1
|
Harsanyi S, Kianickova K, Katrlik J, Danisovic L, Ziaran S. Current look at the most promising proteomic and glycomic biomarkers of bladder cancer. J Cancer Res Clin Oncol 2024; 150:96. [PMID: 38372785 PMCID: PMC10876723 DOI: 10.1007/s00432-024-05623-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/12/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Bladder cancer (BC) belongs to the most frequent cancer types. The diagnostic process is still long and costly, with a high percentage of false-positive or -negative results. Due to the cost and lack of effectiveness, older methods need to be supplemented or replaced by a newer more reliable method. In this regard, proteins and glycoproteins pose high potential. METHODS We performed an online search in PubMed/Medline, Scopus, and Web of Science databases to find relevant studies published in English up until May 2023. If applicable, we set the AUC threshold to 0.90 and sensitivity/specificity (SN/SP) to 90%. FINDINGS Protein and glycoprotein biomarkers are a demonstrably viable option in BC diagnostics. Cholinesterase shows promise in progression-free survival. BLCA-4, ORM-1 along with HTRA1 in the detection of BC. Matrix metallopeptidase 9 exhibits potential for stratification of muscle-invasive subtypes with high negative predictive value for aggressive phenotypes. Distinguishing non-muscle invasive subtypes benefits from Keratin 17. Neu5Gc-modified UMOD glycoproteins pose potential in BC diagnosis, while fibronectin, laminin-5, collagen type IV, and lamprey immunity protein in early detection of BC.
Collapse
Affiliation(s)
- Stefan Harsanyi
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| | | | - Jaroslav Katrlik
- Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Stanislav Ziaran
- Department of Urology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
2
|
Guo F, Tao X, Wu Y, Dong D, Zhu Y, Shang D, Xiang H. Carfilzomib relieves pancreatitis-initiated pancreatic ductal adenocarcinoma by inhibiting high-temperature requirement protein A1. Cell Death Discov 2024; 10:58. [PMID: 38287020 PMCID: PMC10825157 DOI: 10.1038/s41420-024-01806-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 01/31/2024] Open
Abstract
Pancreatitis is a crucial risk factor for pancreatic ductal adenocarcinoma (PDAC), and our previous study had proved high-temperature requirement protein A1 (HTRA1) exacerbates pancreatitis insult; however, the function and mechanism of HTRA1 in pancreatitis-initiated PDAC is still unclear. In the present paper, we clarified the expression of HTRA1 in PDAC using bioinformatics and immunohistochemistry of tissue chip, and found that HTRA1 is significantly upregulated in PDAC. Moreover, the proliferation, migration, invasion and adhesion of PANC-1 and SW1990 cells were promoted by overexpression of HTRA1, but inhibited by knockdown of HTRA1. Meanwhile, we found that HTRA1 arrested PANC-1 and SW1990 cells at G2/M phase. Mechanistically, HTRA1 interacted with CDK1 protein, and CDK1 inhibitor reversed the malignant phenotype of PANC-1 and pancreatitis-initiated PDAC activated by HTRA1 overexpression. Finally, we discovered a small molecule drug that can inhibit HTRA1, carfilzomib, which has been proven to inhibit the biological functions of tumor cells in vitro and intercept the progression of pancreatitis-initiated PDAC in vivo. In conclusion, the activation of HTRA1-CDK1 pathway promotes the malignant phenotype of tumor cells by blocking the cell cycle at the G2/M phase, thereby accelerating pancreatitis-initiated PDAC. Carfilzomib is an innovative candidate drug that can inhibit pancreatitis-initiated PDAC through targeted inhibition of HTRA1.
Collapse
Affiliation(s)
- Fangyue Guo
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Yu Wu
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Yanna Zhu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Dong Shang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China.
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Hong Xiang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
3
|
Gao M, Qiu Y, Cao T, Li D, Wang J, Jiao Y, Chen Z, Huang J. Insufficient HtrA2 causes meiotic defects in aging germinal vesicle oocytes. Reprod Biol Endocrinol 2022; 20:173. [PMID: 36539842 PMCID: PMC9764539 DOI: 10.1186/s12958-022-01048-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND High-temperature requirement protease A2 (HtrA2/Omi) is a mitochondrial chaperone that is highly conserved from bacteria to humans. It plays an important role in mitochondrial homeostasis and apoptosis. In this study, we investigated the role of HtrA2 in mouse oocyte maturation. METHODS The role of HtrA2 in mouse oocyte maturation was investigated by employing knockdown (KD) or overexpression (OE) of HtrA2 in young or old germinal vesicle (GV) oocytes. We employed immunoblotting, immunostaining, fluorescent intensity quantification to test the HtrA2 knockdown on the GV oocyte maturation progression, spindle assembly checkpoint, mitochondrial distribution, spindle organization, chromosome alignment, actin polymerization, DNA damage and chromosome numbers and acetylated tubulin levels. RESULTS We observed a significant reduction in HtrA2 protein levels in aging germinal vesicle (GV) oocytes. Young oocytes with low levels of HtrA2 due to siRNA knockdown were unable to complete meiosis and were partially blocked at metaphase I (MI). They also displayed significantly more BubR1 on kinetochores, indicating that the spindle assembly checkpoint was triggered at MI. Extrusion of the first polar body (Pb1) was significantly less frequent and oocytes with large polar bodies were observed when HtrA2 was depleted. In addition, HtrA2 knockdown induced meiotic spindle/chromosome disorganization, leading to aneuploidy at metaphase II (MII), possibly due to the elevated level of acetylated tubulin. Importantly, overexpression of HtrA2 partially rescued spindle/chromosome disorganization and reduced the rate of aneuploidy in aging GV oocytes. CONCLUSIONS Collectively, our data suggest that HtrA2 is a key regulator of oocyte maturation, and its deficiency with age appears to contribute to reproduction failure in females.
Collapse
Affiliation(s)
- Min Gao
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 51000, China
- Key Laboratory of Reproductive Medicine of Guangdong Province, School of Life Sciences and the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yanling Qiu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- Key Laboratory of Reproductive Medicine of Guangdong Province, School of Life Sciences and the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510275, China
| | - Tianqi Cao
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- Key Laboratory of Reproductive Medicine of Guangdong Province, School of Life Sciences and the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510275, China
| | - Dungao Li
- The Reproduction Medicine Center of Hui Zhou Municipal Central Hospital, Huizhou, 516001, China
| | - Jingwen Wang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- Key Laboratory of Reproductive Medicine of Guangdong Province, School of Life Sciences and the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yiren Jiao
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- Key Laboratory of Reproductive Medicine of Guangdong Province, School of Life Sciences and the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510275, China
| | - Zhiyun Chen
- The Reproduction Medicine Center of Hui Zhou Municipal Central Hospital, Huizhou, 516001, China.
| | - Junjiu Huang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 51000, China.
- Key Laboratory of Reproductive Medicine of Guangdong Province, School of Life Sciences and the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510275, China.
| |
Collapse
|
4
|
Singh T, Kaur P, Singh P, Singh S, Munshi A. Differential molecular mechanistic behavior of HDACs in cancer progression. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:171. [PMID: 35972597 DOI: 10.1007/s12032-022-01770-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/10/2022] [Indexed: 12/13/2022]
Abstract
Genetic aberration including mutation in oncogenes and tumor suppressor genes transforms normal cells into tumor cells. Epigenetic modifications work concertedly with genetic factors in controlling cancer development. Histone acetyltransferases (HATs), histone deacetylases (HDACs), DNA methyltransferases (DNMTs) and chromatin structure modifier are prospective epigenetic regulators. Specifically, HDACs are histone modifiers regulating the expression of genes implicated in cell survival, growth, apoptosis, and metabolism. The majority of HDACs are highly upregulated in cancer, whereas some have a varied function and expression in cancer progression. Distinct HDACs have a positive and negative role in controlling cancer progression. HDACs are also significantly involved in tumor cells acquiring metastatic and angiogenic potential in order to withstand the anti-tumor microenvironment. HDACs' role in modulating metabolic genes has also been associated with tumor development and survival. This review highlights and discusses the molecular mechanisms of HDACs by which they regulate cell survival, apoptosis, metastasis, invasion, stemness potential, angiogenesis, and epithelial to mesenchymal transitions (EMT) in tumor cells. HDACs are the potential target for anti-cancer drug development and various inhibitors have been developed and FDA approved for a variety of cancers. The primary HDAC inhibitors with proven anti-cancer efficacy have also been highlighted in this review.
Collapse
Affiliation(s)
- Tashvinder Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India
| | - Prabhsimran Kaur
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India
| | | | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India.
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
5
|
Tossetta G, Fantone S, Licini C, Marzioni D, Mattioli-Belmonte M. The multifaced role of HtrA1 in the development of joint and skeletal disorders. Bone 2022; 157:116350. [PMID: 35131488 DOI: 10.1016/j.bone.2022.116350] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/21/2022] [Accepted: 02/01/2022] [Indexed: 12/15/2022]
Abstract
HtrA1 (High temperature requirement A1) family proteins include four members, widely conserved from prokaryotes to eukaryotes, named HtrA1, HtrA2, HtrA3 and HtrA4. HtrA1 is a serine protease involved in a variety of biological functions regulating many signaling pathways degrading specific components and playing key roles in many human diseases such as neurodegenerative disorders, pregnancy complications and cancer. Due to its role in the breakdown of many ExtraCellular Matrix (ECM) components of articular cartilage such as fibronectin, decorin and aggrecan, HtrA1 encouraged many researches on studying its role in several skeletal diseases (SDs). These studies were further inspired by the fact that HtrA1 is able to regulate the signaling of one of the most important cytokines involved in SDs, the TGFβ-1. This review aims to summarize the data currently available on the role of HtrA1 in skeletal diseases such as Osteoporosis, Rheumatoid Arthritis, Osteoarthritis and Intervertebral Disc Degeneration (IDD). The use of HtrA1 as a marker of frailty in geriatric medicine would represent a powerful tool for identifying older individuals at risk of developing skeletal disorders, evaluating an appropriate intervention to improve quality care in these people avoiding or improving age-related SDs in the elderly population.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, Ancona, Italy.
| | - Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Caterina Licini
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60126, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60126, Italy
| |
Collapse
|
6
|
May A, Su F, Dinh B, Ehlen R, Tran C, Adivikolanu H, Shaw PX. Ongoing controversies and recent insights of the ARMS2-HTRA1 locus in age-related macular degeneration. Exp Eye Res 2021; 210:108605. [PMID: 33930395 DOI: 10.1016/j.exer.2021.108605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 01/10/2021] [Accepted: 04/21/2021] [Indexed: 01/17/2023]
Abstract
Age-related macular degeneration (AMD) is the most common cause of central vision loss among elderly populations in industrialized countries. Genome-wide association studies have consistently associated two genomic loci with progression to late-stage AMD: the complement factor H (CFH) locus on chromosome 1q31 and the age-related maculopathy susceptibility 2-HtrA serine peptidase 1 (ARMS2-HTRA1) locus on chromosome 10q26. While the CFH risk variant has been shown to alter complement activity, the ARMS2-HTRA1 risk haplotype remains enigmatic due to high linkage disequilibrium and inconsistent functional findings spanning two genes that are plausibly causative for AMD risk. In this review, we detail the genetic and functional evidence used to support either ARMS2 or HTRA1 as the causal gene for AMD risk, emphasizing both the historical development and the current understanding of the ARMS2-HTRA1 locus in AMD pathogenesis. We conclude by summarizing the evidence in favor of HTRA1 and present our hypothesis whereby HTRA1-derived ECM fragments mediate AMD pathogenesis.
Collapse
Affiliation(s)
- Adam May
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, 9415 Campus Point Drive, La Jolla, CA 92093-0946, USA; Altman Clinical and Translational Research Institute, University of California, San Diego, 9452 Medical Center Drive, La Jolla, CA 92093-0990, USA.
| | - Fei Su
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, 9415 Campus Point Drive, La Jolla, CA 92093-0946, USA; Altman Clinical and Translational Research Institute, University of California, San Diego, 9452 Medical Center Drive, La Jolla, CA 92093-0990, USA.
| | - Brian Dinh
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, 9415 Campus Point Drive, La Jolla, CA 92093-0946, USA; Altman Clinical and Translational Research Institute, University of California, San Diego, 9452 Medical Center Drive, La Jolla, CA 92093-0990, USA.
| | - Rachael Ehlen
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, 9415 Campus Point Drive, La Jolla, CA 92093-0946, USA; Altman Clinical and Translational Research Institute, University of California, San Diego, 9452 Medical Center Drive, La Jolla, CA 92093-0990, USA.
| | - Christina Tran
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, 9415 Campus Point Drive, La Jolla, CA 92093-0946, USA; Altman Clinical and Translational Research Institute, University of California, San Diego, 9452 Medical Center Drive, La Jolla, CA 92093-0990, USA.
| | - Harini Adivikolanu
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, 9415 Campus Point Drive, La Jolla, CA 92093-0946, USA; Altman Clinical and Translational Research Institute, University of California, San Diego, 9452 Medical Center Drive, La Jolla, CA 92093-0990, USA.
| | - Peter X Shaw
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, 9415 Campus Point Drive, La Jolla, CA 92093-0946, USA; Altman Clinical and Translational Research Institute, University of California, San Diego, 9452 Medical Center Drive, La Jolla, CA 92093-0990, USA.
| |
Collapse
|
7
|
Lu ZG, May A, Dinh B, Lin V, Su F, Tran C, Adivikolanu H, Ehlen R, Che B, Wang ZH, Shaw DH, Borooah S, Shaw PX. The interplay of oxidative stress and ARMS2-HTRA1 genetic risk in neovascular AMD. ACTA ACUST UNITED AC 2021; 5. [PMID: 34017939 PMCID: PMC8133762 DOI: 10.20517/2574-1209.2020.48] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of vision loss in adults over 60 years old globally. There are two forms of advanced AMD: “dry” and “wet”. Dry AMD is characterized by geographic atrophy of the retinal pigment epithelium and overlying photoreceptors in the macular region; whereas wet AMD is characterized by vascular penetrance from the choroid into the retina, known as choroidal neovascularization (CNV). Both phenotypes eventually lead to loss of central vision. The pathogenesis of AMD involves the interplay of genetic polymorphisms and environmental risk factors, many of which elevate retinal oxidative stress. Excess reactive oxygen species react with cellular macromolecules, forming oxidation-modified byproducts that elicit chronic inflammation and promote CNV. Additionally, genome-wide association studies have identified several genetic variants in the age-related maculopathy susceptibility 2/high-temperature requirement A serine peptidase 1 (ARMS2-HTRA1) locus associated with the progression of late-stage AMD, especially the wet subtype. In this review, we will focus on the interplay of oxidative stress and HTRA1 in drusen deposition, chronic inflammation, and chronic angiogenesis. We aim to present a multifactorial model of wet AMD progression, supporting HTRA1 as a novel therapeutic target upstream of vascular endothelial growth factor (VEGF), the conventional target in AMD therapeutics. By inhibiting HTRA1’s proteolytic activity, we can reduce pro-angiogenic signaling and prevent proteolytic breakdown of the blood-retina barrier. The anti-HTRA1 approach offers a promising alternative treatment option to wet AMD, complementary to anti-VEGF therapy.
Collapse
Affiliation(s)
- Zhi-Gang Lu
- Department of Neurology, First People's Hospital of Jingmen, Jingchu University of Technology, Jingmen 448000, Hubei, China.,Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adam May
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brian Dinh
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Victor Lin
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fei Su
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christina Tran
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Harini Adivikolanu
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Rachael Ehlen
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Briana Che
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zhi-Hao Wang
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel H Shaw
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Westview High School, San Diego, CA 92131, USA
| | - Shyamanga Borooah
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Peter X Shaw
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
8
|
Zurawa-Janicka D, Kobiela J, Slebioda T, Peksa R, Stanislawowski M, Wierzbicki PM, Wenta T, Lipinska B, Kmiec Z, Biernat W, Lachinski AJ, Sledzinski Z. Expression of HTRA Genes and Its Association with Microsatellite Instability and Survival of Patients with Colorectal Cancer. Int J Mol Sci 2020; 21:E3947. [PMID: 32486357 PMCID: PMC7312515 DOI: 10.3390/ijms21113947] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
HtrA proteases regulate cellular homeostasis and cell death. Their dysfunctions have been correlated with oncogenesis and response to therapeutic treatment. We investigated the relation between HtrA1-3 expression and clinicopathological, and survival data, as well as the microsatellite status of tumors. Sixty-five colorectal cancer patients were included in the study. The expression of HTRA1-3 was estimated at the mRNA and protein levels by quantitative PCR and immunoblotting. Microsatellite status was determined by high-resolution-melting PCR. We found that the HTRA1 mRNA level was higher in colorectal cancer tissue as compared to the unchanged mucosa, specifically in primary lesions of metastasizing cancer. The levels of HtrA1 and HtrA2 proteins were reduced in tumor tissue when compared to unchanged mucosa, specifically in primary lesions of metastasizing disease. Moreover, a decrease in HTRA1 and HTRA2 transcripts' levels in cancers with a high level of microsatellite instability compared to microsatellite stable ones has been observed. A low level of HtrA1 or/and HtrA2 in cancer tissue correlated with poorer patient survival. The expression of HTRA1 and HTRA2 changes during colorectal carcinogenesis and microsatellite instability may be, at least partially, associated with these changes. The alterations in the HTRA1/2 genes' expression are connected with metastatic potential of colorectal cancer and may affect patient survival.
Collapse
Affiliation(s)
- Dorota Zurawa-Janicka
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (T.W.); (B.L.)
| | - Jarek Kobiela
- Department of General, Endocrine and Transplant Surgery, Faculty of Medicine, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214 Gdansk, Poland; (J.K.); (A.J.L.); (Z.S.)
| | - Tomasz Slebioda
- Department of Histology, Faculty of Medicine, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (T.S.); (M.S.); (P.M.W.); (Z.K.)
| | - Rafal Peksa
- Department of Pathomorphology, Faculty of Medicine, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214 Gdansk, Poland; (R.P.); (W.B.)
| | - Marcin Stanislawowski
- Department of Histology, Faculty of Medicine, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (T.S.); (M.S.); (P.M.W.); (Z.K.)
| | - Piotr Mieczyslaw Wierzbicki
- Department of Histology, Faculty of Medicine, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (T.S.); (M.S.); (P.M.W.); (Z.K.)
| | - Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (T.W.); (B.L.)
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (T.W.); (B.L.)
| | - Zbigniew Kmiec
- Department of Histology, Faculty of Medicine, Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (T.S.); (M.S.); (P.M.W.); (Z.K.)
| | - Wojciech Biernat
- Department of Pathomorphology, Faculty of Medicine, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214 Gdansk, Poland; (R.P.); (W.B.)
| | - Andrzej Jacek Lachinski
- Department of General, Endocrine and Transplant Surgery, Faculty of Medicine, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214 Gdansk, Poland; (J.K.); (A.J.L.); (Z.S.)
| | - Zbigniew Sledzinski
- Department of General, Endocrine and Transplant Surgery, Faculty of Medicine, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214 Gdansk, Poland; (J.K.); (A.J.L.); (Z.S.)
| |
Collapse
|
9
|
Yao Y, Li N. Effect of HtrA1 Polymorphism on Sensitivity to Chemotherapy in Patients with Colon Cancer. Med Sci Monit 2020; 26:e921933. [PMID: 32218415 PMCID: PMC7133446 DOI: 10.12659/msm.921933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 01/03/2020] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND This study was performed to estimate the genetic effects of HtrA1 polymorphisms rs1049331 and rs11200638 on treatment response in stage III colon cancer patients receiving 5-FU-based chemotherapy. MATERIAL AND METHODS A total of 105 stage III colon cancer patients who received postoperative 5-FU based adjuvant chemotherapy were included in our study. Chemotherapy was performed in 3 cycles for the patients. HtrA1 rs1049331 and rs11200638 polymorphisms were genotyped via polymerase chain reaction with sequencing method. The treatment response was estimated according to the RECIST guidelines. RESULTS The response rate of the eligible patients was 53.33%. For rs1049331, the presences of TT genotype and T allele indicted reduced chemotherapy sensitivity (adjusted TT: OR=1.736, 95%CI: 1.001-3.011, P=0.049; T: OR=1.801, 95%CI: 1.054-2.932, P=0.039). The rs11200638 polymorphism had no significant association with chemotherapy sensitivity in the study population (P>0.05 for all). CONCLUSIONS HtrA1 rs1049331 polymorphism, but not rs11200638 polymorphism, can influence individual sensitivity to 5-FU-based treatment in stage III colon cancer patients.
Collapse
Affiliation(s)
- Yi Yao
- Medical School of Chinese PLA, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, P.R. China
- Department of Gastroenterology, Eighth Medical Center of People’s Liberation Army (PLA) General Hospital, Beijing, P.R. China
| | - Nan Li
- Medical School of Chinese PLA, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, P.R. China
| |
Collapse
|
10
|
Hao Y, Xiao Y, Zhang Z, Zhao H, Zhou W, Guo X, Lu A, Li X. Transition of the abnormal Savda syndrome to the hepatic carcinoma shifted unfolded protein response to autophagy was partly reversed by Savda Munziq in a rat model. Biomed Pharmacother 2019; 121:109643. [PMID: 31810133 DOI: 10.1016/j.biopha.2019.109643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 10/18/2019] [Accepted: 10/26/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES In Uighur medicine, abnormal Savda Munziq (AMSq) is an adjuvant therapy for cancer patients with abnormal Savda syndrome (ASS) who exhibit the highest degree of pathogenicity and malignancy. The aim of the study was to understand the role(s) of AMSq in cancer patients with ASS. METHODS A total of 125 rats were divided into groups: control (NC) (n = 15), ASS (n = 25), ASS with hepatic carcinoma (ASSHC) (n = 25) as well as ASSHC treated with low dose (ASSHC-L, n = 20), medium dose (ASSHC-M, n = 20) and high dose (ASSHC-H, n = 20) AMSq. Changes in the unfolded protein response (UPR) and autophagy were analyzed by RT profiler PCR array kits, which covered 84 UPR and 84 autophagy related genes. Protein expression analyses of LC3B, ATG8, GRP78 and CHOP were carried out using western blotting. RESULTS CHOP and GRP78 expression was enhanced in ASS and ASSHC compared to NC rats and further increased AMSq dose-dependently, indicating an UPR triggering effect of AMSq. The ratios of ATG8/LCB3II-LCB3I protein were reduced in ASSHC rats, an effect which was partly reversed by AMSq. Compared to NC rats in the ASS group, 24 UPR genes were significantly upregulated and 3 downregulated, whereas only 5 autophagy genes were significantly upregulated and 5 downregulated. Compared to NC rats in the ASSHC group, 15 UPR genes were significantly upregulated and 10 downregulated, whereas 16 autophagy genes were significantly upregulated and 8 downregulated. The RT profiler data indicated a shift from UPR in the ASS to the autophagy response in ASSHC rats. ASMq effects on ASSHC rats comprised significant downregulation of 10 autophagy and 2 UPR genes. CONCLUSION The transformation into hepatic cancer cells included a shift from endoplasmic reticulum stress-related UPR to autophagy gene activation, an effect which could be partly reversed by ASMq.
Collapse
Affiliation(s)
- Yi Hao
- Department of Ultrasound, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China
| | - Yue Xiao
- Shenzhen Key Laboratory of Viral Oncology, Center for Clinical Research and Innovation (CCRI), Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China
| | - Zhihong Zhang
- Shenzhen Key Laboratory of Viral Oncology, Center for Clinical Research and Innovation (CCRI), Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China
| | - Hongli Zhao
- Shenzhen Key Laboratory of Viral Oncology, Center for Clinical Research and Innovation (CCRI), Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China
| | - Wenyan Zhou
- Shenzhen Key Laboratory of Viral Oncology, Center for Clinical Research and Innovation (CCRI), Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China
| | - Xia Guo
- Shenzhen Key Laboratory of Viral Oncology, Center for Clinical Research and Innovation (CCRI), Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Anwei Lu
- Department of Gynecology, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Xin Li
- Shenzhen Key Laboratory of Viral Oncology, Center for Clinical Research and Innovation (CCRI), Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| |
Collapse
|
11
|
Wu H, Ma S, Xiang M, Tong S. HTRA1 promotes transdifferentiation of normal fibroblasts to cancer-associated fibroblasts through activation of the NF-κB/bFGF signaling pathway in gastric cancer. Biochem Biophys Res Commun 2019; 514:933-939. [PMID: 31088682 DOI: 10.1016/j.bbrc.2019.05.076] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/30/2022]
Abstract
Cancer-associated fibroblasts comprise the major stromal cell populations in gastric cancer, which is a significant contributor to cancer-related death worldwide. As a member of the serine protease family, HTRA1 is reportedly involved in malignant transformation of various tumor types. In the present study, we observed that HTRA1 is positively correlated with α-SMA expression in gastric cancer tissues, which was also confirmed by correlation analysis and Gene Set Enrichment Analysis (GSEA) using the GEO database. Upregulation of HTRA1 in gastric cancer cell lines induces expression of α-SMA in normal fibroblasts. To explore how HTRA1 activates normal fibroblasts, an ELISA assay was performed. Secretion of bFGF/FGF2 from gastric cancer cells was significantly increased in response to HTRA1 overexpression. However, upreguation of α-SMA in normal fibroblasts induced by HTRA1 was restored by inhibiting the expression of bFGF. Furthermore, HTRA1 promotes bFGF/FGF2 expression through activation of NF-κB signaling in gastric cancer cells. Inhibition of the NF-κB signaling pathway partially restored baseline expression levels of α-SMA induced by HTRA1. In conclusion, HTRA1 promotes transdifferentiation of normal fibroblasts to cancer-associated fibroblasts by increasing bFGF/FGF2 expression, which is dependent upon activation of NF-κB signaling in gastric cancer.
Collapse
Affiliation(s)
- Hongxue Wu
- Department of Gastrointestinal Surgery, Wuhan University, Renmin Hospital, No. 99 Zhang Zhidong Road, Wuhan, 430060, Hubei Province, PR China
| | - Shujin Ma
- Department of Gastrointestinal Surgery, Wuhan University, Renmin Hospital, No. 99 Zhang Zhidong Road, Wuhan, 430060, Hubei Province, PR China
| | - Mingwei Xiang
- Department of Gastrointestinal Surgery, Wuhan University, Renmin Hospital, No. 99 Zhang Zhidong Road, Wuhan, 430060, Hubei Province, PR China
| | - Shilun Tong
- Department of Gastrointestinal Surgery, Wuhan University, Renmin Hospital, No. 99 Zhang Zhidong Road, Wuhan, 430060, Hubei Province, PR China.
| |
Collapse
|
12
|
Klose R, Adam MG, Weis EM, Moll I, Wüstehube-Lausch J, Tetzlaff F, Oka C, Ehrmann M, Fischer A. Inactivation of the serine protease HTRA1 inhibits tumor growth by deregulating angiogenesis. Oncogene 2018; 37:4260-4272. [PMID: 29713059 DOI: 10.1038/s41388-018-0258-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 02/26/2018] [Accepted: 03/27/2018] [Indexed: 11/09/2022]
Abstract
The serine protease HTRA1 is involved in several vascular diseases and its expression is often deregulated in cancer. We aimed at identifying how HTRA1 in the vasculature affects tumor growth. Here we report that silencing of HTRA1 in cultured endothelial cells increased migration rate and tube formation, whereas forced HTRA1 expression impaired sprouting angiogenesis. Mechanistically, endothelial HTRA1 expression enhanced Delta/Notch signaling by reducing the amount of the weak Notch ligand JAG1. HTRA1 physically interacted with JAG1 and cleaved it within the intracellular domain, leading to protein degradation. Expression of a constitutive active Notch1 prevented the hypersprouting phenotype upon silencing of HTRA1. In HtrA1-deficient mice, endothelial Notch signaling was diminished and isolated endothelial cells had increased expression of VEGF receptor-2. Growth of syngeneic tumors was strongly impaired in HtrA1-/- mice. The tumor vasculature was much denser in HtrA1-/- mice and less covered with mural cells. This chaotic and immature vascular network was poorly functional as indicated by large hypoxic tumor areas and low tumor cell proliferation rates. In summary, inhibition of HTRA1 in the tumor stroma impaired tumor progression by deregulating angiogenesis.
Collapse
Affiliation(s)
- Ralph Klose
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - M Gordian Adam
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany.,Metanomics Health GmbH, Berlin, Germany
| | - Eva-Maria Weis
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - Joycelyn Wüstehube-Lausch
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany.,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.,BioNTech AG, Mainz, Germany
| | - Fabian Tetzlaff
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - Chio Oka
- Laboratory of Gene Function in Animals, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara, 630-0192, Japan
| | - Michael Ehrmann
- Centre of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, 45117, Germany
| | - Andreas Fischer
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany. .,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany. .,Medical Clinic I, Endocrinology and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, 69120, Germany.
| |
Collapse
|