1
|
Huang RH, Zeng QM, Jiang B, Xu G, Xiao GC, Xia W, Liao YF, Wu YT, Zou JR, Qian B, Xiao RH, Yuan YH, Zhang GX, Zou XF. Overexpression of DUSP26 gene suppressed the proliferation, migration, and invasion of human prostate cancer cells. Exp Cell Res 2024; 442:114231. [PMID: 39222869 DOI: 10.1016/j.yexcr.2024.114231] [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: 02/06/2024] [Revised: 07/30/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Prostate cancer (PCa) is threatening the health of millions of people, the pathological mechanism of prostate cancer has not been fully elaborated, and needs to be further explored. Here, we found that the expression of DUSP26 is dramatically suppressed, and a positive connection of its expression with PCa prognosis was also observed. In vitro, overexpression of DUSP26 significantly inhibited the proliferative, migrative, and invasive capacities of PC3 cells, DUSP26 silencing presented opposite results. Tumor formation experiments in subcutaneous nude mice demonstrated that DUSP26 overexpression could significantly suppress PC3 growth in vivo. Moreover, the mechanism of DUSP26 gene and PCa was discovered by RNA-Seq analysis. We found that DUSP26 significantly inhibited MAPK signaling pathway activation, and further experiments displayed that DUSP26 could impair TAK1, p38, and JNK phosphorylation. Interestingly, treatment with the TAK1 inhibitor (iTAK1) attenuated the effect of DUSP26 on PC3 cells. Together, these results suggested that DUSP26 may serve as a novel therapeutic target for PC3 cell type PCa, the underlying mechanism may be through TAK1-JNK/p38 signaling.
Collapse
Affiliation(s)
- Ruo-Hui Huang
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Medical College of Soochow University, Suzhou, Jiangsu, 215006, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China.
| | - Qing-Ming Zeng
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Bo Jiang
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Gang Xu
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Guan-Cheng Xiao
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Wei Xia
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Yun-Feng Liao
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Yu-Ting Wu
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Jun-Rong Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Biao Qian
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Ri-Hai Xiao
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Yuan-Hu Yuan
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Guo-Xi Zhang
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China
| | - Xiao-Feng Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Gan Zhou, Jiang Xi, 341000, China; Jiangxi Stone Prevention Engineering Technology Research Center, Gan Zhou, Jiang Xi, 341000, China.
| |
Collapse
|
2
|
Xiang H, Li Z, Li Y, Zheng J, Dou M, Xue W, Wu X. Dual-specificity phosphatase 26 protects against kidney injury caused by ischaemia-reperfusion through restraint of TAK1-JNK/p38-mediated apoptosis and inflammation of renal tubular epithelial cells. Toxicol Appl Pharmacol 2024; 487:116954. [PMID: 38705402 DOI: 10.1016/j.taap.2024.116954] [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: 01/03/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Dual-specificity phosphatase 26 (DUSP26) acts as a pivotal player in the transduction of signalling cascades with its dephosphorylating activity. Currently, DUSP26 attracts extensive attention due to its particular function in several pathological conditions. However, whether DUSP26 plays a role in kidney ischaemia-reperfusion (IR) injury is unknown. Aims of the current work were to explore the relevance of DUSP26 in kidney IR damage. DUSP26 levels were found to be decreased in renal tubular epithelial cells following hypoxia-reoxygenation (HR) and kidney samples subjected to IR treatments. DUSP26-overexpressed renal tubular epithelial cells exhibited protection against HR-caused apoptosis and inflammation, while DUSP26-depleted renal tubular epithelial cells were more sensitive to HR damage. Upregulation of DUSP26 in rat kidneys by infecting adenovirus expressing DUSP26 markedly ameliorated kidney injury caused by IR, while also effectively reducing apoptosis and inflammation. The mechanistic studies showed that the activation of transforming growth factor-β-activated kinase 1 (TAK1)-JNK/p38 MAPK, contributing to kidney injury under HR or IR conditions, was restrained by increasing DUSP26 expression. Pharmacological restraint of TAK1 markedly diminished DUSP26-depletion-exacebated effects on JNK/p38 activation and HR injury of renal tubular cells. The work reported a renal-protective function of DUSP26, which protects against IR-related kidney damage via the intervention effects on the TAK1-JNK/p38 axis. The findings laid a foundation for understanding the molecular pathogenesis of kidney IR injury and provide a prospective target for treating this condition.
Collapse
Affiliation(s)
- Heli Xiang
- Department of Kidney Transplant, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 71006, China
| | - Zepeng Li
- Department of Kidney Transplant, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 71006, China
| | - Yang Li
- Department of Kidney Transplant, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 71006, China
| | - Jin Zheng
- Department of Kidney Transplant, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 71006, China
| | - Meng Dou
- Department of Kidney Transplant, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 71006, China
| | - Wujun Xue
- Department of Kidney Transplant, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 71006, China
| | - Xiaoyan Wu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
3
|
Deng Y, Deng X, Zhao J, Ning S, Gu A, Chen Q, Qu Y. Revealing the Complete Bispecific Phosphatase Genes (DUSPs) across the Genome and Investigating the Expression Patterns of GH_A11G3500 Resistance against Verticillium wilt. Int J Mol Sci 2024; 25:4500. [PMID: 38674085 PMCID: PMC11050305 DOI: 10.3390/ijms25084500] [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: 02/26/2024] [Revised: 04/07/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
DUSPs, a diverse group of protein phosphatases, play a pivotal role in orchestrating cellular growth and development through intricate signaling pathways. Notably, they actively participate in the MAPK pathway, which governs crucial aspects of plant physiology, including growth regulation, disease resistance, pest resistance, and stress response. DUSP is a key enzyme, and it is the enzyme that limits the rate of cell metabolism. At present, complete understanding of the DUSP gene family in cotton and its specific roles in resistance to Verticillium wilt (VW) remains elusive. To address this knowledge gap, we conducted a comprehensive identification and analysis of four key cotton species: Gossypium arboreum, Gossypium barbadense, Gossypium hirsutum, and Gossypium raimondii. The results revealed the identification of a total of 120 DUSP genes in the four cotton varieties, which were categorized into six subgroups and randomly distributed at both ends of 26 chromosomes, predominantly localized within the nucleus. Our analysis demonstrated that closely related DUSP genes exhibited similarities in terms of the conserved motif composition and gene structure. A promoter analysis performed on the GhDUSP gene promoter revealed the presence of several cis-acting elements, which are associated with abiotic and biotic stress responses, as well as hormone signaling. A tissue expression pattern analysis demonstrated significant variations in GhDUSP gene expression under different stress conditions, with roots exhibiting the highest levels, followed by stems and leaves. In terms of tissue-specific detection, petals, leaves, stems, stamens, and receptacles exhibited higher expression levels of the GhDUSP gene. The gene expression analysis results for GhDUSPs under stress suggest that DUSP genes may have a crucial role in the cotton response to stress in cotton. Through Virus-Induced Gene Silencing (VIGS) experiments, the silencing of the target gene significantly reduced the resistance efficiency of disease-resistant varieties against Verticillium wilt (VW). Consequently, we conclude that GH_A11G3500-mediated bispecific phosphorylated genes may serve as key regulators in the resistance of G. hirsutum to Verticillium wilt (VW). This study presents a comprehensive structure designed to provide an in-depth understanding of the potential biological functions of cotton, providing a strong foundation for further research into molecular breeding and resistance to plant pathogens.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yanying Qu
- College of Agronomy, Xinjiang Agricultural University, 311 Nongda East Road, Urumqi 830052, China; (Y.D.); (X.D.); (J.Z.); (S.N.); (A.G.); (Q.C.)
| |
Collapse
|
4
|
Liu H, Li X, Shi Y, Ye Z, Cheng X. Protein Tyrosine Phosphatase PRL-3: A Key Player in Cancer Signaling. Biomolecules 2024; 14:342. [PMID: 38540761 PMCID: PMC10967961 DOI: 10.3390/biom14030342] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 07/02/2024] Open
Abstract
Protein phosphatases are primarily responsible for dephosphorylation modification within signal transduction pathways. Phosphatase of regenerating liver-3 (PRL-3) is a dual-specific phosphatase implicated in cancer pathogenesis. Understanding PRL-3's intricate functions and developing targeted therapies is crucial for advancing cancer treatment. This review highlights its regulatory mechanisms, expression patterns, and multifaceted roles in cancer progression. PRL-3's involvement in proliferation, migration, invasion, metastasis, angiogenesis, and drug resistance is discussed. Regulatory mechanisms encompass transcriptional control, alternative splicing, and post-translational modifications. PRL-3 exhibits selective expressions in specific cancer types, making it a potential target for therapy. Despite advances in small molecule inhibitors, further research is needed for clinical application. PRL-3-zumab, a humanized antibody, shows promise in preclinical studies and clinical trials. Our review summarizes the current understanding of the cancer-related cellular function of PRL-3, its prognostic value, and the research progress of therapeutic inhibitors.
Collapse
Affiliation(s)
- Haidong Liu
- Zhejiang Cancer Hospital, Hangzhou 310022, China;
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China
| | - Xiao Li
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Yin Shi
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, China;
| | - Zu Ye
- Zhejiang Cancer Hospital, Hangzhou 310022, China;
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Xiangdong Cheng
- Zhejiang Cancer Hospital, Hangzhou 310022, China;
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| |
Collapse
|
5
|
Patysheva MR, Prostakishina EA, Budnitskaya AA, Bragina OD, Kzhyshkowska JG. Dual-Specificity Phosphatases in Regulation of Tumor-Associated Macrophage Activity. Int J Mol Sci 2023; 24:17542. [PMID: 38139370 PMCID: PMC10743672 DOI: 10.3390/ijms242417542] [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: 11/22/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
The regulation of protein kinases by dephosphorylation is a key mechanism that defines the activity of immune cells. A balanced process of the phosphorylation/dephosphorylation of key protein kinases by dual-specificity phosphatases is required for the realization of the antitumor immune response. The family of dual-specificity phosphatases is represented by several isoforms found in both resting and activated macrophages. The main substrate of dual-specificity phosphatases are three components of mitogen-activated kinase signaling cascades: the extracellular signal-regulated kinase ERK1/2, p38, and Janus kinase family. The results of the study of model tumor-associated macrophages supported the assumption of the crucial role of dual-specificity phosphatases in the formation and determination of the outcome of the immune response against tumor cells through the selective suppression of mitogen-activated kinase signaling cascades. Since mitogen-activated kinases mostly activate the production of pro-inflammatory mediators and the antitumor function of macrophages, the excess activity of dual-specificity phosphatases suppresses the ability of tumor-associated macrophages to activate the antitumor immune response. Nowadays, the fundamental research in tumor immunology is focused on the search for novel molecular targets to activate the antitumor immune response. However, to date, dual-specificity phosphatases received limited discussion as key targets of the immune system to activate the antitumor immune response. This review discusses the importance of dual-specificity phosphatases as key regulators of the tumor-associated macrophage function.
Collapse
Affiliation(s)
- Marina R. Patysheva
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia; (M.R.P.); (E.A.P.); (A.A.B.)
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Elizaveta A. Prostakishina
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia; (M.R.P.); (E.A.P.); (A.A.B.)
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Arina A. Budnitskaya
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia; (M.R.P.); (E.A.P.); (A.A.B.)
- Laboratory of Genetic Technologies, Siberian State Medical University, 634050 Tomsk, Russia
| | - Olga D. Bragina
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Julia G. Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia; (M.R.P.); (E.A.P.); (A.A.B.)
- Laboratory of Genetic Technologies, Siberian State Medical University, 634050 Tomsk, Russia
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Mannheim Institute of Innate Immunosciences (MI3), University of Heidelberg, 68167 Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, 69117 Mannheim, Germany
| |
Collapse
|
6
|
Zhao H, Lv J, Meng L, Lv J, Li Z. Dual-specificity phosphatase 26-dificient neurons are susceptible to oxygen-glucose deprivation/reoxygenation-evoked apoptosis and proinflammatory response by affecting the TAK1-medaited JNK/P38 MAPK pathway. Int Immunopharmacol 2023; 117:109980. [PMID: 37012870 DOI: 10.1016/j.intimp.2023.109980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/08/2023] [Accepted: 02/28/2023] [Indexed: 03/17/2023]
Abstract
Dual-specificity phosphatase 26 (DUSP26) is linked to a broad range of human disorders as it affects numerous signaling cascades. However, the involvement of DUSP26 in ischemic stroke has not been explored. Here, we investigated DUSP26 as a key mediator of oxygen-glucose deprivation/reoxygenation (OGD/R)-associated neuronal injury, an in vitro model for investigating ischemic stroke. A decline in DUSP26 occurred in neurons suffering from OGD/R. A deficiency in DUSP26 rendered neurons more susceptible to OGD/R by aggravating neuronal apoptosis and inflammation, while the overexpression of DUSP26 blocked OGD/R-evoked neuronal apoptosis and inflammation. Mechanistically, enhanced phosphorylation of transforming growth factor-β-activated kinase 1 (TAK1), c-Jun N-terminal kinase (JNK) and P38 mitogen-activated protein kinase (MAPK) was evidenced in DUSP26-deficient neurons suffering from OGD/R, whereas the opposite effects were observed in DUSP26-overexpressed neurons. Moreover, the inhibition of TAK1 abolished the DUSP26-deficiency-elicited activation of JNK and P38 MAPK and exhibited anti-OGD/R injury effects in DUSP26-deficiency neurons. Results from these experiments show that DUSP26 is essential for neurons in defending against OGD/R insult, while neuroprotection is achieved by restraining the TAK1-mediated JNK/P38 MAPK pathway. Therefore, DUSP26 may serve as a therapeutic target for the management of ischemic stroke.
Collapse
|
7
|
Using Single-Cell RNA Sequencing and MicroRNA Targeting Data to Improve Colorectal Cancer Survival Prediction. Cells 2023; 12:cells12020228. [PMID: 36672162 PMCID: PMC9856396 DOI: 10.3390/cells12020228] [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: 12/01/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023] Open
Abstract
Colorectal cancer has proven to be difficult to treat as it is the second leading cause of cancer death for both men and women worldwide. Recent work has shown the importance of microRNA (miRNA) in the progression and metastasis of colorectal cancer. Here, we develop a metric based on miRNA-gene target interactions, previously validated to be associated with colorectal cancer. We use this metric with a regularized Cox model to produce a small set of top-performing genes related to colon cancer. We show that using the miRNA metric and a Cox model led to a meaningful improvement in colon cancer survival prediction and correct patient risk stratification. We show that our approach outperforms existing methods and that the top genes identified by our process are implicated in NOTCH3 signaling and general metabolism pathways, which are essential to colon cancer progression.
Collapse
|
8
|
Toomer G, Workman A, Harrison KS, Stayton E, Hoyt PR, Jones C. Stress Triggers Expression of Bovine Herpesvirus 1 Infected Cell Protein 4 (bICP4) RNA during Early Stages of Reactivation from Latency in Pharyngeal Tonsil. J Virol 2022; 96:e0101022. [PMID: 36416585 PMCID: PMC9749472 DOI: 10.1128/jvi.01010-22] [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: 06/29/2022] [Accepted: 10/31/2022] [Indexed: 11/24/2022] Open
Abstract
Bovine herpesvirus 1 (BoHV-1), an important pathogen of cattle, establishes lifelong latency in sensory neurons within trigeminal ganglia (TG) after acute infection. The BoHV-1 latency-reactivation cycle, like other alphaherpesvirinae subfamily members, is essential for viral persistence and transmission. Notably, cells within pharyngeal tonsil (PT) also support a quiescent or latent BoHV-1 infection. The synthetic corticosteroid dexamethasone, which mimics the effects of stress, consistently induces BoHV-1 reactivation from latency allowing early stages of viral reactivation to be examined in the natural host. Based on previous studies, we hypothesized that stress-induced cellular factors trigger expression of key viral transcriptional regulatory genes. To explore this hypothesis, RNA-sequencing studies compared viral gene expression in PT during early stages of dexamethasone-induced reactivation from latency. Strikingly, RNA encoding infected cell protein 4 (bICP4), which is translated into an essential viral transcriptional regulatory protein, was detected 30 min after dexamethasone treatment. Ninety minutes after dexamethasone treatment bICP4 and, to a lesser extent, bICP0 RNA were detected in PT. All lytic cycle viral transcripts were detected within 3 h after dexamethasone treatment. Surprisingly, the latency related (LR) gene, the only viral gene abundantly expressed in latently infected TG neurons, was not detected in PT during latency. In TG neurons, bICP0 and the viral tegument protein VP16 are expressed before bICP4 during reactivation, suggesting distinct viral regulatory genes mediate reactivation from latency in PT versus TG neurons. Finally, these studies confirm PT is a biologically relevant site for BoHV-1 latency, reactivation from latency, and virus transmission. IMPORTANCE BoHV-1, a neurotropic herpesvirus, establishes, maintains, and reactivates from latency in neurons. BoHV-1 DNA is also detected in pharyngeal tonsil (PT) from latently infected calves. RNA-sequencing studies revealed the viral infected cell protein 4 (bICP4) RNA was expressed in PT of latently infected calves within 30 min after dexamethasone was used to initiate reactivation. As expected, bICP4 RNA was not detected during latency. All lytic cycle viral genes were expressed within 3 h after dexamethasone treatment. Conversely, bICP0 and the viral tegument protein VP16 are expressed prior to bICP4 in trigeminal ganglionic neurons during reactivation. The viral latency related gene, which is abundantly expressed in latently infected neurons, was not abundantly expressed in PT during latency. These studies provide new evidence PT is a biologically relevant site for BoHV-1 latency and reactivation. Finally, we predict other alphaherpesvirinae subfamily members utilize PT as a site for latency and reactivation.
Collapse
Affiliation(s)
- Gabriela Toomer
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, Oklahoma, USA
| | - Aspen Workman
- United States Department of Agriculture, Agricultural Research Service, U.S. Meat Animal Research Center, Clay Center, Nebraska, USA
| | - Kelly S. Harrison
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, Oklahoma, USA
| | - Erin Stayton
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, Oklahoma, USA
| | - Peter R. Hoyt
- Oklahoma State University, Department of Biochemistry and Molecular Biology, Stillwater, Oklahoma, USA
| | - Clinton Jones
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, Oklahoma, USA
| |
Collapse
|
9
|
Tubular IKKβ Deletion Alleviates Acute Ischemic Kidney Injury and Facilitates Tissue Regeneration. Int J Mol Sci 2022; 23:ijms231710199. [PMID: 36077596 PMCID: PMC9456401 DOI: 10.3390/ijms231710199] [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: 07/06/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/30/2022] Open
Abstract
Acute kidney injury (AKI) is a common renal injury leading to relevant morbidity and mortality worldwide. Most of the clinical cases of AKI are caused by ischemia reperfusion (I/R) injury with renal ischemia injury followed by reperfusion injury and activation of the innate immune response converging to NF-ĸB pathway induction. Despite the clear role of NF-ĸB in inflammation, it has recently been acknowledged that NF-ĸB may impact other cell functions. To identify NF-ĸB function with respect to metabolism, vascular function and oxidative stress after I/R injury and to decipher in detail the underlying mechanism, we generated a transgenic mouse model with targeted deletion of IKKβ along the tubule and applied I/R injury followed by its analysis after 2 and 14 days after I/R injury. Tubular IKKβ deletion ameliorated renal function and reduced tissue damage. RNAseq data together with immunohistochemical, biochemical and morphometric analysis demonstrated an ameliorated vascular organization and mRNA expression profile for increased angiogenesis in mice with tubular IKKβ deletion at 2 days after I/R injury. RNAseq and protein analysis indicate an ameliorated metabolism, oxidative species handling and timely-adapted cell proliferation and apoptosis as well as reduced fibrosis in mice with tubular IKKβ deletion at 14 days after I/R injury. In conclusion, mice with tubular IKKβ deletion upon I/R injury display improved renal function and reduced tissue damage and fibrosis in association with improved vascularization, metabolism, reactive species disposal and fine-tuned cell proliferation.
Collapse
|
10
|
Guler S, Zik B, Yalcin A. Upregulation of dual-specificity phosphatase-26 is required for transforming growth factor β1(TGFβ1)-induced Epithelial-mesenchymal transition in A549 and PANC1 cells. Mol Biol Rep 2022; 49:10195-10204. [PMID: 36053282 DOI: 10.1007/s11033-022-07893-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/19/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND Transforming Growth Factor β (TGFβ) proteins are potent inducers of the epithelial-mesenchymal transition (EMT) in tumor cells. Although mitogen-activated protein kinase (MAPK) family has been shown to be involved in TGFβ-induced EMT, role of Dual Specificity Phosphatases (DUSP), key regulators of MAPK activity, in TGFβ-induced EMT is largely unkonwn. METHODS AND RESULTS Real-time qPCR analyses were performed to determine the effect of TGFβ1 on expression of EMT genes and DUSP proteins in the non-small cell lung cancer model A549 and pancreatic adenocarcinoma model PANC1 cells. Western blot analyses were conducted to study the changes in protein levels of EMT proteins and select DUSP proteins, as well as phosphorylations of MAPK proteins upon TGFβ1 stimulation. Small interfering RNA (siRNA) was utilized to reduce expressions of DUSP genes. We observed that the EMT phenotype coincided with increases in phosphorylations of the MAPK proteins ERK1/2, p38MAPK, and JNK upon TGFβ1 stimulation. Real-time qPCR analysis showed increases in DUSP15 and DUSP26 mRNA levels and Western blot analysis confirmed the increase in DUSP26 protein levels in both A549 and PANC1 cells treated with TGFβ1 relative to control. Silencing of DUSP26 expression by siRNA markedly suppressed the effect of TGFβ1 on E-cadherin and mesenchymal genes in the cells. CONCLUSIONS Data provided suggest that TGFβ1 modulates the expression of DUSP genes and that upregulation of DUSP26 may be required for TGFβ1-promoted EMT in A549 and PANC1 cells. Further studies should be carried out to elucidate the requirement of individual DUSPs in TGFβ1-associated EMT in tumor cells.
Collapse
Affiliation(s)
- Sabire Guler
- Department of Histology & Embryology, School of Veterinary Medicine, Bursa Uludag University, 16059, Bursa, Turkey
| | - Berrin Zik
- Department of Histology & Embryology, School of Veterinary Medicine, Bursa Uludag University, 16059, Bursa, Turkey
| | - Abdullah Yalcin
- Department of Biochemistry, School of Veterinary Medicine, Bursa Uludag University, 16059, Bursa, Turkey.
| |
Collapse
|
11
|
Cui Z, Li D, Zhao J, Chen K. Falnidamol and cisplatin combinational treatment inhibits non-small cell lung cancer (NSCLC) by targeting DUSP26-mediated signal pathways. Free Radic Biol Med 2022; 183:106-124. [PMID: 35278641 DOI: 10.1016/j.freeradbiomed.2022.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022]
Abstract
Non-small-cell lung cancer (NSCLC) is one of the most commonly diagnosed cancers worldwide with limited effective therapies. Cisplatin (DDP), as the first-line treatment, is always served as a mainstay of chemotherapeutic agents in combination with other drugs for NSCLC treatment. Nevertheless, DDP-based therapy is limited due to the frequent development of chemoresistance and adverse effects. Herein, it is necessary to find a more effective therapeutic approach with less toxicity. Falnidamol (FLD) is a pyrimido-pyrimidine compound and exerts anti-cancer activity. In the present study, we found that FLD could strongly promote the cytotoxicity of DDP and markedly reduce the IC50 values to restrain the proliferation of NSCLC cells. Furthermore, combination of FLD and DDP remarkably induced G2/M cell cycle arrest, DNA damage and mitochondrial apoptosis, which was largely through the induction of reactive oxygen species (ROS). Additionally, FLD/DDP in combination greatly triggered ferroptosis, along with free iron accumulation and enhanced lipid peroxidation. Epithelial to mesenchymal transition (EMT) and epidermal growth factor receptor (EGFR) phosphorylation were also considerably restrained in NSCLC cells co-treated with FLD/DDP. Mechanistically, the combinative treatment significantly reduced DUSP26 expression in NSCLC cells. More studies showed that DUSP26 was strongly up-regulated in human NSCLC samples compared with the paired normal tissues, and high DUSP26 predicted poor overall survival rate among patients. Importantly, we found that DUSP26 suppression intensively reduced the proliferation, EMT process and pEGFR expression in NSCLC cells, whereas facilitated ROS production, DNA damage and cell death; however, opposite phenotype was observed in NSCLC cells over-expressing DUSP26. More importantly, DUSP26 over-expression completely abolished the anti-cancer function of FLD/DDP in NSCLC cells. Animal studies finally confirmed that FLD/DDP in combination efficiently reduced tumor growth and lung metastasis in mice with ameliorated side effects. In conclusion, all these data illustrated that FLD and DDP combinational treatment effectively restrained NSCLC progression, and thus can be served as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Zihan Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Dapeng Li
- Department of Oncology, The First Hospital Affiliated to Soochow University, Suzhou, 215000, China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
| | - Kai Chen
- Department of Oncology, The First Hospital Affiliated to Soochow University, Suzhou, 215000, China.
| |
Collapse
|
12
|
Eroglu B, Jin X, Deane S, Öztürk B, Ross OA, Moskophidis D, Mivechi NF. Dusp26 phosphatase regulates mitochondrial respiration and oxidative stress and protects neuronal cell death. Cell Mol Life Sci 2022; 79:198. [PMID: 35313355 PMCID: PMC10601927 DOI: 10.1007/s00018-022-04162-z] [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: 10/19/2021] [Revised: 01/04/2022] [Accepted: 01/21/2022] [Indexed: 11/29/2022]
Abstract
The dual specificity protein phosphatases (Dusps) control dephosphorylation of mitogen-activated protein kinases (MAPKs) as well as other substrates. Here, we report that Dusp26, which is highly expressed in neuroblastoma cells and primary neurons is targeted to the mitochondrial outer membrane via its NH2-terminal mitochondrial targeting sequence. Loss of Dusp26 has a significant impact on mitochondrial function that is associated with increased levels of reactive oxygen species (ROS), reduction in ATP generation, reduction in mitochondria motility and release of mitochondrial HtrA2 protease into the cytoplasm. The mitochondrial dysregulation in dusp26-deficient neuroblastoma cells leads to the inhibition of cell proliferation and cell death. In vivo, Dusp26 is highly expressed in neurons in different brain regions, including cortex and midbrain (MB). Ablation of Dusp26 in mouse model leads to dopaminergic (DA) neuronal cell loss in the substantia nigra par compacta (SNpc), inflammatory response in MB and striatum, and phenotypes that are normally associated with Neurodegenerative diseases. Consistent with the data from our mouse model, Dusp26 expressing cells are significantly reduced in the SNpc of Parkinson's Disease patients. The underlying mechanism of DA neuronal death is that loss of Dusp26 in neurons increases mitochondrial ROS and concurrent activation of MAPK/p38 signaling pathway and inflammatory response. Our results suggest that regulation of mitochondrial-associated protein phosphorylation is essential for the maintenance of mitochondrial homeostasis and dysregulation of this process may contribute to the initiation and development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Binnur Eroglu
- Molecular Chaperone Biology, Georgia Cancer Center, Medical College of Georgia at Augusta University, 1120 15th St., CN3153, Augusta, GA, 30912, USA
| | - Xiongjie Jin
- Molecular Chaperone Biology, Georgia Cancer Center, Medical College of Georgia at Augusta University, 1120 15th St., CN3153, Augusta, GA, 30912, USA
| | - Sadiki Deane
- Molecular Chaperone Biology, Georgia Cancer Center, Medical College of Georgia at Augusta University, 1120 15th St., CN3153, Augusta, GA, 30912, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Bahadır Öztürk
- Molecular Chaperone Biology, Georgia Cancer Center, Medical College of Georgia at Augusta University, 1120 15th St., CN3153, Augusta, GA, 30912, USA
- Medical Biochemistry Department, Selcuk University Medical Faculty, Konya, Turkey
| | - Owen A Ross
- Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL, 32224, USA
| | - Demetrius Moskophidis
- Molecular Chaperone Biology, Georgia Cancer Center, Medical College of Georgia at Augusta University, 1120 15th St., CN3153, Augusta, GA, 30912, USA.
- Department of Medicine, Medical College of Georgia at Augusta University, 1120 15th St., CN3153, Augusta, GA, 30912, USA.
| | - Nahid F Mivechi
- Molecular Chaperone Biology, Georgia Cancer Center, Medical College of Georgia at Augusta University, 1120 15th St., CN3153, Augusta, GA, 30912, USA.
- Departments of Radiation Oncology, Medical College of Georgia at Augusta University, 1120 15th St., CN3153, Augusta, GA, 30912, USA.
- Charlie Norwood VAMC, One Freedom Way, Augusta, GA, 30904, USA.
| |
Collapse
|
13
|
Chou MY, Appan D, Chang KW, Chou CH, Lin CY, Gau SSF, Huang HS. Mouse hybrid genome mediates diverse brain phenotypes with the specificity of reciprocal crosses. FASEB J 2022; 36:e22232. [PMID: 35199866 DOI: 10.1096/fj.202101624r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/27/2022] [Accepted: 02/14/2022] [Indexed: 12/13/2022]
Abstract
Hybrid species have more genetic diversity than their parents. However, the impact of the hybrid genome of reciprocal crosses on brain function remains largely unknown. We performed behavioral, molecular, and neuronal analyses on C57BL/6J mice (B6), CAST/EiJ mice (CAST), and hybrid mice resulting from reciprocal crosses of the two strains, B6/CAST F1i and B6/CAST F1r, respectively. Hybrid mice displayed greater motor strength and coordination, food grinding, social dominance, and less sociability compared to their parental strains. Parental origin influenced body weight, locomotor speed, and heat nociception of hybrid mice. Parental origin, cell type, and the interaction of both affected expression patterns of hybrid genomes including imprinted genes. There was a correlation between affected genes and corresponding behavioral phenotypes. Hybrid genomes mediated neuronal activity in the locus coeruleus, a brain region implicated in arousal, adaptive behaviors, and sleep-wake cycle due to its norepinephrine projections throughout the central nervous system. The comprehensive brain phenotypes in these hybrid mice reveal important functional readouts associated with interactions of hybrid genomes and impacts of parental genomes.
Collapse
Affiliation(s)
- Ming-Yi Chou
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Dhivya Appan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kai-Wei Chang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Hsuan Chou
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Yi Lin
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Susan Shur-Fen Gau
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Hsien-Sung Huang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|