflammation, The University of Manchester, Manchester, UK.,Manchester Collaborative Centre for Inflammation Research, The University of Manchester, Manchester, UK
| | - Angela Simpson
- Division of Infection, Immunity and Respiratory Medicine, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Rajesh Shah
- Department of Thoracic Surgery, University Hospital of South Manchester, Manchester, UK
| | - Oliver J Brand
- The Lydia Becker Institute for Immunology and Inflammation, The University of Manchester, Manchester, UK.,Manchester Collaborative Centre for Inflammation Research, The University of Manchester, Manchester, UK
| | - Christopher P Jagger
- The Lydia Becker Institute for Immunology and Inflammation, The University of Manchester, Manchester, UK.,Manchester Collaborative Centre for Inflammation Research, The University of Manchester, Manchester, UK
| | - Joshua Casulli
- The Lydia Becker Institute for Immunology and Inflammation, The University of Manchester, Manchester, UK.,Manchester Collaborative Centre for Inflammation Research, The University of Manchester, Manchester, UK
| | - Karishma Mohamed
- The Lydia Becker Institute for Immunology and Inflammation, The University of Manchester, Manchester, UK.,Manchester Collaborative Centre for Inflammation Research, The University of Manchester, Manchester, UK
| | - Aleksander M Grabiec
- Manchester Collaborative Centre for Inflammation Research, The University of Manchester, Manchester, UK
| | - Tracy Hussell
- The Lydia Becker Institute for Immunology and Inflammation, The University of Manchester, Manchester, UK .,Manchester Collaborative Centre for Inflammation Research, The University of Manchester, Manchester, UK
| |
Collapse
|
20
|
Li J, Guan X, Fan Z, Ching LM, Li Y, Wang X, Cao WM, Liu DX. Non-Invasive Biomarkers for Early Detection of Breast Cancer. Cancers (Basel) 2020; 12:E2767. [PMID: 32992445 PMCID: PMC7601650 DOI: 10.3390/cancers12102767] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most common cancer in women worldwide. Accurate early diagnosis of breast cancer is critical in the management of the disease. Although mammogram screening has been widely used for breast cancer screening, high false-positive and false-negative rates and radiation from mammography have always been a concern. Over the last 20 years, the emergence of "omics" strategies has resulted in significant advances in the search for non-invasive biomarkers for breast cancer diagnosis at an early stage. Circulating carcinoma antigens, circulating tumor cells, circulating cell-free tumor nucleic acids (DNA or RNA), circulating microRNAs, and circulating extracellular vesicles in the peripheral blood, nipple aspirate fluid, sweat, urine, and tears, as well as volatile organic compounds in the breath, have emerged as potential non-invasive diagnostic biomarkers to supplement current clinical approaches to earlier detection of breast cancer. In this review, we summarize the current progress of research in these areas.
Collapse
Affiliation(s)
- Jiawei Li
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand; (J.L.); (X.G.); (Y.L.)
| | - Xin Guan
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand; (J.L.); (X.G.); (Y.L.)
- Department of Breast Surgery, the First Hospital of Jilin University, Jilin University, Changchun 130021, China;
| | - Zhimin Fan
- Department of Breast Surgery, the First Hospital of Jilin University, Jilin University, Changchun 130021, China;
| | - Lai-Ming Ching
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand;
| | - Yan Li
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand; (J.L.); (X.G.); (Y.L.)
| | - Xiaojia Wang
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital & Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China;
| | - Wen-Ming Cao
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital & Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China;
| | - Dong-Xu Liu
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand; (J.L.); (X.G.); (Y.L.)
| |
Collapse
|
21
|
Zhou W, Tahir F, Wang JCY, Woodson M, Sherman MB, Karim S, Neelakanta G, Sultana H. Discovery of Exosomes From Tick Saliva and Salivary Glands Reveals Therapeutic Roles for CXCL12 and IL-8 in Wound Healing at the Tick-Human Skin Interface. Front Cell Dev Biol 2020; 8:554. [PMID: 32766239 PMCID: PMC7378379 DOI: 10.3389/fcell.2020.00554] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 06/10/2020] [Indexed: 12/30/2022] Open
Abstract
Ticks secrete various anti-coagulatory, anti-vasoconstrictory, anti-inflammatory, and anti-platelet aggregation factors in their saliva at the bite site during feeding to evade host immunological surveillance and responses. For the first time, we report successful isolation of exosomes (small membrane-bound extracellular signaling vesicles) from saliva and salivary glands of partially fed or unfed ixodid ticks. Our data showed a novel role of these in vivo exosomes in the inhibition of wound healing via downregulation of C-X-C motif chemokine ligand 12 (CXCL12) and upregulation of interleukin-8 (IL-8). Cryo-electron microscopy (cryo-EM) analysis revealed that tick saliva and salivary glands are composed of heterogeneous populations of in vivo exosomes with sizes ranging from 30 to 200 nm. Enriched amounts of tick CD63 ortholog protein and heat shock protein 70 (HSP70) were evident in these exosomes. Treatment of human skin keratinocytes (HaCaT cells) with exosomes derived from tick saliva/salivary glands or ISE6 cells dramatically delayed cell migration, wound healing, and repair process. Wound healing is a highly dynamic process with several individualized processes including secretion of cytokines. Cytokine array profiling followed by immunoblotting and quantitative-PCR analysis revealed that HaCaT cells treated with exosomes derived from tick saliva/salivary glands or ISE6 cells showed enhanced IL-8 levels and reduced CXCL12 loads. Inhibition of IL-8 or CXCL12 further delayed exosome-mediated cell migration, wound healing, and repair process, suggesting a skin barrier protection role for these chemokines at the tick bite site. In contrast, exogenous treatment of CXCL12 protein completely restored this delay and enhanced the repair process. Taken together, our study provides novel insights on how tick salivary exosomes secreted in saliva can delay wound healing at the bite site to facilitate successful blood feeding.
Collapse
Affiliation(s)
- Wenshuo Zhou
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States
| | - Faizan Tahir
- Center for Molecular and Cellular Biosciences, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Joseph Che-Yen Wang
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, United States.,Electron Microscopy Center, Indiana University, Bloomington, IN, United States
| | - Michael Woodson
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Michael B Sherman
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Shahid Karim
- Center for Molecular and Cellular Biosciences, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, United States
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, United States.,Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
22
|
Gattelli A, Hynes NE, Schor IE, Vallone SA. Ret Receptor Has Distinct Alterations and Functions in Breast Cancer. J Mammary Gland Biol Neoplasia 2020; 25:13-26. [PMID: 32080788 DOI: 10.1007/s10911-020-09445-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/06/2020] [Indexed: 12/15/2022] Open
Abstract
Ret receptor tyrosine kinase is a proto-oncogene that participates in development of various cancers. Several independent studies have recently identified Ret as a key player in breast cancer. Although Ret overexpression and function have been under investigation, mainly in estrogen receptor positive breast cancer, a more comprehensive analysis of the impact of recurring Ret alterations in breast cancer is needed. This review consolidates the current knowledge of Ret alterations and their potential effects in breast cancer. We discuss and integrate data on Ret changes in different breast cancer subtypes and potential function in progression, as well as the participation of distinct Ret network signaling partners in these processes. We propose that it will be essential to define a shared molecular feature of tumors with alteration in Ret receptor, be this at the genetic level or via overexpression in order to design effective therapies to target the Ret pathway. Here we review experimental evidence from basic research and pre-clinical studies concentrating on Ret alterations as potential biomarkers for recurrence, and we discuss the possibility that targeting the Ret pathway might in the future become a treatment for breast cancer.
Collapse
Affiliation(s)
- Albana Gattelli
- CONICET-UBA, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Ciudad Universitaria, C1428EGA CABA, Buenos Aires, Argentina.
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria C1428EGA CABA, Buenos Aires, Argentina.
| | - Nancy E Hynes
- Friedrich Miescher Institute for Biomedical Research (FMI), Maulbeerstrasse 66, CH-4058, Basel, Switzerland
- University of Basel, CH-4002, Basel, Switzerland
| | - Ignacio E Schor
- CONICET-UBA, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Ciudad Universitaria, C1428EGA CABA, Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), Ciudad Universitaria, C1428EGA, CABA, Argentina
| | - Sabrina A Vallone
- CONICET-UBA, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Ciudad Universitaria, C1428EGA CABA, Buenos Aires, Argentina
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria C1428EGA CABA, Buenos Aires, Argentina
| |
Collapse
|
23
|
Enhancement of HGF-induced tubulogenesis by endothelial cell-derived GDNF. PLoS One 2019; 14:e0212991. [PMID: 30845150 PMCID: PMC6405134 DOI: 10.1371/journal.pone.0212991] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/13/2019] [Indexed: 12/22/2022] Open
Abstract
Tubulogenesis, the organization of epithelial cells into tubular structures, is an essential step during renal organogenesis as well as during the regeneration process of renal tubules after injury. In the present study, endothelial cell-derived factors that modulate tubule formation were examined using an in vitro human tubulogenesis system. When human renal proximal tubular epithelial cells (RPTECs) were cultured in gels, tubular structures with lumens were induced in the presence of hepatocyte growth factor (HGF). Aquaporin 1 was localized in the apical membrane of these tubular structures, suggesting that these structures are morphologically equivalent to renal tubules in vivo. HGF-induced tubule formation was significantly enhanced when co-cultured with human umbilical vein endothelial cells (HUVECs) or in the presence of HUVEC-conditioned medium (HUVEC-CM). Co-culture with HUVECs did not induce tubular structures in the absence of HGF. A phospho-receptor tyrosine kinase array revealed that HUVEC-CM markedly enhanced phosphorylation of Ret, glial cell-derived neurotrophic factor (GDNF) receptor, in HGF-induced tubular structures compared to those without HUVEC-CM. HUVECs produced GDNF, and RPTECs expressed both Ret and GDNF family receptor alpha1 (co-receptor). HGF-induced tubule formation was significantly enhanced by addition of GDNF. Interestingly, not only HGF but also GDNF significantly induced phosphorylation of the HGF receptor, Met. These data indicate that endothelial cell-derived GDNF potentiates the tubulogenic properties of HGF and may play a critical role in the epithelial-endothelial crosstalk during renal tubulogenesis as well as tubular regeneration after injury.
Collapse
|
24
|
Amit M, Na'ara S, Fridman E, Vladovski E, Wasserman T, Milman N, Gil Z. RET, a targetable driver of pancreatic adenocarcinoma. Int J Cancer 2019; 144:3014-3022. [DOI: 10.1002/ijc.32040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 11/14/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Moran Amit
- Head and Neck SurgeryHouston Methodist Hospital Houston TX USA
- The Laboratory for Applied Cancer Research, The TechnionIsrael Institute of Technology Haifa Israel
- Department of Otolaryngology Head and Neck Surgery, the Head and Neck Center, Rambam Healthcare CampusClinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology Haifa Israel
| | - Shorook Na'ara
- The Laboratory for Applied Cancer Research, The TechnionIsrael Institute of Technology Haifa Israel
- Department of Otolaryngology Head and Neck Surgery, the Head and Neck Center, Rambam Healthcare CampusClinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology Haifa Israel
| | - Eran Fridman
- The Laboratory for Applied Cancer Research, The TechnionIsrael Institute of Technology Haifa Israel
- Department of Otolaryngology Head and Neck Surgery, the Head and Neck Center, Rambam Healthcare CampusClinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology Haifa Israel
| | - Euvgeni Vladovski
- Department of Pathology, Rambam Healthcare Campus, The TechnionIsrael Institute of Technology Haifa Israel
| | - Tanya Wasserman
- Department of Physiology, Biophysics and Systems Biology, Faculty of MedicineTechnion Haifa Israel
| | - Neta Milman
- The Laboratory for Applied Cancer Research, The TechnionIsrael Institute of Technology Haifa Israel
| | - Ziv Gil
- The Laboratory for Applied Cancer Research, The TechnionIsrael Institute of Technology Haifa Israel
- Department of Otolaryngology Head and Neck Surgery, the Head and Neck Center, Rambam Healthcare CampusClinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology Haifa Israel
| |
Collapse
|
25
|
Mulligan LM. GDNF and the RET Receptor in Cancer: New Insights and Therapeutic Potential. Front Physiol 2019; 9:1873. [PMID: 30666215 PMCID: PMC6330338 DOI: 10.3389/fphys.2018.01873] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022] Open
Abstract
The Glial cell line-derived neurotrophic Family Ligands (GFL) are soluble neurotrophic factors that are required for development of multiple human tissues, but which are also important contributors to human cancers. GFL signaling occurs through the transmembrane RET receptor tyrosine kinase, a well-characterized oncogene. GFL-independent RET activation, through rearrangement or point mutations occurs in thyroid and lung cancers. However, GFL-mediated activation of wildtype RET is an increasingly recognized mechanism promoting tumor growth and dissemination of a much broader group of cancers. RET and GFL expression have been implicated in metastasis or invasion in diverse human cancers including breast, pancreatic, and prostate tumors, where they are linked to poorer patient prognosis. In addition to directly inducing tumor growth in these diseases, GFL-RET signaling promotes changes in the tumor microenvironment that alter the surrounding stroma and cellular composition to enhance tumor invasion and metastasis. As such, GFL RET signaling is an important target for novel therapeutic approaches to limit tumor growth and spread and improve disease outcomes.
Collapse
Affiliation(s)
- Lois M. Mulligan
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Cancer Research Institute, Queen’s University, Kingston, ON, Canada
| |
Collapse
|
26
|
Nahari E, Razi M. Silymarin amplifies apoptosis in ectopic endometrial tissue in rats with endometriosis; implication on growth factor GDNF, ERK1/2 and Bcl-6b expression. Acta Histochem 2018; 120:757-767. [PMID: 30195499 DOI: 10.1016/j.acthis.2018.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/29/2018] [Accepted: 08/16/2018] [Indexed: 12/25/2022]
Abstract
The present prospective study was done to evaluate the effect of silymarin (SMN) on endometriotic-like legions establishment and growth in experimentally-induced endometriosis. For this purpose, the experimental endometriosis was induced in 12 rats and then the animals subdivided into endometriosis-sole and SMN (50 mg kg-1, orally)+endometriosis groups. Following 28 days, the legions establishment, size, Glial cell line-derived neurotrophic factor (GDNF), gfrα1, B Cell Lymphoma 6 (Bcl-6b), Bcl-2, extracellular regulator kinase (ERK1/2) expression ratios, angiogenesis, the apoptosis and fibrosis indices were investigated. The SMN significantly (P < 0.05) decreased the enometriotic-like legions establishment and size, decreased mRNA levels of GDNF, gfrα1, Bcl-6b and Bcl-2 and remarkably diminished GDNF, gfrα1, Bcl-6b and Bcl-2-positive cells distribution/mm2 of tissue versus endometriosis-sole group. The SMN + endometriosis group exhibited a significant (P < 0.05) enhancement in ERK1/2 expression and represented diminished vascularized area and increased apoptosis and fibrosis indices, as well. In conclusion, the SMN by down-regulating GDNF and its receptor gfrα1 expression inhibits GDNF-gfrα1 complex generation and consequently suppresses Bcl-6b expression. Moreover, the SMN by enhancing the ERK1/2 expression and by suppressing the Bcl-2 expression promotes the apoptosis pathway. Finally, the SMN by down-regulating the angiogenesis ratio accelerates apoptosis and consequently induces severe fibrosis in endometriotic-like legions.
Collapse
Affiliation(s)
- Elaheh Nahari
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Mazdak Razi
- Department of Basic Sciences, Faculty of Veterinary Medicine, P.O. BOC: 1177, Urmia University, Urmia, Iran.
| |
Collapse
|
27
|
Tumor-Induced Generation of Splenic Erythroblast-like Ter-Cells Promotes Tumor Progression. Cell 2018; 173:634-648.e12. [DOI: 10.1016/j.cell.2018.02.061] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/22/2018] [Accepted: 02/26/2018] [Indexed: 12/19/2022]
|
28
|
Song Z, Yang F, Du H, Li X, Liu J, Dong M, Xu X. Role of artemin in non-small cell lung cancer. Thorac Cancer 2018; 9:555-562. [PMID: 29575549 PMCID: PMC5928368 DOI: 10.1111/1759-7714.12615] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/01/2018] [Accepted: 02/02/2018] [Indexed: 02/06/2023] Open
Abstract
Background In this study, we investigated the role of artemin, a member of the glial cell‐derived neurotrophic factor of ligands, in the malignant phenotype of lung cancer. Methods Artemin expression was examined in various types of lung cancer and normal lung tissues, as well as in lung cancer cell lines by immunohistochemistry and semi‐quantitative PCR. Functional studies were performed using artemin overexpression or knockdown vectors in lung cancer cell lines. Methyl thiazolyl tetrazolium, flow cytometry, wound healing, and transwell assays were conducted to evaluate the contribution of artemin on tumor cell proliferation, migration, and invasion. Results Artemin is broadly expressed in lung cancer tissues, and is associated with tumor staging. Overexpression of artemin in NL9980 large cell lung cancer cells increased proliferating cells and enhanced migrating capability in wound healing and transwell assays, as well as demonstrating enhanced invasion capability. Silencing artemin in LTEP‐α‐2 adenocarcinoma cell lines decreased cellular proliferation, migration, and invasion capabilities. Conclusion Artemin could promote the proliferation and invasiveness of lung cancer cells in vitro and therefore could be a new potential target to combat lung cancer.
Collapse
Affiliation(s)
- Zuoqing Song
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Fan Yang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hui Du
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinghao Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ming Dong
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaohong Xu
- College of Nursing, Tianjin Medical University, Tianjin, China.,Institute of Acupuncture, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
29
|
Wang J, Wang H, Cai J, Du S, Xin B, Wei W, Zhang T, Shen X. Artemin regulates CXCR4 expression to induce migration and invasion in pancreatic cancer cells through activation of NF-κB signaling. Exp Cell Res 2018; 365:12-23. [PMID: 29453972 DOI: 10.1016/j.yexcr.2018.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/09/2018] [Accepted: 02/13/2018] [Indexed: 12/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most lethal human malignant tumor because of the early onset of local invasion and distant metastasis. Perineural invasion is a prominent characteristic of pancreatic adenocarcinoma, which is a multifactorial process that involves various signaling molecules from different signaling pathways. The glial cell line-derived neurotrophic factor family of ligands was reported to be involved in perineural invasion in pancreatic cancer. Artemin is one member of the glial cell line-derived neurotrophic factor family of ligands. Although Artemin has previously been demonstrated to promote invasiveness of pancreatic cancer, the mechanisms remain poorly understood. In this study, we performed an analysis to determine the effects of Artemin on modulating tumor cell metastatic potential and invasion activity and explored its mechanisms in pancreatic cancer. We indicated that Artemin and CXCR4 were overexpressed in cancer tissues and widely expressed in pancreatic cancer cell lines. We observed that activation of ERK1/2 and Akt in Artemin-treated cells led to enhanced nuclear accumulation of NF-κB, which then induced CXCR4 expression. Through regulation of the expression of CXCR4, Artemin functionally promoted the migration and invasion in pancreatic cancer cells. The present study indicated that Artemin induced CXCR4 expression by activating Akt and ERK 1/2/NF-κB signaling, thereby modulating tumor cell metastatic potential and invasion activity in pancreatic cancer by regulating SDF-1α/CXCR4 axis. Artemin might be an effective and potent therapeutic target for pancreatic cancer metastasis, especially in perineural invasion.
Collapse
Affiliation(s)
- Juan Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Hui Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jun Cai
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shaoxia Du
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Beibei Xin
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Wei Wei
- Tianjin Medical University Cancer Institute and Hospital, Huanhu West Road, Tianjin 300060, China
| | - Ti Zhang
- Tianjin Medical University Cancer Institute and Hospital, Huanhu West Road, Tianjin 300060, China
| | - Xiaohong Shen
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| |
Collapse
|
30
|
Fielder GC, Yang TWS, Razdan M, Li Y, Lu J, Perry JK, Lobie PE, Liu DX. The GDNF Family: A Role in Cancer? Neoplasia 2018; 20:99-117. [PMID: 29245123 PMCID: PMC5730419 DOI: 10.1016/j.neo.2017.10.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 10/31/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023]
Abstract
The glial cell line-derived neurotrophic factor (GDNF) family of ligands (GFLs) comprising of GDNF, neurturin, artemin, and persephin plays an important role in the development and maintenance of the central and peripheral nervous system, renal morphogenesis, and spermatogenesis. Here we review our current understanding of GFL biology, and supported by recent progress in the area, we examine their emerging role in endocrine-related and other non-hormone-dependent solid neoplasms. The ability of GFLs to elicit actions that resemble those perturbed in an oncogenic phenotype, alongside mounting evidence of GFL involvement in tumor progression, presents novel opportunities for therapeutic intervention.
Collapse
Affiliation(s)
| | | | - Mahalakshmi Razdan
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Yan Li
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Jun Lu
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Jo K Perry
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore; Tsinghua Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, Guangdong, P. R. China
| | - Dong-Xu Liu
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand.
| |
Collapse
|
31
|
Hezam K, Jiang J, Sun F, Zhang X, Zhang J. Artemin promotes oncogenicity, metastasis and drug resistance in cancer cells. Rev Neurosci 2017; 29:93-98. [DOI: 10.1515/revneuro-2017-0029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 06/02/2017] [Indexed: 01/06/2023]
Abstract
Abstract
Artemin (ARTN) is a member of glial cell line-derived neurotrophic factor (GDNF) family of ligands, and its signaling is mediated via a multi-component receptor complex including the glycosylphosphatidylinositol-anchored GDNF family receptors a (GFRa1, GFRa3) and RET receptor tyrosine kinase. The major mechanism of ARTN action is via binding to a non-signaling co-receptor. The major function of ARTN is to drive the molecule to induce migration and axonal projection from sympathetic neurons. It also promotes the survival, proliferation and neurite outgrowth of sympathetic neurons in vitro. ARTN triggers oncogenicity and metastasis by the activation of the AKT signaling pathway. Recent studies have reported that the expression of ARTN in hepatocellular carcinoma is associated with increased tumor size, quick relapse and shorter survival. Furthermore, ARTN promotes drug resistance such as antiestrogens, doxorubicin, fulvestrant, paclitaxel, tamoxifen and trastuzumab. Moreover, ARTN also stimulates the radio-therapeutic resistance. This review highlights the proposed roles of ARTN in cancer cells and discusses recent results supporting its emerging role as an oncogenic, metastatic and drug-resisting agent with a special focus on how these new insights may facilitate rational development of ARTN for targeted therapies in the future.
Collapse
|
32
|
Zhang J, Liu Y, Yu CJ, Dai F, Xiong J, Li HJ, Wu ZS, Ding R, Wang H. Role of ARPC2 in Human Gastric Cancer. Mediators Inflamm 2017; 2017:5432818. [PMID: 28694563 PMCID: PMC5485321 DOI: 10.1155/2017/5432818] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 03/06/2017] [Indexed: 01/01/2023] Open
Abstract
Gastric cancer continues to be the second most frequent cause of cancer deaths worldwide. However, the exact molecular mechanisms are still unclear. Further research to find potential targets for therapy is critical and urgent. In this study, we found that ARPC2 promoted cell proliferation and invasion in the human cancer cell line MKN-28 using a cell total number assay, MTT (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide) assay, cell colony formation assay, migration assay, invasion assay, and wound healing assay. For downstream pathways, CTNND1, EZH2, BCL2L2, CDH2, VIM, and EGFR were upregulated by ARPC2, whereas PTEN, BAK, and CDH1 were downregulated by ARPC2. In a clinical study, we examined the expression of ARPC2 in 110 cases of normal human gastric tissues and 110 cases of human gastric cancer tissues. ARPC2 showed higher expression in gastric cancer tissues than in normal gastric tissues. In the association analysis of 110 gastric cancer tissues, ARPC2 showed significant associations with large tumor size, lymph node invasion, and high tumor stage. In addition, ARPC2-positive patients exhibited lower RFS and OS rates compared with ARPC2-negative patients. We thus identify that ARPC2 plays an aneretic role in human gastric cancer and provided a new target for gastric cancer therapy.
Collapse
Affiliation(s)
- Jun Zhang
- Department of General Surgery, Third Affiliated Hospital (Hefei First People's Hospital) of Anhui Medical University, Hefei, China
| | - Yi Liu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chang-Jun Yu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fu Dai
- Department of General Surgery, Third Affiliated Hospital (Hefei First People's Hospital) of Anhui Medical University, Hefei, China
| | - Jie Xiong
- Department of General Surgery, Third Affiliated Hospital (Hefei First People's Hospital) of Anhui Medical University, Hefei, China
| | - Hong-Jun Li
- Department of General Surgery, Third Affiliated Hospital (Hefei First People's Hospital) of Anhui Medical University, Hefei, China
| | - Zheng-Sheng Wu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rui Ding
- Department of General Surgery, Third Affiliated Hospital (Hefei First People's Hospital) of Anhui Medical University, Hefei, China
| | - Hong Wang
- Department of General Surgery, Third Affiliated Hospital (Hefei First People's Hospital) of Anhui Medical University, Hefei, China
| |
Collapse
|
33
|
Ding K, Yuan Y, Chong QY, Yang Y, Li R, Li X, Kong X, Qian P, Xiong Z, Pandey V, Ma L, Wu Z, Lobie PE, Zhu T. Autocrine Prolactin Stimulates Endometrial Carcinoma Growth and Metastasis and Reduces Sensitivity to Chemotherapy. Endocrinology 2017; 158:1595-1611. [PMID: 28204229 DOI: 10.1210/en.2016-1903] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/06/2017] [Indexed: 12/29/2022]
Abstract
Advanced and recurrent endometrial carcinoma (EC) exhibits a poor response to chemotherapy and low survival rates. It has been previously reported that human prolactin (hPRL) is upregulated in endometrial cancer and is associated with worse survival outcomes. We provide evidence for the functional role of hPRL in EC progression. We generated a model for the study of autocrine hPRL-mediated cell functional effects through the forced expression of hPRL in human EC cells. Autocrine hPRL expression stimulated cell proliferation, anchorage-independent growth, migration, and invasion of EC cells and promoted tumor growth, local invasion, and metastatic colonization in xenograft models. In addition, forced expression of hPRL decreased sensitivity of EC cells to chemotherapeutic drugs (i.e., doxorubicin and paclitaxel), both in vitro and in vivo. Consistently, small interfering RNA-mediated depletion of hPRL significantly reduced oncogenicity and enhanced the chemosensitivity of EC cells. As CD24 is hPRL-regulated and has been implicated in drug resistance in EC, we further showed that CD24 is a critical mediator of hPRL-stimulated reduced sensitivity to doxorubicin and paclitaxel in EC cells. Therefore, inhibition of hPRL signaling is a potential therapeutic strategy for the treatment of late-stage EC, which can be used in combination with chemotherapy to improve the chemotherapeutic response.
Collapse
Affiliation(s)
- Keshuo Ding
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230000, China
| | - Yan Yuan
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Qing-Yun Chong
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore 117599
| | - Yulu Yang
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Rui Li
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiaoni Li
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Xiangjun Kong
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Pengxu Qian
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| | - Zirui Xiong
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Vijay Pandey
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore 117599
| | - Lan Ma
- Tsinghua-Berkeley Shenzhen Institute and Division of Life Sciences and Health, Tsinghua University Graduate School, Shenzhen 518055, China
| | - Zhengsheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230000, China
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore 117599
- Tsinghua-Berkeley Shenzhen Institute and Division of Life Sciences and Health, Tsinghua University Graduate School, Shenzhen 518055, China
| | - Tao Zhu
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
| |
Collapse
|
34
|
Simvastatin down-regulates differential genetic profiles produced by organochlorine mixtures in primary breast cell (HMEC). Chem Biol Interact 2017; 268:85-92. [PMID: 28263720 DOI: 10.1016/j.cbi.2017.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/21/2017] [Accepted: 03/01/2017] [Indexed: 11/22/2022]
Abstract
Women all over the world are exposed to an unavoidable contamination by organochlorine pesticides and other chemical pollutants. Many of them are considered as xenoestrogens and have been associated with the development and progression of breast cancer. We have demonstrated that the most prevalent pesticide mixtures found in healthy women and in women diagnosed with breast cancer modulates the gene expression in human epithelial mammary cells. Statins are well-known cholesterol-depleting agents acting as inhibitors of cholesterol synthesis. Since the early 1990s, it has been known that statins could be successfully used in cancer therapy, including breast cancer, but the exact mechanism behind anti-tumor activity of the statins remains unclear. In the present study we evaluated the effect of simvastatin in the gene expression pattern induced by realistic organochlorine mixtures found in breast cancer patients. The gene expression of 94 genes related with the cell signaling pathways were assessed. Our results indicate that simvastatin exerts a global down regulating effect on successfully determined genes (78.7%), thus attenuating the effects induced by organochlorine mixtures on the gene profile of human mammary epithelial cells. This effect was more evident on genes whose function is the ATP-binding process (that also were particularly up-regulated by pesticide mixtures). We also found that MERTK (a proto-oncogene which is overexpressed in several malignancies) and PDGFRB (a member of the platelet-derived growth factor family whose expression is high in breast-cancer cells that have become resistant to endocrine therapy) were among the genes with a higher differential regulation by simvastatin. Since resistance to treatment with tyrosine kinase inhibitors is closely related to MERKT, our findings would enhance the possible utility of statins in breast cancer treatment, i.e. improving therapeutic results combining statins with tyrosine Kinase inhibitors.
Collapse
|
35
|
Gao Y, Lin L, Li T, Yang J, Wei Y. The role of miRNA-223 in cancer: Function, diagnosis and therapy. Gene 2017; 616:1-7. [PMID: 28322994 DOI: 10.1016/j.gene.2017.03.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 01/28/2017] [Accepted: 03/16/2017] [Indexed: 01/01/2023]
Abstract
MicroRNAs (miRNAs) constitute a large family of small, non-coding RNAs with the capacity to regulate gene expression post-transcriptionally. miRNAs appear to hold promise of mechanistic explanations for various physiological and pathological processes. miRNA-223 is highly conserved and preferentially expressed in the hematopoietic system in regulation of myeloid differentiation. Recently, increasing evidence suggests that miRNA-223 may also play an essential part in both hematological malignancies and solid tumors. miRNA-223 can function as either an oncogene or a tumor suppressor gene, which is achieved by targeting a wide range of genes and regulating downstream signal transduction. As yet, the function of miR-223 in cancer has not been fully characterized and understood. To make it more clear, this review firstly summarizes the present understanding of the regulation of miR-223 at the molecular level, its crucial role in oncogenesis, development, and metastasis, its function as a diagnostic and prognostic biomarker and finally, its potential applications in monitoring and therapy of diverse types of malignancies.
Collapse
Affiliation(s)
- Yunliang Gao
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, United States
| | - Le Lin
- Department of Urology, Fujian Medical University Teaching Hospital, Fujian Provincial Hospital, Fuzhou 350001, PR China
| | - Tao Li
- Department of Urology, Fujian Medical University Teaching Hospital, Fujian Provincial Hospital, Fuzhou 350001, PR China
| | - Jinrui Yang
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, PR China.
| | - Yongbao Wei
- Department of Urology, Fujian Medical University Teaching Hospital, Fujian Provincial Hospital, Fuzhou 350001, PR China.
| |
Collapse
|
36
|
Amit M, Na'ara S, Leider-Trejo L, Binenbaum Y, Kulish N, Fridman E, Shabtai-Orbach A, Wong RJ, Gil Z. Upregulation of RET induces perineurial invasion of pancreatic adenocarcinoma. Oncogene 2017; 36:3232-3239. [PMID: 28092668 DOI: 10.1038/onc.2016.483] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 10/14/2016] [Accepted: 11/17/2016] [Indexed: 02/06/2023]
Abstract
Tumor spread along nerves, a phenomenon known as perineurial invasion, is common in various cancers including pancreatic ductal adenocarcinoma (PDAC). Neural invasion is associated with poor outcome, yet its mechanism remains unclear. Using the transgenic Pdx-1-Cre/KrasG12D /p53R172H (KPC) mouse model, we investigated the mechanism of neural invasion in PDAC. To detect tissue-specific factors that influence neural invasion by cancer cells, we characterized the perineurial microenvironment using a series of bone marrow transplantation (BMT) experiments in transgenic mice expressing single mutations in the Cx3cr1, GDNF and CCR2 genes. Immunolabeling of tumors in KPC mice of different ages and analysis of human cancer specimens revealed that RET expression is upregulated during PDAC tumorigenesis. BMT experiments revealed that BM-derived macrophages expressing the RET ligand GDNF are highly abundant around nerves invaded by cancer. Inhibition of perineurial macrophage recruitment, using the CSF-1R antagonist GW2580 or BMT from CCR2-deficient donors, reduced perineurial invasion. Deletion of GDNF expression by perineurial macrophages, or inhibition of RET with shRNA or a small-molecule inhibitor, reduced perineurial invasion in KPC mice with PDAC. Taken together, our findings show that RET is upregulated during pancreas tumorigenesis and its activation induces cancer perineurial invasion. Trafficking of BM-derived macrophages to the perineurial microenvironment and secretion of GDNF are essential for pancreatic cancer neural spread.
Collapse
Affiliation(s)
- M Amit
- Head and Neck Surgery Department, MD Anderson Cancer Center University of Texas, Houston, TX, USA.,The Laboratory for Applied Cancer Research, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology, Haifa, Israel.,Department of Otolaryngology Head and Neck Surgery, The Head and Neck Center, Rambam Healthcare Campus, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, Rambam Medical Center, The Technion, Israel Institute of Technology, Haifa, Israel
| | - S Na'ara
- The Laboratory for Applied Cancer Research, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology, Haifa, Israel.,Department of Otolaryngology Head and Neck Surgery, The Head and Neck Center, Rambam Healthcare Campus, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, Rambam Medical Center, The Technion, Israel Institute of Technology, Haifa, Israel
| | - L Leider-Trejo
- Department of Pathology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Y Binenbaum
- The Laboratory for Applied Cancer Research, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology, Haifa, Israel
| | - N Kulish
- The Laboratory for Applied Cancer Research, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology, Haifa, Israel
| | - E Fridman
- The Laboratory for Applied Cancer Research, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology, Haifa, Israel.,Department of Otolaryngology Head and Neck Surgery, The Head and Neck Center, Rambam Healthcare Campus, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, Rambam Medical Center, The Technion, Israel Institute of Technology, Haifa, Israel
| | - A Shabtai-Orbach
- The Laboratory for Applied Cancer Research, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology, Haifa, Israel
| | - R J Wong
- Department of Surgery Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Z Gil
- The Laboratory for Applied Cancer Research, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology, Haifa, Israel.,Department of Otolaryngology Head and Neck Surgery, The Head and Neck Center, Rambam Healthcare Campus, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, Rambam Medical Center, The Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
37
|
Histological and Pathological Assessment of miR-204 and SOX4 Levels in Gastric Cancer Patients. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6894675. [PMID: 28133610 PMCID: PMC5241485 DOI: 10.1155/2017/6894675] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/15/2016] [Accepted: 11/29/2016] [Indexed: 01/05/2023]
Abstract
Gastric cancer is one of the most common cancers and the efficient therapeutic methods are limited. Further study of the exact molecular mechanism of gastric cancer to develop novel targeted therapies is necessary and urgent. We herein systematically examined that miR-204 suppressed both proliferation and metastasis of gastric cancer AGS cells. miR-204 directly targeted SOX4. In clinical tissue research, we determined that miR-204 was expressed much lower and SOX4 expressed much higher in gastric cancer tissues compared with normal gastric tissues. Associated analysis with clinicopathological parameters in gastric cancer patients showed miR-204 was associated with no lymph node metastasis and early tumor stages whereas SOX4 was associated with lymph node metastasis and advanced tumor stages. In addition, miR-204 and SOX4 were negatively correlated in tissues from gastric cancer patients. Our findings examined the important role of miR-204 and SOX4 played in gastric cancer, and they could be used as candidate therapeutic targets for gastric cancer therapy.
Collapse
|
38
|
Lian EY, Maritan SM, Cockburn JG, Kasaian K, Crupi MJF, Hurlbut D, Jones SJM, Wiseman SM, Mulligan LM. Differential roles of RET isoforms in medullary and papillary thyroid carcinomas. Endocr Relat Cancer 2017; 24:53-69. [PMID: 27872141 DOI: 10.1530/erc-16-0393] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 12/25/2022]
Abstract
The RET receptor tyrosine kinase mediates cell proliferation, survival and migration in embryogenesis and is implicated in the transformation and tumour progression in multiple cancers. RET is frequently mutated and constitutively activated in familial and sporadic thyroid carcinomas. As a result of alternative splicing, RET is expressed as two protein isoforms, RET9 and RET51, which differ in their unique C-terminal amino acids. These isoforms have distinct intracellular trafficking and associated signalling complexes, but functional differences are not well defined. We used shRNA-mediated knockdown (KD) of individual RET isoforms or of total RET to evaluate their functional contributions in thyroid carcinoma cells. We showed that RET is required for cell survival in medullary (MTC) but not papillary thyroid carcinoma (PTC) cells. In PTC cells, RET depletion reduced cell migration and induced a flattened epithelial-like morphology. RET KD decreased the expression of mesenchymal markers and matrix metalloproteinases and reduced anoikis resistance and invasive potential. Further, we showed that RET51 depletion had significantly greater effects on each of these processes than RET9 depletion in both MTC and PTC cells. Finally, we showed that expression of RET, particularly RET51, was correlated with malignancy in a panel of human thyroid tumour tissues. Together, our data show that RET expression promotes a more mesenchymal phenotype with reduced cell-cell adhesion and increased invasiveness in PTC cell models, but is more important for tumour cell survival, proliferation and anoikis resistance in MTC models. Our data suggest that the RET51 isoform plays a more prominent role in mediating these processes compared to RET9.
Collapse
Affiliation(s)
- Eric Y Lian
- Division of Cancer Biology and GeneticsCancer Research Institute, Queen's University, Kingston, Ontario, Canada
- Department of Pathology & Molecular MedicineQueen's University, Kingston, Ontario, Canada
| | - Sarah M Maritan
- Division of Cancer Biology and GeneticsCancer Research Institute, Queen's University, Kingston, Ontario, Canada
- Department of Pathology & Molecular MedicineQueen's University, Kingston, Ontario, Canada
| | - Jessica G Cockburn
- Division of Cancer Biology and GeneticsCancer Research Institute, Queen's University, Kingston, Ontario, Canada
- Department of Pathology & Molecular MedicineQueen's University, Kingston, Ontario, Canada
| | - Katayoon Kasaian
- Michael Smith Genome Sciences CentreBritish Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Mathieu J F Crupi
- Division of Cancer Biology and GeneticsCancer Research Institute, Queen's University, Kingston, Ontario, Canada
- Department of Pathology & Molecular MedicineQueen's University, Kingston, Ontario, Canada
| | - David Hurlbut
- Department of Pathology & Molecular MedicineQueen's University, Kingston, Ontario, Canada
| | - Steven J M Jones
- Michael Smith Genome Sciences CentreBritish Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Medical GeneticsUniversity of British Columbia, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Sam M Wiseman
- Department of SurgerySt Paul's Hospital & University of British Columbia, Vancouver, British Columbia, Canada
| | - Lois M Mulligan
- Division of Cancer Biology and GeneticsCancer Research Institute, Queen's University, Kingston, Ontario, Canada
- Department of Pathology & Molecular MedicineQueen's University, Kingston, Ontario, Canada
| |
Collapse
|
39
|
Sebastian A, Pandey V, Mohan CD, Chia YT, Rangappa S, Mathai J, Baburajeev CP, Paricharak S, Mervin LH, Bulusu KC, Fuchs JE, Bender A, Yamada S, Lobie PE, Rangappa KS. Novel Adamantanyl-Based Thiadiazolyl Pyrazoles Targeting EGFR in Triple-Negative Breast Cancer. ACS OMEGA 2016; 1:1412-1424. [PMID: 30023509 PMCID: PMC6044684 DOI: 10.1021/acsomega.6b00251] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 12/15/2016] [Indexed: 06/08/2023]
Abstract
The epidermal growth factor receptor (EGFR) is a validated therapeutic target for triple-negative breast cancer (TNBC). In the present study, we synthesize novel adamantanyl-based thiadiazolyl pyrazoles by introducing the adamantane ring to thiazolopyrazoline. On the basis of loss of cell viability in TNBC cells, 4-(adamantan-1-yl)-2-(3-(2,4-dichlorophenyl)-5-phenyl-4,5-dihydro-1H-pyrazol-1-yl)thiazole (APP) was identified as a lead compound. Using a Parzen-Rosenblatt Window classifier, APP was predicted to target the EGFR protein, and the same was confirmed by surface plasmon resonance. Further analysis revealed that APP suppressed the phosphorylation of EGFR at Y992, Y1045, Y1068, Y1086, Y1148, and Y1173 in TNBC cells. APP also inhibited the phosphorylation of ERK at Y204 and of STAT3 at Y705, implying that APP downregulates the activity of EGFR downstream effectors. Small interfering RNA mediated depletion of EGFR expression prevented the effect of APP in BT549 and MDA-MB-231 cells, indicating that APP specifically targets the EGFR. Furthermore, APP modulated the expression of the proteins involved in cell proliferation and survival. In addition, APP altered the expression of epithelial-mesenchymal transition related proteins and suppressed the invasion of TNBC cells. Hence, we report a novel and specific inhibitor of the EGFR signaling cascade.
Collapse
Affiliation(s)
- Anusha Sebastian
- Laboratory
of Chemical Biology, Department of Chemistry, Bangalore University, Central College campus, Palace Road, Bangalore560001, India
| | - Vijay Pandey
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, 14 Medical Drive #11-02, MD6, Singapore 117599, Singapore
| | - Chakrabhavi Dhananjaya Mohan
- Department of Studies in Chemistry and Department of Studies in Molecular
Biology, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Yi Ting Chia
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, 14 Medical Drive #11-02, MD6, Singapore 117599, Singapore
| | - Shobith Rangappa
- Frontier
Research Center for Post-Genome Science and Technology, Hokkaido University, Sapporo 060-0808, Japan
| | - Jessin Mathai
- Centre
for Advanced Biomedical Research and Innovation, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - C. P. Baburajeev
- Laboratory
of Chemical Biology, Department of Chemistry, Bangalore University, Central College campus, Palace Road, Bangalore560001, India
| | - Shardul Paricharak
- Department
of Chemistry, Centre for Molecular Informatics, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
- Division
of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, P.O.
Box 9502, Leiden 2300 RA, The Netherlands
| | - Lewis H. Mervin
- Department
of Chemistry, Centre for Molecular Informatics, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Krishna C. Bulusu
- Department
of Chemistry, Centre for Molecular Informatics, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Julian E. Fuchs
- Department
of Chemistry, Centre for Molecular Informatics, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Andreas Bender
- Department
of Chemistry, Centre for Molecular Informatics, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Shuhei Yamada
- Department
of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoyo 468-8503, Japan
| | - Peter E. Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, 14 Medical Drive #11-02, MD6, Singapore 117599, Singapore
| | - Kanchugarakoppal S. Rangappa
- Department of Studies in Chemistry and Department of Studies in Molecular
Biology, University of Mysore, Manasagangotri, Mysore 570006, India
| |
Collapse
|
40
|
Zhang M, Zhang W, Wu Z, Liu S, Sun L, Zhong Y, Zhang X, Kong X, Qian P, Zhang H, Lobie PE, Zhu T. Artemin is hypoxia responsive and promotes oncogenicity and increased tumor initiating capacity in hepatocellular carcinoma. Oncotarget 2016; 7:3267-82. [PMID: 26675549 PMCID: PMC4823105 DOI: 10.18632/oncotarget.6572] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/21/2015] [Indexed: 12/28/2022] Open
Abstract
Hypoxia has been reported to regulate the cancer stem cell (CSC) population yet the underlying mechanism is poorly characterized. Herein, we show that Artemin (ARTN), a member of the glial cell derived neurotrophic factor family of ligands, is a hypoxia-responsive factor and is essential for hypoxia-induced CSC expansion in hepatocellular carcinoma (HCC). Clinically, elevated expression of ARTN in HCC was associated with larger tumor size, faster relapse and shorter survival. In vitro, HCC cells with forced expression of ARTN exhibited reduced apoptosis, increased proliferation, epithelial-mesenchymal transition (EMT) and enhanced motility. Additionally, ARTN dramatically increased xenograft tumor size and metastasis in vivo. Moreover, ARTN also enhanced tumorsphere formation and the tumor initiating capacity of HCC cells, consequent to expansion of the CD133+ CSC population. ARTN transcription was directly activated by hypoxia-induced factor-1α (HIF-1α) and hypoxia induced ARTN promoted EMT and increased the CSC population via AKT signaling. We herein identify a novel HIF-1α/ARTN axis promoting CSC-like behavior in hypoxic environments which implicates ARTN as a valuable therapeutic target for HCC.
Collapse
Affiliation(s)
- Min Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Weijie Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Zhengsheng Wu
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Shumin Liu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Linchong Sun
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Yanghao Zhong
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Xiao Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Xiangjun Kong
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Pengxu Qian
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Huafeng Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore
| | - Tao Zhu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| |
Collapse
|
41
|
|
42
|
Rivero J, Henríquez-Hernández LA, Luzardo OP, Pestano J, Zumbado M, Boada LD, Valerón PF. Differential gene expression pattern in human mammary epithelial cells induced by realistic organochlorine mixtures described in healthy women and in women diagnosed with breast cancer. Toxicol Lett 2016; 246:42-8. [DOI: 10.1016/j.toxlet.2016.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 02/05/2023]
|
43
|
Crupi MJF, Yoganathan P, Bone LN, Lian E, Fetz A, Antonescu CN, Mulligan LM. Distinct Temporal Regulation of RET Isoform Internalization: Roles of Clathrin and AP2. Traffic 2015; 16:1155-73. [DOI: 10.1111/tra.12315] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 08/19/2015] [Accepted: 08/19/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Mathieu J. F. Crupi
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine; Queen's University; Kingston Ontario K7L 3N6 Canada
| | - Piriya Yoganathan
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine; Queen's University; Kingston Ontario K7L 3N6 Canada
| | - Leslie N. Bone
- Department of Chemistry and Biology; Ryerson University; Toronto Ontario M5B 2K3 Canada
| | - Eric Lian
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine; Queen's University; Kingston Ontario K7L 3N6 Canada
| | - Andrew Fetz
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine; Queen's University; Kingston Ontario K7L 3N6 Canada
| | - Costin N. Antonescu
- Department of Chemistry and Biology; Ryerson University; Toronto Ontario M5B 2K3 Canada
| | - Lois M. Mulligan
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine; Queen's University; Kingston Ontario K7L 3N6 Canada
| |
Collapse
|
44
|
Pundavela J, Roselli S, Faulkner S, Attia J, Scott RJ, Thorne RF, Forbes JF, Bradshaw RA, Walker MM, Jobling P, Hondermarck H. Nerve fibers infiltrate the tumor microenvironment and are associated with nerve growth factor production and lymph node invasion in breast cancer. Mol Oncol 2015; 9:1626-35. [PMID: 26009480 DOI: 10.1016/j.molonc.2015.05.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 05/01/2015] [Accepted: 05/04/2015] [Indexed: 01/15/2023] Open
Abstract
Infiltration of the tumor microenvironment by nerve fibers is an understudied aspect of breast carcinogenesis. In this study, the presence of nerve fibers was investigated in a cohort of 369 primary breast cancers (ductal carcinomas in situ, invasive ductal and lobular carcinomas) by immunohistochemistry for the neuronal marker PGP9.5. Isolated nerve fibers (axons) were detected in 28% of invasive ductal carcinomas as compared to only 12% of invasive lobular carcinomas and 8% of ductal carcinomas in situ (p = 0.0003). In invasive breast cancers, the presence of nerve fibers was observed in 15% of lymph node negative tumors and 28% of lymph node positive tumors (p = 0.0031), indicating a relationship with the metastatic potential. In addition, there was an association between the presence of nerve fibers and the expression of nerve growth factor (NGF) in cancer cells (p = 0.0001). In vitro, breast cancer cells were able to induce neurite outgrowth in PC12 cells, and this neurotrophic activity was partially inhibited by anti-NGF blocking antibodies. In conclusion, infiltration by nerve fibers is a feature of the tumor microenvironment that is associated with aggressiveness and involves NGF production by cancer cells. The potential participation of nerve fibers in breast cancer progression needs to be further considered.
Collapse
Affiliation(s)
- Jay Pundavela
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Severine Roselli
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Sam Faulkner
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - John Attia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Rick F Thorne
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - John F Forbes
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Ralph A Bradshaw
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA
| | - Marjorie M Walker
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Phillip Jobling
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
45
|
Lin L, Yuan J, Sander B, Golas MM. In Vitro Differentiation of Human Neural Progenitor Cells Into Striatal GABAergic Neurons. Stem Cells Transl Med 2015; 4:775-88. [PMID: 25972145 DOI: 10.5966/sctm.2014-0083] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 03/05/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED : Huntington's disease (HD) results from a CAG repeat expansion in the gene encoding the huntingtin protein. This inherited disorder is characterized by progressive neurodegeneration. In particular, HD progression involves the loss of striatal projection neurons. The limited availability of reliable sources of human striatal projection neurons currently hampers our understanding of HD mechanisms and hinders the development of novel HD treatments. In this paper, we described two- and three-step methods for differentiating human neural progenitor cells toward striatal projection neurons. In the two-step differentiation protocol, 90%, 54%, and 6% of MAP2-positive cells were immunopositive for GABA, calbindin (CALB1), and DARPP-32/PPP1R1B, respectively. In the three-step differentiation protocol, 96%, 84%, and 21% of MAP2-positive cells were immunopositive for GABA, calbindin, and DARPP-32/PPP1R1B, respectively. In line with a striatal projection neuron phenotype, cells differentiated with our protocols displayed significantly increased expression of MAP2, CALB1, DARPP-32/PPP1R1B, ARPP21, and CTIP2. Application of glutamate receptor agonists induced calcium influx; accordingly, the cells also expressed various ionotropic glutamate receptor subunits. Differentiated cells also released GABA on stimulation. We suggest that our three-step differentiation protocol presents a reliable and simplified method for the generation of striatal projection neurons, yielding a critical resource for neuronal physiology and neurodegenerative disorder studies. SIGNIFICANCE The earliest changes in the neurodegenerative disorder Huntington's disease affect a specific type of brain neurons, the so-called medium spiny neurons of the striatum. In this study, two protocols were developed for the differentiation of neural progenitor cells into striatal medium spiny neurons, and the differentiated neurons were extensively characterized. The data indicate that the three-step differentiation protocol presents a reliable and simplified method for the generation of striatal medium spiny neurons. The generated striatal medium spiny neurons could represent a critical resource for the study of neurodegenerative disorders, a model system for drug discovery, and a step toward cell-based regeneration therapies.
Collapse
Affiliation(s)
- Lin Lin
- Department of Biomedicine, Stereology and Electron Microscopy Laboratory, Department of Clinical Medicine, and Center for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Juan Yuan
- Department of Biomedicine, Stereology and Electron Microscopy Laboratory, Department of Clinical Medicine, and Center for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Bjoern Sander
- Department of Biomedicine, Stereology and Electron Microscopy Laboratory, Department of Clinical Medicine, and Center for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Monika M Golas
- Department of Biomedicine, Stereology and Electron Microscopy Laboratory, Department of Clinical Medicine, and Center for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| |
Collapse
|
46
|
Abstract
OBJECTIVES The aim of this study was to investigate the effect of the neurotrophic factor Artemin on neuroplasticity and perineural invasion of pancreatic adenocarcinoma. METHODS Artemin expressions were detected in human pancreatic adenocarcinoma tissues by Western blot and immunohistochemistry. Artemin overexpression and RNA interference in the pancreatic cancer cell lines were performed to evaluate the effects of Artemin on cell proliferation, invasion, and neurotrophic activity in vitro and in nude orthotopic transplantation tumor models. RESULTS Artemin expression in pancreatic cancer tissues was related to the incidence of lymphatic metastasis and perineural invasion as well as the mean density and total area of nerve fibers. Overexpression of Artemin in pancreatic cancer cell lines improved colony formation, cell migration, matrigel invasion, and neurotrophic activity in vitro. This overexpression also increased the volume of nude orthotopic transplantation tumors; promoted cancer cell invasion of the peripheral organs, nerves, vessels, and lymph nodes; and stimulated the proliferation of peritumoral nerve fibers. Artemin depletion by RNA interference had an inhibitory effect mentioned previously. CONCLUSIONS Artemin could promote invasiveness and neurotrophic function of pancreatic adenocarcinoma in vivo and in vitro. Therefore, Artemin could be used as a new therapeutic target of pancreatic carcinoma.
Collapse
|
47
|
Jiménez A, López-Ornelas A, Estudillo E, González-Mariscal L, González RO, Segovia J. A soluble form of GAS1 inhibits tumor growth and angiogenesis in a triple negative breast cancer model. Exp Cell Res 2014; 327:307-17. [DOI: 10.1016/j.yexcr.2014.06.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 06/04/2014] [Accepted: 06/22/2014] [Indexed: 12/18/2022]
|
48
|
Pallanti S, Tofani T, Zanardelli M, Di Cesare Mannelli L, Ghelardini C. BDNF and Artemin are increased in drug-naïve non-depressed GAD patients: preliminary data. Int J Psychiatry Clin Pract 2014; 18:255-60. [PMID: 24994477 DOI: 10.3109/13651501.2014.940051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE While the role of neuronal and glial plasticity are well established in the pathophysiology of mood disorders, the pattern and measures of neuronal and glial cell line-derived neurotrophic factors are unknown in generalized anxiety disorder (GAD). The present study evaluates brain-derived neurotrophic factor (BDNF) and Artemin (ARTN) plasma levels in GAD patients. METHODS Fourteen drug-naïve GAD patients without major depression were enrolled and plasmatic levels of BDNF and ARTN mRNA were measured by RT-PCR, and compared to matched healthy controls. RESULTS The results showed an unexpected increase in mRNA levels of both BDNF and ARTN in patients with GAD, that appeared almost doubled when compared to healthy controls. In comparison, both BDNF and ARTN are reduced in patients with major depressive disorder. Further, the results are intriguing and might involve distinguishing pathophysiological pathways. CONCLUSIONS This is the first report of increased levels of a neurotrophic factor and of a glial cell line-derived neurotrophic factor family member in GAD patients. While further studies to confirm these results and the functional meaning in terms of pathophysiology of GAD are needed, the potential conceptual and clinical meanings are discussed.
Collapse
Affiliation(s)
- Stefano Pallanti
- UC Davis Department of Psychiatry and Behavioral Sciences , Sacramento, CA , USA
| | | | | | | | | |
Collapse
|
49
|
Gao C, Li X, Tong B, Wu K, Liu Y, Anniko M, Duan M. Up-regulated expression of Dicer reveals poor prognosis in laryngeal squamous cell carcinoma. Acta Otolaryngol 2014; 134:959-63. [PMID: 25123850 DOI: 10.3109/00016489.2014.920962] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CONCLUSIONS Increased expression of Dicer may be a prognostic biomarker for patients with laryngeal squamous cell carcinoma (LSCC). OBJECTIVES Recent studies have shown that many microRNAs (miRNAs) play an important role in the development and progression of human cancers. Dicer, one of the most important enzymes of the miRNA machinery, performs the final step of biogenesis of miRNAs. This study aimed to investigate the impact of Dicer expression on patient survival in human LSCC. METHODS We detected the expression of Dicer in larynx tissue specimens from 76 LSCC samples and 26 polyps by immunohistochemistry. The clinicopathological and prognostic significance of Dicer expression was investigated in LSCC. RESULTS Our data showed that the expression of Dicer was significantly higher in the LSCC than in the polyp tissue specimens. Moreover, the expression level of Dicer was significantly associated with the pTNM stage and tumor lymph node metastasis. Kaplan-Meier survival analyses revealed a strong association between tumor Dicer expression and the survival of the patients with LSCC.
Collapse
Affiliation(s)
- Chaobing Gao
- Department of Otorhinolaryngology, Head and Neck Surgery, First Affiliated Hospital of AnHui Medical University , Hefei , China
| | | | | | | | | | | | | |
Collapse
|
50
|
Gao C, Cheng X, Li X, Tong B, Wu K, Liu Y. Prognostic significance of artemin and GFRα1 expression in laryngeal squamous cell carcinoma. Exp Ther Med 2014; 8:818-822. [PMID: 25120606 PMCID: PMC4113528 DOI: 10.3892/etm.2014.1821] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 06/05/2014] [Indexed: 12/13/2022] Open
Abstract
Artemin (ARTN) has been implicated in the development and progression of several human malignancies. However, the clinical and prognostic significance of ARTN and its receptors has not yet been investigated in human laryngeal squamous cell carcinoma (LSCC). Therefore, in the present study, the protein expression of ARTN and its receptor, namely GFRα1, was determined in 76 LSCC and 26 laryngeal polyp tissue samples using immunohistochemistry. Furthermore, the clinicopathological and prognostic significance of ARTN and GFRα1 expression was analyzed in patients with LSCC. The results revealed that the expression of ARTN and GFRα1 was significantly increased in LSCC compared with polyp tissue samples. Furthermore, the expression of ARTN and GFRα1 was positively associated with pTNM stage in LSCC. Kaplan-Meier survival analyses revealed a strong association between the expression of ARTN or GFRα1 and the survival of patients with LSCC. Correlation analysis demonstrated that the expression of ARTN was significantly correlated with the expression GFRα1. In conclusion, the results demonstrated that ARTN and GFRα1 may be useful predictors of disease progression and outcome in patients with LSCC.
Collapse
Affiliation(s)
- Chaobing Gao
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xingwang Cheng
- Department of Emergency, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233030, P.R. China
| | - Xiaohong Li
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Busheng Tong
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Kaile Wu
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yehai Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|