1
|
Repasky E, Mohammadpour H. Targeting nerve growth factor: an Achilles' heel for tumors? J Immunother Cancer 2025; 13:e011609. [PMID: 40316307 PMCID: PMC12049907 DOI: 10.1136/jitc-2025-011609] [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: 01/27/2025] [Accepted: 04/01/2025] [Indexed: 05/04/2025] Open
Abstract
A tumor's ability to attract innervation is a critical factor in tumor progression and immune escape, with the sympathetic nervous system playing a major role. Catecholamines released by sympathetic nerves activate adrenergic receptors on tumor cells, enhancing growth and resistance to therapies, while activation of adrenergic receptors on immune cells triggers immunosuppressive activity in the tumor microenvironment. Nerve growth factor (NGF) produced by tumor cells is a key driver of tumor innervation, making it a promising target for novel therapeutic strategies. In this commentary, we highlight a recent study by Yang et al, which examines NGF single-chain variable fragment (scFv)-secreting chimeric antigen receptor(CAR) T cells and the impact of NGF neutralization by CAR T cells on CAR T-cell function and the remodeling of the tumor microenvironment. This work shows that we may be able to exploit a tumor-derived survival factor as a vulnerability and a means to enhance antitumor immune activity.
Collapse
Affiliation(s)
- Elizabeth Repasky
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Hemn Mohammadpour
- Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| |
Collapse
|
2
|
Li C, Cheng D, Li P. Androgen receptor dynamics in prostate cancer: from disease progression to treatment resistance. Front Oncol 2025; 15:1542811. [PMID: 40008000 PMCID: PMC11850250 DOI: 10.3389/fonc.2025.1542811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Prostate cancer is the most common cancer among men worldwide, especially in those over 65, and is a leading cause of cancer-related mortality. The disease typically advances from an androgen-dependent state to castration-resistant prostate cancer (CRPC), which poses significant treatment challenges. The androgen receptor (AR) on the X chromosome is a central driver in this process, activating genes that govern proliferation and survival. Mutations and amplifications of the AR are closely associated with disease progression and treatment resistance. While traditional therapies such as androgen deprivation therapy (ADT) and AR antagonists like enzalutamide have been effective, resistance persists due to reactivation of AR signaling through mechanisms like ligand-independent activation. Recent research highlights the role of epigenetic modifications in enhancing AR activity and drug resistance. The tumor microenvironment, particularly interactions with cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), further complicates treatment by promoting aggressive tumor behavior and immune evasion. Future directions include developing next-generation AR antagonists, identifying AR-related biomarkers for personalized therapy, and exploring combinations with immune checkpoint inhibitors. Additionally, basal cell-lumen-derived organoids provide innovative models that can enhance understanding and treatment strategies in prostate cancer.
Collapse
Affiliation(s)
| | | | - Peng Li
- Center for Reproductive Medicine, Shenyang Jinghua Hospital, Shenyang, China
| |
Collapse
|
3
|
Di Donato M, Cristiani CM, Capone M, Garofalo C, Madonna G, Passacatini LC, Ottaviano M, Ascierto PA, Auricchio F, Carbone E, Migliaccio A, Castoria G. Role of the androgen receptor in melanoma aggressiveness. Cell Death Dis 2025; 16:34. [PMID: 39837817 PMCID: PMC11751086 DOI: 10.1038/s41419-025-07350-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Malignant melanoma represents the fifth most common cancer in the world and its incidence is rising. Novel therapies targeting receptor tyrosine kinases, kinases and immune checkpoints have been employed with a significant improvement of the overall survival and long-term disease containment. Nevertheless, the disease often progresses and becomes resistant to the therapies. As such, the discovery of new targets and drugs for advanced melanoma still remains a difficult task. Gender disparities, with a female advantage in melanoma incidence and outcome, have been reported. Although emerging studies support the pro-tumorigenic role of androgen/androgen receptor axis in melanoma, the molecular bases of such evidence are still under intense investigation. We now report that ligand activation of the androgen receptor drives melanoma invasiveness and its escape from natural killer-mediated cytotoxic effect. By combining different experimental approaches, we observe that melanoma escape is mediated by the androgen-triggered shedding of the surface molecule MICA. Specific blockade of ADAM10 or androgen receptor impairs the androgen-induced MICA shedding and melanoma immune-escape. Further, the increase in MICA serum levels correlates with a poor outcome in melanoma patients treated with the anti-PD-1 monoclonal antibody, pembrolizumab. At last, melanoma cells depleted of the androgen receptor become more responsive to the most commonly used immunocheckpoint inhibitors, suggesting that the receptor dampens the immunotherapy efficacy. Taken together, our findings identify the androgen receptor as a diagnostic guidance in melanoma and support the repositioning of AR blockers in clinical management of patients.
Collapse
Affiliation(s)
- Marzia Di Donato
- Department of Precision Medicine, University of Campania 'L. Vanvitelli'- Via L. De Crecchio 7, 80138, Naples, Italy
| | - Costanza Maria Cristiani
- Neuroscience Research Center, Department of Medical and Surgical Sciences - 'Magna Graecia' University of Catanzaro, 88100, Catanzaro, Italy
| | - Mariaelena Capone
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS- Fondazione "G. Pascale", Napoli, Italy
| | - Cinzia Garofalo
- Department of Experimental and Clinical Medicine, 'Magna Graecia' University of Catanzaro, 88100, Catanzaro, Italy
| | - Gabriele Madonna
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS- Fondazione "G. Pascale", Napoli, Italy
| | | | - Margaret Ottaviano
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS- Fondazione "G. Pascale", Napoli, Italy
| | - Paolo Antonio Ascierto
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS- Fondazione "G. Pascale", Napoli, Italy
| | - Ferdinando Auricchio
- Department of Precision Medicine, University of Campania 'L. Vanvitelli'- Via L. De Crecchio 7, 80138, Naples, Italy
| | - Ennio Carbone
- Department of Precision Medicine, University of Campania 'L. Vanvitelli'- Via L. De Crecchio 7, 80138, Naples, Italy
| | - Antimo Migliaccio
- Department of Precision Medicine, University of Campania 'L. Vanvitelli'- Via L. De Crecchio 7, 80138, Naples, Italy.
| | - Gabriella Castoria
- Department of Precision Medicine, University of Campania 'L. Vanvitelli'- Via L. De Crecchio 7, 80138, Naples, Italy.
| |
Collapse
|
4
|
Morado-Urbina CE, Kato J, Sandor K, Vazquez-Mora JA, Ängeby Möller K, Simon N, Salcido J, Martinez-Martinez A, Munoz-Islas E, Jimenez-Andrade JM, Svensson CI. Sex-dependent effects of the targeted nerve growth factor mutation (R100E) on pain behavior, joint inflammation, and bone erosion in mice. Pain 2024; 165:2814-2828. [PMID: 39324959 PMCID: PMC11562760 DOI: 10.1097/j.pain.0000000000003343] [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: 10/20/2023] [Revised: 05/10/2024] [Accepted: 06/07/2024] [Indexed: 09/27/2024]
Abstract
ABSTRACT Nerve growth factor (NGF)-R100E is a mutated form of human recombinant NGF that reduces the binding of NGF to its p75NTR receptor while retaining its affinity toward the TrkA receptor. Here, we used human wild type NGF and NGF-R100E knock-in mice to investigate the effects of this NGF mutation on inflammation-induced pain-related behaviors and bone loss. The hNGF-R100E mutation did not alter the nerve fiber density in the sciatic nerve, ankle joint synovium, and skin of naïve mice. Withdrawal responses to mechanical, thermal, and cold stimuli before and after joint inflammation induced by intra-articular injection of complete Freund adjuvant (CFA) were similar between human recombinant nerve growth factor-wild type and hNGF-R100E male and female mice while weight bearing and gait analysis revealed significant differences. Intriguingly, hNGF-R100E male and female mice showed only mild changes, indicating lower degrees of deep joint-related pain compared to their wild type counterparts. Furthermore, micro-CT analysis demonstrated that hNGF-R100E female mice, but not males, were protected from CFA-induced bone loss, and mRNA analysis showed a different gene regulation indicating a sex-dependent relationship between NGF, inflammation, and bone loss. In conclusion, our study reveals that the hNGF-R100E mutation renders mice insensitive to inflammation-induced impact on joint loading and gait while preserving the development of the peripheral nociceptive neurons and sensitivity to punctate stimulation of the skin. Notably, the mutation uncovers a sex-dependent relationship between NGF and inflammation-induced bone loss. These findings offer valuable insights into NGF as a target for pain management and the interplay between NGF and bone architecture.
Collapse
Affiliation(s)
- Carlos E. Morado-Urbina
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Jungo Kato
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
- Department of Anesthesiology, Keio University School of Medicine, Tokyo, Japan
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Juan Antonio Vazquez-Mora
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Kristina Ängeby Möller
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Nils Simon
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Jaira Salcido
- Unidad Académica Multidisciplinaria Reynosa Aztlán, Universidad Autónoma de Tamaulipas, Reynosa, México
| | - Arisai Martinez-Martinez
- Unidad Académica Multidisciplinaria Reynosa Aztlán, Universidad Autónoma de Tamaulipas, Reynosa, México
| | - Enriqueta Munoz-Islas
- Unidad Académica Multidisciplinaria Reynosa Aztlán, Universidad Autónoma de Tamaulipas, Reynosa, México
| | | | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
5
|
Agbana S, McIlroy M. Extra-nuclear and cytoplasmic steroid receptor signalling in hormone dependent cancers. J Steroid Biochem Mol Biol 2024; 243:106559. [PMID: 38823459 DOI: 10.1016/j.jsbmb.2024.106559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/03/2024]
Abstract
Steroid hormone receptors are key mediators in the execution of hormone action through a combination of genomic and non-genomic action. Since their isolation and characterisation in the early 20th Century much of our understanding of the biological actions of steroid hormones are underpinned by their activated receptor activity. Over the past two decades there has been an acceleration of more omics-based research which has resulted in a major uptick in our comprehension of genomic steroid action. However, it is well understood that steroid hormones can induce very rapid signalling events in tandem with their genomic actions wherein they exert their influence through alterations in gene expression. Thus the totality of genomic and non-genomic steroid action occurs in a simultaneous and reciprocal manner and a greater appreciation of whole cell action is required to fully evaluate steroid hormone activity in vivo. In this mini-review we outline the most recent developments in non-genomic steroid action and cytoplasmic steroid hormone receptor biology in endocrine-related cancers with a focus on the 3-keto steroid receptors, in particular the androgen receptor.
Collapse
Affiliation(s)
- Stephanie Agbana
- Androgens in Health and Disease research group, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Department of Surgery, RCSI University of Medicine and Health Sciences, Ireland
| | - Marie McIlroy
- Androgens in Health and Disease research group, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Department of Surgery, RCSI University of Medicine and Health Sciences, Ireland.
| |
Collapse
|
6
|
Andolfi C, Bartolini C, Morales E, Gündoğdu B, Puhr M, Guzman J, Wach S, Taubert H, Aigner A, Eder IE, Handle F, Culig Z. MED12 and CDK8/19 Modulate Androgen Receptor Activity and Enzalutamide Response in Prostate Cancer. Endocrinology 2024; 165:bqae114. [PMID: 39253786 PMCID: PMC11398899 DOI: 10.1210/endocr/bqae114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/16/2024] [Accepted: 09/08/2024] [Indexed: 09/11/2024]
Abstract
Prostate cancer progression is driven by androgen receptor (AR) activity, which is a target for therapeutic approaches. Enzalutamide is an AR inhibitor that prolongs the survival of patients with advanced prostate cancer. However, resistance mechanisms arise and impair its efficacy. One of these mechanisms is the expression of AR-V7, a constitutively active AR splice variant. The Mediator complex is a multisubunit protein that modulates gene expression on a genome-wide scale. MED12 and cyclin-dependent kinase (CDK)8, or its paralog CDK19, are components of the kinase module that regulates the proliferation of prostate cancer cells. In this study, we investigated how MED12 and CDK8/19 influence cancer-driven processes in prostate cancer cell lines, focusing on AR activity and the enzalutamide response. We inhibited MED12 expression and CDK8/19 activity in LNCaP (AR+, enzalutamide-sensitive), 22Rv1 (AR-V7+, enzalutamide-resistant), and PC3 (AR-, enzalutamide-insensitive) cells. Both MED12 and CDK8/19 inhibition reduced cell proliferation in all cell lines, and MED12 inhibition reduced proliferation in the respective 3D spheroids. MED12 knockdown significantly inhibited c-Myc protein expression and signaling pathways. In 22Rv1 cells, it consistently inhibited the AR response, prostate-specific antigen (PSA) secretion, AR target genes, and AR-V7 expression. Combined with enzalutamide, MED12 inhibition additively decreased the AR activity in both LNCaP and 22Rv1 cells. CDK8/19 inhibition significantly decreased PSA secretion in LNCaP and 22Rv1 cells and, when combined with enzalutamide, additively reduced proliferation in 22Rv1 cells. Our study revealed that MED12 and CDK8/19 regulate AR activity and that their inhibition may modulate response to enzalutamide in prostate cancer.
Collapse
Affiliation(s)
- Chiara Andolfi
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Caterina Bartolini
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- University of Florence, 50 121 Florence, Italy
| | - Elisa Morales
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Johannes Gutenberg University Mainz, 55122 Mainz, Germany
| | - Büşra Gündoğdu
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Graudate School of Science and Engineering, Yıldız Technical University, 34220 Istanbul, Turkey
| | - Martin Puhr
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Juan Guzman
- Department of Urology and Pediatric Urology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Sven Wach
- Department of Urology and Pediatric Urology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Helge Taubert
- Department of Urology and Pediatric Urology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, 04107 Leipzig, Germany
| | - Iris E Eder
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Florian Handle
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Institute of Pathology, Neuropathology & Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Zoran Culig
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
7
|
Gebrael G, Jo Y, Swami U, Plets M, Hage Chehade C, Narang A, Gupta S, Myint ZW, Sayegh N, Tangen CM, Hussain M, Dorff T, Lara PN, Lerner SP, Thompson I, Agarwal N. Bone Pain and Survival Among Patients With Metastatic, Hormone-Sensitive Prostate Cancer: A Secondary Analysis of the SWOG-1216 Trial. JAMA Netw Open 2024; 7:e2419966. [PMID: 38980676 PMCID: PMC11234233 DOI: 10.1001/jamanetworkopen.2024.19966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/01/2024] [Indexed: 07/10/2024] Open
Abstract
Importance The presence of bone pain is significantly associated with worse overall survival (OS) in patients with castration-resistant prostate cancer. However, there are few data regarding bone pain and survival outcomes in the context of metastatic, hormone-sensitive prostate cancer (MHSPC). Objective To compare survival outcomes among patients with MHSPC by presence or absence of baseline bone pain at diagnosis. Design, Setting, and Participants This post hoc secondary analysis, conducted from September 1 to December 31, 2023, used patient-level data from SWOG-1216, a phase 3, prospective randomized clinical trial that enrolled patients with newly diagnosed MHSPC from 248 academic and community centers across the US from March 1, 2013, to July 15, 2017. All patients in the intention-to-treat population who had available bone pain status were eligible and included in this secondary analysis. Interventions In the SWOG-1216 trial, patients were randomized (1:1) to receive either androgen deprivation therapy (ADT) with orteronel, 300 mg orally twice daily (experimental group), or ADT with bicalutamide, 50 mg orally daily (control group), until disease progression, unacceptable toxic effects, or patient withdrawal. Main Outcomes and Measures Overall survival was the primary end point; progression-free survival (PFS) and prostate-specific antigen (PSA) response were secondary end points. Cox proportional hazards regression models were used for both univariable and multivariable analyses adjusting for age, treatment type, Gleason score, disease volume, Zubrod performance status, and PSA level. Results Of the 1279 male study participants, 301 (23.5%) had baseline bone pain at MHSPC diagnosis and 896 (70.1%) did not. Bone pain status was unavailable in 82 patients (6.4%). The median age of the 1197 patients eligible and included in this secondary analysis was 67.6 years (IQR, 61.8-73.6 years). Compared with patients who did not experience bone pain, those with baseline bone pain were younger (median age, 66.0 [IQR, 60.1-73.4] years vs 68.2 [IQR, 62.4-73.7] years; P = .02) and had a higher incidence of high-volume disease (212 [70.4%] vs 373 [41.6%]; P < .001). After adjustment, bone pain was associated with shorter PFS and OS. At a median follow-up of 4.0 years (IQR, 2.5-5.4 years), patients with bone pain had median PFS of 1.3 years (95% CI, 1.1-1.7 years) vs 3.7 years (95% CI, 3.3-4.2 years) in patients without initial bone pain (adjusted hazard ratio [AHR], 1.46; 95% CI, 1.22-1.74; P < .001) and OS of 3.9 years (95% CI, 3.3-4.8 years) vs not reached (NR) (95% CI, 6.6 years to NR) in patients without initial bone pain (AHR, 1.66; 95% CI, 1.34-2.05; P < .001). Conclusions and Relevance In this post hoc secondary analysis of the SWOG-1216 randomized clinical trial, patients with baseline bone pain at MHSPC diagnosis had worse survival outcomes than those without bone pain. These data suggest prioritizing these patients for enrollment in clinical trials, may aid patient counseling, and indicate that the inclusion of bone pain in prognostic models of MHSPC may be warranted. Trial Registration ClinicalTrials.gov Identifier: NCT01809691.
Collapse
Affiliation(s)
- Georges Gebrael
- Huntsman Cancer Institute at the University of Utah, Salt Lake City
| | - Yeonjung Jo
- Huntsman Cancer Institute at the University of Utah, Salt Lake City
| | - Umang Swami
- Huntsman Cancer Institute at the University of Utah, Salt Lake City
| | - Melissa Plets
- SWOG Statistics and Data Management Center, Seattle, Washington
| | | | - Arshit Narang
- Huntsman Cancer Institute at the University of Utah, Salt Lake City
| | - Shilpa Gupta
- Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| | - Zin W. Myint
- Department of Internal Medicine–Division of Medical Oncology, University of Kentucky, Lexington
| | - Nicolas Sayegh
- Huntsman Cancer Institute at the University of Utah, Salt Lake City
| | | | - Maha Hussain
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Tanya Dorff
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Primo N. Lara
- University of California, Davis Comprehensive Cancer Center, Sacramento
| | | | - Ian Thompson
- University of Texas Health Science Center at San Antonio, San Antonio
| | - Neeraj Agarwal
- Huntsman Cancer Institute at the University of Utah, Salt Lake City
| |
Collapse
|
8
|
Qian X, Liu E, Zhang C, Feng R, Tran N, Zhai W, Wang F, Qin Z. Promotion of perineural invasion of cholangiocarcinoma by Schwann cells via nerve growth factor. J Gastrointest Oncol 2024; 15:1198-1213. [PMID: 38989424 PMCID: PMC11231841 DOI: 10.21037/jgo-24-309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024] Open
Abstract
Background Cholangiocarcinoma (CCA), a highly lethal tumor of the hepatobiliary system originating from bile duct epithelium, can be divided into the intrahepatic, hilar, and extrahepatic types. Due to its insidious onset and atypical early clinical symptoms, the overall prognosis is poor. One of the important factors contributing to the poor prognosis of CCA is the occurrence of perineural invasion (PNI), but the specific mechanisms regarding how it contributes to the occurrence of PNI are still unclear. The main purpose of this study is to explore the molecular mechanism leading to the occurrence of PNI and provide new ideas for clinical treatment. Methods CCA cell lines and Schwann cells (SCs) were stimulated to observe the changes in cell behavior. SCs cocultured with tumor supernatant and SCs cultured in normal medium were subjected to transcriptome sequencing to screen the significantly upregulated genes. Following this, the two types of tumor cells were cultured with SC supernatant, and the changes in behavior of the tumor cells were observed. Nonobese diabetic-severe combined immunodeficiency disease (NOD-SCID) mice were injected with cell suspension supplemented with nerve growth factor (NGF) via the sciatic nerve. Four weeks later, the mice were euthanized and the tumor sections were removed and stained. Results Nerve invasion by tumor cells was common in CCA tissues. SCs were observed in tumor tissues, and the number of SCs in tumor tissues and the degree of PNI were much higher than were those in normal tissues or tissues without PNI. The overall survival time was shorter in patients with CCA with PNI than in patients without PNI. SCs were enriched in CCA tissues, indicating the presence of PNI and associated with poor prognosis in CCA patients. CCA was found to promote NGF secretion from SCs in vitro. After the addition of exogenous NGF in CCA cell culture medium, the proliferation activity and migration ability of CCA cells were significantly increased, suggesting that SCs can promote the proliferation and migration of CCA through the secretion of NGF. NGF, in turn, was observed to promote epithelial-mesenchymal transition in CCA through tropomyosin receptor kinase A (TrkA), thus promoting its progression. Tumor growth in mice shows that NGF can promote PNI in CCA. Conclusions In CCA, tumor cells can promote the secretion of NGF by SCs, which promotes the progression of CCA and PNI by binding to its high-affinity receptor TrkA, leading to poor prognosis.
Collapse
Affiliation(s)
- Xingwang Qian
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Enchi Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chong Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruo Feng
- Department of Histology and Embryology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Nguyen Tran
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Wenlong Zhai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Digestive Organs, Zhengzhou University, Zhengzhou, China
| | - Fazhan Wang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Fan Y, Zhang B, Du X, Wang B, Yan Q, Guo L, Yao W. Regulating Tumorigenicity and Cancer Metastasis through TRKA Signaling. Curr Cancer Drug Targets 2024; 24:271-287. [PMID: 37670705 DOI: 10.2174/1568009623666230904150957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/15/2023] [Accepted: 07/25/2023] [Indexed: 09/07/2023]
Abstract
Tropomyosin receptor kinase (TRK) A, TRKA, is a specific binding receptor of nerve growth factor (NGF), which plays an essential role in the occurrence and progression of human cancers. TRKA overexpression has been proven to be a powerful carcinogenic driver and has been verified in many tumors. The TRKA receptor kinase domain is over-activated in an NGF-dependent manner, accompanied by activation of downstream signal pathways, such as RAS-MAPK, PI3K-AKT, JAK2-STAT3 pathway, PLC γ pathway, and Hippo pathway, which participate in tumor cell proliferation, invasion, epithelial-mesenchymal transition (EMT), perineural invasion (PNI), drug resistance, and cancer pain. In addition, chimeric oncogenes produced by the fusion of NTRK1 and other genes are also the direct cause of tumorigenesis and cancer development. The newly developed TRK inhibitors can improve symptoms and tumor regression in cancer patients with overexpression of TRKA or NTRK1 fusion gene. With the emergence of drug resistance, next generation of TRK inhibitors can still maintain strong clinical efficacy in the case of TRK kinase domain mutations, and these inhibitors are in clinical trials. This review summarizes the characteristics and research progress of TRKA, focusing on the regulatory role of the TRKA signal pathway in different tumors. In addition, we have summarized the clinical significance of TRKA and the TRK inhibitors. This review may provide a new reference for the study of the mechanism of TRKA in different tumors, and also provide a new perspective for the in-depth understanding of the role of TRKA as a biomarker and therapeutic target in human cancer.
Collapse
Affiliation(s)
- Yichao Fan
- Henan Cancer Hospital, Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Boya Zhang
- Henan Cancer Hospital, Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xinhui Du
- Henan Cancer Hospital, Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Bangmin Wang
- Henan Cancer Hospital, Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Qiang Yan
- Henan Cancer Hospital, Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Liangyu Guo
- Henan Cancer Hospital, Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Weitao Yao
- Henan Cancer Hospital, Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
10
|
Di Donato M, Di Zazzo E, Salvati A, Sorrentino C, Giurato G, Fiore D, Proto MC, Rienzo M, Casamassimi A, Gazzerro P, Bifulco M, Castoria G, Weisz A, Nassa G, Abbondanza C. RIZ2 at the crossroad of the EGF/EGFR signaling in colorectal cancer. J Transl Med 2023; 21:736. [PMID: 37853459 PMCID: PMC10585774 DOI: 10.1186/s12967-023-04621-6] [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: 08/03/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most deadly and fourth most diagnosed cancer worldwide. Despite the progress in early diagnosis and advanced therapeutic options, CRC shows a poor prognosis with a 5 year survival rate of ~ 45%. PRDM2/RIZ, a member of PR/SET domain family (PRDM), expresses two main molecular variants, the PR-plus isoform (RIZ1) and the PR-minus (RIZ2). The imbalance in their expression levels in favor of RIZ2 is observed in many cancer types. The full length RIZ1 has been extensively investigated in several cancers where it acts as a tumor suppressor, whereas few studies have explored the RIZ2 oncogenic properties. PRDM2 is often target of frameshift mutations and aberrant DNA methylation in CRC. However, little is known about its role in CRC. METHODS We combined in-silico investigation of The Cancer Genome Atlas (TCGA) CRC datasets, cellular and molecular assays, transcriptome sequencing and functional annotation analysis to assess the role of RIZ2 in human CRC. RESULTS Our in-silico analysis on TCGA datasets confirmed that PRDM2 gene is frequently mutated and transcriptionally deregulated in CRC and revealed that a RIZ2 increase is highly correlated with a significant RIZ1 downregulation. Then, we assayed several CRC cell lines by qRT-PCR analysis for the main PRDM2 transcripts and selected DLD1 cell line, which showed the lowest RIZ2 levels. Therefore, we overexpressed RIZ2 in these cells to mimic TCGA datasets analysis results and consequently to assess the PRDM2/RIZ2 role in CRC. Data from RNA-seq disclosed that RIZ2 overexpression induced profound changes in CRC cell transcriptome via EGF pathway deregulation, suggesting that RIZ2 is involved in the EGF autocrine regulation of DLD1 cell behavior. Noteworthy, the forced RIZ2 expression increased cell viability, growth, colony formation, migration and organoid formation. These effects could be mediated by the release of high EGF levels by RIZ2 overexpressing DLD1 cells. CONCLUSIONS Our findings add novel insights on the putative RIZ2 tumor-promoting functions in CRC, although additional efforts are warranted to define the underlying molecular mechanism.
Collapse
Affiliation(s)
- Marzia Di Donato
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Erika Di Zazzo
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081, Baronissi, Italy
| | - Carmela Sorrentino
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081, Baronissi, Italy
- CRGS-Genome Research Center for Health, University of Salerno Campus of Medicine, 84081, Baronissi, Italy
| | - Donatella Fiore
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | | | - Monica Rienzo
- Department of Environmental, Biological, and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Amelia Casamassimi
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | | | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Gabriella Castoria
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081, Baronissi, Italy
- CRGS-Genome Research Center for Health, University of Salerno Campus of Medicine, 84081, Baronissi, Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081, Baronissi, Italy
- CRGS-Genome Research Center for Health, University of Salerno Campus of Medicine, 84081, Baronissi, Italy
| | - Ciro Abbondanza
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
11
|
Zhang Y, Li W, Guo S, Wu Z, Zhang L, Liu Y, Li X, Guo X, Cao J, Yang C, Wang Z. FBXO22 Mediates the NGF/TRKA Signaling Pathway in Bone Metastases in Prostate Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1248-1266. [PMID: 37301536 DOI: 10.1016/j.ajpath.2023.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/06/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023]
Abstract
Prostate cancer (PC) is a malignancy with high morbidity and mortality. Bone metastasis is the main driver of short survival time and difficulties in the treatment and prevention of PC. The goal of this study was to explore the biological function of E3 ubiquitin ligase F-box only protein 22 (FBXO22) in PC metastasis and its specific regulation mechanism. According to transcriptome sequencing, FBXO22 was overexpressed in PC tissues (versus adjacent tissues) and bone tissues (versus biopsied bone tissues without bone metastases). Fbxo22 down-regulation reduced bone metastases and macrophage M2 polarization in mice. FBXO22 was down-regulated in macrophages, and polarization was observed by flow cytometry. Macrophages were co-cultured with PC cells and osteoblasts to assess PC cell and osteoblast activity. FBXO22 knockdown restored osteoblast capacity. FBXO22 ubiquitinated and degraded Krüppel-like factor 4 (KLF4), which regulated the nerve growth factor (NGF)/tropomyosin receptor kinase A pathway by repressing NGF transcription. Silencing of KLF4 mitigated the metastasis-suppressing properties of FBXO22 knockdown, whereas NGF reversed the metastasis-suppressing properties of KLF4 in vitro and in vivo. Cumulatively, these data indicate that FBXO22 promotes PC cell activity and osteogenic lesions by stimulating macrophage M2 polarization. It also degrades KLF4 in macrophages and promotes NGF transcription, thereby activating the NGF/tropomyosin receptor kinase A pathway.
Collapse
Affiliation(s)
- Yuehua Zhang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wen Li
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shenghu Guo
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zheng Wu
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Zhang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ya Liu
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xing Li
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaojin Guo
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Cao
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chunwang Yang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhiyu Wang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
12
|
Thakur N, Quazi S, Naik B, Jha SK, Singh P. New insights into molecular signaling pathways and current advancements in prostate cancer diagnostics & therapeutics. Front Oncol 2023; 13:1193736. [PMID: 37664036 PMCID: PMC10469924 DOI: 10.3389/fonc.2023.1193736] [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: 03/25/2023] [Accepted: 07/18/2023] [Indexed: 09/05/2023] Open
Abstract
Prostate adenocarcinoma accounts for more than 20% of deaths among males due to cancer. It is the fifth-leading cancer diagnosed in males across the globe. The mortality rate is quite high due to prostate cancer. Despite the fact that advancements in diagnostics and therapeutics have been made, there is a lack of effective drugs. Metabolic pathways are altered due to the triggering of androgen receptor (AR) signaling pathways, and elevated levels of dihydrotestosterone are produced due to defects in AR signaling that accelerate the growth of prostate cancer cells. Further, PI3K/AKT/mTOR pathways interact with AR signaling pathway and act as precursors to promote prostate cancer. Prostate cancer therapy has been classified into luminal A, luminal B, and basal subtypes. Therapeutic drugs inhibiting dihydrotestosterone and PI3K have shown to give promising results to combat prostate cancer. Many second-generation Androgen receptor signaling antagonists are given either as single agent or with the combination of other drugs. In order to develop a cure for metastasized prostate cancer cells, Androgen deprivation therapy (ADT) is applied by using surgical or chemical methods. In many cases, Prostatectomy or local radiotherapy are used to control metastasized prostate cancer. However, it has been observed that after 1.5 years to 2 years of Prostatectomy or castration, there is reoccurrence of prostate cancer and high incidence of castration resistant prostate cancer is seen in population undergone ADT. It has been observed that Androgen derivation therapy combined with drugs like abiraterone acetate or docetaxel improve overall survival rate in metastatic hormone sensitive prostate cancer (mHSPC) patients. Scientific investigations have revealed that drugs inhibiting poly ADP Ribose polymerase (PARP) are showing promising results in clinical trials in the prostate cancer population with mCRPC and DNA repair abnormalities. Recently, RISUG adv (reversible inhibition of sperm under guidance) has shown significant results against prostate cancer cell lines and MTT assay has validated substantial effects of this drug against PC3 cell lines. Current review paper highlights the advancements in prostate cancer therapeutics and new drug molecules against prostate cancer. It will provide detailed insights on the signaling pathways which need to be targeted to combat metastasized prostate cancer and castration resistant prostate cancer.
Collapse
Affiliation(s)
- Neha Thakur
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, India
| | - Sameer Quazi
- Department of Chemistry, Akshara First Grade College, Bengaluru, India
- GenLab Biosolutions Private Limited, Bangalore, Karnataka, India
- Department of Biomedical Sciences, School of Life Sciences, Anglia Ruskin University, Cambridge, United Kingdom
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Solution Chemistry of Advanced Materials and Technologies (SCAMT) Institute, ITMO University, St. Petersburg, Russia
| | - Bindu Naik
- Department of Food Science and Technology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, India
| | - Pallavi Singh
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, India
| |
Collapse
|
13
|
Di Donato M, Giovannelli P, Migliaccio A, Castoria G. The nerve growth factor-delivered signals in prostate cancer and its associated microenvironment: when the dialogue replaces the monologue. Cell Biosci 2023; 13:60. [PMID: 36941697 PMCID: PMC10029315 DOI: 10.1186/s13578-023-01008-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/06/2023] [Indexed: 03/22/2023] Open
Abstract
Prostate cancer (PC) represents the most diagnosed and the second most lethal cancer in men worldwide. Its development and progression occur in concert with alterations in the surrounding tumor microenvironment (TME), made up of stromal cells and extracellular matrix (ECM) that dynamically interact with epithelial PC cells affecting their growth and invasiveness. PC cells, in turn, can functionally sculpt the TME through the secretion of various factors, including neurotrophins. Among them, the nerve growth factor (NGF) that is released by both epithelial PC cells and carcinoma-associated fibroblasts (CAFs) triggers the activation of various intracellular signaling cascades, thereby promoting the acquisition of a metastatic phenotype. After many years of investigation, it is indeed well established that aberrations and/or derangement of NGF signaling are involved not only in neurological disorders, but also in the pathogenesis of human proliferative diseases, including PC. Another key feature of cancer progression is the nerve outgrowth in TME and the concept of nerve dependence related to perineural invasion is currently emerging. NGF released by cancer cells can be a driver of tumor neurogenesis and nerves infiltrated in TME release neurotransmitters, which might stimulate the growth and sustainment of tumor cells.In this review, we aim to provide a snapshot of NGF action in the interactions between TME, nerves and PC cells. Understanding the molecular basis of this dialogue might expand the arsenal of therapeutic strategies against this widespread disease.
Collapse
Affiliation(s)
- Marzia Di Donato
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy.
| | - Pia Giovannelli
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy.
| | - Antimo Migliaccio
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy
| | - Gabriella Castoria
- Department of Precision Medicine, University of Campania "L.Vanvitelli", 80138, Naples, Italy
| |
Collapse
|
14
|
Roy S, Morgan SC, Wallis CJD, Sun Y, Spratt DE, Malone J, Grimes S, Mukherjee D, Kishan AU, Saad F, Malone S. Association of dynamic change in patient-reported pain with survival in metastatic castrate sensitive prostate cancer-exploratory analysis of LATITUDE study. Prostate Cancer Prostatic Dis 2023; 26:96-104. [PMID: 36097167 DOI: 10.1038/s41391-022-00529-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Pain is an important dimension of quality-of-life in patients with metastatic castrate-sensitive prostate cancer (mCSPC). However, it is unclear if dynamic change in pain over time can predict for overall survival (OS) or progression-free survival (PFS) in these patients. METHODS This is an exploratory analysis of LATITUDE, a phase III randomized study, in which men with de novo mCSPC were randomized to receive either ADT plus abiraterone versus ADT alone. Information was collected on patient-reported worst pain score (WPS) and pain-interference score (PIS) from the Brief Pain Inventory-Short Form. A Bayesian joint modelling approach was used determine the association of dynamic change in WPS and PIS with OS and PFS. RESULTS Overall, 1125 patients with at least 3 measurements on pain scores were eligible. On Cox multivariable regression, increase in baseline WPS was associated with inferior OS (hazard ratio [HR] 1.049 [95% confidence intervals [CI] 1.015-1.085]; time dependent area under curve [tAUC] 0.64) and PFS (HR 1.045 [1.011-1.080]; tAUC: 0.64). Increase in baseline PIS was associated with inferior OS (HR 1.062 [1.020-1.105]; tAUC: 0.63) but not with PFS (HR 1.038 [0.996-1.08]). On independent joint models, an increase in the current value of WPS by 1-unit was associated with inferior OS (HR 1.316 [1.258-1.376]; tAUC 0.74) and PFS (HR 1.319 [1.260-1.382]; tAUC 0.70). Similar association was seen for increase in the current value of PIS with OS (HR 1.319 [1.261-1.381]; tAUC 0.73) and PFS (HR 1.282 [1.224-1.344]; tAUC 0.73). CONCLUSIONS The above findings highlight the potential dynamic interplay between patient-reported pain with OS and PFS in mCSPC. Compared to baseline pain, such dynamic assessment of pain was found to have superior predictive ability and thus has the potential to tailor subsequent treatment based on response to initial therapy beyond its role as a very important dimension of quality-of-life.
Collapse
Affiliation(s)
- Soumyajit Roy
- Department of Radiation Oncology, Rush University Medical Center, Chicago, IL, USA.
| | - Scott C Morgan
- Radiation Medicine Program, The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, ON, Canada
| | | | - Yilun Sun
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Daniel E Spratt
- University Hospital Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | | | - Scott Grimes
- Radiation Medicine Program, The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, ON, Canada
| | | | - Amar U Kishan
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Fred Saad
- Department of surgery, Université de Montréal, Montreal, QC, Canada
| | - Shawn Malone
- Radiation Medicine Program, The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
15
|
Wang J, Yang Z, Liu Y, Li H, Yang X, Gao W, Zhao Q, Yang X, Wei J. The GAL/GALR2 axis promotes the perineural invasion of salivary adenoid cystic carcinoma via epithelial-to-mesenchymal transition. Cancer Med 2023; 12:4496-4509. [PMID: 36039037 PMCID: PMC9972115 DOI: 10.1002/cam4.5181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Perineural invasion (PNI) is a typical pathological characteristic of salivary adenoid cystic carcinoma (SACC) and other neurotrophic cancers. The mechanism of the neural microenvironment controlling tumor progression during the PNI process is unclear. In the present study, we investigated the role and molecular mechanisms of nerve-derived neuropeptide galanin (GAL) and its receptor (GALR2) in the regulation of PNI in SACC. METHODS Immunohistochemistry staining and clinical association studies were performed to analyze the expression of GAL and GALR2 in SACC tissues and their clinical value. Dorsal root ganglion or SH-SY5Y cells were co-cultured with SACC cells in vitro to simulate the interactions between the neural microenvironment and tumor cells, and a series of assays including transcriptome sequencing, Western blot, and Transwell were performed to investigate the role and molecular mechanism of GAL and GALR2 in the regulation of SACC cells. Moreover, both the in vitro and in vivo PNI models were established to assess the potential PNI-specific therapeutic effects by blocking the GAL/GALR2 axis. RESULTS GAL and GALR2 were highly expressed in SACC tissues, and were associated with PNI and poor prognosis in SACC patients (p < 0.05). Nerve-derived GAL activated GALR2 expression in SACC cells and induced epithelial-to-mesenchymal transition (EMT) in SACC cells. Adding human recombinant GAL to the co-culture system promoted the proliferation, migration, and invasion of SACC cells significantly, but inhibited the apoptosis of SACC cells. Adding M871, a specific antagonist of GALR2, significantly blocked the above effects (p < 0.05) and inhibited the PNI of SACC cells in vitro and in vivo (p < 0.05). CONCLUSIONS This study demonstrated that nerve-derived GAL activated GALR2 expression, and promoted EMT in SACC cells, thereby enhancing the PNI process. Interruption of the GAL/GALR2 axis might be a novel strategy for anti-PNI therapy for SACC.
Collapse
Affiliation(s)
- Jun Wang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Zihui Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Yuanyang Liu
- Senior Department of Neurosurgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Huan Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Xiangming Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Wanpeng Gao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Qi Zhao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Xinjie Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Jianhua Wei
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
16
|
Su X, Wang B, Zhou Z, Li Z, Tong S, Chen S, Zhang N, Liu S, Zhang M. A positive feedback loop of heparanase/syndecan1/nerve growth factor regulates cancer pain progression. Korean J Pain 2023; 36:60-71. [PMID: 36536517 PMCID: PMC9812689 DOI: 10.3344/kjp.22277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 12/24/2022] Open
Abstract
Background The purpose of this research was to assess the role of heparanase (HPSE)/syndecan1 (SDC1)/nerve growth factor (NGF) on cancer pain from melanoma. Methods The influence of HPSE on the biological function of melanoma cells and cancer pain in a mouse model was evaluated. Immunohistochemical staining was used to analyze HPSE and SDC1. HPSE, NGF, and SDC1 were detected using western blot. Inflammatory factors were detected using ELISA assay. Results HPSE promoted melanoma cell viability, proliferation, migration, invasion, and tumor growth, as well as cancer pain, while SST0001 treatment reversed the promoting effect of HPSE. HPSE up-regulated NGF, and NGF feedback promoted HPSE. High expression of NGF reversed the inhibitory effect of HPSE down-regulation on melanoma cell phenotype deterioration, including cell viability, proliferation, migration, and invasion. SST0001 down-regulated SDC1 expression. SDC1 reversed the inhibitory effect of SST0001 on cancer pain. Conclusions The results showed that HPSE promoted melanoma development and cancer pain by interacting with NGF/SDC1. It provides new insights to better understand the role of HPSE in melanoma and also provides a new direction for cancer pain treatment.
Collapse
Affiliation(s)
- Xiaohu Su
- Department of Anesthesiology, Suqian First People’s Hospital, Suqian City, Jiangsu Province, China
| | - Bingwu Wang
- Cancer Institute, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Zhaoyun Zhou
- Department of Anesthesiology, Tai’an Central Hospital, Tai’an City, Shandong Province, China
| | - Zixian Li
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Song Tong
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Simin Chen
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Nan Zhang
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Su Liu
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Maoyin Zhang
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China,Correspondence: Maoyin Zhang Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, No. 99, Huaihai West Road, Quanshan District, Xuzhou City, Jiangsu Province 221002, China, Tel: +86-18168777315, Fax: +86-0516-85805911, E-mail:
| |
Collapse
|
17
|
Han S, Wang D, Huang Y, Zeng Z, Xu P, Xiong H, Ke Z, Zhang Y, Hu Y, Wang F, Wang J, Zhao Y, Zhuo W, Zhao G. A reciprocal feedback between colon cancer cells and Schwann cells promotes the proliferation and metastasis of colon cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:348. [PMID: 36522730 PMCID: PMC9753336 DOI: 10.1186/s13046-022-02556-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Research has indicated that the emergence of Schwann cells around premalignant lesions of colon cancer might be an early indicator promoting the onset of tumorigenesis. The present study explored the communication between colon cancer cells and Schwann cells. METHODS Immunofluorescence analyses were conducted to examine the differential distribution of Schwann cells within colon cancer tissues and normal colon tissues. CCK8 assay, colony formation assay, wound healing assay, and transwell assay were performed to investigate the interaction between colon cancer cells and Schwann cells. Exosomes derived from colon cancer cells were isolated to further explore the effect of colon cancer cells on Schwann cells. Gain- and loss-of function experiments, luciferase reporter assays, chromatin immunoprecipitation assays, and immunohistochemistry assays were performed to reveal the cross-talk between colon cancer cells and Schwann cells. Furthermore, colon cancer cells co-cultured with Schwann cells were transplanted into nude mice for evaluating their effect on tumor proliferation and metastasis in vivo. RESULTS The clinicopathological characteristics indicated that Schwann cells were enriched in colon cancer tissues and were associated with tumor metastasis and poor prognosis. The co-culture of Schwann cells with colon cancer cells promoted the proliferation and migration of colon cancer cells and Schwann cells, which was mediated by nerve growth factor (NGF) secreted from Schwann cells. Exosomal miR-21-5p released by colon cancer cells inhibited VHL expression in Schwann cells, which in turn stabilized the HIF-1α protein and increased the transcription of NGF. Meanwhile, the Schwann cells-derived NGF activated TrkA/ERK/ELK1/ZEB1 signaling pathway in colon cancer cells, which further enhanced the expression of exosomal miR-21-5p. Inhibition of either NGF or miR-21-5p significantly inhibited the proliferation and metastasis of transplanted colon cancer cells in nude mice. Coincidently, miR-21-5p was positively associated with the expression of NGF, p-ERK, p-ELK1, and ZEB1 in human colon cancer tissues. CONCLUSIONS Our results implicated a reciprocal communication between colon cancer cells and Schwan cells that promoted the proliferation and metastasis of colon cancer, and identified NGF and exosomal miR-21-5p as potential therapeutic targets for the treatment of colon cancer.
Collapse
Affiliation(s)
- Shengbo Han
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Decai Wang
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Yan Huang
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Zhu Zeng
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Peng Xu
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Hewei Xiong
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Zunxiang Ke
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Ya Zhang
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Yuhang Hu
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Fan Wang
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Jie Wang
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Yong Zhao
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Wenfeng Zhuo
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Gang Zhao
- grid.33199.310000 0004 0368 7223Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| |
Collapse
|
18
|
Agri-Food By-Products in Cancer: New Targets and Strategies. Cancers (Basel) 2022; 14:cancers14225517. [PMID: 36428610 PMCID: PMC9688227 DOI: 10.3390/cancers14225517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/27/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
The globalization and the changes in consumer lifestyles are forcing us to face a deep transformation in food demand and in the organization of the entire food production system. In this new era, the food-loss and food-waste security nexus is relevant in the global debate and avoiding unsustainable waste in agri-food systems as well as the supply chain is a big challenge. "Food waste" is useful for the recovery of its valuable components, thus it can assume the connotation of a "food by-product". Sustainable utilization of agri-food waste by-products provides a great opportunity. Increasing evidence shows that agri-food by-products are a source of different bioactive molecules that lower the inflammatory state and, hence, the aggressiveness of several proliferative diseases. This review aims to summarize the effects of agri-food by-products derivatives, already recognized as promising therapeutics in human diseases, including different cancer types, such as breast, prostate, and colorectal cancer. Here, we examine products modulating or interfering in the signaling mediated by the epidermal growth factor receptor.
Collapse
|
19
|
Krause W. Resistance to prostate cancer treatments. IUBMB Life 2022; 75:390-410. [PMID: 35978491 DOI: 10.1002/iub.2665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/09/2022] [Indexed: 12/14/2022]
Abstract
A review of the current treatment options for prostate cancer and the formation of resistance to these regimens has been compiled including primary, acquired, and cross-resistance. The diversification of the pathways involved and the escape routes the tumor is utilizing have been addressed. Whereas early stages of tumor can be cured, there is no treatment available after a point of no return has been reached, leaving palliative treatment as the only option. The major reasons for this outcome are the heterogeneity of tumors, both inter- and intra-individually and the nearly endless number of escape routes, which the tumor can select to overcome the effects of treatment. This means that more focus should be applied to the individualization of both diagnosis and therapy of prostate cancer. In addition to current treatment options, novel drugs and ongoing clinical trials have been addressed in this review.
Collapse
|
20
|
Di Donato M, Giovannelli P, Migliaccio A, Bilancio A. Inhibition of Vps34 and p110δ PI3K Impairs Migration, Invasion and Three-Dimensional Spheroid Growth in Breast Cancer Cells. Int J Mol Sci 2022; 23:9008. [PMID: 36012280 PMCID: PMC9409264 DOI: 10.3390/ijms23169008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is a heterogeneous disease that represents the most common cancer around the world; it comprises 12% of new cases according to the World Health Organization. Despite new approaches in early diagnosis and current treatment, breast cancer is still the leading cause of death for cancer mortality. New targeted therapies against key signalling transduction molecules are required. Phosphoinositide 3-kinase (PI3K) regulates multiple biological functions such as proliferation, survival, migration, and growth. It is well established that PI3K isoform-selective inhibitors show fewer toxic side effects compared to broad spectrum inhibition of PI3K (pan-PI3K inhibitors). Therefore, we tested the PI3K p110δ-selective inhibitor, IC87114, and Vps34-selective inhibitor, Vps34-IN1, on the breast cancer cell lines MCF-7 and MDA-MB-231, representing hormone-responsive and triple-negative breast cancer cells, respectively. Our data show that both inhibitors decreased migration of MCF-7 and MDA-MB-231 cells, and Vps34 also significantly impacted MCF-7 cell proliferation. Three-dimensional (3D) in vitro culture models show that IC87114 and Vps34-IN1 treatment reduced the growth of MCF-7 and MDA-MB-231 cells in 3D tumour spheroid cultures. This study identifies IC87114 and Vps34-IN1 as potential therapeutic approaches in breast cancer.
Collapse
Affiliation(s)
| | | | | | - Antonio Bilancio
- Department of Medicine Precision, “Luigi Vanvitelli”, Affiliation University of Campania, Via L. De Crecchio 7, 80138 Naples, Italy
| |
Collapse
|
21
|
Di Sarno V, Giovannelli P, Medina-Peris A, Ciaglia T, Di Donato M, Musella S, Lauro G, Vestuto V, Smaldone G, Di Matteo F, Bifulco G, Castoria G, Migliaccio A, Fernandez-Carvajal A, Campiglia P, Gomez-Monterrey I, Ostacolo C, Bertamino A. New TRPM8 blockers exert anticancer activity over castration-resistant prostate cancer models. Eur J Med Chem 2022; 238:114435. [DOI: 10.1016/j.ejmech.2022.114435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/04/2022]
|
22
|
Epithelial-Mesenchymal Plasticity Induced by Discontinuous Exposure to TGFβ1 Promotes Tumour Growth. BIOLOGY 2022; 11:biology11071046. [PMID: 36101425 PMCID: PMC9312510 DOI: 10.3390/biology11071046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary In this manuscript, we used a non-genetically manipulated EMT/MET cell line model to demonstrate that epithelial mesenchymal plasticity occurring in normal cells generates co-existing phenotypically and functionally divergent cell subpopulations which result in fast growing tumours in vivo. Abstract Transitions between epithelial and mesenchymal cellular states (EMT/MET) contribute to cancer progression. We hypothesize that EMT followed by MET promotes cell population heterogeneity, favouring tumour growth. We developed an EMT model by on and off exposure of epithelial EpH4 cells (E-cells) to TGFβ1 that mimics phenotypic EMT (M-cells) and MET. We aimed at understanding whether phenotypic MET is accompanied by molecular and functional reversion back to epithelia by using RNA sequencing, immunofluorescence (IF), proliferation, wound healing, focus formation and mamosphere formation assays as well as cell xenografts in nude mice. Phenotypic reverted epithelial cells (RE-cells) obtained after MET induction presented epithelial morphologies and proliferation rates resembling E cells. However, the RE transcriptomic profile and IF staining of epithelial and mesenchymal markers revealed a uniquely heterogeneous mixture of cell subpopulations with a high self-renewal ability. RE cell heterogeneity was stably maintained for long periods after TGFβ1 removal both in vitro and in large tumours derived from the nude mice. Overall, we show that phenotypic reverted epithelial cells (RE cells) do not return to the molecular and functional epithelial state and present mesenchymal features related to aggressiveness and cellular heterogeneity that favour tumour growth in vivo. This work strengthens epithelial cell reprogramming and cellular heterogeneity fostered by inflammatory cues as a tumour growth-promoting factor in vivo.
Collapse
|
23
|
Boccellino M, Ambrosio P, Ballini A, De Vito D, Scacco S, Cantore S, Feola A, Di Donato M, Quagliuolo L, Sciarra A, Galasso G, Crocetto F, Imbimbo C, Boffo S, Di Zazzo E, Di Domenico M. The Role of Curcumin in Prostate Cancer Cells and Derived Spheroids. Cancers (Basel) 2022; 14:3348. [PMID: 35884410 PMCID: PMC9320241 DOI: 10.3390/cancers14143348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
A major challenge in the clinical management of prostate cancer (PC) is to inhibit tumor growth and prevent metastatic spreading. In recent years, considerable efforts have been made to discover new compounds useful for PC therapy, and promising advances in this field were reached. Drugs currently used in PC therapy frequently induce resistance and PC progresses toward metastatic castration-resistant forms (mCRPC), making it virtually incurable. Curcumin, a commercially available nutritional supplement, represents an attractive therapeutic agent for mCRPC patients. In the present study, we compared the effects of chemotherapeutic drugs such as docetaxel, paclitaxel, and cisplatin, to curcumin, on two PC cell lines displaying a different metastatic potential: DU145 (moderate metastatic potential) and PC-3 (high metastatic potential). Our results revealed a dose-dependent reduction of DU145 and PC-3 cell viability upon treatment with curcumin similar to chemotherapeutic agents (paclitaxel, cisplatin, and docetaxel). Furthermore, we explored the EGFR-mediated signaling effects on ERK activation in DU145 and PC-3 cells. Our results showed that DU145 and PC-3 cells overexpress EGFR, and the treatment with chemotherapeutic agents or curcumin reduced EGFR expression levels and ERK activation. Finally, chemotherapeutic agents and curcumin reduced the size of DU145 and PC-3 spheroids and have the potential to induce apoptosis and also in Matrigel. In conclusion, despite different studies being carried out to identify the potential synergistic curcumin combinations with chemopreventive/therapeutic efficacy for inhibiting PC growth, the results show the ability of curcumin used alone, or in combinatorial approaches, to impair the size and the viability of PC-derived spheroids.
Collapse
Affiliation(s)
- Mariarosaria Boccellino
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Pasqualina Ambrosio
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Andrea Ballini
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Danila De Vito
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy; (D.D.V.); (S.S.)
| | - Salvatore Scacco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy; (D.D.V.); (S.S.)
| | | | - Antonia Feola
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Marzia Di Donato
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Lucio Quagliuolo
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Antonella Sciarra
- Department of Biology, University of Naples “Federico II”, 80126 Naples, Italy;
| | - Giovanni Galasso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Felice Crocetto
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (C.I.)
| | - Ciro Imbimbo
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (C.I.)
| | - Silvia Boffo
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122-6078, USA;
| | - Erika Di Zazzo
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
| | - Marina Di Domenico
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (P.A.); (A.F.); (M.D.D.); (L.Q.); (G.G.); (E.D.Z.); (M.D.D.)
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122-6078, USA;
| |
Collapse
|
24
|
Enamel Matrix Derivative Enhances the Odontoblastic Differentiation of Dental Pulp Stem Cells via Activating MAPK Signaling Pathways. Stem Cells Int 2022; 2022:2236250. [PMID: 35530415 PMCID: PMC9071913 DOI: 10.1155/2022/2236250] [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: 11/13/2021] [Revised: 03/27/2022] [Accepted: 04/05/2022] [Indexed: 12/03/2022] Open
Abstract
The odontoblastic differentiation of dental pulp stem cells (DPSCs) contributes to pulp-dentin regeneration. Enamel matrix derivative (EMD) is considered to be a critical epithelial signal to induce cell differentiation during odontogenesis and has been widely applied to clinical periodontal tissue regeneration. The purpose of this study was to explore the effect of EMD on DPSCs proliferation and odontoblastic differentiation, as well as the underlying mechanisms. We conducted in vitro and in vivo researches to get a comprehensive understanding of EMD. In vitro phase: cell proliferation was assessed by a cell counting kit-8 (CCK-8) assay; then, alkaline phosphatase (ALP) activity and staining, alizarin red staining, real-time RT-PCR, and western blot analysis were conducted to determine the odontoblastic potential and involvement of MAPK signaling pathways. In vivo phase: after ensuring the biocompatibility of VitroGel 3D-RGD via scanning electron microscopy (SEM), the hydrogel mixture was subcutaneously injected into nude mice followed by histological and immunohistochemical analyses. The results revealed that EMD did not interfere with DPSCs proliferation but promoted the odontoblastic differentiation of DPSCs in vitro and in vivo. Furthermore, blocking the MAPK pathways suppressed the EMD-enhanced differentiation of DPSCs. Finally, VitroGel 3D-RGD could well support the proliferation, differentiation, and regeneration of DPSCs. Overall, this study demonstrates that EMD enhances the odontoblastic differentiation of DPSCs through triggering MAPK signaling pathways. The findings provide a new insight into the mechanism by which EMD affects DPSCs differentiation and proposes EMD as a promising candidate for future stem cell therapy in endodontics.
Collapse
|
25
|
Qin T, Xiao Y, Qian W, Wang X, Gong M, Wang Q, An R, Han L, Duan W, Ma Q, Wang Z. HGF/c-Met pathway facilitates the perineural invasion of pancreatic cancer by activating the mTOR/NGF axis. Cell Death Dis 2022; 13:387. [PMID: 35449152 PMCID: PMC9023560 DOI: 10.1038/s41419-022-04799-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 12/13/2022]
Abstract
Perineural invasion (PNI) is a pathologic feature of pancreatic cancer and is associated with poor outcomes, metastasis, and recurrence in pancreatic cancer patients. However, the molecular mechanism of PNI remains unclear. The present study aimed to investigate the mechanism that HGF/c-Met pathway facilitates the PNI of pancreatic cancer. In this study, we confirmed that c-Met expression was correlated with PNI in pancreatic cancer tissues. Activating the HGF/c-Met signaling pathway potentiated the expression of nerve growth factor (NGF) to recruit nerves and promote the PNI. Activating the HGF/c-Met signaling pathway also enhanced the migration and invasion ability of cancer cells to facilitate cancer cells invading nerves. Mechanistically, HGF/c-Met signaling pathway can active the mTOR/NGF axis to promote the PNI of pancreatic cancer. Additionally, we found that knocking down c-Met expression inhibited cancer cell migration along the nerve, reduced the damage of the sciatic nerve caused by cancer cells and protected the function of the sciatic nerve in vivo. Taken together, our findings suggest a supportive mechanism of the HGF/c-Met signaling pathway in promoting PNI by activating the mTOR/NGF axis in pancreatic cancer. Blocking the HGF/c-Met signaling pathway may be an effective target for the treatment of PNI.
Collapse
Affiliation(s)
- Tao Qin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Xiao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xueni Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengyuan Gong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiqi Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui An
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Centre for Pancreatic Diseases of Xi'an Jiaotong University, Xi'an, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Centre for Pancreatic Diseases of Xi'an Jiaotong University, Xi'an, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Centre for Pancreatic Diseases of Xi'an Jiaotong University, Xi'an, China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, China.
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Centre for Pancreatic Diseases of Xi'an Jiaotong University, Xi'an, China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
26
|
Nayeem MJ, Yamamura A, Hayashi H, Muramatsu H, Nakamura K, Sassa N, Sato M. Imatinib mesylate inhibits androgen-independent PC-3 cell viability, proliferation, migration, and tumor growth by targeting platelet-derived growth factor receptor-α. Life Sci 2022; 288:120171. [PMID: 34822800 DOI: 10.1016/j.lfs.2021.120171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/08/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022]
Abstract
AIM The abnormal expression of oncogenic tyrosine kinase receptors such as platelet-derived growth factor receptors (PDGFRs) has been reported in cancer progression. However, the role of PDGFRs in the human androgen-independent prostate cancer PC-3 cell line is not well understood. Thus, this study examined the role of PDGFRs in androgen-independent PC-3 cells. MAIN METHODS PDGFR mRNA and protein expression was determined by quantitative real-time PCR and western blotting, respectively. The effects of the tyrosine kinase inhibitor imatinib (imatinib mesylate) and small interfering RNAs (siRNAs) were determined by a Cell Counting Kit-8 assay, bromodeoxyuridine assay, and Transwell migration assay. The in vivo effect of imatinib was analyzed using a tumor formation assay in nude mice. KEY FINDINGS PDGFRα was upregulated in androgen-independent PC-3 cells compared with normal prostate epithelial cells. PDGF-BB induced the phosphorylation of PDGFRα and downstream signaling molecules, including Akt, in a dose-dependent manner. Imatinib reduced the phosphorylation of the PDGFRα/Akt axis. Imatinib also suppressed the viability, proliferation, migration, and tumor growth of PC-3 cells. PDGFRα knockdown by siRNA decreased the viability and migration of PC-3 cells. SIGNIFICANCE These results demonstrated the distinct contribution of PDGFRα signaling to the proliferation and migration of PC-3 cells and suggested the potential for PDGFRα as a therapeutic target for metastatic and androgen-independent prostate cancer.
Collapse
Affiliation(s)
| | - Aya Yamamura
- Department of Physiology, Aichi Medical University, Japan
| | - Hisaki Hayashi
- Department of Physiology, Aichi Medical University, Japan
| | | | | | - Naoto Sassa
- Department of Urology, Aichi Medical University, Japan
| | - Motohiko Sato
- Department of Physiology, Aichi Medical University, Japan.
| |
Collapse
|
27
|
Arman T, Nelson PS. Endocrine and paracrine characteristics of neuroendocrine prostate cancer. Front Endocrinol (Lausanne) 2022; 13:1012005. [PMID: 36440195 PMCID: PMC9691667 DOI: 10.3389/fendo.2022.1012005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/24/2022] [Indexed: 11/12/2022] Open
Abstract
Prostate cancer is a common malignancy affecting men worldwide. While the vast majority of newly diagnosed prostate cancers are categorized as adenocarcinomas, a spectrum of uncommon tumor types occur including those with small cell and neuroendocrine cell features. Benign neuroendocrine cells exist in the normal prostate microenvironment, and these cells may give rise to primary neuroendocrine carcinomas. However, the more common development of neuroendocrine prostate cancer is observed after therapeutics designed to repress the signaling program regulated by the androgen receptor which is active in the majority of localized and metastatic adenocarcinomas. Neuroendocrine tumors are identified through immunohistochemical staining for common markers including chromogranin A/B, synaptophysin and neuron specific enolase (NSE). These markers are also common to neuroendocrine tumors that arise in other tissues and organs such as the gastrointestinal tract, pancreas, lung and skin. Notably, neuroendocrine prostate cancer shares biochemical features with nerve cells, particularly functions involving the secretion of a variety of peptides and proteins. These secreted factors have the potential to exert local paracrine effects, and distant endocrine effects that may modulate tumor progression, invasion, and resistance to therapy. This review discusses the spectrum of factors derived from neuroendocrine prostate cancers and their potential to influence the pathophysiology of localized and metastatic prostate cancer.
Collapse
Affiliation(s)
- Tarana Arman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, United States
- *Correspondence: Peter S. Nelson,
| |
Collapse
|
28
|
Licitra F, Giovannelli P, Di Donato M, Monaco A, Galasso G, Migliaccio A, Castoria G. New Insights and Emerging Therapeutic Approaches in Prostate Cancer. Front Endocrinol (Lausanne) 2022; 13:840787. [PMID: 35222290 PMCID: PMC8873523 DOI: 10.3389/fendo.2022.840787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Prostate cancer is the second most frequently diagnosed cancer in men and several therapeutic approaches are currently available for patient's care. Although the androgen receptor status represents a good predictor of response to androgen deprivation therapy, prostate cancer frequently becomes resistant to this approach and spreads. The molecular mechanisms that contribute to progression and drug-resistance of this cancer remain still debated. However, few therapeutic options are available for patient's management, at this stage. Recent years have seen a great expansion of the studies concerning the role of stromal-epithelial interactions and tumor microenvironment in prostate cancer progression. The findings so far collected have provided new insights into diagnostic and clinical management of prostate cancer patients. Further, new fascinating aspects concerning the intersection of the androgen receptor with survival factors as well as calcium channels have been reported in cultured prostate cancer cells and mouse models. The results of these researches have opened the way for a better understanding of the basic mechanisms involved in prostate cancer invasion and drug-resistance. They have also significantly expanded the list of new biomarkers and druggable targets in prostate cancer. The primary aim of this manuscript is to provide an update of these issues, together with their translational aspects. Exploiting the power of novel promising therapeutics would increase the success rate in the diagnostic path and clinical management of patients with advanced disease.
Collapse
|
29
|
A Small Peptide Targeting the Ligand-Induced Androgen Receptor/Filamin a Interaction Inhibits the Invasive Phenotype of Prostate Cancer Cells. Cells 2021; 11:cells11010014. [PMID: 35011576 PMCID: PMC8750472 DOI: 10.3390/cells11010014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/16/2021] [Accepted: 12/18/2021] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer (PC) is one of the most widespread malignancies among males worldwide. The androgen receptor (AR) plays a major role in prostate cancer development and progression and is the main target of PC therapy. Nonetheless, its action is not yet fully elucidated. We report here that the AR associates with Filamin A (FlnA) promoting migration and invasiveness of various PC-derived cells after androgen challenging. Inhibition of the AR/FlnA complex assembly by a very low concentration of Rh-2025u, an AR-derived peptide specifically interfering with this association, impairs such phenotype in monolayer cells and in 3D models. This study, together with our recent data in cancer-associated fibroblasts (CAFs), indicates that targeting the AR/FlnA complex could improve the clinical management of invasive PC, as the limited number of new drugs reaching the market suggests that we must re-examine the way invasive PC is currently treated. In this context, the synthesis of new biologically active molecules, such as the Rh-2025u peptide, which has been shown to efficiently interfere in the complex assembly in CAFs and PC cells, should overcome the limits of current available therapies, mostly based on hormone antagonists.
Collapse
|
30
|
Slabáková E, Kahounová Z, Procházková J, Souček K. Regulation of Neuroendocrine-like Differentiation in Prostate Cancer by Non-Coding RNAs. Noncoding RNA 2021; 7:ncrna7040075. [PMID: 34940756 PMCID: PMC8704250 DOI: 10.3390/ncrna7040075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPC) represents a variant of prostate cancer that occurs in response to treatment resistance or, to a much lesser extent, de novo. Unravelling the molecular mechanisms behind transdifferentiation of cancer cells to neuroendocrine-like cancer cells is essential for development of new treatment opportunities. This review focuses on summarizing the role of small molecules, predominantly microRNAs, in this phenomenon. A published literature search was performed to identify microRNAs, which are reported and experimentally validated to modulate neuroendocrine markers and/or regulators and to affect the complex neuroendocrine phenotype. Next, available patients’ expression datasets were surveyed to identify deregulated microRNAs, and their effect on NEPC and prostate cancer progression is summarized. Finally, possibilities of miRNA detection and quantification in body fluids of prostate cancer patients and their possible use as liquid biopsy in prostate cancer monitoring are discussed. All the addressed clinical and experimental contexts point to an association of NEPC with upregulation of miR-375 and downregulation of miR-34a and miR-19b-3p. Together, this review provides an overview of different roles of non-coding RNAs in the emergence of neuroendocrine prostate cancer.
Collapse
|
31
|
Di Donato M, Ostacolo C, Giovannelli P, Di Sarno V, Monterrey IMG, Campiglia P, Migliaccio A, Bertamino A, Castoria G. Therapeutic potential of TRPM8 antagonists in prostate cancer. Sci Rep 2021; 11:23232. [PMID: 34853378 PMCID: PMC8636514 DOI: 10.1038/s41598-021-02675-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/22/2021] [Indexed: 12/27/2022] Open
Abstract
Transient receptor potential melastatin-8 (TRPM8) represents an emerging target in prostate cancer, although its mechanism of action remains unclear. Here, we have characterized and investigated the effects of TRPM8 modulators in prostate cancer aggressiveness disclosing the molecular mechanism underlying their biological activity. Patch-clamp and calcium fluorometric assays were used to characterize the synthesized compounds. Androgen-stimulated prostate cancer-derived cells were challenged with the compounds and the DNA synthesis was investigated in a preliminary screening. The most effective compounds were then employed to inhibit the pro-metastatic behavior of in various PC-derived cells, at different degree of malignancy. The effect of the compounds was then assayed in prostate cancer cell-derived 3D model and the molecular targets of selected compounds were lastly identified using transcriptional and non-transcriptional reporter assays. TRPM8 antagonists inhibit the androgen-dependent prostate cancer cell proliferation, migration and invasiveness. They are highly effective in reverting the androgen-induced increase in prostate cancer cell spheroid size. The compounds also revert the proliferation of castrate-resistant prostate cancer cells, provided they express the androgen receptor. In contrast, no effects were recorded in prostate cancer cells devoid of the receptor. Selected antagonists interfere in non-genomic androgen action and abolish the androgen-induced androgen receptor/TRPM8 complex assembly as well as the increase in intracellular calcium levels in prostate cancer cells. Our results shed light in the processes controlling prostate cancer progression and make the transient receptor potential melastatin-8 as a ‘druggable’ target in the androgen receptor-expressing prostate cancers.
Collapse
Affiliation(s)
- Marzia Di Donato
- Department of Precision Medicine, School of Medicine, University of Campania 'L. Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131, Naples, Italy
| | - Pia Giovannelli
- Department of Precision Medicine, School of Medicine, University of Campania 'L. Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G.Paolo II, 84084, Fisciano, SA, Italy
| | - Isabel M Gomez Monterrey
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131, Naples, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G.Paolo II, 84084, Fisciano, SA, Italy
| | - Antimo Migliaccio
- Department of Precision Medicine, School of Medicine, University of Campania 'L. Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G.Paolo II, 84084, Fisciano, SA, Italy.
| | - Gabriella Castoria
- Department of Precision Medicine, School of Medicine, University of Campania 'L. Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy.
| |
Collapse
|
32
|
Wang L, Li J, Wang R, Chen H, Wang R, Wang W, Yang X. NGF Signaling Interacts With the Hippo/YAP Pathway to Regulate Cervical Cancer Progression. Front Oncol 2021; 11:688794. [PMID: 34722240 PMCID: PMC8552705 DOI: 10.3389/fonc.2021.688794] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/21/2021] [Indexed: 12/18/2022] Open
Abstract
Nerve growth factor (NGF) is increasingly implicated in cervical cancer progression, but its mechanism in cervical cancer is unclear. Here, studies demonstrate that NGF inhibits the Hippo signaling pathway and activates Yes-associated protein (YAP) to induce cervical cancer cell proliferation and migration. Our results suggested that stimulation of NGF promoted cell growth and migration and activated YAP in HeLa and C-33A cell lines. The expression of YAP target genes (CTGF and ANKRD1) was upregulated after NGF treatment. The NGF inhibitor Ro 08-2750 and siRNA-mediated NGF receptor gene silencing suppressed HeLa and C-33A cells proliferation and migration, activated large suppressor kinase 1 (LATS1) kinase activity, and suppressed YAP function. In addition, the expression of YAP target genes (CTGF and ANKRD1) was suppressed by Ro 08-2750 treatment in HeLa and C-33A cells. Interestingly, proliferation was significantly higher in NGF-treated cells than in control cells, and this effect was completely reversed by the YAP small molecule inhibitor-verteporfin. Furthermore, the mouse xenograft model shows that NGF regulates YAP oncogenic activity in vivo. Mechanistically, NGF stimulation inactivates LATS1 and activates YAP, and NGF inhibition was found to induce large suppressor kinase 1 (LATS1) phosphorylation. Taken together, these data provide the first direct evidence of crosstalk between the NGF signaling and Hippo cancer pathways, an interaction that affects cervical cancer progression. Our study indicates that combined targeting of the NGF signaling and the Hippo pathway represents a novel therapeutic strategy for treatment of cervical cancer.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Li
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rongli Wang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - He Chen
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ruiqi Wang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wei Wang
- Department of Anesthesiology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyuan Yang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
33
|
Di Donato M, Galasso G, Giovannelli P, Sinisi AA, Migliaccio A, Castoria G. Targeting the Nerve Growth Factor Signaling Impairs the Proliferative and Migratory Phenotype of Triple-Negative Breast Cancer Cells. Front Cell Dev Biol 2021; 9:676568. [PMID: 34268306 PMCID: PMC8275826 DOI: 10.3389/fcell.2021.676568] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer is a heterogeneous disease that still lacks specific therapeutic approaches. The identification of new biomarkers, predictive of the disease's aggressiveness and pharmacological response, is a challenge for a more tailored approach in the clinical management of patients. Nerve growth factor, initially identified as a key factor for neuronal survival and differentiation, turned out to be a multifaceted molecule with pleiotropic effects in quite divergent cell types, including cancer cells. Many solid tumors exhibit derangements of the nerve growth factor and its receptors, including the tropomyosin receptor kinase A. This receptor is expressed in triple-negative breast cancer, although its role in the pathogenesis and aggressiveness of this disease is still under investigation. We now report that triple-negative breast cancer-derived MDA-MB-231 and MDA-MB-453 cells express appreciable levels of tropomyosin receptor kinase A and release a biologically active nerve growth factor. Activation of tropomyosin receptor kinase by nerve growth factor treatment positively affects the migration, invasion, and proliferation of triple-negative breast cancer cells. An increase in the size of triple-negative breast cancer cell spheroids is also detected. This latter effect might occur through the nerve growth factor-induced release of matrix metalloproteinase 9, which contributes to the reorganization of the extracellular matrix and cell invasiveness. The tropomyosin receptor kinase A inhibitor GW441756 reverses all these responses. Co-immunoprecipitation experiments in both cell lines show that nerve growth factor triggers the assembly of the TrkA/β1-integrin/FAK/Src complex, thereby activating several downstream effectors. GW441756 prevents the complex assembly induced by nerve growth factor as well as the activation of its dependent signaling. Pharmacological inhibition of the tyrosine kinases Src and FAK (focal adhesion kinase), together with the silencing of β1-integrin, shows that the tyrosine kinases impinge on both proliferation and motility, while β1-integrin is needed for motility induced by nerve growth factor in triple-negative breast cancer cells. The present data support the key role of the nerve growth factor/tropomyosin receptor kinase A pathway in triple-negative breast cancer and offer new hints in the diagnostic and therapeutic management of patients.
Collapse
Affiliation(s)
- Marzia Di Donato
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Galasso
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Pia Giovannelli
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonio A Sinisi
- Dipartimento di Scienze Mediche e Chirurgiche Avanzate, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Antimo Migliaccio
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Gabriella Castoria
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
34
|
Ehsani M, David FO, Baniahmad A. Androgen Receptor-Dependent Mechanisms Mediating Drug Resistance in Prostate Cancer. Cancers (Basel) 2021; 13:1534. [PMID: 33810413 PMCID: PMC8037957 DOI: 10.3390/cancers13071534] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 12/16/2022] Open
Abstract
Androgen receptor (AR) is a main driver of prostate cancer (PCa) growth and progression as well as the key drug target. Appropriate PCa treatments differ depending on the stage of cancer at diagnosis. Although androgen deprivation therapy (ADT) of PCa is initially effective, eventually tumors develop resistance to the drug within 2-3 years of treatment onset leading to castration resistant PCa (CRPC). Castration resistance is usually mediated by reactivation of AR signaling. Eventually, PCa develops additional resistance towards treatment with AR antagonists that occur regularly, also mostly due to bypass mechanisms that activate AR signaling. This tumor evolution with selection upon therapy is presumably based on a high degree of tumor heterogenicity and plasticity that allows PCa cells to proliferate and develop adaptive signaling to the treatment and evolve pathways in therapy resistance, including resistance to chemotherapy. The therapy-resistant PCa phenotype is associated with more aggressiveness and increased metastatic ability. By far, drug resistance remains a major cause of PCa treatment failure and lethality. In this review, various acquired and intrinsic mechanisms that are AR‑dependent and contribute to PCa drug resistance will be discussed.
Collapse
Affiliation(s)
| | | | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Am Klinikum 1, 07740 Jena, Germany; (M.E.); (F.O.D.)
| |
Collapse
|
35
|
Rienzo M, Sorrentino A, Di Zazzo E, Di Donato M, Carafa V, Marino MM, De Rosa C, Gazzerro P, Castoria G, Altucci L, Casamassimi A, Abbondanza C. Searching for a Putative Mechanism of RIZ2 Tumor-Promoting Function in Cancer Models. Front Oncol 2021; 10:583533. [PMID: 33585202 PMCID: PMC7880127 DOI: 10.3389/fonc.2020.583533] [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: 07/15/2020] [Accepted: 12/08/2020] [Indexed: 12/16/2022] Open
Abstract
Positive Regulatory Domain (PRDM) gene family members commonly express two main molecular variants, the PR-plus isoform usually acting as tumor suppressor and the PR-minus one functioning as oncogene. Accordingly, PRDM2/RIZ encodes for RIZ1 (PR-plus) and RIZ2 (PR-minus). In human cancers, genetic or epigenetic modifications induce RIZ1 silencing with an expression level imbalance in favor of RIZ2 that could be relevant for tumorigenesis. Additionally, in estrogen target cells and tissues, estradiol increases RIZ2 expression level with concurrent increase of cell proliferation and survival. Several attempts to study RIZ2 function in HeLa or MCF-7 cells by its over-expression were unsuccessful. Thus, we over-expressed RIZ2 in HEK-293 cells, which are both RIZ1 and RIZ2 positive but unresponsive to estrogens. The forced RIZ2 expression increased cell viability and growth, prompted the G2-to-M phase transition and organoids formation. Accordingly, microarray analysis revealed that RIZ2 regulates several genes involved in mitosis. Consistently, RIZ silencing in both estrogen-responsive MCF-7 and -unresponsive MDA-MB-231 cells induced a reduction of cell proliferation and an increase of apoptosis rate. Our findings add novel insights on the putative RIZ2 tumor-promoting functions, although additional attempts are warranted to depict the underlying molecular mechanism.
Collapse
Affiliation(s)
- Monica Rienzo
- Department of Environmental, Biological, and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Anna Sorrentino
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Erika Di Zazzo
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.,Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Marzia Di Donato
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vincenzo Carafa
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Michela Marino
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Caterina De Rosa
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Gabriella Castoria
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Amelia Casamassimi
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Ciro Abbondanza
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
36
|
The androgen receptor/filamin A complex as a target in prostate cancer microenvironment. Cell Death Dis 2021; 12:127. [PMID: 33500395 PMCID: PMC7838283 DOI: 10.1038/s41419-021-03402-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
Prostate cancer represents the major cause of cancer-related death in men and patients frequently develop drug-resistance and metastatic disease. Most studies focus on hormone-resistance mechanisms related to androgen receptor mutations or to the acquired property of prostate cancer cells to over-activate signaling pathways. Tumor microenvironment plays a critical role in prostate cancer progression. However, the mechanism involving androgen/androgen receptor signaling in cancer associated fibroblasts and consequences for prostate cancer progression still remains elusive. We now report that prostate cancer associated fibroblasts express a transcriptional-incompetent androgen receptor. Upon androgen challenging, the receptor co-localizes with the scaffold protein filamin A in the extra-nuclear compartment of fibroblasts, thus mediating their migration and invasiveness. Cancer-associated fibroblasts move towards epithelial prostate cancer cells in 2D and 3D cultures, thereby inducing an increase of the prostate cancer organoid size. Androgen enhances both these effects through androgen receptor/filamin A complex assembly in cancer-associated fibroblasts. An androgen receptor-derived stapled peptide, which disrupts the androgen receptor/filamin A complex assembly, abolishes the androgen-dependent migration and invasiveness of cancer associated fibroblasts. Notably, the peptide impairs the androgen-induced invasiveness of CAFs in 2D models and reduces the overall tumor area in androgen-treated 3D co-culture. The androgen receptor in association with β1 integrin and membrane type-matrix metalloproteinase 1 activates a protease cascade triggering extracellular matrix remodeling. The peptide also impairs the androgen activation of this cascade. This study offers a potential new marker, the androgen receptor/filamin A complex, and a new therapeutic approach targeting intracellular pathways activated by the androgen/androgen receptor axis in prostate cancer-associated fibroblasts. Such a strategy, alone or in combination with conventional therapies, may allow a more efficient treatment of prostate cancer.
Collapse
|
37
|
Nerve growth factor interacts with CHRM4 and promotes neuroendocrine differentiation of prostate cancer and castration resistance. Commun Biol 2021; 4:22. [PMID: 33398073 PMCID: PMC7782543 DOI: 10.1038/s42003-020-01549-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
Nerve growth factor (NGF) contributes to the progression of malignancy. However, the functional role and regulatory mechanisms of NGF in the development of neuroendocrine prostate cancer (NEPC) are unclear. Here, we show that an androgen-deprivation therapy (ADT)-stimulated transcription factor, ZBTB46, upregulated NGF via ZBTB46 mediated-transcriptional activation of NGF. NGF regulates NEPC differentiation by physically interacting with a G-protein-coupled receptor, cholinergic receptor muscarinic 4 (CHRM4), after ADT. Pharmacologic NGF blockade and NGF knockdown markedly inhibited CHRM4-mediated NEPC differentiation and AKT-MYCN signaling activation. CHRM4 stimulation was associated with ADT resistance and was significantly correlated with increased NGF in high-grade and small-cell neuroendocrine prostate cancer (SCNC) patient samples. Our results reveal a role of the NGF in the development of NEPC that is linked to ZBTB46 upregulation and CHRM4 accumulation. Our study provides evidence that the NGF-CHRM4 axis has potential to be considered as a therapeutic target to impair NEPC progression. Here, the authors discover that NGF, upregulated by transcription factor ZBTB46 in prostate cancer exposed to androgen therapy, promotes neuroendocrine differentiation. They show that NGF interacts with the GPCR CHRM4, that both NGF and CHRM4 are upregulated in highly metastatic prostate cancer and that targeting NGF reduces therapy resistance in a mouse xenograft model.
Collapse
|
38
|
Formaggio N, Rubin MA, Theurillat JP. Loss and revival of androgen receptor signaling in advanced prostate cancer. Oncogene 2021; 40:1205-1216. [PMID: 33420371 PMCID: PMC7892335 DOI: 10.1038/s41388-020-01598-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/20/2020] [Accepted: 11/30/2020] [Indexed: 02/08/2023]
Abstract
Targeting the androgen receptor (AR) signaling axis has been, over decades, the mainstay of prostate cancer therapy. More potent inhibitors of androgen synthesis and antiandrogens have emerged and have been successfully implemented in clinical practice. That said, the stronger inhibition of the AR signaling axis has led in recent years to an increase of prostate cancers that de-differentiate into AR-negative disease. Unfortunately, this process is intimately linked with a poor prognosis. Here, we review the molecular mechanisms that enable cancer cells to switch from an AR-positive to an AR-negative disease and efforts to prevent/revert this process and thereby maintain/restore AR-dependence.
Collapse
Affiliation(s)
- Nicolò Formaggio
- grid.29078.340000 0001 2203 2861Institute of Oncology Research, Università della Svizzera italiana, Lugano, Switzerland
| | - Mark A. Rubin
- grid.5734.50000 0001 0726 5157Department for BioMedical Research and Bern Center of Precision Medicine, University of Bern and Inselspital, Bern, Switzerland
| | - Jean-Philippe Theurillat
- grid.29078.340000 0001 2203 2861Institute of Oncology Research, Università della Svizzera italiana, Lugano, Switzerland
| |
Collapse
|
39
|
Dahl HC, Kanchwala M, Thomas-Jardin SE, Sandhu A, Kanumuri P, Nawas AF, Xing C, Lin C, Frigo DE, Delk NA. Chronic IL-1 exposure drives LNCaP cells to evolve androgen and AR independence. PLoS One 2020; 15:e0242970. [PMID: 33326447 PMCID: PMC7743957 DOI: 10.1371/journal.pone.0242970] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/12/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic inflammation promotes prostate cancer (PCa) initiation and progression. We previously reported that acute intereluekin-1 (IL-1) exposure represses androgen receptor (AR) accumulation and activity, providing a possible mechanism for IL-1-mediated development of androgen- and AR-independent PCa. Given that acute inflammation is quickly resolved, and chronic inflammation is, instead, co-opted by cancer cells to promote tumorigenicity, we set out to determine if chronic IL-1 exposure leads to similar repression of AR and AR activity observed for acute IL-1 exposure and to determine if chronic IL-1 exposure selects for androgen- and AR-independent PCa cells. We generated isogenic sublines from LNCaP cells chronically exposed to IL-1α or IL-1β. Cells were treated with IL-1α, IL-1β, TNFα or HS-5 bone marrow stromal cells conditioned medium to assess cell viability in the presence of cytotoxic inflammatory cytokines. Cell viability was also assessed following serum starvation, AR siRNA silencing and enzalutamide treatment. Finally, RNA sequencing was performed for the IL-1 sublines. MTT, RT-qPCR and western blot analysis show that the sublines evolved resistance to inflammation-induced cytotoxicity and intracellular signaling and evolved reduced sensitivity to siRNA-mediated loss of AR, serum deprivation and enzalutamide. Differential gene expression reveals that canonical AR signaling is aberrant in the IL-1 sublines, where the cells show constitutive PSA repression and basally high KLK2 and NKX3.1 mRNA levels and bioinformatics analysis predicts that pro-survival and pro-tumorigenic pathways are activated in the sublines. Our data provide evidence that chronic IL-1 exposure promotes PCa cell androgen and AR independence and, thus, supports CRPCa development.
Collapse
Affiliation(s)
- Haley C. Dahl
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX, United States of America
| | - Mohammed Kanchwala
- McDermott Center of Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Shayna E. Thomas-Jardin
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX, United States of America
| | - Amrit Sandhu
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX, United States of America
| | - Preethi Kanumuri
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX, United States of America
| | - Afshan F. Nawas
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX, United States of America
| | - Chao Xing
- McDermott Center of Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
- Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
- Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Chenchu Lin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Daniel E. Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, United States of America
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States of America
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- The Houston Methodist Research Institute, Houston, TX, United States of America
| | - Nikki A. Delk
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX, United States of America
| |
Collapse
|
40
|
Wei J, Huang J, Kuang Y, Li Y, Zhong D, Song J. Metformin inhibits proliferation of oral squamous cell carcinoma cells by suppressing proteolysis of nerve growth factor receptor. Arch Oral Biol 2020; 121:104971. [PMID: 33220581 DOI: 10.1016/j.archoralbio.2020.104971] [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: 02/27/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE We aimed to explore the effects of metformin on oral squamous cell carcinoma (OSCC) cell proliferation and the associated molecular mechanisms. METHODS We established an OSCC model in SCC15 cells overexpressing nerve growth factor receptor (NGFR) or the N-terminal region (aa 1-250; NGFR-N), and assessed cell proliferation by CCK-8 assay, colony formation assay, and cell cycle analysis. Levels of NGFR and related genes and proteins were detected by qPCR and western blotting, and NGFR and NGFR-N affinity for p53 was assessed by immunoprecipitation assay. Additionally, the effects of NGFR and NGFR-N on p53 binding with its downstream gene promoters were analyzed by chromatin immunoprecipitation. RESULTS Metformin inhibited OSCC cell proliferation and blocked NGFR proteolysis, thereby reducing the generation of its intracellular domain and NGFR-N. Moreover, compared with NGFR, NGFR-N showed higher affinity for p53 and more strongly inactivated p53 to promote cell proliferation. Furthermore, upregulation of NGFR-N downregulated levels of p53-specific downstream transcripts and proteins, whereas these levels were significantly upregulated in metformin-treated cells overexpressing NGFR. CONCLUSIONS These results showed that metformin inhibited cell proliferation by suppressing NGFR proteolysis, thereby promoting its antitumor effect in OSCC and offering novel insight into a role for metformin in OSCC treatment.
Collapse
Affiliation(s)
- Jingjing Wei
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Jiao Huang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Yunchun Kuang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Yongkai Li
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Daiqin Zhong
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China.
| |
Collapse
|
41
|
Ng CF, Chiu PKF, Yee CH, Lau BSY, Leung SCH, Teoh JYC. Effect of androgen deprivation therapy on cardiovascular function in Chinese patients with advanced prostate cancer: a prospective cohort study. Sci Rep 2020; 10:18060. [PMID: 33093594 PMCID: PMC7582924 DOI: 10.1038/s41598-020-75139-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/12/2020] [Indexed: 11/09/2022] Open
Abstract
Androgen deprivation therapy (ADT) is the standard treatment for advanced prostate cancer, but its effect on cardiovascular and metabolic function in Asian patients is still inconclusive. We prospectively assess the effects of ADT on 36 patients with advanced prostate cancer, with reference to another 24 prostate cancer patients not requiring ADT, for 2 years. Patients’ anthropometric, metabolic and vascular parameters were assessed every six-monthly. The baseline parameters of the two groups were comparable. There was a significant negative effect of the usage of ADT on the changes in BMI (p = 0.020), waist to hip ratio (p = 0.005), body fat percentage (p = 0.012), and high-density-lipoprotein (p = 0.012). ADT-patients were 4.9 times more likely to have metabolic syndrome at 24 months. (CI 0.889–27.193, p = 0.068). The Framingham risk score (p = 0.018) and pulse-wave-velocity (p = 0.024) for ADT-group were also significantly higher than controls, which signified increase in cardiovascular risk. Although there was no statistically significant difference in ischemic cardiovascular events between two groups, a trend for more events in ADT-group was observed. Therefore, Asian patients have increased cardiovascular and metabolic risks after being treated with ADT for two years. Appropriate counselling and monitoring of associated complications with ADT are essential.
Collapse
Affiliation(s)
- Chi-Fai Ng
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | - Peter K F Chiu
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chi-Hang Yee
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Becky S Y Lau
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Steven C H Leung
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jeremy Y C Teoh
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
42
|
Fontana F, Raimondi M, Marzagalli M, Sommariva M, Gagliano N, Limonta P. Three-Dimensional Cell Cultures as an In Vitro Tool for Prostate Cancer Modeling and Drug Discovery. Int J Mol Sci 2020; 21:E6806. [PMID: 32948069 PMCID: PMC7554845 DOI: 10.3390/ijms21186806] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
In the last decade, three-dimensional (3D) cell culture technology has gained a lot of interest due to its ability to better recapitulate the in vivo organization and microenvironment of in vitro cultured cancer cells. In particular, 3D tumor models have demonstrated several different characteristics compared with traditional two-dimensional (2D) cultures and have provided an interesting link between the latter and animal experiments. Indeed, 3D cell cultures represent a useful platform for the identification of the biological features of cancer cells as well as for the screening of novel antitumor agents. The present review is aimed at summarizing the most common 3D cell culture methods and applications, with a focus on prostate cancer modeling and drug discovery.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Androgens
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Culture Techniques/instrumentation
- Cell Culture Techniques/methods
- Cell Hypoxia
- Drug Discovery/methods
- Drug Screening Assays, Antitumor/instrumentation
- Drug Screening Assays, Antitumor/methods
- Energy Metabolism
- Epithelial-Mesenchymal Transition
- Extracellular Matrix/metabolism
- Humans
- Inflammation
- Male
- Molecular Targeted Therapy
- Monitoring, Immunologic
- Neoplasm Metastasis
- Neoplasm Proteins/metabolism
- Neoplasms, Hormone-Dependent/drug therapy
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Neoplastic Stem Cells/cytology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neovascularization, Pathologic/drug therapy
- Oxidation-Reduction
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/therapy
- Spheroids, Cellular/drug effects
- Therapies, Investigational
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy; (M.R.); (M.M.); (P.L.)
| | - Michela Raimondi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy; (M.R.); (M.M.); (P.L.)
| | - Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy; (M.R.); (M.M.); (P.L.)
| | - Michele Sommariva
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, via Mangiagalli 31, 20133 Milan, Italy; (M.S.); (N.G.)
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, via Mangiagalli 31, 20133 Milan, Italy; (M.S.); (N.G.)
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy; (M.R.); (M.M.); (P.L.)
| |
Collapse
|
43
|
Tang L, Xu M, Zhang L, Qu L, Liu X. Role of αVβ3 in Prostate Cancer: Metastasis Initiator and Important Therapeutic Target. Onco Targets Ther 2020; 13:7411-7422. [PMID: 32801764 PMCID: PMC7395689 DOI: 10.2147/ott.s258252] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Abstract
In prostate cancer, distant organ metastasis is the leading cause of patient death. Although the mechanism of malignant tumor metastasis is unclear, studies have confirmed that integrin αVβ3 plays an important role in this process. In prostate cancer, αVβ3 mediates adhesion, invasion, immune escape and neovascularization through interactions with different ligands. Among these ligands and in addition to proteins that are directly related to tumor invasion, other proteins that contain the RGD structure could also bind to αVβ3 and cause a number of biological effects. In this article, we summarized the ligand and downstream proteins related to αVβ3-mediated prostate cancer metastasis as well as some diagnostic and therapeutic measures targeting αVβ3.
Collapse
Affiliation(s)
- Lin Tang
- College of Mathematics and Computer Science, Chifeng University, Chifeng, The Inner Mongol Autonomous Region 024005, People's Republic of China
| | - Meng Xu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, People's Republic of China.,R&D Department, Seekgene Technology Co., Ltd, Beijing 100000, People's Republic of China
| | - Long Zhang
- Department of Hepatobiliary Surgery, Yidu Central Hospital, Weifang, Shandong 262500, People's Republic of China
| | - Lin Qu
- Department of Orthopaedic Surgery, Anshan Hospital of the First Hospital of China Medical University, Anshan, Liaoning 114000, People's Republic of China
| | - Xiaoyan Liu
- Department of Pathology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100000, People's Republic of China
| |
Collapse
|
44
|
TGFB2 serves as a link between epithelial-mesenchymal transition and tumor mutation burden in gastric cancer. Int Immunopharmacol 2020; 84:106532. [PMID: 32388013 DOI: 10.1016/j.intimp.2020.106532] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/19/2020] [Accepted: 04/19/2020] [Indexed: 02/06/2023]
Abstract
Immune checkpoint blockade (ICB) has been a major breakthrough in various cancers including gastric cancer (GC), yet the clinical outcomes remain poor. Currently, epithelial-mesenchymal transition (EMT) has been reported to be associated with tumor mutational burden (TMB), which can cause lack of response to ICB. However, the underlying mechanism remains unknown. Members of the transforming growth factorβ (TGFB) family are regarded as the main mediators of EMT, yet how TGFB2 drives EMT in GC is not fully understood. In this study, we found that overexpression of TGFB2 was correlated with poor prognosis in TGCA-STAD and four GEO GC datasets.Gene set enrichment analysis revealed that the EMT pathway was significantly enriched in the high TGFB2 expression group, whilst the TMB-related pathways including mismatch repair, base excision repair, and DNA replication were strongly enriched in the low expression group. Furthermore, EMT score analysis, WGCNA and functional analysis showed that TGFB2 was co-expressed with neurite-related pathways that might drive EMT. Also, CIBERSORT analysis revealed that tumor-infiltrating immune cells like T follicular helper cells might participate in the process of TGFB2 affecting TMB levels in GC. Moreover, in other various cancers, TGFB2 was also negatively correlated with TMB levels as well as ICB response. Overall, these results revealed that TGFB2 could play a vital role in linking EMT and TMB in GC, suggesting that TGFB2 may be a predictive therapeutic target for GC.
Collapse
|
45
|
ADAMTS-15 Has a Tumor Suppressor Role in Prostate Cancer. Biomolecules 2020; 10:biom10050682. [PMID: 32354091 PMCID: PMC7277637 DOI: 10.3390/biom10050682] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/23/2020] [Accepted: 04/25/2020] [Indexed: 12/18/2022] Open
Abstract
Extracellular matrix remodeling has emerged as an important factor in many cancers. Proteoglycans, including versican (VCAN), are regulated via cleavage by the proteolytic actions of A Disintegrin-like And Metalloproteinase domain with Thrombospondin-1 motif (ADAMTS) family members. Alterations in the balance between Proteoglycans and ADAMTS enzymes have been proposed to contribute to cancer progression. Here, we analyzed the expression of ADAMTS-15 in human prostate cancer, and investigated the effects of enforced expression in prostate cancer cell lines. ADAMTS-15 was found to be expressed in human prostate cancer biopsies with evidence of co-localization with VCAN and its bioactive cleavage fragment versikine. Enforced expression of ADAMTS-15, but not a catalytically-inactive version, decreased cell proliferation and migration of the ‘castrate-resistant’ PC3 prostate cancer cell line in vitro, with survival increased. Analysis of ‘androgen-responsive’ LNCaP prostate cancer cells in vivo in NOD/SCID mice revealed that ADAMTS-15 expression caused slower growing tumors, which resulted in increased survival. This was not observed in castrated mice or with cells expressing catalytically-inactive ADAMTS-15. Collectively, this research identifies the enzymatic function of ADAMTS-15 as having a tumor suppressor role in prostate cancer, possibly in concert with androgens, and that VCAN represents a likely key substrate, highlighting potential new options for the clinic.
Collapse
|
46
|
The new biocompatible material for mouse ovarian follicle development in three-dimensional in vitro culture systems. Theriogenology 2020; 144:33-40. [DOI: 10.1016/j.theriogenology.2019.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/15/2019] [Accepted: 12/15/2019] [Indexed: 12/30/2022]
|
47
|
Lin C, Ren Z, Yang X, Yang R, Chen Y, Liu Z, Dai Z, Zhang Y, He Y, Zhang C, Wang X, Cao W, Ji T. Nerve growth factor (NGF)-TrkA axis in head and neck squamous cell carcinoma triggers EMT and confers resistance to the EGFR inhibitor erlotinib. Cancer Lett 2020; 472:81-96. [PMID: 31838083 DOI: 10.1016/j.canlet.2019.12.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 02/08/2023]
Abstract
Understanding the molecular mechanisms regulating tumor dissemination and therapeutic resistance is of central importance for effective cancer therapies. Here, we report that nerve growth factor (NGF) and its receptor TrkA facilitate epithelial-mesenchymal transition (EMT) and EGFR inhibitor resistance via STAT3 activation in head and neck squamous cell carcinoma (HNSCC). Both NGF and TrkA expression were elevated in HNSCC, indicating poor clinical outcomes. NGF was highly expressed in cancer cells and nerves in perineural niche, whereas TrkA expression was higher in cancer cells with perineural invasion. The NGF/TrkA axis could promote HNSCC cell dissemination and trigger EMT via STAT3 activation. Moreover, we discovered that the NGF/TrkA axis conferred resistance to the EGFR inhibitor erlotinib via EMT processes in HNSCC cells. Blocking TrkA signaling markedly reversed EMT and sensitized HNSCC cells to erlotinib in both in vitro and in vivo models. Overall, our results demonstrate novel evidence that the paracrine NGF/TrkA axis favors EMT and confers EGFR-targeted therapeutic resistance in HNSCC.
Collapse
Affiliation(s)
- Chengzhong Lin
- The 2nd Dental Center, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China; National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Zhenhu Ren
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Xi Yang
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Rong Yang
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Yiming Chen
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Zheqi Liu
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Zhenlin Dai
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Yu Zhang
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Youya He
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Chunye Zhang
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral Pathology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xu Wang
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China.
| | - Wei Cao
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China.
| | - Tong Ji
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
48
|
Zhang T, Sun Y, Zheng T, Wang R, Jia D, Zhang W. MLPH Accelerates the Epithelial-Mesenchymal Transition in Prostate Cancer. Onco Targets Ther 2020; 13:701-708. [PMID: 32158222 PMCID: PMC6986253 DOI: 10.2147/ott.s225023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 11/20/2019] [Indexed: 01/15/2023] Open
Abstract
INTRODUCTION Prostate cancer (PC) is the second greatest cause of cancer deaths globally. PC presents a poor prognosis once it metastasizes. There is considerable proof of vital epithelial-mesenchymal transition (EMT) functionality in PC metastasis. Previous studies revealed that melanophilin (MLPH) is associated with PC; however, its role in PC remains poorly understood. METHODS Bioinformatics analyses were performed. The cellular responses to MLPH knockdown were examined in HCC cell lines via wound healing assay, migration and invasion assay, Western blotting. RESULTS Analysis of the PROGgeneV2 database revealed that high MLPH expression might indicate poor overall survival. MLPH knockdown reduced PC cell migration, proliferation, and invasion. MLPH downregulation in vivo resulted in a lower growth rate and fewer metastatic nodules in lung tissues. Furthermore, MLPH knockdown recovered downregulated expression of the mesenchymal marker N-cadherin and the epithelial marker E-cadherin following a decrease in β-catenin. CONCLUSION These results indicate that progression of PC is stimulated via MLPH-dependent initiation of the EMT.
Collapse
Affiliation(s)
- Tianbiao Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou450052, People’s Republic of China
| | - Yangyang Sun
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou450052, People’s Republic of China
| | - Tao Zheng
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou450052, People’s Republic of China
| | - Rui Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou450052, People’s Republic of China
| | - Donghui Jia
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou450052, People’s Republic of China
| | - Weixing Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou450052, People’s Republic of China
| |
Collapse
|
49
|
Itami Y, Miyake M, Ohnishi S, Tatsumi Y, Gotoh D, Hori S, Morizawa Y, Iida K, Ohnishi K, Nakai Y, Inoue T, Anai S, Tanaka N, Fujii T, Shimada K, Furuya H, Khadka VS, Deng Y, Fujimoto K. Disabled Homolog 2 (DAB2) Protein in Tumor Microenvironment Correlates with Aggressive Phenotype in Human Urothelial Carcinoma of the Bladder. Diagnostics (Basel) 2020; 10:E54. [PMID: 31968685 PMCID: PMC7168324 DOI: 10.3390/diagnostics10010054] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/14/2020] [Accepted: 01/17/2020] [Indexed: 12/14/2022] Open
Abstract
Disabled homolog-2 (DAB2) has been reported to be a tumor suppressor gene. However, a number of contrary studies suggested that DAB2 promotes tumor invasion in urothelial carcinoma of the bladder (UCB). Here, we investigated the clinical role and biological function of DAB2 in human UCB. Immunohistochemical staining analysis for DAB2 was carried out on UCB tissue specimens. DAB2 expression levels were compared with clinicopathological factors. DAB2 was knocked-down by small interfering RNA (siRNA) transfection, and then its effects on cell proliferation, invasion, and migration, and changes to epithelial-mesenchymal transition (EMT)-related proteins were evaluated. In our in vivo assays, tumor-bearing athymic nude mice subcutaneously inoculated with human UCB cells (MGH-U-3 or UM-UC-3) were treated by DAB2-targeting siRNA. Higher expression of DAB2 was associated with higher clinical T category, high tumor grade, and poor oncological outcome. The knock-down of DAB2 decreased both invasion and migration ability and expression of EMT-related proteins. Significant inhibitory effects on tumor growth and invasion were observed in xenograft tumors of UM-UC-3 treated by DAB2-targeting siRNA. Our findings suggested that DAB2 expression was associated with poor prognosis through increased oncogenic properties including tumor proliferation, migration, invasion, and enhancement of EMT in human UCB.
Collapse
Affiliation(s)
- Yoshitaka Itami
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Makito Miyake
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Sayuri Ohnishi
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Yoshihiro Tatsumi
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Daisuke Gotoh
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Shunta Hori
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Yousuke Morizawa
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Kota Iida
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Kenta Ohnishi
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Yasushi Nakai
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Takeshi Inoue
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Satoshi Anai
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Nobumichi Tanaka
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| | - Tomomi Fujii
- Department of Pathology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan;
| | - Keiji Shimada
- Department of Pathology, Nara City Hospital, 1-50-1 Higashi kidera-cho, Nara 630-8305, Japan;
| | - Hideki Furuya
- Division of Urology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Vedbar S. Khadka
- Bioinformatics Core, Department of Complementary and Integrative Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI 96813, USA; (V.S.K.); (Y.D.)
| | - Youping Deng
- Bioinformatics Core, Department of Complementary and Integrative Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI 96813, USA; (V.S.K.); (Y.D.)
| | - Kiyohide Fujimoto
- Department of Urology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan; (Y.I.); (M.M.); (S.O.); (Y.T.); (D.G.); (S.H.); (Y.M.); (K.I.); (K.O.); (Y.N.); (T.I.); (S.A.); (N.T.)
| |
Collapse
|
50
|
Relevance of Neurotrophin Receptors CD271 and TrkC for Prognosis, Migration, and Proliferation in Head and Neck Squamous Cell Carcinoma. Cells 2019; 8:cells8101167. [PMID: 31569361 PMCID: PMC6830344 DOI: 10.3390/cells8101167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/16/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide and often has a poor prognosis. The present study investigated the role of the low affinity nerve growth factor receptor CD271 as a putative therapy target in HNSCC. Neurotrophins that bind to CD271 also have a high affinity for the tropomyosin receptor kinase family (Trk), consisting of TrkA, TrkB, and TrkC, which must also be considered in addition to CD271. A retrospective study and functional in vitro cell line tests (migration assay and cell sorting) were conducted in order to evaluate the relevance of CD271 expression alone and with regard to Trk expression. CD271 and Trks were heterogeneously expressed in human HNSCC. The vast majority of tumors exhibited CD271 and TrkA, whereas only half of the tumors expressed TrkB and TrkC. High expression of CD271-positive cells predicted a bad clinical outcome of patients with HNSCC and was associated with distant metastases. However, the human carcinomas that also expressed TrkC had a reduced correlation with distant metastases and better survival rates. In vitro, CD271 expression marked a subpopulation with higher proliferation rates, but proliferation was lower in tumor cells that co-expressed CD271 and TrkC. The CD271 inhibitor LM11A 31 suppressed cell motility in vitro. However, neither TrkA nor TrkB expression were linked to prognosis or cell proliferation. We conclude that CD271 is a promising candidate that provides prognostic information for HNSCC and could be a putative target for HNSCC treatment.
Collapse
|