1
|
Chen H, Zhang L, Liu M, Li Y, Chi Y. Multi-Omics Research on Angina Pectoris: A Novel Perspective. Aging Dis 2024:AD.2024.1298. [PMID: 39751862 DOI: 10.14336/ad.2024.1298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Angina pectoris (AP), a clinical syndrome characterized by paroxysmal chest pain, is caused by insufficient blood supply to the coronary arteries and sudden temporary myocardial ischemia and hypoxia. Long-term AP typically induces other cardiovascular events, including myocardial infarction and heart failure, posing a serious threat to patient safety. However, AP's complex pathological mechanisms and developmental processes introduce significant challenges in the rapid diagnosis and accurate treatment of its different subtypes, including stable angina pectoris (SAP), unstable angina pectoris (UAP), and variant angina pectoris (VAP). Omics research has contributed significantly to revealing the pathological mechanisms of various diseases with the rapid development of high-throughput sequencing approaches. The application of multi-omics approaches effectively interprets systematic information on diseases from the perspective of genes, RNAs, proteins, and metabolites. Integrating multi-omics research introduces novel avenues for identifying biomarkers to distinguish different AP subtypes. This study reviewed articles related to multi-omics and AP to elaborate on the research progress in multi-omics approaches (including genomics, transcriptomics, proteomics, and metabolomics), summarized their applications in screening biomarkers employed to discriminate multiple AP subtypes, and delineated integration methods for multi-omics approaches. Finally, we discussed the advantages and disadvantages of applying a single-omics approach in distinguishing diverse AP subtypes. Our review demonstrated that the integration of multi-omics technologies is preferable for quick and precise diagnosis of the three AP types, namely SAP, UAP, and VAP.
Collapse
Affiliation(s)
- Haiyang Chen
- Department of Psycho-cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lijun Zhang
- Department of Psycho-cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Meiyan Liu
- Department of Psycho-cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yanwei Li
- Department of Psycho-cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Yunpeng Chi
- Department of Psycho-cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Pasmans K, Goossens GH, Groenhuijzen E, Kemper EJ, Reijnders D, Most J, Blaak EE, Watt MJ, Meex RCR. Fetuin B in white adipose tissue induces inflammation and is associated with peripheral insulin resistance in mice and humans. Obesity (Silver Spring) 2024; 32:517-527. [PMID: 38112242 DOI: 10.1002/oby.23961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/13/2023] [Accepted: 10/22/2023] [Indexed: 12/21/2023]
Abstract
OBJECTIVE Fetuin B is a steatosis-responsive hepatokine that causes glucose intolerance in mice, but the underlying mechanisms remain incompletely described. This study aimed to elucidate the mechanisms of action of fetuin B by investigating its putative effects on white adipose tissue metabolism. METHODS First, fetuin B gene and protein expression was measured in multiple organs in mice and in cultured adipocytes. Next, the authors performed a hyperinsulinemic-euglycemic clamp in mice and in humans to examine the link between white adipose tissue fetuin B content and indices of insulin sensitivity. Finally, the effect of fetuin B on inflammation was investigated in cultured adipocytes by quantitative polymerase chain reaction and full RNA sequencing. RESULTS This study demonstrated in adipocytes and mice that fetuin B was produced and secreted by the liver and taken up by adipocytes and adipose tissue. There was a strong negative correlation between white adipose tissue fetuin B content and peripheral insulin sensitivity in mice and in humans. RNA sequencing and polymerase chain reaction analysis revealed that fetuin B induced an inflammatory response in adipocytes. CONCLUSIONS Fetuin B content in white adipose tissue strongly associated with peripheral insulin resistance in mice and humans. Furthermore, fetuin B induced a proinflammatory response in adipocytes, which might drive peripheral insulin resistance.
Collapse
Affiliation(s)
- Kenneth Pasmans
- NUTRIM School of Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Gijs H Goossens
- NUTRIM School of Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Evi Groenhuijzen
- NUTRIM School of Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Esther J Kemper
- NUTRIM School of Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Dorien Reijnders
- NUTRIM School of Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jasper Most
- NUTRIM School of Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Orthopedics, Zuyderland Medical Center, Sittard-Geleen, The Netherlands
| | - Ellen E Blaak
- NUTRIM School of Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Matthew J Watt
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, Australia
- Department of Physiology, Monash University, Clayton, Australia
| | - Ruth C R Meex
- NUTRIM School of Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Physiology, Monash University, Clayton, Australia
| |
Collapse
|
3
|
Zhang W, Yao J, Chen C, Wang J, Zhou A. Fetuin-B Overexpression Promotes Inflammation in Diabetic Retinopathy Through Activating Microglia and the NF-κB Signaling Pathway. Curr Eye Res 2024; 49:168-179. [DOI: https:/doi.org/10.1080/02713683.2023.2276683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/21/2023] [Indexed: 02/14/2024]
Affiliation(s)
- Wenyi Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jing Yao
- Department of Ophthalmology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chen Chen
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Jianming Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Aiyi Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
4
|
Zhang W, Yao J, Chen C, Wang J, Zhou A. Fetuin-B Overexpression Promotes Inflammation in Diabetic Retinopathy Through Activating Microglia and the NF-κB Signaling Pathway. Curr Eye Res 2024; 49:168-179. [PMID: 37883127 DOI: 10.1080/02713683.2023.2276683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/21/2023] [Indexed: 10/27/2023]
Abstract
PURPOSE To investigate the expression, source, role, and mechanism of Fetuin-B (FETUB) in diabetic retinopathy (DR). METHODS ELISA and immunofluorescence were used to analyze the concentration of FETUB in plasma, aqueous fluid, and tissue specimens of patients with DR and healthy controls. Immunofluorescence, q-PCR, and western blotting were used to examine the expression of FETUB in DR mice and cells cultured with different concentrations of glucose. BV2 microglia cell line and DR mice were treated using FETUB recombination protein and FETUB shRNA to explore the function of FETUB in DR by q-PCR, western blotting, and immunofluorescence. RESULTS FETUB concentrations in plasma, aqueous fluid, and tissue specimens were significantly increased in DR patients. The mice in DR group had a higher concentration of FETUB in the retina and liver tissues than those in the control group, and the expression of FETUB was increased in both ARPE19 and BV2 cells under a high-glucose environment. The ratio of p-P65 (Phospho-P65)/P65 and the expression levels of TNF-α, VEGF, and ionized calcium binding adaptor molecule (IBA)-1 were increased in BV2 cells cultured with FETUB recombinant protein, while they were decreased in BV2 cells transfected with FETUB shRNA. Immunofluorescence staining showed that there were more IBA-1+ activated microglia in the retinas of the FETUB recombination protein group than in the retinas of the DR group, and there were fewer IBA-1+ activated microglia in the retinas of the FETUB shRNA group than in the retinas of the DR group. CONCLUSIONS FETUB sourced from endocrine, autocrine, and paracrine pathways could promote inflammation in DR by activating the NF-κB pathway and microglia.
Collapse
Affiliation(s)
- Wenyi Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Yao
- Department of Ophthalmology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chen Chen
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Jianming Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Aiyi Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
5
|
Hou J, Cao Y, Deng Q, Zhang Q, Deng X, Chen Z, Zhong Z. A fluorescence-based immunochromatographic assay using quantum dot-encapsulated nanoparticles for the rapid and sensitive detection of fetuin-B. Anal Chim Acta 2024; 1288:342143. [PMID: 38220278 DOI: 10.1016/j.aca.2023.342143] [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: 10/28/2023] [Accepted: 12/14/2023] [Indexed: 01/16/2024]
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. Earlier detection of CAD improves treatment outcomes and secondary prevention. The circulating fetuin-B protein is considered to be a promising biomarker for the early detection of CAD. However, a facile and reliable clinical test for fetuin-B is still lacking. Herein, we describe a reliable fluorescent biosensor for detecting fetuin-B in plasma that combines quantum dots-doped polystyrene nanoparticles with an immunochromatographic assay strip (QNPs-ICAS). The QNPs served as detection signals in the QNPs-ICAS sensor system, which was based on a double-antibody sandwich structure. Under optimum experimental conditions, the biosensor exhibited a broad linear range of 1-200 ng mL-1 and a low detection limit of 0.299 ng mL-1. Furthermore, the proposed immunosensor demonstrated high sensitivity, satisfactory selectivity, good reproducibility, and excellent recovery. Finally, the performance and applicability of our QNPs-based ICAS system were validated in clinical samples using a commercial ELISA kit with excellent correlations (r = 0.98451, n = 116). To conclude, the proposed sensor served as a rapid, sensitive, and accurate method for detecting fetuin-B in actual clinical samples, thereby demonstrating its potential for preliminary CAD screening and diagnosis.
Collapse
Affiliation(s)
- Jingyuan Hou
- Center for Cardiovascular Diseases, Meizhou Clinical Institute of Shantou University Medical College, Meizhou, Guangdong, 514031, China; GuangDong Engineering Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, Guangdong, 514031, China
| | - Yue Cao
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510180, China
| | - Qiaoting Deng
- Center for Cardiovascular Diseases, Meizhou Clinical Institute of Shantou University Medical College, Meizhou, Guangdong, 514031, China
| | - Qunji Zhang
- Center for Cardiovascular Diseases, Meizhou Clinical Institute of Shantou University Medical College, Meizhou, Guangdong, 514031, China
| | - Xunwei Deng
- Center for Cardiovascular Diseases, Meizhou Clinical Institute of Shantou University Medical College, Meizhou, Guangdong, 514031, China
| | - Zhenhua Chen
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China.
| | - Zhixiong Zhong
- Center for Cardiovascular Diseases, Meizhou Clinical Institute of Shantou University Medical College, Meizhou, Guangdong, 514031, China; GuangDong Engineering Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, Guangdong, 514031, China.
| |
Collapse
|
6
|
Ploypetch S, Wongbandue G, Roytrakul S, Phaonakrop N, Prapaiwan N. Comparative Serum Proteome Profiling of Canine Benign Prostatic Hyperplasia before and after Castration. Animals (Basel) 2023; 13:3853. [PMID: 38136890 PMCID: PMC10740436 DOI: 10.3390/ani13243853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/18/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
BPH is the most prevalent prostatic condition in aging dogs. Nevertheless, clinical diagnosis and management remain inconsistent. This study employed in-solution digestion coupled with nano-liquid chromatography tandem mass spectrometry to assess serum proteome profiling of dogs with BPH and those dogs after castration. Male dogs were divided into two groups; control and BPH groups. In the BPH group, each dog was evaluated at two time points: Day 0 (BF subgroup) and Day 30 after castration (AT subgroup). In the BF subgroup, three proteins were significantly upregulated and associated with dihydrotestosterone: solute carrier family 5 member 5, tyrosine-protein kinase, and FRAT regulator of WNT signaling pathway 1. Additionally, the overexpression of polymeric immunoglobulin receptors in the BF subgroup hints at its potential as a novel protein linked to the BPH development process. Conversely, alpha-1-B glycoprotein (A1BG) displayed significant downregulation in the BF subgroup, suggesting A1BG's potential as a predictive protein for canine BPH. Finasteride was associated with increased proteins in the AT subgroup, including apolipoprotein C-I, apolipoprotein E, apolipoprotein A-II, TAO kinase 1, DnaJ homolog subfamily C member 16, PH domain and leucine-rich repeat protein phosphatase 1, neuregulin 1, and pseudopodium enriched atypical kinase 1. In conclusion, this pilot study highlighted alterations in various serum proteins in canine BPH, reflecting different pathological changes occurring in this condition. These proteins could be a source of potential non-invasive biomarkers for diagnosing this disease.
Collapse
Affiliation(s)
- Sekkarin Ploypetch
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom 73170, Thailand; (S.P.); (G.W.)
| | - Grisnarong Wongbandue
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom 73170, Thailand; (S.P.); (G.W.)
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand; (S.R.); (N.P.)
| | - Narumon Phaonakrop
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand; (S.R.); (N.P.)
| | - Nawarus Prapaiwan
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom 73170, Thailand; (S.P.); (G.W.)
| |
Collapse
|
7
|
Liu X, Shao Y, Han L, Zhang R, Chen J. Emerging Evidence Linking the Liver to the Cardiovascular System: Liver-derived Secretory Factors. J Clin Transl Hepatol 2023; 11:1246-1255. [PMID: 37577236 PMCID: PMC10412704 DOI: 10.14218/jcth.2022.00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/19/2022] [Accepted: 02/27/2023] [Indexed: 07/03/2023] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide. Recently, accumulating evidence has revealed hepatic mediators, termed as liver-derived secretory factors (LDSFs), play an important role in regulating CVDs such as atherosclerosis, coronary artery disease, thrombosis, myocardial infarction, heart failure, metabolic cardiomyopathy, arterial hypertension, and pulmonary hypertension. LDSFs presented here consisted of microbial metabolite, extracellular vesicles, proteins, and microRNA, they are primarily or exclusively synthesized and released by the liver, and have been shown to exert pleiotropic actions on cardiovascular system. LDSFs mainly target vascular endothelial cell, vascular smooth muscle cells, cardiomyocytes, fibroblasts, macrophages and platelets, and further modulate endothelial nitric oxide synthase/nitric oxide, endothelial function, energy metabolism, inflammation, oxidative stress, and dystrophic calcification. Although some LDSFs are known to be detrimental/beneficial, controversial findings were also reported for many. Therefore, more studies are required to further explore the causal relationships between LDSFs and CVDs and uncover the exact mechanisms, which is expected to extend our understanding of the crosstalk between the liver and cardiovascular system and identify potential therapeutic targets. Furthermore, in the case of patients with liver disease, awareness should be given to the implications of these abnormalities in the cardiovascular system. These studies also underline the importance of early recognition and intervention of liver abnormalities in the practice of cardiovascular care, and a multidisciplinary approach combining hepatologists and cardiologists would be more preferable for such patients.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| | - Yijia Shao
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linjiang Han
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| | - Ruyue Zhang
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| | - Jimei Chen
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Murota Y, Nagane M, Wu M, Santra M, Venkateswaran S, Tanaka S, Bradley M, Taga T, Tabu K. A niche-mimicking polymer hydrogel-based approach to identify molecular targets for tackling human pancreatic cancer stem cells. Inflamm Regen 2023; 43:46. [PMID: 37759310 PMCID: PMC10523636 DOI: 10.1186/s41232-023-00296-0] [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: 06/19/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) is one of the most fatal human cancers, but effective therapies remain to be established. Cancer stem cells (CSCs) are highly resistant to anti-cancer drugs and a deeper understanding of their microenvironmental niche has been considered important to provide understanding and solutions to cancer eradication. However, as the CSC niche is composed of a wide variety of biological and physicochemical factors, the development of multidisciplinary tools that recapitulate their complex features is indispensable. Synthetic polymers have been studied as attractive biomaterials due to their tunable biofunctionalities, while hydrogelation technique further renders upon them a diversity of physical properties, making them an attractive tool for analysis of the CSC niche. METHODS To develop innovative materials that recapitulate the CSC niche in pancreatic cancers, we performed polymer microarray analysis to identify niche-mimicking scaffolds that preferentially supported the growth of CSCs. The niche-mimicking activity of the identified polymers was further optimized by polyethylene glycol (PEG)-based hydrogelation. To reveal the biological mechanisms behind the activity of the optimized hydrogels towards CSCs, proteins binding onto the hydrogel were analyzed by liquid chromatography with tandem mass spectrometry (LC-MS/MS), and the potential therapeutic targets were validated by looking at gene expression and patients' outcome in the TCGA database. RESULTS PA531, a heteropolymer composed of 2-methoxyethyl methacrylate (MEMA) and 2-(diethylamino)ethyl methacrylate (DEAEMA) (5.5:4.5) that specifically supports the growth and maintenance of CSCs was identified by polymer microarray screening using the human PAAD cell line KLM1. The polymer PA531 was converted into five hydrogels (PA531-HG1 to HG5) and developed to give an optimized scaffold with the highest CSC niche-mimicking activities. From this polymer that recapitulated CSC binding and control, the proteins fetuin-B and angiotensinogen were identified as candidate target molecules with clinical significance due to the correlation between gene expression levels and prognosis in PAAD patients and the proteins associated with the niche-mimicking polymer. CONCLUSION This study screened for biofunctional polymers suitable for recapitulation of the pancreatic CSC niche and one hydrogel with high niche-mimicking abilities was successfully fabricated. Two soluble factors with clinical significance were identified as potential candidates for biomarkers and therapeutic targets in pancreatic cancers. Such a biomaterial-based approach could be a new platform in drug discovery and therapy development against CSCs, via targeting of their niche.
Collapse
Affiliation(s)
- Yoshitaka Murota
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8510, Japan
| | - Mariko Nagane
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8510, Japan
| | - Mei Wu
- School of Chemistry, University of Edinburgh, Joseph Black Building, West Mains Road, Edinburgh, EH9 3FJ, UK
| | - Mithun Santra
- School of Chemistry, University of Edinburgh, Joseph Black Building, West Mains Road, Edinburgh, EH9 3FJ, UK
| | - Seshasailam Venkateswaran
- School of Chemistry, University of Edinburgh, Joseph Black Building, West Mains Road, Edinburgh, EH9 3FJ, UK
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Mark Bradley
- School of Chemistry, University of Edinburgh, Joseph Black Building, West Mains Road, Edinburgh, EH9 3FJ, UK
| | - Tetsuya Taga
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8510, Japan
| | - Kouichi Tabu
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
9
|
Jeong HY, An HJ, Sung MJ, Ha MH, Lee YH, Yang DH, Yang TY, Han D, Lee SY. Proteomic profiling of protein expression changes after 3 months-exercise in ESRD patients on hemodialysis. BMC Nephrol 2023; 24:102. [PMID: 37085769 PMCID: PMC10122383 DOI: 10.1186/s12882-023-03146-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: 12/14/2022] [Accepted: 03/31/2023] [Indexed: 04/23/2023] Open
Abstract
The prevalence of chronic kidney disease (CKD) is steadily increasing, and it is a global health burden. Exercise has been suggested to improve physical activity and the quality of life in patients with CKD, eventually reducing mortality. This study investigated the change in physical performance after exercise in dialysis-dependent patients with CKD and analyzed differentially expressed proteins before and after the exercise. Plasma samples were collected at enrollment and after 3 months of exercise. Liquid chromatography with tandem mass spectrometry analysis and data-independent acquisition results were analyzed to determine the significantly regulated proteins. A total of 37 patients on dialysis were recruited, and 16 were randomized to exercise for 3 months. The hand grip strength and the walking speed significantly improved in the exercise group. Proteome analysis revealed 60 significantly expressed proteins after 3 months of exercise. In the protein functional analysis, the significantly expressed proteins were involved in the immune response. Also, some of the key significantly expressed proteins [(M Matrix metallopeptidase 9 (MMP-9), Activin A Receptor Type 1B (ACVR1B), Fetuin B (FETUB)] were validated via an enzyme-linked immunosorbent assay. Our results showed that exercise in dialysis-dependent patients with CKD could improve their physical performance. These results indicated that this beneficial effect of exercise in these populations could be associated with immune response.
Collapse
Affiliation(s)
- Hye Yun Jeong
- Division of Nephrology, Department of Internal Medicine, CHA University School of Medicine, CHA Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam-si, 13496, Republic of Korea
| | - Hyun-Ju An
- Division of Nephrology, Department of Internal Medicine, CHA University School of Medicine, CHA Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam-si, 13496, Republic of Korea
| | - Min Ji Sung
- Division of Nephrology, Department of Internal Medicine, CHA University School of Medicine, CHA Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam-si, 13496, Republic of Korea
| | - Min Heui Ha
- Division of Nephrology, Department of Internal Medicine, CHA University School of Medicine, CHA Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam-si, 13496, Republic of Korea
| | - Yu Ho Lee
- Division of Nephrology, Department of Internal Medicine, CHA University School of Medicine, CHA Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam-si, 13496, Republic of Korea
| | - Dong Ho Yang
- Division of Nephrology, Department of Internal Medicine, CHA University School of Medicine, CHA Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam-si, 13496, Republic of Korea
| | - Tae Young Yang
- Division of Nephrology, Department of Internal Medicine, CHA University School of Medicine, CHA Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam-si, 13496, Republic of Korea
| | - Dohyun Han
- Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, 101 Daehak-ro, Seoul, 03080, Republic of Korea.
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| | - So-Young Lee
- Division of Nephrology, Department of Internal Medicine, CHA University School of Medicine, CHA Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam-si, 13496, Republic of Korea.
| |
Collapse
|
10
|
Paul Owens E, Grania Healy H, Andrew Vesey D, Elizabeth Hoy W, Carolyn Gobe G. Targeted biomarkers of progression in chronic kidney disease. Clin Chim Acta 2022; 536:18-28. [PMID: 36041551 DOI: 10.1016/j.cca.2022.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Chronic kidney disease (CKD) is an increasingly significant health issue worldwide. Early stages of CKD can be asymptomatic and disease trajectory difficult to predict. Not everyone with CKD progresses to kidney failure, where kidney replacement therapy is the only life-sustaining therapy. Predicting which patients will progress to kidney failure would allow better use of targeted treatments and more effective allocation of health resources. Current diagnostic tests to identify patients with progressive disease perform poorly but there is a suite of new and emerging predictive biomarkers with great clinical promise. METHODS This narrative review describes new and emerging biomarkers of pathophysiologic processes of CKD development and progression, accessible in blood or urine liquid biopsies. Biomarkers were selected based on their reported pathobiological functions in kidney injury, inflammation, oxidative stress, repair and fibrosis. Biomarker function and evidence of involvement in CKD development and progression are reported. CONCLUSION Many biomarkers reviewed here have received little attention to date, perhaps because of conflicting conclusions of their utility in CKD. The functional roles of the selected biomarkers in the underlying pathobiology of progression of CKD are a powerful rationale for advancing and validating these molecules as prognosticators and predictors of CKD trajectory.
Collapse
Affiliation(s)
- Evan Paul Owens
- NHMRC CKD CRE (CKD.QLD), The University of Queensland, Brisbane 4072, Australia; Faculty of Medicine, The University of Queensland, Brisbane 4072, Australia; Kidney Disease Research Collaborative, Translational Research Institute, Brisbane 4102, Australia
| | - Helen Grania Healy
- NHMRC CKD CRE (CKD.QLD), The University of Queensland, Brisbane 4072, Australia; Faculty of Medicine, The University of Queensland, Brisbane 4072, Australia; Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane 4029, Australia
| | - David Andrew Vesey
- NHMRC CKD CRE (CKD.QLD), The University of Queensland, Brisbane 4072, Australia; Faculty of Medicine, The University of Queensland, Brisbane 4072, Australia
| | - Wendy Elizabeth Hoy
- NHMRC CKD CRE (CKD.QLD), The University of Queensland, Brisbane 4072, Australia; Faculty of Medicine, The University of Queensland, Brisbane 4072, Australia; Centre for Chronic Disease, The University of Queensland, Brisbane 4072, Australia
| | - Glenda Carolyn Gobe
- NHMRC CKD CRE (CKD.QLD), The University of Queensland, Brisbane 4072, Australia; Faculty of Medicine, The University of Queensland, Brisbane 4072, Australia; Kidney Disease Research Collaborative, Translational Research Institute, Brisbane 4102, Australia.
| |
Collapse
|
11
|
Serum Metabolomics and Proteomics to Study the Antihypertensive Effect of Protein Extracts from Tenebrio molitor. Nutrients 2022; 14:nu14163288. [PMID: 36014793 PMCID: PMC9413627 DOI: 10.3390/nu14163288] [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: 07/08/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 12/04/2022] Open
Abstract
Hypertension is the leading risk factor for premature death worldwide and significantly contributes to the development of all major cardiovascular disease events. The management of high blood pressure includes lifestyle changes and treatment with antihypertensive drugs. Recently, it was demonstrated that a diet supplemented with Tenebrio molitor (TM) extracts is useful in the management of numerous pathologies, including hypertension. This study is aimed at unveiling the underlying mechanism and the molecular targets of intervention of TM dietary supplementation in hypertension treatment by means of proteomics and metabolomics techniques based on liquid chromatography coupled with high-resolution mass spectrometry. We demonstrate that serum proteome and metabolome of spontaneously hypertensive rats are severely altered with respect to their normotensive counterparts. Additionally, our results reveal that a diet enriched with TM extracts restores the expression of 15 metabolites and 17 proteins mainly involved in biological pathways associated with blood pressure maintenance, such as the renin–angiotensin and kallikrein–kinin systems, serin protease inhibitors, reactive oxygen scavenging, and lipid peroxidation. This study provides novel insights into the molecular pathways that may underlie the beneficial effects of TM, thus corroborating that TM could be proposed as a helpful functional food supplement in the treatment of hypertension.
Collapse
|
12
|
Hou J, Deng Q, Liu S, Qiu X, Deng X, Zhong W, Zhong Z. Plasma Proteome Profiling of Patients With In-stent Restenosis by Tandem Mass Tag-Based Quantitative Proteomics Approach. Front Cardiovasc Med 2022; 9:793405. [PMID: 35265678 PMCID: PMC8899613 DOI: 10.3389/fcvm.2022.793405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Background Despite the widespread application of new drug-eluting stents, a considerable portion of patients experience in-stent restenosis (ISR). To date, the pathophysiologic mechanisms of ISR remain poorly understood. Methods In this study, we collected plasma samples from ISR patients (n = 29) and non-ISR patients (n = 36) after drug-eluting stent implantation, as well as from healthy controls (HCs) (n = 32). Our goal was to investigate differences in plasma protein profiles using tandem mass tag (TMT) labeling coupled with liquid chromatography and tandem mass spectrometry. The proteomic data were validated by enzyme-linked immunosorbent assay (ELISA). Bioinformatic analyses were conducted to analyze potential pathways and protein-protein interaction (PPI) involved in ISR. Results A total of 1,696 proteins were identified, of which 278 differed in protein abundance between non-ISR and HCs, 497 between ISR and HCs, and 387 between ISR and non-ISR, respectively. Bioinformatic analyses, including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and PPI, further demonstrated that differentially abundant proteins between ISR and non-ISR are involved in several crucial biological processes and signaling pathways, such as focal adhesion, platelet activation, Rap1 signaling, regulation of actin cytoskeleton, and cholesterol metabolism. Among the identified differentially abundant proteins in ISR, 170 were increased in abundance relative to both non-ISR patients and HCs. Some of these proteins were identified to have critical functions for atherosclerosis development and might be involved in ISR pathology. Among these proteins, 3 proteins with increased abundance including fetuin-B, apolipoprotein C-III (APOC3), and cholesteryl ester transfer protein (CETP) were confirmed by ELISA. Conclusions This is the first study provided a comprehensive proteomic profile to understand ISR pathology, which may help identify early diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jingyuan Hou
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China
| | - Qiaoting Deng
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China
| | - Sudong Liu
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China
| | - Xiaohong Qiu
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China
| | - Xunwei Deng
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China
- Guangdong Provincial Engineering and Technological Research Center for Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, China
| | - Wei Zhong
- Center for Cardiovascular Diseases, Meizhou People's Hospital, Meizhou, China
| | - Zhixiong Zhong
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China
- *Correspondence: Zhixiong Zhong
| |
Collapse
|
13
|
Li L, Spranger L, Stobäus N, Beer F, Decker AM, Wernicke C, Brachs S, Brachs M, Spranger J, Mai K. Fetuin-B, a potential link of liver-adipose tissue cross talk during diet-induced weight loss-weight maintenance. Nutr Diabetes 2021; 11:31. [PMID: 34611132 PMCID: PMC8492646 DOI: 10.1038/s41387-021-00174-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/28/2021] [Accepted: 07/09/2021] [Indexed: 11/29/2022] Open
Abstract
Background/objectives Numerous hepatokines are involved in inter-organ cross talk regulating tissue-specific insulin sensitivity. Adipose tissue lipolysis represents a crucial element of adipose insulin sensitivity and is substantially involved in long-term body weight regulation after dietary weight loss. Thus, we aimed to analyze the impact of the hepatokine Fetuin-B in the context of weight loss induced short- and long-term modulation of adipose insulin sensitivity. Subjects/methods 143 subjects (age > 18; BMI ≥ 27 kg/m2) were analyzed before (T-3) and after (T0) a standardized 12-week dietary weight reduction program. Afterward, subjects were randomized to a 12-month lifestyle intervention or a control group. After 12 months (T12) no further intervention was performed until 6 months later (T18) (Maintain-Adults trial). Tissue-specific insulin sensitivity was estimated by HOMA-IR (predominantly liver), ISIClamp (predominantly skeletal muscle), and free fatty acid suppression during hyperinsulinemic-euglycemic clamp (FFASupp) (predominantly adipose tissue). Fetuin-B was measured at all concomitant time points. Results Circulating Fetuin-B levels correlated significantly with estimates of obesity, hepatic steatosis as well as HOMA-IR, ISIClamp, FFASupp at baseline. Fetuin-B decreased during dietary weight loss (4.2 (3.5–4.9) vs. 3.8 (3.2–4.6) µg/ml; p = 2.1 × 10−5). This change was associated with concomitant improvement of HOMA-IR (r = 0.222; p = 0.008) and FFASupp (r = −0.210; p = 0.013), suggesting a particular relationship to hepatic and adipose tissue insulin sensitivity. Weight loss induced improvements of insulin resistance were almost completely preserved until months 12 and 18 and most interestingly, the short and long-term improvement of FFASupp was partially predicted by baseline level of Fetuin-B. Conclusions Our data suggest that Fetuin-B might be a potential mediator of liver-adipose cross talk involved in short- and long-term regulation of adipose insulin sensitivity, especially in the context of diet-induced weight changes. Trial registration ClinicalTrials.gov number: NCT00850629, https://clinicaltrials.gov/ct2/show/NCT00850629, date of registration: February 25, 2009.
Collapse
Affiliation(s)
- Linna Li
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Clinical Research Unit, 10117, Berlin, Germany
| | - Leonard Spranger
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany
| | - Nicole Stobäus
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Clinical Research Unit, 10117, Berlin, Germany
| | - Finja Beer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany
| | - Anne-Marie Decker
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany
| | - Charlotte Wernicke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Clinical Research Unit, 10117, Berlin, Germany
| | - Sebastian Brachs
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Maria Brachs
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany
| | - Joachim Spranger
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Knut Mai
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany. .,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Clinical Research Unit, 10117, Berlin, Germany. .,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
14
|
Bahniuk MS, Ortega VA, Alshememry AK, Stafford JL, Goss GG, Unsworth LD. Effect of amino acid composition of elastin-like polypeptide nanoparticles on nonspecific protein adsorption, macrophage cell viability and phagocytosis. Biopolymers 2021; 112:e23468. [PMID: 34363693 DOI: 10.1002/bip.23468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 02/05/2023]
Abstract
Development of elastin-like polypeptide (ELP) biomaterials is widespread, but information critical for clinical deployment is limited, with biocompatibility studies focused on a narrow cross-section of ELP sequences. Macrophages can impair biomaterial systems by degrading or isolating the biomaterial and by activating additional immune functions. Their phagocytic response will reveal early immune biocompatibility of ELP nanoparticles (NPs). This study examines that response, induced by the adsorbed protein corona, as a function of ELP guest amino acid, chain length and NP diameter. The breadth of proteins adsorbed to ELP NPs varied, with valine-containing ELP NPs adsorbing fewer types of proteins than leucine-containing constructs. Particle diameter was also a factor, with smaller leucine-containing ELP NPs adsorbing the broadest range of proteins. Macrophage viability was unaffected by the ELP NPs, and their phagocytic capabilities were unimpeded except when incubated with a 500 nm valine-containing 40-mer. This NP significantly decreased the phagocytic capacity of macrophages relative to the control and to a corresponding 500 nm leucine-containing 40-mer. NP size and the proportion of opsonin to dysopsonin proteins likely influenced this outcome. These results suggest that certain combinations of ELP sequence and particle size can result in an adsorbed protein corona, which may hinder macrophage function.
Collapse
Affiliation(s)
- Markian S Bahniuk
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Van A Ortega
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Abdullah K Alshememry
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - James L Stafford
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Greg G Goss
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Larry D Unsworth
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
15
|
Xing W, Tan Y, Li K, Tian P, Tian F, Zhang H. Upregulated hepatokine fetuin B aggravates myocardial ischemia/reperfusion injury through inhibiting insulin signaling in diabetic mice. J Mol Cell Cardiol 2021; 151:163-172. [PMID: 32147518 DOI: 10.1016/j.yjmcc.2020.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/11/2020] [Accepted: 03/03/2020] [Indexed: 12/27/2022]
Abstract
Patients with type 2 diabetes mellitus (T2DM) are more susceptible to acute myocardial ischemia/reperfusion (MI/R) injury. However, the mechanism remains largely elusive. Clinical observation showed that high levels of hepatokine fetuin-B (FetB) in plasma are significantly associated with both diabetes and coronary artery diseases. This study was aimed to determine whether FetB mostly derived from liver exacerbates MI/R-induced injury and the underlying mechanisms in T2DM. Mice were given high-fat diet and streptozotocin to induce T2DM model and subjected to 30 min MI followed by reperfusion. Diabetes caused increased hepatic FetB expression and greater myocardial injury as evidenced by increased apoptosis and myocardial enzymes release following MI/R. In T2DM hearts, insulin-induced phosphorylations of insulin receptor substrate 1 at Tyr608 site and Akt at Ser473 site and glucose transporter 4 membrane translocation were markedly reduced. Interaction between FetB and insulin receptor-β subunit (IRβ) was enhanced assessed by immunoprecipitation analysis. More importantly, FetB knockdown via AAV9 alleviated MI/R injury and improved cardiac insulin-induced signaling in T2DM mice. Conversely, upregulation of FetB in normal mice caused exacerbated MI/R injury and impairment of insulin-mediated signaling. In cultured neonatal mouse cardiomyocytes, incubation of FetB significantly reduced tyrosine kinase activity of IR and insulin-induced glucose uptake, and increased hypoxia/reoxygenation-induced apoptosis. Furthermore, FoxO1 knockdown by siRNA suppressed FetB expressions in hepatocytes treated with palmitic acid. In conclusion, upregulated FetB in diabetic liver contributes to increased MI/R injury and cardiac dysfunction via directly interacting with IRβ and consequently impairing cardiac insulin signaling.
Collapse
Affiliation(s)
- Wenjuan Xing
- Department of Aerospace Medicine, Fourth Military Medical University, Xi'an, China; State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yanzhen Tan
- Department of Aerospace Medicine, Fourth Military Medical University, Xi'an, China; Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kaifeng Li
- Teaching Experiment Center, Fourth Military Medical University, Xi'an, China
| | - Pei Tian
- Department of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Fei Tian
- Teaching Experiment Center, Fourth Military Medical University, Xi'an, China.
| | - Haifeng Zhang
- Teaching Experiment Center, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
16
|
Olkowicz M, Czyzynska-Cichon I, Szupryczynska N, Kostogrys RB, Kochan Z, Debski J, Dadlez M, Chlopicki S, Smolenski RT. Multi-omic signatures of atherogenic dyslipidaemia: pre-clinical target identification and validation in humans. J Transl Med 2021; 19:6. [PMID: 33407555 PMCID: PMC7789501 DOI: 10.1186/s12967-020-02663-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Dyslipidaemia is a major risk factor for atherosclerosis and cardiovascular diseases. The molecular mechanisms that translate dyslipidaemia into atherogenesis and reliable markers of its progression are yet to be fully elucidated. To address this issue, we conducted a comprehensive metabolomic and proteomic analysis in an experimental model of dyslipidaemia and in patients with familial hypercholesterolemia (FH). METHODS Liquid chromatography/mass spectrometry (LC/MS) and immunoassays were used to find out blood alterations at metabolite and protein levels in dyslipidaemic ApoE-/-/LDLR-/- mice and in FH patients to evaluate their human relevance. RESULTS We identified 15 metabolites (inhibitors and substrates of nitric oxide synthase (NOS), low-molecular-weight antioxidants (glutamine, taurine), homocysteine, methionine, 1-methylnicotinamide, alanine and hydroxyproline) and 9 proteins (C-reactive protein, proprotein convertase subtilisin/kexin type 9, apolipoprotein C-III, soluble intercellular adhesion molecule-1, angiotensinogen, paraoxonase-1, fetuin-B, vitamin K-dependent protein S and biglycan) that differentiated FH patients from healthy controls. Most of these changes were consistently found in dyslipidaemic mice and were further amplified if mice were fed an atherogenic (Western or low-carbohydrate, high-protein) diet. CONCLUSIONS The alterations highlighted the involvement of an immune-inflammatory response system, oxidative stress, hyper-coagulation and impairment in the vascular function/regenerative capacity in response to dyslipidaemia that may also be directly engaged in development of atherosclerosis. Our study further identified potential biomarkers for an increased risk of atherosclerosis that may aid in clinical diagnosis or in the personalized treatment.
Collapse
Affiliation(s)
- Mariola Olkowicz
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 1 Debinki St, 80-211, Gdansk, Poland. .,Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego St., 30-348, Krakow, Poland.
| | - Izabela Czyzynska-Cichon
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego St., 30-348, Krakow, Poland
| | - Natalia Szupryczynska
- Department of Nutritional Biochemistry, Faculty of Health Sciences, Medical University of Gdansk, 7 Debinki St., 80-211, Gdansk, Poland
| | - Renata B Kostogrys
- Department of Human Nutrition and Dietetics, Faculty of Food Technology, University of Agriculture in Krakow, 122 Balicka St., 30-149, Krakow, Poland
| | - Zdzislaw Kochan
- Department of Nutritional Biochemistry, Faculty of Health Sciences, Medical University of Gdansk, 7 Debinki St., 80-211, Gdansk, Poland
| | - Janusz Debski
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 5a Pawinskiego St., 02-106, Warsaw, Poland
| | - Michal Dadlez
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 5a Pawinskiego St., 02-106, Warsaw, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego St., 30-348, Krakow, Poland.,Chair of Pharmacology, Jagiellonian University Medical College, 16 Grzegorzecka St., 31-531, Krakow, Poland
| | - Ryszard T Smolenski
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 1 Debinki St, 80-211, Gdansk, Poland.
| |
Collapse
|
17
|
Zhan K, Liu R, Tong H, Gao S, Yang G, Hossain A, Li T, He W. Fetuin B overexpression suppresses proliferation, migration, and invasion in prostate cancer by inhibiting the PI3K/AKT signaling pathway. Biomed Pharmacother 2020; 131:110689. [PMID: 32892030 DOI: 10.1016/j.biopha.2020.110689] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022] Open
Abstract
Fetuin B (FETUB) is a glycoprotein that is a member of the cysteine protease inhibitor family, and it is associated with cancer. However, the role of FETUB in prostate carcinogenesis is unknown. In this study, we overexpressed FETUB in prostate cancer cells by using lentivirus and then studied the impacts on cell apoptosis, migration and invasion. We found that apoptosis was increased and the migration and invasion of prostate cancer cells were significantly inhibited after overexpression. Then, we performed experiments in vivo and found that there were fewer tumors in the overexpression groups than in the control groups. In addition, we demonstrated that overexpression of FETUB inactivates the PI3K/AKT signaling pathway. Rescue assays revealed that intervention of 740Y-P reversed the anti-tumor effect of FETUB on prostate cancer cells. Taken together, our results revealed that FETUB may act as a novel regulator to promote apoptosis and inhibit the migration and invasion of prostate cancer cells and that FETUB is related to the inactivation of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Kai Zhan
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Rui Liu
- Department of Oncology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Hang Tong
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Shun Gao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Guang Yang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Arman Hossain
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Tinghao Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
18
|
Jeong H, Vacanti NM. Systemic vitamin intake impacting tissue proteomes. Nutr Metab (Lond) 2020; 17:73. [PMID: 32863845 PMCID: PMC7449053 DOI: 10.1186/s12986-020-00491-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
The kinetics and localization of the reactions of metabolism are coordinated by the enzymes that catalyze them. These enzymes are controlled via a myriad of mechanisms including inhibition/activation by metabolites, compartmentalization, thermodynamics, and nutrient sensing-based transcriptional or post-translational regulation; all of which are influenced as a network by the activities of metabolic enzymes and have downstream potential to exert direct or indirect control over protein abundances. Considering many of these enzymes are active only when one or more vitamin cofactors are present; the availability of vitamin cofactors likely yields a systems-influence over tissue proteomes. Furthermore, vitamins may influence protein abundances as nuclear receptor agonists, antioxidants, substrates for post-translational modifications, molecular signal transducers, and regulators of electrolyte homeostasis. Herein, studies of vitamin intake are explored for their contribution to unraveling vitamin influence over protein expression. As a body of work, these studies establish vitamin intake as a regulator of protein abundance; with the most powerful demonstrations reporting regulation of proteins directly related to the vitamin of interest. However, as a whole, the field has not kept pace with advances in proteomic platforms and analytical methodologies, and has not moved to validate mechanisms of regulation or potential for clinical application.
Collapse
Affiliation(s)
- Heesoo Jeong
- Division of Nutritional Sciences, Cornell University, Ithaca, NY USA
| | | |
Collapse
|
19
|
Olausson N, Mobarrez F, Zubarev R, Chernobrovkin A, Rutishauser D, Bremme K, Westerlund E, Hovatta O, Wallén H, Henriksson P. Changes in the plasma microvesicle proteome during the ovarian hyperstimulation phase of assisted reproductive technology. Sci Rep 2020; 10:13645. [PMID: 32788624 PMCID: PMC7423945 DOI: 10.1038/s41598-020-70541-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 07/22/2020] [Indexed: 11/26/2022] Open
Abstract
The incidence of pulmonary and venous thromboembolism is increased during the first trimester of pregnancies after assisted reproductive technology (ART) compared to spontaneous conception. We previously found that haemostatic plasma variables changed but within normal limits during controlled ovarian hyperstimulation (COH) concomitant with a major increase in plasma microvesicles (MVs) and markers indicating cell activation. We now explored the proteome of these MVs. Thirty-one women undergoing ART were blood sampled at down-regulation (DR) of oestrogen and at high level stimulation (HLS) with its 10-100-fold increased oestrogen level. Samples were analysed by liquid chromatography and tandem mass spectrometry to identify and quantify the proteome. We identified 306 proteins in the MVs and 72 had changed significantly at HLS compared to DR and more than 20% of them were associated with haemostasis. Thus, proteins related to both haemostasis and complement activation altered in plasma MVs in parallel with MV activation during COH. This needs to be further explored in the clinical context.
Collapse
Affiliation(s)
- Nina Olausson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, 18288, Stockholm, Sweden.
| | - Fariborz Mobarrez
- Department of Medical Sciences, Uppsala University, 75185, Uppsala, Sweden
| | - Roman Zubarev
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Alexey Chernobrovkin
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Dorothea Rutishauser
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Katarina Bremme
- Department of Women's and Children's Health, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Eli Westerlund
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, 18288, Stockholm, Sweden
| | - Outi Hovatta
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Håkan Wallén
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, 18288, Stockholm, Sweden
| | - Peter Henriksson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, 18288, Stockholm, Sweden
| |
Collapse
|
20
|
Maglinger B, Frank JA, McLouth CJ, Trout AL, Roberts JM, Grupke S, Turchan-Cholewo J, Stowe AM, Fraser JF, Pennypacker KR. Proteomic changes in intracranial blood during human ischemic stroke. J Neurointerv Surg 2020; 13:395-399. [PMID: 32641418 PMCID: PMC7982920 DOI: 10.1136/neurintsurg-2020-016118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 01/29/2023]
Abstract
Background Since 2015, mechanical thrombectomy has been the standard treatment for emergent large vessel occlusion ischemic stroke. Objective To investigate, using the previously published Blood and Clot Thrombectomy Registry and Collaboration (BACTRAC) protocol (clinicaltrials.gov NCT03153683), how the protein expression of a patient’s intracranial blood during ischemic stroke compares with the protein expression of their systemic arterial blood in order to better understand and treat stroke. Methods Plasma samples from 25 subjects underwent proteomic analysis, where intracranial protein expression was compared with systemic protein levels. Data including sex, comorbidities, infarct volume, and infarct time were included for each subject. Results A majority of important proteins had a lower expression in intracranial blood than in systemic arterial blood. Proteins with the most significant changes in expression were: endopeptidase at −0.26 (p<0.0001), phospholipid transfer protein (PLTP) at −0.26 (p=0.0005), uromodulin (UMOD) at −0.14 (p=0.002), ficolin-2 (FCN2) at −0.46 (p=0.005), C-C motif chemokine 19 (CCL19) at −0.51 (p<0.0001), C-C motif chemokine 20 (CCL20) at −0.40 (p<0.0001), fibroblast growth factor 21 at −0.37 (p=0.0002), and C-C motif chemokine (CCL23) at −0.43 (p=0.0003). Conclusions Evaluation of proteomic changes in the intravascular space of a cerebral infarct in progress in human subjects suggested that changes in proteins such PLTP, fetuin-B (FETUB), and FCN2 may be involved in atherosclerotic changes, and chemokines such as CCL23 are known to play a role in the Th2 autoimmune response. These data provide a scientific springboard for identifying clinically relevant biomarkers for diagnosis/prognosis, and targets for much needed neuroprotective/neuroreparative pharmacotherapies.
Collapse
Affiliation(s)
- Benton Maglinger
- Department of Neurology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Jacqueline A Frank
- Department of Neurology; Department of Neuroscience; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | | | - Amanda L Trout
- Department of Neurology; Department of Neuroscience; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Jill Marie Roberts
- Department of Neurosurgery; Department of Neuroscience; Department of Neurology; Department of Radiology; Center for Advanced Translational Stroke Science, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Stephen Grupke
- Department of Neurosurgery; Department of Radiology; Department of Neurology, University of Kentucky, Lexington, Kentucky, USA
| | - Jadwiga Turchan-Cholewo
- Department of Neurology; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Ann M Stowe
- Department of Neurology; Department of Neuroscience; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Justin F Fraser
- Department of Neurosurgery; Department of Radiology; Department of Neuroscience; Department of Neurology; Department of Radiology; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Keith R Pennypacker
- Department of Neurology; Department of Neuroscience; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
21
|
Jung SH, Lee D, Jin H, Lee HM, Ko HM, Lee KJ, Kim SJ, Ryu Y, Choi WS, Kim B, Won KJ. Fetuin-B regulates vascular plaque rupture via TGF-β receptor-mediated Smad pathway in vascular smooth muscle cells. Pflugers Arch 2020; 472:571-581. [PMID: 32382986 DOI: 10.1007/s00424-020-02385-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 11/28/2022]
Abstract
Fetuin-B is a serum protein linked to the regulation of physiological or pathophysiological events such as fertility, energy metabolism, and liver disease. Recently, fetuin-B has been reported to be involved in the modulation of the rupture of atherosclerotic plaques associated with acute myocardial infarction. However, the exact mechanism involved in the modulation of atherosclerotic plaque rupture event by fetuin-B is not fully elucidated yet. In the present study, we investigated whether fetuin-B could influence atherosclerotic plaque rupture through vascular smooth muscle cells (VSMCs). Immunoprecipitation assay using membrane proteins from VSMCs revealed that fetuin-B tightly bound to transforming growth factor-β receptor (TGF-βR). Fetuin-B treatment elevated TGF-βR signals (e.g., phosphorylation of Smad2 and Smad3) in VSMCs. Fetuin-B also stimulated nuclear translocation of phosphorylated Smads. Phosphorylation of Smad and its nuclear translocation by treatment with fetuin-B were inhibited in VSMCs by treatment with SB431542, a selective inhibitor of TGF-βR. Fetuin-B enhanced expression levels of plasminogen activator inhibitor-1 (PAI-1) and matrix metalloproteinase-2 (MMP-2) in VSMCs through its epigenetic modification including recruitments of both histone deacetylase 1 and RNA polymerase II. These epigenetic alterations in VSMCs were also inhibited by treatment with SB431542. In vivo administration of fetuin-B protein increased expression levels of PAI-1 and MMP-2 in the vascular plaque. However, these increases in expression were inhibited by the administration of SB43154. These results indicate that fetuin-B may modulate vascular plaque rupture by promoting expression of PAI-1 and MMP-2 in VSMCs via TGF-βR-mediated Smad pathway.
Collapse
Affiliation(s)
- Seung Hyo Jung
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea
| | - Donghyen Lee
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea
| | - Hengzhe Jin
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea
| | - Hwan Myung Lee
- Department of Cosmetic Science, College of Life and Health Science, Hoseo University, Asan, 31499, South Korea
| | - Hyun Myung Ko
- Department of Life Sciences, College of Science and Technology, Woosuk University, Jincheon, 27841, South Korea
| | - Kyung-Jin Lee
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea
| | - Su Jung Kim
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea
| | - Yunkyoung Ryu
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea
| | - Wahn Soo Choi
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea
| | - Bokyung Kim
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea
| | - Kyung-Jong Won
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, South Korea.
| |
Collapse
|
22
|
Serum and follicular fluid fetuin-B levels are correlated with fertilization rates in conventional IVF cycles. J Assist Reprod Genet 2019; 36:1101-1107. [PMID: 31073723 DOI: 10.1007/s10815-019-01454-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 04/16/2019] [Indexed: 12/22/2022] Open
Abstract
PURPOSE To investigate the relationship between serum/follicular fluid fetuin-B levels and fertilization outcomes in conventional IVF cycles. METHODS A prospective cohort study of conventional IVF treatments including 78 cycles with low fertilization rates (two pronuclei [2PN] rate < 30%; LF group) and 104 cycles performed during the same period with 2PN rate > 70% (high fertilization group, HF). To calculate the required sample size, a two-sample t test power analysis was applied to data from our pilot study, using PASS 11.0 software. Fetuin-B was measured using a commercial sandwich enzyme-linked immunosorbent assay. RESULTS Serum fetuin-B and follicular fluid fetuin-B were positively correlated (r = 0.703, P < 0.001). Compared to the HF group, the LF group had significantly lower levels of fetuin-B, both in serum (5.81 ± 1.53 vs. 7.19 ± 1.42, P < 0.001) and follicular fluid (5.06 ± 1.29 vs. 6.16 ± 1.52, P < 0.001). The serum fetuin-B level from cycles with polypronuclear (PPN) zygotes was significantly lower when compared to cycles without PPN zygotes (6.82 ± 1.65 vs. 6.10 ± 1.43, P = 0.006). However, serum fetuin-B level was not correlated with preimplantation embryo development or clinical pregnancy. CONCLUSION Serum fetuin-B level is correlated with fertilization rate in conventional IVF and it may be used as a predictive marker of fertilization in IVF treatment.
Collapse
|
23
|
Montgomery MK, De Nardo W, Watt MJ. Impact of Lipotoxicity on Tissue "Cross Talk" and Metabolic Regulation. Physiology (Bethesda) 2019; 34:134-149. [PMID: 30724128 DOI: 10.1152/physiol.00037.2018] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Obesity-associated comorbidities include non-alcoholic fatty liver disease, Type 2 diabetes, and cardiovascular disease. These diseases are associated with accumulation of lipids in non-adipose tissues, which can impact many intracellular cellular signaling pathways and functions that have been broadly defined as "lipotoxic." This review moves beyond understanding intracellular lipotoxic outcomes and outlines the consequences of lipotoxicity on protein secretion and inter-tissue "cross talk," and the impact this exerts on systemic metabolism.
Collapse
Affiliation(s)
| | - William De Nardo
- Department of Physiology, The University of Melbourne , Melbourne, Victoria , Australia
| | - Matthew J Watt
- Department of Physiology, The University of Melbourne , Melbourne, Victoria , Australia
| |
Collapse
|
24
|
Martínez PJ, Baldán-Martín M, López JA, Martín-Lorenzo M, Santiago-Hernández A, Agudiez M, Cabrera M, Calvo E, Vázquez J, Ruiz-Hurtado G, Vivanco F, Ruilope LM, Barderas MG, Alvarez-Llamas G. Identification of six cardiovascular risk biomarkers in the young population: A promising tool for early prevention. Atherosclerosis 2019; 282:67-74. [PMID: 30690299 DOI: 10.1016/j.atherosclerosis.2019.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/26/2018] [Accepted: 01/10/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS The predictive value of traditional CV risk calculators is limited. Novel indicators of CVD progression are needed particularly in the young population. The main aim of this study was the identification of a molecular profile with added value to classical CV risk estimation. METHODS Eighty-one subjects (30-50 years) were classified in 3 groups according to their CV risk: healthy subjects; individuals with CV risk factors; and those who had suffered a previous CV event. The urine proteome was quantitatively analyzed and significantly altered proteins were identified between patients' groups, either related to CV risk or established organ damage. Target-MS and ELISA were used for confirmation in independent patients' cohorts. Systems Biology Analysis (SBA) was carried out to identify functional categories behind CVD. RESULTS 4309 proteins were identified, 75 of them differentially expressed. ADX, ECP, FETUB, GDF15, GUAD and NOTCH1 compose a fingerprint positively correlating with lifetime risk estimate (LTR QRISK). Best performance ROC curve was obtained when ECP, GDF15 and GUAD were combined (AUC = 0.96). SBA revealed oxidative stress response, dilated cardiomyopathy, signaling by Wnt and proteasome, as main functional processes related to CV risk. CONCLUSIONS A novel urinary protein signature is shown, which correlates with CV risk estimation in young individuals. Pending further confirmation, this six-protein-panel could help in CV risk assessment.
Collapse
Affiliation(s)
- Paula J Martínez
- Immunoallergy and Proteomics Laboratory, Department of Immunology, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | | | - Juan A López
- Laboratory of Cardiovascular Proteomics CNIC, Madrid, Spain
| | - Marta Martín-Lorenzo
- Immunoallergy and Proteomics Laboratory, Department of Immunology, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - Aránzazu Santiago-Hernández
- Immunoallergy and Proteomics Laboratory, Department of Immunology, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - Marta Agudiez
- Immunoallergy and Proteomics Laboratory, Department of Immunology, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | | | | | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics CNIC, Madrid, Spain
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Instituto de Investigación I+12, Hospital Universitario 12 de Octubre/CIBER-CV, Madrid, Spain
| | - Fernando Vivanco
- Department of Biochemistry and Molecular Biology, I Universidad Complutense, Madrid, Spain
| | - Luis M Ruilope
- Cardiorenal Translational Laboratory, Instituto de Investigación I+12, Hospital Universitario 12 de Octubre/CIBER-CV, Madrid, Spain; Hypertension Unit, Hospital Universitario 12 de Octubre, Madrid, Spain; School of Doctoral Studies and Research, Universidad Europea de Madrid, Madrid, Spain.
| | - María G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos SESCAM, Toledo, Spain
| | - Gloria Alvarez-Llamas
- Immunoallergy and Proteomics Laboratory, Department of Immunology, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain; REDINREN, Madrid, Spain.
| |
Collapse
|
25
|
Rabadi MM, Abdulmahdi W, Nesi L, Jules E, Marghani Y, Sheinin E, Tilzer J, Gupta S, Chen S, Cassimatis ND, Lipphardt M, Kozlowski PB, Ratliff BB. Maternal malnourishment induced upregulation of fetuin-B blunts nephrogenesis in the low birth weight neonate. Dev Biol 2018; 443:78-91. [DOI: 10.1016/j.ydbio.2018.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/21/2018] [Accepted: 09/01/2018] [Indexed: 11/16/2022]
|
26
|
Walker GE, Follenzi A, Bruscaggin V, Manfredi M, Bellone S, Marengo E, Maiuri L, Prodam F, Bona G. Fetuin B links vitamin D deficiency and pediatric obesity: Direct negative regulation by vitamin D. J Steroid Biochem Mol Biol 2018; 182:37-49. [PMID: 29684480 PMCID: PMC6092561 DOI: 10.1016/j.jsbmb.2018.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/30/2018] [Accepted: 04/17/2018] [Indexed: 12/13/2022]
Abstract
Vitamin D (VD) deficiency (VDD) correlates to obesity, with VD a recognized mediator of metabolic diseases. From a previous proteomic study identifying adiponectin as a link between VDD and pediatric obesity, herein we analysed another protein (SSP2301) increased with VDD. A focused 2D-electrophoretic analysis identified 4 corresponding plasma proteins, with one predicted to be fetuin B (FETUB). FETUB was studied due to its emerging role in metabolic diseases and cytogenetic location (3q27.3) with adiponectin. Results were confirmed in obese children, where plasma FETUB was higher with VDD. A direct effect by 1α,25-(OH)2D3 on hepatocellular FETUB synthesis was observed, with a time and dose dependent reduction. Further, we demonstrated the VD-receptor (VDR) is key, with FETUB "released" with VDR silencing. Finally, VD supplementation (6weeks) to juvenile mice fed a standard diet, reduced plasma FETUB. Only at 22weeks did liver FETUB correspond to plasma FETUB, highlighting the contribution of other VD-responsive tissues. Overall, FETUB is a key protein linking VDD to pediatric obesity. With an emerging role in metabolic diseases, we demonstrate that VD/VDR directly regulate FETUB.
Collapse
Affiliation(s)
- Gillian E Walker
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.
| | - Antonia Follenzi
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | | | - Marcello Manfredi
- Isalit S.R.L., Department of Science Innovation and Technology, Università del Piemonte Orientale, Novara, Italy
| | - Simonetta Bellone
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy; Division of Pediatrics, Università del Piemonte Orientale, Novara, Italy
| | - Emilio Marengo
- Isalit S.R.L., Department of Science Innovation and Technology, Università del Piemonte Orientale, Novara, Italy
| | - Luigi Maiuri
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy; Division of Pediatrics, Università del Piemonte Orientale, Novara, Italy
| | - Flavia Prodam
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy; Division of Pediatrics, Università del Piemonte Orientale, Novara, Italy
| | - Gianni Bona
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy; Division of Pediatrics, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
27
|
Borlak J, Länger F, Chatterji B. Serum proteome mapping of EGF transgenic mice reveal mechanistic biomarkers of lung cancer precursor lesions with clinical significance for human adenocarcinomas. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3122-3144. [PMID: 29960043 DOI: 10.1016/j.bbadis.2018.06.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/12/2018] [Accepted: 06/25/2018] [Indexed: 12/18/2022]
Abstract
Atypical adenomatous hyperplasia (AAH) of the lung is a pre-invasive lesion (PL) with high risk of progression to lung cancer (LC). However, the pathways involved are uncertain. We searched for novel mechanistic biomarkers of AAH in an EGF transgenic disease model of lung cancer. Disease regulated proteins were validated by Western immunoblotting and immunohistochemistry (IHC) of control and morphologically altered respiratory epithelium. Translational work involved clinical resection material. Collectively, 68 unique serum proteins were identified by 2DE-MALDI-TOF mass spectrometry and 13 reached statistical significance (p < 0.05). EGF, amphiregulin and the EGFR endosomal sorting protein VPS28 were induced up to 5-fold while IHC confirmed strong induction of these proteins. Furthermore, ApoA1, α-2-macroglobulin, and vitamin-D binding protein were nearly 6- and 2-fold upregulated in AAH; however, ApoA1 was oppositely regulated in LC to evidence disease stage dependent regulation of this tumour suppressor. Conversely, plasminogen and transthyretin were highly significantly repressed by 3- and 20-fold. IHC confirmed induced ApoA1, Fetuin-B and transthyretin expression to influence calcification, inflammation and tumour-infiltrating macrophages. Moreover, serum ApoA4, ApoH and ApoM were 2-, 2- and 6-fold repressed; however tissue ApoM and sphingosine-1-phosphate receptor expression was markedly induced to suggest a critical role of sphingosine-1-phosphate signalling in PL and malignant transformation. Finally, a comparison of three different LC models revealed common and unique serum biomarkers mechanistically linked to EGFR, cMyc and cRaf signalling. Their validation by IHC on clinical resection material established relevance for distinct human lung pathologies. In conclusion, we identified mechanistic biomarker candidates recommended for in-depth clinical evaluation.
Collapse
Affiliation(s)
- Jürgen Borlak
- Hannover Medical School, Centre for Pharmacology and Toxicology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Florian Länger
- Hannover Medical School, Institute of Pathology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Bijon Chatterji
- Hannover Medical School, Centre for Pharmacology and Toxicology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
28
|
Zhu K, Wang Y, Shu P, Zhou Q, Zhu J, Zhou W, Du C, Xu C, Liu X, Tang L. Increased serum levels of fetuin B in patients with coronary artery disease. Endocrine 2017; 58:97-105. [PMID: 28822077 DOI: 10.1007/s12020-017-1387-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/27/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND Recent evidence indicates a pivotal role for fetuin B, one of the cystatin superfamily of cysteine protease inhibitors, in the pathogenesis of metabolic diseases. This study investigated whether serum fetuin B levels are associated with the presence of coronary artery disease. METHODS Serum fetuin B levels were assessed in 87 patients with coronary artery disease (41 with acute coronary syndromes and 46 with stable angina pectoris) and 87 healthy controls using an enzyme-linked immunosorbent assay. The association of serum fetuin B levels with cardiac risk factors was analyzed. RESULTS Serum fetuin B levels were significantly higher in patients with coronary artery disease than those in healthy controls (90.7 ± 32.1 vs. 110.0 ± 32.7 μg/ml, P < 0.001), extremely elevated in group with acute coronary syndromes (115.0 ± 35.2 μg/ml). Pearson correlation analysis showed that serum fetuin B levels were positively associated with the levels of total cholesterol (r = 0.276, P < 0.001), low-density lipoprotein cholesterol (r = 0.363, P < 0.001), and fasting blood glucose (r = 0.159, P < 0.05). In addition, multiple logistic regression analyses revealed that fetuin B was independently associated with the presence of coronary artery disease (OR, 1.019; 95% CI, 1.009 to 1.029; P < 0.001) and acute coronary syndromes (OR, 1.017; 95% CI, 1.006 to 1.028; P < 0.01). CONCLUSIONS Our data revealed that high fetuin B levels are associated with the presence of coronary artery disease and acute coronary syndromes, and that fetuin B may serve as a potential biomarker for coronary artery disease.
Collapse
Affiliation(s)
- Kefu Zhu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, 310013, Zhejiang Province, China
| | - Yuming Wang
- Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang Province, China
| | - Pengqin Shu
- Department of Cardiology, Hangzhou First People's Hospital, Hangzhou, 310006, Zhejiang Province, China
| | - Qinyi Zhou
- Department of Mathematical Science, The University of Texas at Dallas, Dallas, TX, 75080, USA
| | - Jinzhou Zhu
- School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang Province, China
| | - Wenjing Zhou
- Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang Province, China
| | - Changqing Du
- Department of Cardiology, Zhejiang Hospital, Hangzhou, 310013, Zhejiang Province, China
| | - Chenkai Xu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, 310013, Zhejiang Province, China
| | - Xiaowei Liu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, 310013, Zhejiang Province, China
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, 310013, Zhejiang Province, China.
- School of Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang Province, China.
| |
Collapse
|
29
|
Desoubeaux G, Piqueras MDC, Pantin A, Bhattacharya SK, Peschke R, Joachim A, Cray C. Application of mass spectrometry to elucidate the pathophysiology of Encephalitozoon cuniculi infection in rabbits. PLoS One 2017; 12:e0177961. [PMID: 28723944 PMCID: PMC5516978 DOI: 10.1371/journal.pone.0177961] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/05/2017] [Indexed: 11/30/2022] Open
Abstract
Encephalitozoon cuniculi is a microsporidian species which can induce subclinical to serious disease in mammals including rabbits, a definitive natural host. The pathophysiology of infection has not been comprehensively elucidated. In this exploratory study, we utilized two mass spectrometry approaches: first, the analysis of the humoral response by profiling the microsporidian antigens as revealed by Western blot screening, and second, implementing the iTRAQ®-labeling protocol to focus on the changes within the host proteome during infection. Seven E. cuniculi proteins were identified at one-dimensional gel regions where specific seropositive reaction was observed by Western blot, including polar tube protein 3, polar tube protein 2, and for the first time reported: heat shock related 70kDa protein, polysaccharide deacetylase domain-containing protein, zinc finger protein, spore wall and anchoring disk complex protein EnP1, and translation elongation factor 1 alpha. In addition, there was a significant increase of nine host proteins in blood samples from E. cuniculi-diseased rabbits in comparison with non-diseased control subjects undergoing various inflammatory processes. This included serum paraoxonase, alpha-1-antiproteinase F precursor and alpha-1-antiproteinase S-1 which have presumptive catalytic activity likely related to infection control, and cystatin fetuin-B-type, an enzyme regulator that has been poorly studied to date. Notably, 11 proteins were found to be statistically increased in rabbits with neurological versus renal clinical presentation of E. cuniculi infection. Overall, this novel analysis based on mass spectrometry has provided new insights on the inflammatory and humoral responses during E. cuniculi infection in rabbits.
Collapse
Affiliation(s)
- Guillaume Desoubeaux
- University of Miami - Miller School of Medicine, Division of Comparative Pathology, Department of Pathology & Laboratory Medicine, Miami, Florida, United States of America
- CHU de Tours, Service de Parasitologie – Mycologie – Médecine tropicale, Tours, France
- Université François-Rabelais, Faculté de Médecine, CEPR - INSERM U1100 / Équipe 3, Tours, France
| | - Maria del Carmen Piqueras
- University of Miami, Mass Spectrometry Core Facility, Miller School of Medicin–, Miami, Florida, United States of America
| | - Ana Pantin
- University of Miami - Miller School of Medicine, Division of Comparative Pathology, Department of Pathology & Laboratory Medicine, Miami, Florida, United States of America
| | - Sanjoy K. Bhattacharya
- University of Miami, Mass Spectrometry Core Facility, Miller School of Medicin–, Miami, Florida, United States of America
| | - Roman Peschke
- University of Veterinary Medicine, Institute of Parasitology, Department of Pathobiology, Vienna, Austria
| | - Anja Joachim
- University of Veterinary Medicine, Institute of Parasitology, Department of Pathobiology, Vienna, Austria
| | - Carolyn Cray
- University of Miami - Miller School of Medicine, Division of Comparative Pathology, Department of Pathology & Laboratory Medicine, Miami, Florida, United States of America
| |
Collapse
|
30
|
Zhu J, Wan X, Wang Y, Zhu K, Li C, Yu C, Li Y. Serum fetuin B level increased in subjects of nonalcoholic fatty liver disease: a case-control study. Endocrine 2017; 56:208-211. [PMID: 27628583 DOI: 10.1007/s12020-016-1112-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 09/01/2016] [Indexed: 11/28/2022]
Abstract
Fetuin is an endogenous inhibitor of the insulin receptor tyrosine kinase. Recent studies supported the possible role of fetuin B in metabolic diseases. This study is to evaluate the role of serum fetuin B in nonalcoholic fatty liver disease (NAFLD). A hospital-based case-control study of 184 subjects was conducted. Serum level of fetuin B was measured by enzyme-linked immunosorbent assay. The serum level of fetuin B in the control (91.0 ± 36.9 μg/ml) was lower than it in NAFLD (108.7 ± 38.5 μg/ml, P < 0.001). The percentage of NAFLD increased (42.9%, 58.7% and 60.2%; P < 0.001; linear-by-linear association: P < 0.001), as fetuin B concentration elevated in its tertiles, after adjustment of body mass index (BMI). Furthermore, compared with the 1st tertile, the 3rd tertile of fetuin B indicated an association with the presence of NAFLD (adjusted odds ratio = 2.087, 95% confidence interval [1.016 - 3.937], P = 0.023), after controlling age, sex, BMI, diabetes, hypertension and hypertriglyceridemia. Lastly, fetuin B correlated with diastolic blood pressure, serum alanine transaminase and triglycerides, among the controls. It suggested a potential association between serum fetuin B and the presence of NAFLD.
Collapse
Affiliation(s)
- Jinzhou Zhu
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang Province, China
| | - Xingyong Wan
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang Province, China
| | - Yuming Wang
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang Province, China
| | - Kefu Zhu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310003, Zhejiang Province, China
| | - Chunxiao Li
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang Province, China
| | - Chaohui Yu
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang Province, China
| | - Youming Li
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang Province, China.
| |
Collapse
|