1
|
Avasthi KK, Choi JW, Glushko T, Manley BJ, Yu A, Park JY, Brown JS, Pow-Sang J, Gantenby R, Wang L, Balagurunathan Y. Extracellular Microvesicle MicroRNAs and Imaging Metrics Improve the Detection of Aggressive Prostate Cancer: A Pilot Study. Cancers (Basel) 2025; 17:835. [PMID: 40075682 PMCID: PMC11898942 DOI: 10.3390/cancers17050835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Prostate cancer (PCa) is the most diagnosed cancer in men worldwide. Early diagnosis of the disease provides better treatment options for these patients. Recent studies have demonstrated that plasma-based extracellular vesicle microRNAs (miRNAs) are functionally linked to cancer progression, metastasis, and aggressiveness. The use of magnetic resonance imaging (MRI) as the standard of care provides an overall assessment of prostate disease. Quantitative metrics (radiomics) from the MRI provide a better evaluation of the tumor and have been shown to improve disease detection. Methods: We conducted a study on prostate cancer patients, analyzing baseline blood plasma and MRI data. Exosomes were isolated from blood plasma samples to quantify miRNAs, while MRI scans provided detailed tumor morphology. Radiomics features from MRI and miRNA expression data were integrated to develop predictive models, which were evaluated using ROC curve analysis, highlighting the multivariable model's effectiveness. Results: Our findings indicate that the univariate feature-based model with the highest Youden's index achieved average areas under the receiver operating characteristic (ROC) curve of 0.76, 0.82, and 0.84 for miRNA, MR-T2W, and MR-ADC features, respectively, in identifying clinically aggressive (Gleason grade) disease. The multivariable feature-based model yielded an average area under the curve (AUC) of 0.88 and 0.95 using combinations of miRNA markers with imaging features in MR-ADC and MR-T2W, respectively. Conclusions: Our study demonstrates that combining miRNA markers with MRI-based radiomics improves the identification of clinically aggressive prostate cancer.
Collapse
Affiliation(s)
- Kapil K. Avasthi
- Department of Tumor Microenvironment and Metastasis, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Jung W. Choi
- Department of Diagnostic & Interventional Radiology, Moffitt Cancer Center, Tampa, FL 33612, USA; (J.W.C.); (R.G.)
| | - Tetiana Glushko
- Department of Diagnostic & Interventional Radiology, Moffitt Cancer Center, Tampa, FL 33612, USA; (J.W.C.); (R.G.)
| | - Brandon J. Manley
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (B.J.M.); (A.Y.); (J.P.-S.)
| | - Alice Yu
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (B.J.M.); (A.Y.); (J.P.-S.)
| | - Jong Y. Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Joel S. Brown
- Department of Mathematical Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Julio Pow-Sang
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (B.J.M.); (A.Y.); (J.P.-S.)
| | - Robert Gantenby
- Department of Diagnostic & Interventional Radiology, Moffitt Cancer Center, Tampa, FL 33612, USA; (J.W.C.); (R.G.)
| | - Liang Wang
- Department of Tumor Microenvironment and Metastasis, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | | |
Collapse
|
2
|
Li P, Gong Q, Wang D, Zhang Z, Zhang W. Clusterin mediates hydroquinone-induced cytotoxic responses in HL-60 differentiated cells. Sci Rep 2024; 14:30402. [PMID: 39638872 PMCID: PMC11621340 DOI: 10.1038/s41598-024-82140-0] [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: 08/04/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024] Open
Abstract
Benzene is a crucial industrial hydrocarbon, posing significant health risks due to its toxic metabolites like hydroquinone (HQ). This study investigates the role of clusterin (CLU) in benzene toxicity by analyzing its protein and mRNA levels, as well as the expression of Bcl-2 and Bax, to evaluate the feasibility of CLU as a biomarker for chronic benzene poisoning. HL-60 cells were induced to differentiate into neutrophil-like cells using 1% Dimethyl Sulfoxide (DMSO). Enzyme-linked immunosorbent assay (ELISA) and RT-PCR were used to analyze CLU protein and mRNA levels. ELISA was employed to detect sCLU protein content in cell culture supernatants, and western blot was used to assess Bcl-2 and Bax expression. The optimal time for 1% DMSO to induce HL-60 cells into neutrophil-like cells was 48 h. As HQ concentration increased, HL-60 cell viability decreased, CLU protein and sCLU protein levels in the supernatant decreased, CLU mRNA levels decreased, Bcl-2 protein expression decreased, and Bax expression increased. HQ exposure reduces CLU protein concentration and mRNA levels in neutrophil-like cells induced from HL-60 cells, indicating that CLU could be a potential biomarker for chronic benzene poisoning.
Collapse
Affiliation(s)
- Peimao Li
- Medical Laboratory, Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, 518020, China.
| | - Qirui Gong
- Medical Laboratory College, Hebei North University in China, Zhangjiakou, 075000, Hebei, China
| | - Dianpeng Wang
- Medical Laboratory, Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, 518020, China
| | - Zhimin Zhang
- Medical Laboratory, Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, 518020, China
| | - Wen Zhang
- Medical Laboratory, Shenzhen Prevention and Treatment Center for Occupational Diseases, Shenzhen, 518020, China
| |
Collapse
|
3
|
Mamun M, Zheng YC, Wang N, Wang B, Zhang Y, Pang JR, Shen DD, Liu HM, Gao Y. Decoding CLU (Clusterin): Conquering cancer treatment resistance and immunological barriers. Int Immunopharmacol 2024; 137:112355. [PMID: 38851158 DOI: 10.1016/j.intimp.2024.112355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/10/2024]
Abstract
One major obstacle in the treatment of cancer is the presence of proteins resistant to cancer therapy, which can impede the effectiveness of traditional approaches such as radiation and chemotherapy. This resistance can lead to disease progression and cause treatment failure. Extensive research is currently focused on studying these proteins to create tailored treatments that can circumvent resistance mechanisms. CLU (Clusterin), a chaperone protein, has gained notoriety for its role in promoting resistance to a wide range of cancer treatments, including chemotherapy, radiation therapy, and targeted therapy. The protein has also been discovered to have a role in regulating the immunosuppressive environment within tumors. Its ability to influence oncogenic signaling and inhibit cell death bolster cancer cells resistant against treatments, which poses a significant challenge in the field of oncology. Researchers are actively investigating to the mechanisms by which CLU exerts its resistance-promoting effects, with the ultimate goal of developing strategies to circumvent its impact and enhance the effectiveness of cancer therapies. By exploring CLU's impact on cancer, resistance mechanisms, tumor microenvironment (TME), and therapeutic strategies, this review aims to contribute to the ongoing efforts to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Maa Mamun
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ning Wang
- The School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Bo Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yu Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Jing-Ru Pang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Dan-Dan Shen
- Key Laboratory of Endometrial Disease Prevention and Treatment, Zhengzhou China, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
4
|
Zhu D, Zhang S, Wang X, Xiao C, Cui G, Yang X. Secretory Clusterin Inhibits Dopamine Neuron Apoptosis in MPTP Mice by Preserving Autophagy Activity. Neuroscience 2024; 540:38-47. [PMID: 38242280 DOI: 10.1016/j.neuroscience.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Secretory clusterin (sCLU) plays an important role in the research progress of nervous system diseases. However, the physiological function of sCLU in Parkinson's disease (PD) are unclear. The purpose of this study was to examine the effects of sCLU-mediated autophagy on cell survival and apoptosis inhibition in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. We found that MPTP administration induced prolonged pole-climbing time, shortened traction time and rotarod time, significantly decreased TH protein expression in the SN tissue of mice. In contrast, sCLU -treated mice took less time to climb the pole and had an extended traction time and rotating rod time. Meanwhile, sCLU intervention induced increased expression of the TH protein in the SN of mice. These results indicated that sCLU intervention could reduce the loss of dopamine neurons in the SN area and alleviate dyskinesia in mice. Furthermore, MPTP led to suppressed viability, enhanced apoptosis, an increased Bax/Bcl-2 ratio, and cleaved caspase-3 in the SN of mice, and these effects were abrogated by sCLU intervention. In addition, MPTP increased the levels of P62 protein, decreased Beclin1 protein, decreased the ratio of LC3B-II/LC3B-I, and decreased the numbers of autophagosomes and autophagolysosomes in the SN tissues of mice. These effects were also abrogated by sCLU intervention. Activation of PI3K/AKT/mTOR signaling with MPTP inhibited autophagy in the SN of MPTP mice; however, sCLU treatment activated autophagy in MPTP-induced PD mice by inhibiting PI3K/AKT/mTOR signaling. These data indicated that sCLU treatment had a neuroprotective effect in an MPTP-induced model of PD.
Collapse
Affiliation(s)
- Dongxue Zhu
- Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Shenyang Zhang
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xiaoying Wang
- Department of Ultrasound, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Chenghua Xiao
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Guiyun Cui
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xinxin Yang
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| |
Collapse
|
5
|
Zhang Y, Lv X, Chen L, Liu Y. The role and function of CLU in cancer biology and therapy. Clin Exp Med 2023; 23:1375-1391. [PMID: 36098834 DOI: 10.1007/s10238-022-00885-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/29/2022] [Indexed: 11/03/2022]
Abstract
Clusterin (CLU) is a highly evolutionary conserved glycoprotein with multiple isoform-specific functions and is widely distributed in different species. Accumulated evidence has shown the prominent role of CLU in regulating several essential physiological processes, including programmed cell death, metastasis, invasion, proliferation and cell growth via regulating diverse signaling pathways to mediate cancer progression in various cancers, such as prostate, breast, lung, liver, colon, bladder and pancreatic cancer. Several studies have revealed the potential benefit of inhibiting CLU in CLU inhibition-based targeted cancer therapies in vitro, in vivo or in human, suggesting CLU is a promising therapeutic target. This review discusses the multiple functions and mechanisms of CLU in regulating tumor progression of various cancers and summarizes the inhibitors of CLU used in CLU inhibition-based targeted cancer therapies.
Collapse
Affiliation(s)
- Yefei Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Department of Biochemistry, Institute of Cancer, College of Life Science, Nanjing Normal University, Nanjing, 210023, People's Republic of China
| | - Xiang Lv
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Department of Biochemistry, Institute of Cancer, College of Life Science, Nanjing Normal University, Nanjing, 210023, People's Republic of China
| | - Liming Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Department of Biochemistry, Institute of Cancer, College of Life Science, Nanjing Normal University, Nanjing, 210023, People's Republic of China.
| | - Yan Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Department of Biochemistry, Institute of Cancer, College of Life Science, Nanjing Normal University, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
6
|
Darici S, Jørgensen HG, Huang X, Serafin V, Antolini L, Barozzi P, Luppi M, Forghieri F, Marmiroli S, Zavatti M. Improved efficacy of quizartinib in combination therapy with PI3K inhibition in primary FLT3-ITD AML cells. Adv Biol Regul 2023; 89:100974. [PMID: 37245251 DOI: 10.1016/j.jbior.2023.100974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/09/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Acute myeloid leukemia is a heterogeneous hematopoietic malignancy, characterized by uncontrolled clonal proliferation of abnormal myeloid progenitor cells, with poor outcomes. The internal tandem duplication (ITD) mutation of the Fms-like receptor tyrosine kinase 3 (FLT3) (FLT3-ITD) represents the most common genetic alteration in AML, detected in approximately 30% of AML patients, and is associated with high leukemic burden and poor prognosis. Therefore, this kinase has been regarded as an attractive druggable target for the treatment of FLT3-ITD AML, and selective small molecule inhibitors, such as quizartinib, have been identified and trialled. However, clinical outcomes have been disappointing so far due to poor remission rates, also because of acquired resistance. A strategy to overcome resistance is to combine FLT3 inhibitors with other targeted therapies. In this study, we investigated the preclinical efficacy of the combination of quizartinib with the pan PI3K inhibitor BAY-806946 in FLT3-ITD cell lines and primary cells from AML patients. We show here that BAY-806946 enhanced quizartinib cytotoxicity and, most importantly, that this combination increases the ability of quizartinib to kill CD34+ CD38-leukemia stem cells, whilst sparing normal hematopoietic stem cells. Because constitutively active FLT3 receptor tyrosine kinase is known to boost aberrant PI3K signaling, the increased sensitivity of primary cells to the above combination can be the mechanistic results of the disruption of signaling by vertical inhibition.
Collapse
Affiliation(s)
- Salihanur Darici
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, Largo del Pozzo 71, University of Modena and Reggio Emilia, Modena, 41125, Italy; Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Heather G Jørgensen
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Xu Huang
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Valentina Serafin
- Department of Surgery Oncology and Gastroenterology Oncology and Immunology Section University of Padova, Italy
| | - Ludovica Antolini
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, Largo del Pozzo 71, University of Modena and Reggio Emilia, Modena, 41125, Italy
| | - Patrizia Barozzi
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Di Modena, Via del Pozzo 71, 41124, Modena, Italy
| | - Mario Luppi
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Di Modena, Via del Pozzo 71, 41124, Modena, Italy.
| | - Fabio Forghieri
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria Di Modena, Via del Pozzo 71, 41124, Modena, Italy.
| | - Sandra Marmiroli
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, Largo del Pozzo 71, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Manuela Zavatti
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, Largo del Pozzo 71, University of Modena and Reggio Emilia, Modena, 41125, Italy
| |
Collapse
|
7
|
Sadeghi R, Pirankuraim H, Javanshir ST, Arabi M, Bereimipour A, Javanshir HT, Mahmoodzadeh H, Nayernia K. Risk of secondary tumours in patients with non-metastatic and metastatic human retinoblastoma. Eye (Lond) 2023; 37:2327-2334. [PMID: 36528757 PMCID: PMC10366135 DOI: 10.1038/s41433-022-02345-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Retinoblastoma is an intraocular cancer in children and infants. Despite all the available treatment options and high survival rates in children with retinoblastoma, exposure to secondary tumours in adulthood is one of the concerns that physicians face. In many cases, dysfunction of the RB1 gene is the main cause of secondary tumours due to retinoblastoma. Therefore, the aim of this study was to evaluate the incidence of other secondary tumours in children with retinoblastoma. METHODS In this regard, we performed continuous and integrated bioinformatics analyses to find genes, protein products, and signal pathways involved in other cancers. RESULTS 1170 high-expression genes and 960 low-expression genes between non-invasive and invasive retinoblastoma were isolated. After examining the signal pathways, we observed bladder cancer and small cell lung cancer in the overexpressed genes. We also observed 5 cancers of endometriosis, prostate, non-small cell lung cancer, glioblastoma and renal cell carcinoma in low-expression genes. Based on the P-value index, non-small cell lung cancer, prostate and bladder cancers had the highest risk, and endometriosis cancer showed a lower probability of developing a secondary tumour in patients with retinoblastoma. In addition, the network between proteins also showed us that TP53, CDK2, SRC, MAPK1 proteins with high expression and JUN, HSP90AA1, and UBC proteins with low-expression play a significant role in candidate cancers. CONCLUSION Lastly, we used continuous bioinformatics analysis to show that seven cancers are strongly linked to retinoblastoma cancer. Of course, more research is needed to find the best way to care for children who have been treated for retinoblastoma.
Collapse
Affiliation(s)
- Reza Sadeghi
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hanieh Pirankuraim
- Medical Genomics Research Center, Tehran Medical Sciences Islamic Azad University, Tehran, Iran
| | | | - Maryam Arabi
- Medical Genomics Research Center, Tehran Medical Sciences Islamic Azad University, Tehran, Iran
- Cancer Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Bereimipour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Habibollah Mahmoodzadeh
- Cancer Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Breast Disease Research Center (BDRC), Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| | - Karim Nayernia
- International Center for Personalized Medicine (P7MEDICINE), 40235, Düsseldorf, Germany
| |
Collapse
|
8
|
Pungsrinont T, Kallenbach J, Baniahmad A. Role of PI3K-AKT-mTOR Pathway as a Pro-Survival Signaling and Resistance-Mediating Mechanism to Therapy of Prostate Cancer. Int J Mol Sci 2021; 22:11088. [PMID: 34681745 PMCID: PMC8538152 DOI: 10.3390/ijms222011088] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/27/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Androgen deprivation therapy (ADT) and androgen receptor (AR)-targeted therapy are the gold standard options for treating prostate cancer (PCa). These are initially effective, as localized and the early stage of metastatic disease are androgen- and castration-sensitive. The tumor strongly relies on systemic/circulating androgens for activating AR signaling to stimulate growth and progression. However, after a certain point, the tumor will eventually develop a resistant stage, where ADT and AR antagonists are no longer effective. Mechanistically, it seems that the tumor becomes more aggressive through adaptive responses, relies more on alternative activated pathways, and is less dependent on AR signaling. This includes hyperactivation of PI3K-AKT-mTOR pathway, which is a central signal that regulates cell pro-survival/anti-apoptotic pathways, thus, compensating the blockade of AR signaling. The PI3K-AKT-mTOR pathway is well-documented for its crosstalk between genomic and non-genomic AR signaling, as well as other signaling cascades. Such a reciprocal feedback loop makes it more complicated to target individual factor/signaling for treating PCa. Here, we highlight the role of PI3K-AKT-mTOR signaling as a resistance mechanism for PCa therapy and illustrate the transition of prostate tumor from AR signaling-dependent to PI3K-AKT-mTOR pathway-dependent. Moreover, therapeutic strategies with inhibitors targeting the PI3K-AKT-mTOR signal used in clinic and ongoing clinical trials are discussed.
Collapse
Affiliation(s)
| | | | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, 07747 Jena, Germany; (T.P.); (J.K.)
| |
Collapse
|
9
|
Rigotto G, Montini B, Mattiolo A, Lazzari N, Piano MA, Remondini D, Marmiroli S, Bertacchini J, Chieco-Bianchi L, Calabrò ML. Mechanisms Involved in the Promoting Activity of Fibroblasts in HTLV-1-Mediated Lymphomagenesis: Insights into the Plasticity of Lymphomatous Cells. Int J Mol Sci 2021; 22:10562. [PMID: 34638901 PMCID: PMC8508730 DOI: 10.3390/ijms221910562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022] Open
Abstract
Among the mechanisms leading to progression to Adult T-cell Leukaemia/Lymphoma in Human T-cell Leukaemia Virus type 1 (HTLV-1)-infected subjects, the contribution of stromal components remains poorly understood. To dissect the role of fibroblasts in HTLV-1-mediated lymphomagenesis, transcriptome studies, cytofluorimetric and qRT-PCR analyses of surface and intracellular markers linked to plasticity and stemness in coculture, and in vivo experiments were performed. A transcriptomic comparison between a more lymphomagenic (C91/III) and the parental (C91/PL) cell line evidenced hyperactivation of the PI3K/Akt pathway, confirmed by phospho-ELISA and 2-DE and WB analyses. C91/III cells also showed higher expression of mesenchymal and stemness genes. Short-term coculture with human foreskin fibroblasts (HFF) induced these features in C91/PL cells, and significantly increased not only the cancer stem cells (CSCs)-supporting CD10+GPR77+ HFF subpopulation, but also the percentage of ALDH1bright C91/PL cells. A non-cytotoxic acetylsalicylic acid treatment decreased HFF-induced ALDH1bright C91/PL cells, downregulated mesenchymal and stemness genes in cocultured cells, and delayed lymphoma growth in immunosuppressed mice, thus hindering the supportive activity of HFF on CSCs. These data suggest that crosstalk with HFF significantly intensifies the aggressiveness and plasticity of C91/PL cells, leading to the enrichment in lymphoma-initiating cells. Additional research is needed to better characterize these preliminary findings.
Collapse
Affiliation(s)
- Giulia Rigotto
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV—IRCCS, 35128 Padua, Italy; (G.R.); (B.M.); (A.M.); (N.L.); (M.A.P.)
| | - Barbara Montini
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV—IRCCS, 35128 Padua, Italy; (G.R.); (B.M.); (A.M.); (N.L.); (M.A.P.)
| | - Adriana Mattiolo
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV—IRCCS, 35128 Padua, Italy; (G.R.); (B.M.); (A.M.); (N.L.); (M.A.P.)
| | - Nayana Lazzari
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV—IRCCS, 35128 Padua, Italy; (G.R.); (B.M.); (A.M.); (N.L.); (M.A.P.)
| | - Maria Assunta Piano
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV—IRCCS, 35128 Padua, Italy; (G.R.); (B.M.); (A.M.); (N.L.); (M.A.P.)
| | - Daniel Remondini
- Department of Physics and Astronomy, University of Bologna, and Istituto Nazionale di Fisica Nucleare, INFN, 40127 Bologna, Italy;
| | - Sandra Marmiroli
- Department of Biomedical, Metabolic and Neuronal Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.M.); (J.B.)
| | - Jessika Bertacchini
- Department of Biomedical, Metabolic and Neuronal Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.M.); (J.B.)
| | - Luigi Chieco-Bianchi
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy;
| | - Maria Luisa Calabrò
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV—IRCCS, 35128 Padua, Italy; (G.R.); (B.M.); (A.M.); (N.L.); (M.A.P.)
| |
Collapse
|
10
|
Darici S, Zavatti M, Braglia L, Accordi B, Serafin V, Horne GA, Manzoli L, Palumbo C, Huang X, Jørgensen HG, Marmiroli S. Synergistic cytotoxicity of dual PI3K/mTOR and FLT3 inhibition in FLT3-ITD AML cells. Adv Biol Regul 2021; 82:100830. [PMID: 34555701 DOI: 10.1016/j.jbior.2021.100830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 01/17/2023]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematopoietic malignancy, characterized by a heterogeneous genetic landscape and complex clonal evolution, with poor outcomes. Mutation at the internal tandem duplication of FLT3 (FLT3-ITD) is one of the most common somatic alterations in AML, associated with high relapse rates and poor survival due to the constitutive activation of the FLT3 receptor tyrosine kinase and its downstream effectors, such as PI3K signaling. Thus, aberrantly activated FLT3-kinase is regarded as an attractive target for therapy for this AML subtype, and a number of small molecule inhibitors of this kinase have been identified, some of which are approved for clinical practice. Nevertheless, acquired resistance to these molecules is often observed, leading to severe clinical outcomes. Therapeutic strategies to tackle resistance include combining FLT3 inhibitors with other antileukemic agents. Here, we report on the preclinical activity of the combination of the FLT3 inhibitor quizartinib with the dual PI3K/mTOR inhibitor PF-04691502 in FLT3-ITD cells. Briefly, we show that the association of these two molecules displays synergistic cytotoxicity in vitro in FLT3-ITD AML cells, triggering 90% cell death at nanomolar concentrations after 48 h.
Collapse
Affiliation(s)
- Salihanur Darici
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy; Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Manuela Zavatti
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy
| | - Luca Braglia
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Benedetta Accordi
- Department of Woman and Child Health, Haemato-Oncology Laboratory, University of Padua, Via Giustiniani 3 and IRP Città Della Speranza, Corso Stati Uniti 4, 35128, Padua, Italy
| | - Valentina Serafin
- Department of Woman and Child Health, Haemato-Oncology Laboratory, University of Padua, Via Giustiniani 3 and IRP Città Della Speranza, Corso Stati Uniti 4, 35128, Padua, Italy
| | - Gillian A Horne
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Carla Palumbo
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy
| | - Xu Huang
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK.
| | - Heather G Jørgensen
- Haemato-Oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Sandra Marmiroli
- Cellular Signaling Unit, Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| |
Collapse
|
11
|
Guo L, Liu M, Dou Y, Duan R, Shen L, Jia L, Wang J, Li C, Li X, Liang T. Screening and identification of haptoglobin showing its important role in pathophysiological process of gallbladder carcinoma. Gene 2021; 776:145429. [PMID: 33444685 DOI: 10.1016/j.gene.2021.145429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/17/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
Abstract
Gallbladder cancer (GBC) with poor prognosis has been a major cause of cancer-related deaths worldwide. In this study, we aimed to screen and identify crucial genes in GBC through integrative analysis of multiple datasets and further experimental validation. A candidate crucial gene, up-regulated haptoglobin (HP), was firstly screened, and then further analysis and validation mainly focused on whether higher enrichment level of HP was responsible for pathophysiological process of GBC. HP was found with diverse expression patterns in various cancer types, and the dynamic expression patterns indicated its spatiotemporal characteristics in different tissues and disease stages, implicating its role in multiple biological processes. Further experimental validation showed that HP could promote the GBC-SD cell proliferation, migration and invasion, implying its role in pathophysiological process of GBC. HP may have a crucial role in occurrence and development of GBC, and it provides possibility as a potential biomarker or target in cancer prognosis and treatment.
Collapse
Affiliation(s)
- Li Guo
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Mengting Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, and Changzhou Institute of Innovation and Development, Nanjing Normal University, Nanjing 210023, China
| | - Yuyang Dou
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Rui Duan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, and Changzhou Institute of Innovation and Development, Nanjing Normal University, Nanjing 210023, China
| | - Lulu Shen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, and Changzhou Institute of Innovation and Development, Nanjing Normal University, Nanjing 210023, China
| | - Lin Jia
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, and Changzhou Institute of Innovation and Development, Nanjing Normal University, Nanjing 210023, China
| | - Jun Wang
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Changxian Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiangcheng Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, and Changzhou Institute of Innovation and Development, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
12
|
Dou J, Cánovas A, Brito LF, Yu Y, Schenkel FS, Wang Y. Comprehensive RNA-Seq Profiling Reveals Temporal and Tissue-Specific Changes in Gene Expression in Sprague-Dawley Rats as Response to Heat Stress Challenges. Front Genet 2021; 12:651979. [PMID: 33897767 PMCID: PMC8063118 DOI: 10.3389/fgene.2021.651979] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/02/2021] [Indexed: 12/13/2022] Open
Abstract
Understanding heat stress physiology and identifying reliable biomarkers are paramount for developing effective management and mitigation strategies. However, little is known about the molecular mechanisms underlying thermal tolerance in animals. In an experimental model of Sprague–Dawley rats subjected to temperatures of 22 ± 1°C (control group; CT) and 42°C for 30 min (H30), 60 min (H60), and 120 min (H120), RNA-sequencing (RNA-Seq) assays were performed for blood (CT and H120), liver (CT, H30, H60, and H120), and adrenal glands (CT, H30, H60, and H120). A total of 53, 1,310, and 1,501 differentially expressed genes (DEGs) were significantly identified in the blood (P < 0.05 and |fold change (FC)| >2), liver (P < 0.01, false discovery rate (FDR)–adjusted P = 0.05 and |FC| >2) and adrenal glands (P < 0.01, FDR-adjusted P = 0.05 and |FC| >2), respectively. Of these, four DEGs, namely Junb, P4ha1, Chordc1, and RT1-Bb, were shared among the three tissues in CT vs. H120 comparison. Functional enrichment analyses of the DEGs identified in the blood (CT vs. H120) revealed 12 biological processes (BPs) and 25 metabolic pathways significantly enriched (FDR = 0.05). In the liver, 133 BPs and three metabolic pathways were significantly detected by comparing CT vs. H30, H60, and H120. Furthermore, 237 BPs were significantly (FDR = 0.05) enriched in the adrenal glands, and no shared metabolic pathways were detected among the different heat-stressed groups of rats. Five and four expression patterns (P < 0.05) were uncovered by 73 and 91 shared DEGs in the liver and adrenal glands, respectively, over the different comparisons. Among these, 69 and 73 genes, respectively, were proposed as candidates for regulating heat stress response in rats. Finally, together with genome-wide association study (GWAS) results in cattle and phenome-wide association studies (PheWAS) analysis in humans, five genes (Slco1b2, Clu, Arntl, Fads1, and Npas2) were considered as being associated with heat stress response across mammal species. The datasets and findings of this study will contribute to a better understanding of heat stress response in mammals and to the development of effective approaches to mitigate heat stress response in livestock through breeding.
Collapse
Affiliation(s)
- Jinhuan Dou
- Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, Canada
| | - Angela Cánovas
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, Canada
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Ying Yu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Flavio S Schenkel
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, Canada
| | - Yachun Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
13
|
Abstract
In over two decades since the discovery of phosphatase and tensin homologue deleted on chromosome 10 (PTEN), nearly 18,000 publications have attempted to elucidate its functions and roles in normal physiology and disease. The frequent disruption of PTEN in cancer cells was a strong indication that it had critical roles in tumour suppression. Germline PTEN mutations have been identified in patients with heterogeneous tumour syndromic diseases, known as PTEN hamartoma tumour syndrome (PHTS), and in some individuals with autism spectrum disorders (ASD). Today we know that by limiting oncogenic signalling through the phosphoinositide 3-kinase (PI3K) pathway, PTEN governs a number of processes including survival, proliferation, energy metabolism, and cellular architecture. Some of the most exciting recent advances in the understanding of PTEN biology and signalling have revisited its unappreciated roles as a protein phosphatase, identified non-enzymatic scaffold functions, and unravelled its nuclear function. These discoveries are certain to provide a new perspective on its full tumour suppressor potential, and knowledge from this work will lead to new anti-cancer strategies that exploit PTEN biology. In this review, we will highlight some outstanding questions and some of the very latest advances in the understanding of the tumour suppressor PTEN.
Collapse
Affiliation(s)
- Jonathan Tak-Sum Chow
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Leonardo Salmena
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Cochet MF, Baron F, Bonnassie S, Jan S, Leconte N, Jardin J, Briard-Bion V, Gautier M, Andrews SC, Guérin-Dubiard C, Nau F. Identification of New Antimicrobial Peptides that Contribute to the Bactericidal Activity of Egg White against Salmonella enterica Serovar Enteritidis at 45 °C. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:2118-2128. [PMID: 33561347 DOI: 10.1021/acs.jafc.0c06677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
A recent work revealed that egg white (EW) at 45 °C exhibits powerful bactericidal activity against S. enterica serovar Enteritidis, which is surprisingly little affected by removal of the >10 kDa EW proteins. Here, we sought to identify the major EW factors responsible for this bactericidal activity by fractionating EW using ultrafiltration and nanofiltration and by characterizing the physicochemical and antimicrobial properties of the resulting fractions. In particular, 22 peptides were identified by nano-LC/MS-MS and the bactericidal activities of representative peptides (with predicted antimicrobial activity) were further assessed. Two peptides (FVPPVQR and GDPSAWSWGAEAHS) were found to be bactericidal against S. enterica serovar Enteritidis at 45 °C when provided in an EW environment. Nevertheless, these peptides contribute only part of this bactericidal activity, suggesting other, yet to be determined, antimicrobial factors.
Collapse
Affiliation(s)
| | | | - Sylvie Bonnassie
- UFR Sciences de la Vie et de l'Environnement, Rennes 35700, France
| | - Sophie Jan
- STLO, INRAE, Institut Agro, 35042 Rennes, France
| | | | | | | | | | - Simon C Andrews
- School of Biological Sciences, Knight Building, University of Reading, Reading RG6 6AS, U.K
| | | | | |
Collapse
|
15
|
Praharaj PP, Patra S, Panigrahi DP, Patra SK, Bhutia SK. Clusterin as modulator of carcinogenesis: A potential avenue for targeted cancer therapy. Biochim Biophys Acta Rev Cancer 2020; 1875:188500. [PMID: 33385484 DOI: 10.1016/j.bbcan.2020.188500] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/14/2020] [Accepted: 12/24/2020] [Indexed: 12/30/2022]
Abstract
Clusterin (CLU) is an evolutionary conserved molecular chaperone present in different human tissues and fluids and established to be a significant cancer regulator. It controls several cancer-associated cellular events, including cancer cell proliferation, stemness, survival, metastasis, epithelial-mesenchymal transition, therapy resistance, and inhibition of programmed cell death to support cancer growth and recurrence. This multifunctional role of CLU makes it an ideal target for cancer control. More importantly, genetic and antisense-mediated (OGX-011) inhibition of CLU enhances the anticancer potential of different FDA-approved chemotherapeutic drugs at the clinical level, improving patient's survival. In this review, we have discussed the detailed mechanism of CLU-mediated modulation of different cancer-associated signaling pathways. We have also provided updated information on the current preclinical and clinical findings that drive trials in various cancer types for potential targeted cancer therapy.
Collapse
Affiliation(s)
- Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Debasna Pritimanjari Panigrahi
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| |
Collapse
|
16
|
Peng B, Qiu X, Dong Z, Zhang J, Pei Y, Wang T. Proteomic profiling of biomarkers by MALDI-TOF mass spectrometry for the diagnosis of tracheobronchial stenosis after tracheobronchial tuberculosis. Exp Ther Med 2020; 21:63. [PMID: 33365063 PMCID: PMC7716632 DOI: 10.3892/etm.2020.9495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 07/14/2020] [Indexed: 12/27/2022] Open
Abstract
Tracheobronchial tuberculosis (TB) leads to airway stenosis, irreversible airway damage and even death. The present study aimed to identify biomarkers for the diagnosis of tracheobronchial stenosis (TBS) secondary to tracheobronchial TB. A cohort was recruited, including patients with TBS after tracheobronchial TB, TBS after tracheal intubation or tracheotomy (TIT) and no stenosis of early-stage lung cancer,. Proteomic profiling was performed to gain insight into the mechanisms of the pathological processes. Differentially expressed proteins in the serum and bronchial alveolar lavage fluid (BALF) from patients were detected by matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS). Subsequently, ELISA was performed to validate the changes of protein levels in an additional cohort. MALDI-TOF MS revealed that 8 peptides in the serum, including myeloid-associated differentiation marker, keratin type I cytoskeletal 18, fibrinogen α-chain, angiotensinogen (AGT), apolipoprotein A-I (APOAI), clusterin and two uncharacterized peptides, and nine peptides in BALF, including argininosuccinate lyase, APOAI, AGT and five uncharacterized peptides, were differentially expressed (molecular-weight range, 1,000-10,000 Da) in the TB group compared with the TIT group. The ELISA results indicated that the changes in the protein levels had a similar trend as those identified by proteomic profiling. In conclusion, the present study identified proteins that may serve as potential biomarkers and provide novel insight into the molecular mechanisms underlying TBS after tracheobronchial TB.
Collapse
Affiliation(s)
- Bihao Peng
- The Second Clinical Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Xiaojian Qiu
- Department of Pulmonary Diseases, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Zhiwu Dong
- Department of Laboratory Medicine, Shanghai Sixth People's Hospital Jinshan Branch, Shanghai 201599, P.R. China
| | - Jie Zhang
- Department of Pulmonary Diseases, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Yinghua Pei
- Department of Pulmonary Diseases, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Ting Wang
- Department of Pulmonary Diseases, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
17
|
Naik PP, Mukhopadhyay S, Praharaj PP, Bhol CS, Panigrahi DP, Mahapatra KK, Patra S, Saha S, Panda AK, Panda K, Paul S, Aich P, Patra SK, Bhutia SK. Secretory clusterin promotes oral cancer cell survival via inhibiting apoptosis by activation of autophagy in AMPK/mTOR/ULK1 dependent pathway. Life Sci 2020; 264:118722. [PMID: 33160989 DOI: 10.1016/j.lfs.2020.118722] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/26/2020] [Accepted: 10/31/2020] [Indexed: 02/06/2023]
Abstract
AIMS Secretory clusterin (sCLU) plays an important role in tumor development and cancer progression. However, the molecular mechanisms and physiological functions of sCLU in oral cancer is unclear. We examined the impact of sCLU-mediated autophagy in cell survival and apoptosis inhibition in oral cancer. MAIN METHODS Immunohistochemical analysis was performed to analyze protein expression in patient samples. Autophagy and mitophagy was studied by immunofluorescence microscopy and Western blot. The gain and loss of function was studied by overexpression of plasmid and siRNA approaches respectively. Cellular protection against nutrient starvation and therapeutic stress by sCLU was studied by cell viability, caspase assay and meta-analysis. KEY FINDINGS The data from oral cancer patients showed that the expression levels of sCLU, ATG14, ULK1, and PARKIN increased in grade-wise manners. Interestingly, sCLU overexpression promoted autophagy through AMPK/Akt/mTOR signaling pathway leading to cell survival and protection from long exposure serum starvation induced-apoptosis. Additionally, sCLU was demonstrated to interact with ULK1 and inhibition of ULK1 activity by SBI206965 was found to abolish sCLU-induced autophagy indicating critical role of ULK1 in induction of autophagy. Furthermore, sCLU was observed to promote expression of mitophagy-associated proteins in serum starvation conditions to protect cells from nutrient deprivation. The meta-analysis elucidated that high CLU expression is associated with therapy resistance in cancer and we demonstrated that sCLU-mediated mitophagy was revealed to inhibit cell death by cisplatin. SIGNIFICANCE The present investigation has highlighted the probable implications of the clusterin-induced autophagy in cell survival and inhibition of apoptosis in oral cancer.
Collapse
Affiliation(s)
- Prajna Paramita Naik
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Subhadip Mukhopadhyay
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Chandra Sekhar Bhol
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Debasna Pritimanjari Panigrahi
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Kewal Kumar Mahapatra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Sarbari Saha
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | | | - Krupasindhu Panda
- Panda Curie Cancer Hospital, Telenga Pentha, Cuttack, 753051, Odisha, India
| | - Subhankar Paul
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Palok Aich
- National Institute of Science Education and Research (NISER), HBNI, Bhipmpur-Padanpur, Jatni, Khurda 752050, Odisha, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| |
Collapse
|
18
|
Deregulated PTEN/PI3K/AKT/mTOR signaling in prostate cancer: Still a potential druggable target? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118731. [PMID: 32360668 DOI: 10.1016/j.bbamcr.2020.118731] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 01/13/2023]
Abstract
Although the prognosis of patients with localized prostate cancer is good after surgery, with a favorable response to androgen deprivation therapy, about one third of them invariably relapse, and progress to castration-resistant prostate cancer. Overall, prostate cancer therapies remain scarcely effective, thus it is mandatory to devise alternative treatments enhancing the efficacy of surgical castration and hormone administration. Dysregulation of the phosphoinositide 3-kinase pathway has attracted growing attention in prostate cancer due to the highly frequent association of epigenetic and post-translational modifications as well as to genetic alterations of both phosphoinositide 3-kinase and PTEN to onset and/or progression of this malignancy, and to resistance to canonical androgen-deprivation therapy. Here we provide a summary of the biological functions of the major players of this cascade and their deregulation in prostate cancer, summarizing the results of preclinical and clinical studies with PI3K signaling inhibitors and the reasons of failure independent from genomic changes.
Collapse
|
19
|
Xie M, Zhang Z, Cui Y. Long Noncoding RNA SNHG1 Contributes to the Promotion of Prostate Cancer Cells Through Regulating miR-377-3p/AKT2 Axis. Cancer Biother Radiopharm 2020; 35:109-119. [PMID: 32077748 DOI: 10.1089/cbr.2019.3177] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background: Long noncoding RNAs could serve as a candidate target for prostate cancer (PCa) diagnosis and treatment. The current study aimed to investigate the role and functions of SNHG1 in PCa cells. Materials and Methods: Abnormal expression of SNHG1, survival analysis, and target gene were determined or predicted by bioinformatics techniques. Gene expressions at transcriptional and translational levels were determined by Quantitative Real-time PCR and Western blotting, respectively. Cell viability, growth, and apoptosis rate were detected by Cell Counting Kit-8, colony formation assay and flow cytometry. Results: The results showed that SNHG1 was highly expressed in PCa tissues, which was accompanied by decreased miR-377-3p expression and poor overall survival rate, and that miR-377-3p was predicted as the target of SNHG1 in PCa cells. Moreover, SNHG1 counteracted the effects of miR-377-3p on inhibiting cell growth and promoting apoptosis of PCa cells. Furthermore, miR-377-3p counteracted the effects of AKT2 on promoting cell viability, growth, and suppressing apoptosis of PCa cells. In addition, AKT2 expression was proved to be regulated by miR-377-3p. Conclusions: The SNHG1/miR-377-3p/AKT2 regulatory axis in PCa cells was disclosed. The upregulated AKT2 might be a result of dysregulated interaction balance between the expressions of miR-377-3p and SNHG1. Based on such discoveries, the intervention of SNHG1/miR-377-3p/AKT2 axis could be further explored in the treatment of PCa.
Collapse
Affiliation(s)
- Mao Xie
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao, Yantai, China
| | - Zhiyu Zhang
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao, Yantai, China
| | - Yupeng Cui
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao, Yantai, China
| |
Collapse
|
20
|
Abstract
miRNAs, a major class of small noncoding RNAs approximately 18-25 nucleotides in length, function by repressing the expression of target genes through binding to complementary sequences in the 3'-UTRs of target genes. Emerging evidence has highlighted their important roles in numerous diseases, including human cancers. Recently, miR-190 has been shown to be dysregulated in various types of human cancers that participates in cancer-related biological processes, including proliferation, apoptosis, metastasis, drug resistance, by regulating associated target genes, and to predict cancer diagnosis and prognosis. In this review, we summarized the roles of miR-190-5p in human diseases, especially in human cancers. Then we classified its target genes in tumorigenesis and progression, which might provide evidence for cancer diagnosis and prognosis, promising tools for cancer treatment, or leads for further investigation.
Collapse
Affiliation(s)
- Yue Yu
- 1The First Department of Breast Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huan-Hu-Xi Road, Hexi District, Tianjin, 300060 China.,2Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060 China.,4Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060 China
| | - Xu-Chen Cao
- 1The First Department of Breast Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huan-Hu-Xi Road, Hexi District, Tianjin, 300060 China.,2Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060 China.,4Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060 China
| |
Collapse
|