1
|
Mongkolpathumrat P, Pikwong F, Phutiyothin C, Srisopar O, Chouyratchakarn W, Unnajak S, Nernpermpisooth N, Kumphune S. The secretory leukocyte protease inhibitor (SLPI) in pathophysiology of non-communicable diseases: Evidence from experimental studies to clinical applications. Heliyon 2024; 10:e24550. [PMID: 38312697 PMCID: PMC10835312 DOI: 10.1016/j.heliyon.2024.e24550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 12/13/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
Non-communicable diseases (NCDs) are a worldwide health issue because of their prevalence, negative impacts on human welfare, and economic costs. Protease enzymes play important roles in viral and NCD diseases. Slowing disease progression by inhibiting proteases using small-molecule inhibitors or endogenous inhibitory peptides appears to be crucial. Secretory leukocyte protease inhibitor (SLPI), an inflammatory serine protease inhibitor, maintains protease/antiprotease balance. SLPI is produced by host defense effector cells during inflammation to prevent proteolytic enzyme-induced tissue damage. The etiology of noncommunicable illnesses is linked to SLPI's immunomodulatory and tissue regeneration roles. Disease phases are associated with SLPI levels and activity changes in regional tissue and circulation. SLPI has been extensively evaluated in inflammation, but rarely in NCDs. Unfortunately, the thorough evaluation of SLPI's pathophysiological functions in NCDs in multiple research models has not been published elsewhere. In this review, data from PubMed from 2014 to 2023 was collected, analysed, and categorized into in vitro, in vivo, and clinical studies. According to the review, serine protease inhibitor (SLPI) activity control is linked to non-communicable diseases (NCDs) and other illnesses. Overexpression of the SLPI gene and protein may be a viable diagnostic and therapeutic target for non-communicable diseases (NCDs). SLPI is also cytoprotective, making it a unique treatment. These findings suggest that future research should focus on these pathways using advanced methods, reliable biomarkers, and therapy approaches to assess susceptibility and illness progression. Implications from this review will help pave the way for a new therapeutic target and diagnosis marker for non-communicable diseases.
Collapse
Affiliation(s)
- Podsawee Mongkolpathumrat
- Cardiovascular and Thoracic Technology Program, Chulabhorn International College of Medicine (CICM), Thammasat University (Rangsit Center), Pathumthani 12120, Thailand
| | - Faprathan Pikwong
- Biomedical Engineering and Innovation Research Center, Chiang Mai University, Mueang Chiang Mai District, Chiang Mai, 50200 Thailand
- Biomedical Engineering Institute (BMEI), Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Chayanisa Phutiyothin
- Biomedical Engineering and Innovation Research Center, Chiang Mai University, Mueang Chiang Mai District, Chiang Mai, 50200 Thailand
- Biomedical Engineering Institute (BMEI), Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Onnicha Srisopar
- Biomedical Engineering and Innovation Research Center, Chiang Mai University, Mueang Chiang Mai District, Chiang Mai, 50200 Thailand
- Biomedical Engineering Institute (BMEI), Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Wannapat Chouyratchakarn
- Biomedical Engineering and Innovation Research Center, Chiang Mai University, Mueang Chiang Mai District, Chiang Mai, 50200 Thailand
- Biomedical Engineering Institute (BMEI), Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Sasimanas Unnajak
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, 10900 Thailand
| | - Nitirut Nernpermpisooth
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000 Thailand
| | - Sarawut Kumphune
- Biomedical Engineering and Innovation Research Center, Chiang Mai University, Mueang Chiang Mai District, Chiang Mai, 50200 Thailand
- Biomedical Engineering Institute (BMEI), Chiang Mai University, Chiang Mai, 50200 Thailand
| |
Collapse
|
2
|
Zhang X, Liu SS, Ma J, Qu W. Secretory leukocyte protease inhibitor (SLPI) in cancer pathophysiology: Mechanisms of action and clinical implications. Pathol Res Pract 2023; 248:154633. [PMID: 37356220 DOI: 10.1016/j.prp.2023.154633] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/17/2023] [Accepted: 06/18/2023] [Indexed: 06/27/2023]
Abstract
Cancer is a multifaceted disorder frequently linked to the dysregulation of several biological processes. The SLPI is a multifunctional protein involved in the modulation of immunological response and the inhibition of protease activities. SLPI acts as an inhibitor of proteases, exerts antibacterial properties, and suppresses the transcription of proinflammatory genes through the nuclear factor-kappa B (NF-κB) pathway. The role of this protein as a regulatory agent has been implicated in various types of cancer. Recent research has revealed that SLPI upregulation in cancer cells enhances the metastatic capacity of epithelial malignancies, indicating the deleterious effects of this protein. Furthermore, SLPI interacts intricately with other cancer-promoting factors, including matrix metalloproteinase-2 (MMP-2), MMP-9, the NF-κB and Akt pathways, and the p53-upregulated modulator of apoptosis (PUMA). This review provides an overview of the role of SLPI in cancer pathophysiology, emphasizing its expression in cancer cells and tissues, its potential as a prognostic biomarker, and its therapeutic promise as a target in cancer treatment. The mechanisms of SLPI action in cancer, including its anti-inflammatory effects, regulation of cell proliferation and angiogenesis, and modulation of the tumor microenvironment, have been investigated. The clinical implications of SLPI in cancer have been discussed, including its potential as a diagnostic and prognostic biomarker, its role in chemoresistance, and its therapeutic potential in several types of cancer, such as hepatocellular carcinoma (HCC), colorectal cancer (CRC), pancreatic cancer, head and neck squamous cell carcinoma (HNSCC), ovarian cancer (OvCa), prostate cancer (PC), gastric cancer (GC), breast cancer, and other cancers. In addition, we emphasized the significance of SLPI in cancer, which offers fresh perspectives on potential targets for cancer therapy.
Collapse
Affiliation(s)
- Xiaohua Zhang
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China
| | - Shan Shan Liu
- Department of General Medicine, the Second Hospital of Jilin University, Changchun 130000, China.
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China
| | - Wei Qu
- Department of General Medicine, the Second Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
3
|
Wei Z, Liu G, Jia R, Zhang W, Li L, Zhang Y, Wang Z, Bai X. Inhibition of secretory leukocyte protease inhibitor (SLPI) promotes the PUMA-mediated apoptosis and chemosensitivity to cisplatin in colorectal cancer cells. Discov Oncol 2023; 14:1. [PMID: 36595102 PMCID: PMC9810770 DOI: 10.1007/s12672-022-00535-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/21/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Aberrant expression of Secretory Leukocyte Protease Inhibitor (SLPI) has been associated with human cancer growth and its suppression was identified as a potential target for anti-cancer drugs, particularly in colorectal cancer. However, the underlying mechanism by which SLPI affected the development of drug resistance in CRC remains unclear. OBJECTIVE This study investigated the role of SLPI in the p53-up-regulated modulator of apoptosis (PUMA)-mediated CRC cells' apoptosis and their chemosensitivity to Cisplatin. METHODS A series of qRT-PCR and western blot analyses were performed to characterize the expressions of SLPI, PUMA, and Akt in CRC lines. Tunel, transwell, and CCK-8 analyses were monitored to define the impacts of the siRNA-mediated knockdown of SLPI on CRC cell development. Furthermore, in vivo development of CRC was evaluated in nude mice infected with siSLPI or Cisplatin alone or both, and Ki67 and caspase-3 immunohistochemistry assay was monitored on multiple tissue microarray from the same cohort. RESULTS Our results showed that SLPI inhibition strongly promoted the expressions of the pro-apoptotic protein PUMA, cleaved-caspase3 and Bax and reduced the cell viability of HT29 and HT116 cell lines in vitro. In addition, siSLPI knockdown effectively suppressed both Akt and FoxO3 proteins and improved the sensitivity to cisplatin chemotherapy. Xenograft tumor assay revealed a lowered growth in mice treated with Cisplatin, while combined treatment of siSLPI achieved more significant anticancer effects than Cisplatin alone. CONCLUSIONS Taken together, these findings demonstrated that suppression of SLPI might repress the growth of human colorectal cancer cells both in vitro and in vivo. These results suggested SLPI as a novel resistance factor to Cisplatin, and a combination of Cisplatin and SLPI inhibitor be beneficial for colorectal cancer therapy.
Collapse
Affiliation(s)
- Zhijiang Wei
- The First Department of Tumor Surgery, Cangzhou Central Hospital, Cangzhou, 061001, Hebei, People's Republic of China.
| | - Guiying Liu
- The First Department of Tumor Surgery, Cangzhou Central Hospital, Cangzhou, 061001, Hebei, People's Republic of China
| | - Rufu Jia
- The Brain Science Hospital of CangZhou Central Hospital, Cangzhou, 061001, Hebei, People's Republic of China
| | - Wei Zhang
- The First Department of Tumor Surgery, Cangzhou Central Hospital, Cangzhou, 061001, Hebei, People's Republic of China
| | - Li Li
- The Brain Science Hospital of CangZhou Central Hospital, Cangzhou, 061001, Hebei, People's Republic of China
| | - Yuanyuan Zhang
- The First Department of Tumor Surgery, Cangzhou Central Hospital, Cangzhou, 061001, Hebei, People's Republic of China
| | - Zhijing Wang
- The Brain Science Hospital of CangZhou Central Hospital, Cangzhou, 061001, Hebei, People's Republic of China
| | - Xiyong Bai
- The First Department of Tumor Surgery, Cangzhou Central Hospital, Cangzhou, 061001, Hebei, People's Republic of China
| |
Collapse
|
4
|
Mongkolpathumrat P, Kijtawornrat A, Suwan E, Unajak S, Panya A, Pusadee T, Kumphune S. Anti-Protease Activity Deficient Secretory Leukocyte Protease Inhibitor (SLPI) Exerts Cardioprotective Effect against Myocardial Ischaemia/Reperfusion. Biomedicines 2022; 10:biomedicines10050988. [PMID: 35625725 PMCID: PMC9138276 DOI: 10.3390/biomedicines10050988] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 12/02/2022] Open
Abstract
Inhibition of proteases shows therapeutic potential. Our previous studies demonstrated the cardioprotection by the Secretory Leukocyte Protease Inhibitor (SLPI) against myocardial ischaemia/reperfusion (I/R) injury. However, it is unclear whether the cardioprotective effect of SLPI seen in our previous works is due to the inhibition of protease enzymes. Several studies demonstrate that the anti-protease independent activity of SLPI could provide therapeutic benefits. Here, we show for the first time that recombinant protein of anti-protease deficient mutant SLPI (L72K, M73G, L74G) (mt-SLPI) could significantly reduce cell death and intracellular reactive oxygen species (ROS) production against an in vitro simulated I/R injury. Furthermore, post-ischaemic treatment of mt-SLPI is found to significantly reduce infarct size and cardiac biomarkers lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) activity, improve cardiac functions, attenuate I/R induced-p38 MAPK phosphorylation, and reduce apoptotic regulatory protein levels, including Bax, cleaved-Caspase-3 and total Capase-8, in rats subjected to an in vivo I/R injury. Additionally, the beneficial effect of mt-SLPI was not significantly different from the wildtype (wt-SLPI). In summary, SLPI could provide cardioprotection without anti-protease activity, which could be more clinically beneficial in terms of providing cardioprotection without interfering with basal serine protease activity.
Collapse
Affiliation(s)
- Podsawee Mongkolpathumrat
- Graduate Programs in Biomedical Sciences, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand;
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Biomedical Engineering Institute (BMEI), Chiang Mai University, Chiang Mai 50200, Thailand
| | - Anusak Kijtawornrat
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Eukote Suwan
- Department of Veterinary Technology, Faculty of Veterinary Technology, Kasetsart University, Bangkok 10900, Thailand;
| | - Sasimanas Unajak
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand;
| | - Aussara Panya
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Tonapha Pusadee
- Department of Plant and Soil Science, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Sarawut Kumphune
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Biomedical Engineering Institute (BMEI), Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence: ; Tel.: +66-624-693-987
| |
Collapse
|
5
|
Nugteren S, Samsom JN. Secretory Leukocyte Protease Inhibitor (SLPI) in mucosal tissues: Protects against inflammation, but promotes cancer. Cytokine Growth Factor Rev 2021; 59:22-35. [PMID: 33602652 DOI: 10.1016/j.cytogfr.2021.01.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 01/24/2021] [Indexed: 12/20/2022]
Abstract
The immune system is continuously challenged with large quantities of exogenous antigens at the barriers between the external environment and internal human tissues. Antimicrobial activity is essential at these sites, though the immune responses must be tightly regulated to prevent tissue destruction by inflammation. Secretory Leukocyte Protease Inhibitor (SLPI) is an evolutionarily conserved, pleiotropic protein expressed at mucosal surfaces, mainly by epithelial cells. SLPI inhibits proteases, exerts antimicrobial activity and inhibits nuclear factor-kappa B (NF-κB)-mediated inflammatory gene transcription. SLPI maintains homeostasis at barrier tissues by preventing tissue destruction and regulating the threshold of inflammatory immune responses, while protecting the host from infection. However, excessive expression of SLPI in cancer cells may have detrimental consequences, as recent studies demonstrate that overexpression of SLPI increases the metastatic potential of epithelial tumors. Here, we review the varied functions of SLPI in the respiratory tract, skin, gastrointestinal tract and genitourinary tract, and then discuss the mechanisms by which SLPI may contribute to cancer.
Collapse
Affiliation(s)
- Sandrine Nugteren
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
6
|
Zhu J, Ji Y, Yu Y, Jin Y, Zhang X, Zhou J, Chen Y. Knockdown of serine/threonine protein phosphatase 5 enhances gemcitabine sensitivity by promoting apoptosis in pancreatic cancer cells in vitro. Oncol Lett 2018; 15:8761-8769. [PMID: 29805615 PMCID: PMC5950513 DOI: 10.3892/ol.2018.8363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/22/2017] [Indexed: 12/28/2022] Open
Abstract
The targeting protein of serine/threonine protein phosphatase 5 (PPP5C) has been reported to be present in various malignancies. However, its functional role in pancreatic cancer (PC) remains unknown. In the present study, the function of PPP5C in PC cells treated with the first-line drug gemcitabine (GEM) was investigated. Short hairpin (sh)RNA targeting PPP5C was constructed to knockdown PPP5C in PANC-1 cells. Cell cycle and apoptosis analyses were performed in order to investigate the mechanisms underlying the effects induced by PPP5C silencing combined with GEM treatment. Western blot analysis was applied to detect the expression of certain key regulators of cell apoptosis in PANC-1 cells treated with GEM. shRNA against PPP5C effectively suppressed the proliferation of PANC-1 cells treated with GEM. Additionally, cell cycle analysis indicated that PPP5C knockdown resulted in a higher number of PANC-1 cells treated with GEM in G0/G1 phase arrest. Knockdown of PPP5C increased the expression of associated apoptotic markers, including cleaved caspase 3, poly (ADP-ribose) polymerase and phosphorylated (p)-p53. In addition, the combination of treatment with GEM and PPP5C silencing significantly increased the apoptosis of PANC-1 cells by affecting the expression levels of p-c-Jun N-terminal kinases and p-p38. The present study suggests that PPP5C may be a potential target for the treatment of PC and that it may enhance the gemcitabine sensitivity of PC cells.
Collapse
Affiliation(s)
- Jinhui Zhu
- Department of General Surgery and Laparoscopic Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yun Ji
- Department of General Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yuanquan Yu
- Department of General Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yun Jin
- Department of General Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiaoxiao Zhang
- Department of General Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Jiale Zhou
- Department of General Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yan Chen
- Department of General Surgery and Laparoscopic Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
7
|
Hao S, Du X, Song Y, Ren M, Yang Q, Wang A, Wang Q, Zhao H, Du Z, Zhang G. Targeted gene therapy of the HSV-TK/hIL-12 fusion gene controlled by the hSLPI gene promoter of human non-small cell lung cancer in vitro. Oncol Lett 2018; 15:6503-6512. [PMID: 29731853 DOI: 10.3892/ol.2018.8148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 11/07/2017] [Indexed: 12/12/2022] Open
Abstract
The incidence of lung cancer and lung cancer-associated mortality have markedly increased worldwide, and gene-targeted therapy has emerged as a promising treatment strategy. The present study aimed to explore the targeted antitumor effect of the herpes simplex virus-thymidine kinase/human interleukin-12 (HSV-TK/hIL-12) fusion gene regulated by the human secretory leukocyte protease inhibitor (hSLPI) promoter of human non-small cell lung cancer (hNSCLC). There were four recombinant eukaryotic expression vectors: pcDNA3.1-CMV-TK, pcDNA3.1-CMV-TK/hIL-12, pcDNA3.1-phSLP-TK and pcDNA3.1-phSLP-TK/hIL-12. These were constructed and transfected into the A549, SPC-A1 and HepG2 cell lines in vitro. The expression of the HSV-TK/hIL-12 fusion gene was detected with reverse transcription-polymerase chain reaction (RT-PCR), and the content of hIL-12 was measured using an ELISA. The antitumor effect of the fusion gene on the A549, SPC-A1 and HepG2 cell lines was determined using an MTT assay. Analysis of the experimental data demonstrated that genes regulated by the cytomegalovirus promoter were expressed at the same level in three different tumor cell lines. Genes regulated by the hSLPI promoter were expressed in the A549 and SPC-A1 cell lines, but not in the HepG2 cell line. Coincidentally, the hIL-12 expression levels were similar to those observed in previous RT-PCR findings. In the Pcmv-TK/Pcmv-TK-hIL-12 group for all three cell lines, as well as in the PSLPI-TK/PSLPI-TK-hIL-12 group for the A549 and SPC-A1 cell lines, the cell survival rate declined significantly and the fusion gene transfection group indicated a lower cell survival rate, when compared with single gene transfection group. The present study indicated that the fusion gene regulated by the hSLPI promoter had a targeted antitumor effect on hNSCLC, and that the combined suicide gene and immune gene therapy had a stronger antitumor effect, compared with single gene therapy.
Collapse
Affiliation(s)
- Shuhong Hao
- Department of Hematology and Oncology, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaoyuan Du
- Department of Pathology, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Yang Song
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China
| | - Ming Ren
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China
| | - Qiwei Yang
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China.,Research Center, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ao Wang
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China
| | - Qingyu Wang
- The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China.,Research Center, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Haiyue Zhao
- Research Center, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhenwu Du
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China.,Research Center, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guizhen Zhang
- Department of Orthopedics, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China.,The Center of Molecular Diagnosis and Cellular Treatment for Metabolic Bone Diseases, Changchun, Jilin 130041, P.R. China.,Research Center, The Second Clinical College, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
8
|
Hellwege JN, Russell SB, Williams SM, Edwards TL, Velez Edwards DR. Gene-based evaluation of low-frequency variation and genetically-predicted gene expression impacting risk of keloid formation. Ann Hum Genet 2018; 82:206-215. [PMID: 29484647 DOI: 10.1111/ahg.12245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 12/08/2017] [Accepted: 01/08/2018] [Indexed: 12/31/2022]
Abstract
Keloids are benign dermal tumors occurring approximately 20 times more often in individuals of African descent as compared to individuals of European descent. While most keloids occur sporadically, a genetic predisposition is supported by both familial aggregation of some keloids and large differences in risk among populations. Despite Africans and African Americans being at increased risk over lighter-skinned individuals, little genetic research exists into this phenotype. Using a combination of admixture mapping and exome analysis, we reported multiple common variants within chr15q21.2-22.3 associated with risk of keloid formation in African Americans. Here we describe a gene-based association analysis using 478 African American samples with exome genotyping data to identify genes containing low-frequency variants associated with keloids, with evaluation of genetically-predicted gene expression in skin tissues using association summary statistics. The strongest signal from gene-based association was located in C15orf63 (P-value = 6.6 × 10-6 ) located at 15q15.3. The top result from gene expression was increased predicted DCAF4 expression (P-value = 5.5 × 10-4 ) in non-sun-exposed skin, followed by increased predicted OR10A3 expression in sun-exposed skin (P-value = 6.9 × 10-4 ). Our findings identify variation with putative roles in keloid formation, enhanced by the use of predicted gene expression to support the biological roles of variation identified only though genetic association studies.
Collapse
Affiliation(s)
- Jacklyn N Hellwege
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shirley B Russell
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Dermatology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Scott M Williams
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Digna R Velez Edwards
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
9
|
Elevated Concentrations of SERPINE2/Protease Nexin-1 and Secretory Leukocyte Protease Inhibitor in the Serum of Patients with Papillary Thyroid Cancer. DISEASE MARKERS 2017; 2017:4962137. [PMID: 28255192 PMCID: PMC5306996 DOI: 10.1155/2017/4962137] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/02/2017] [Accepted: 01/16/2017] [Indexed: 01/02/2023]
Abstract
Introduction. SERPINE2 and secretory leukocyte protease inhibitor (SLPI) are proteins with anticoagulant properties which could promote solid tumor growth. However, their role in the pathogenesis of thyroid cancer has not been determined. Materials and Methods. The aim of this study was to assess serum SERPINE2 and SLPI concentrations in a group of 36 patients with papillary thyroid cancer (PTC) and a group of 19 subjects with multinodular nontoxic goiter (MNG). The control group (CG) consisted of 20 healthy volunteers. Blood samples were collected one day before surgery. Serum SERPINE2 and SLPI concentrations were measured using specific ELISA methods. Results. Significantly higher concentrations of SERPINE2 and SLPI were found in patients with PTC as compared with MNG and controls. Positive correlation was found between SERPINE2 and SLPI concentrations in PTC patients. The levels of SERPINE2 and SLPI did not differ significantly between MNG and healthy controls. Conclusions. Our results indicate that SERPINE2 and SLPI play a significant role in the development of papillary thyroid cancer and imply that the evaluation of serum concentrations of both anticoagulant molecules may be considered as additional marker for the differentiation of malignancies during the preoperative diagnosis of patients with thyroid gland tumors.
Collapse
|
10
|
Choi BD, Lee SY, Jeong SJ, Lim DS, Cha HJ, Chung WG, Jeong MJ. Secretory leukocyte protease inhibitor promotes differentiation and mineralization of MC3T3-E1 preosteoblasts on a titanium surface. Mol Med Rep 2016; 14:1241-6. [PMID: 27279420 DOI: 10.3892/mmr.2016.5381] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 03/08/2016] [Indexed: 11/06/2022] Open
Abstract
Mineralized bone matrix constituted with collagenous and non-collagenous proteins was synthesized by osteoblasts differentiated from mesenchymal stem cells. Secretory leukocyte protease inhibitor (SLPI), a serine protease inhibitor, promotes cell migration and proliferation, and suppresses the inflammatory response. Recent studies reported that SLPI regulates the formation of dentin and mineralization by odontoblasts and increases the adhesion and viability of preosteoblasts on a titanium (Ti) surface. Ti and its alloys are widely used implant materials in artificial joints and dental implants owing to their biocompatibility with bone. Therefore, this study aimed to examine whether SLPI can be an effective molecule in promoting differentiation and mineralization of osteoblasts on a Ti surface. In order to investigate the effects of SLPI on osteoblasts, an MTT assay, PCR, western blotting and Alizarin Red S staining were performed. The results demonstrated that SLPI increased the viability of osteoblasts during differentiation on Ti discs compared with that of the control. The expression levels of SLPI mRNA and protein were higher than that of the control after treatment of osteoblasts with SLPI on Ti discs during differentiation. SLPI increased the formation of mineralized nodules and mRNA expression of alkaline phosphatase, dentin sialophosphoprotein, dentin matrix protein 1, bone sialoprotein, and collagen I in osteoblasts on Ti discs compared with that of the control. In conclusion, SLPI increases the viability and promotes the differentiation and mineralization of osteoblasts on Ti surfaces, suggesting that SLPI is an effective molecule for achieving successful osseointegration between osteoblasts and a Ti surface.
Collapse
Affiliation(s)
- Baik-Dong Choi
- Department of Oral Histology and Developmental Biology, School of Dentistry, Chosun University, Gwangju 501‑759, Republic of Korea
| | - Seung-Yeon Lee
- Department of Oral Histology and Developmental Biology, School of Dentistry, Chosun University, Gwangju 501‑759, Republic of Korea
| | - Soon-Jeong Jeong
- Department of Dental Hygiene, Youngsan University, Yangsan, Gyeongsangnam‑do 626‑790, Republic of Korea
| | - Do-Seon Lim
- Department of Dental Hygiene, Eulji University, Seongnam, Gyeonggi 461‑713, Republic of Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, College of Medicine, Kosin University, Busan 602‑072, Republic of Korea
| | - Won-Gyun Chung
- Department of Dental Hygiene, Wonju College of Medicine, Yonsei University, Wonju, Gangwon 220‑701, Republic of Korea
| | - Moon-Jin Jeong
- Department of Oral Histology and Developmental Biology, School of Dentistry, Chosun University, Gwangju 501‑759, Republic of Korea
| |
Collapse
|