1
|
Rai AB, Codi JAK, Suchitha GP, Hemavathi KN, Dagamajalu S, Abhinand CS, Raju R, Prasad TSK. Mapping growth differentiation factor-15 (GDF15)-mediated signaling pathways in cancer: insights into its role across different cancer types. Discov Oncol 2025; 16:386. [PMID: 40128491 PMCID: PMC11933546 DOI: 10.1007/s12672-025-02121-1] [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: 11/20/2024] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
Growth differentiation factor-15 (GDF15) is a cytokine/growth factor that belongs to the Transforming growth factor-ß (TGF-ß) protein family. The expression of GDF15 is low in most human organs under normal conditions. GDF15 is a stress-responsive cytokine primarily produced by macrophages in response to inflammatory stimuli. The altered expression of GDF15 is associated with many cancers due to the inflammation caused by the disease. GDF15 triggers the activity through its receptor Glial-derived neurotrophic factor-family receptor α-like (GFRAL) and mediates multiple downstream signaling cascades, which are involved in the progression of cancers. Considering the biological importance of GDF15 in different cancers, we applied data mining techniques to systematically compile and analyze the signaling events associated with GDF15 using NetPath criteria. This resulted in constructing a detailed GDF15-mediated signaling pathway map, enhancing our understanding of its molecular mechanisms in cancer. Furthermore, proteins linked to colorectal and breast cancer identified in our pathway map were cross-referenced with established cancer pathway databases to identify unannotated proteins, highlighting gaps in the current annotations. To investigate potential therapeutic strategies, we performed molecular docking simulations and identified Vitisifuran B as a novel inhibitor that could block the GDF15-GFRAL interaction. These findings suggest that Vitisifuran B could effectively modulate GDF15 signaling, offering a promising avenue for cancer therapeutics. This study underscores the power of computational approaches, such as data mining and molecular docking, in enhancing our understanding of GDF15 signaling in cancer and identifying potential inhibitors for therapeutic development.
Collapse
Affiliation(s)
- Akhila Balakrishna Rai
- Center for Systems Biology and Molecular Medicine [An ICMR Collaborating Centre of Excellence 2024 (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Jalaluddin Akbar Kandel Codi
- Department of Surgical Oncology, Yenepoya Medical College and Hospital, Yenepoya (Deemed to Be University), Mangalore, 575018, India.
| | - Giridhara Prema Suchitha
- Center for Systems Biology and Molecular Medicine [An ICMR Collaborating Centre of Excellence 2024 (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Kadabagere Narayanaswamy Hemavathi
- Center for Systems Biology and Molecular Medicine [An ICMR Collaborating Centre of Excellence 2024 (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Shobha Dagamajalu
- Center for Systems Biology and Molecular Medicine [An ICMR Collaborating Centre of Excellence 2024 (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India.
| | - Chandran S Abhinand
- Center for Systems Biology and Molecular Medicine [An ICMR Collaborating Centre of Excellence 2024 (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Rajesh Raju
- Center for Systems Biology and Molecular Medicine [An ICMR Collaborating Centre of Excellence 2024 (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
- Center for Integrative Omics Data Science, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, India
| | - Thottethodi Subrahmanya Keshava Prasad
- Center for Systems Biology and Molecular Medicine [An ICMR Collaborating Centre of Excellence 2024 (ICMR-CCoE 2024)], Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India.
| |
Collapse
|
2
|
Wu GT, Tian QY, Xie B, Hu YB, Deng ZH. GDF15 activates human fibroblast MRC5 cells via miR-338/STAT1 in silicosis. Clin Exp Med 2025; 25:91. [PMID: 40111545 PMCID: PMC11925976 DOI: 10.1007/s10238-025-01627-w] [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: 11/29/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
Growth differentiation factor 15 (GDF-15) has been implicated in multiple biological functions. However, the role of GDF15 in silicosis remains unclear. In this study, the serum level of GDF-15 was investigated in 46 patients with silicosis by ELISA and results showed it was higher than that of control patients. The effects of exogenous GDF15 on mRNA and miRNA expression profiles of MRC5 cells were analyzed by RNA sequencing. GDF15 activated human embryonic lung fibroblast MRC5 cells with upregulation of col1a and α-SMA. GDF15 reduced miR-338 expression and increased STAT1 expression in MRC5 cells. The results of the luciferase reporter assay and bioinformatics analysis indicated that STAT1 was a direct target of miR-338. miR-338 mimics down-regulated col1a and α-SMA expression induced by GDF15 with STAT1 overexpression, whereas miR-338 inhibitor up-regulated col1a and α-SMA expression induced by GDF15 with STAT1 knockdown. Those results indicated GDF15 activated MRC5 cells through the miR-338/STAT1 pathway and GDF-15 may play an important role in silicosis.
Collapse
Affiliation(s)
- Ge-Ting Wu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qiu-Yan Tian
- Department of Pathology, Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, 410000, Hunan, China
| | - Bin Xie
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yong-Bin Hu
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Zheng-Hao Deng
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
3
|
Meng X, Li Y, Meng L, Yang C, Xia C, Wang X, Wang F. Growth Differentiation Factor 15 Inhibits Cardiac Fibrosis, Oxidative Stress, Inflammation, and Apoptosis in a Rat Model of Heart Failure with Preserved Ejection Fraction. FRONT BIOSCI-LANDMRK 2025; 30:26857. [PMID: 40018931 DOI: 10.31083/fbl26857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/02/2024] [Accepted: 12/12/2024] [Indexed: 03/01/2025]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a systemic syndrome primarily associated with fibrosis, oxidative stress, inflammation, and cellular apoptosis. Growth differentiation factor 15 (GDF15), a biomarker commonly used in clinical studies, exhibits protective effects on the myocardium. Therefore, the focus of the present study is to determine the mechanism by which GDF15 protects cardiac function in HFpEF. METHODS We conducted functional enrichment analysis and protein-protein interaction network analysis on genes highly expressed in HFpEF but lowly expressed in normal samples. We established an HFpEF rat model by feeding the rats with a high-fat diet and administering N-omega-nitro-l-arginine-methyl ester (L-NAME) in their drinking water and silenced GDF15 by tail vein injection of lentivirus (L3110). After 12 weeks of feeding, echocardiographic examinations were performed. Following euthanasia of the rats, blood and heart tissue samples were collected. Heart tissue sections were stained using Masson's trichrome and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) staining methods. Western blot (WB) analysis was employed to determine the concentrations of relevant proteins. RESULTS The echocardiographic results showed that compared with the HFpEF + MOCK group, the HFpEF+silencing GDF15 (siGDF15) group exhibited more severe cardiac dysfunction, with significant decreases in ejection fraction (p < 0.05) and E/A ratio (p < 0.001). WB results demonstrated that, compared with the HFpEF + MOCK group, the HFpEF+siGDF15 group exhibited increased expression of cardiac fibrosis-associated proteins, including collagen I (p < 0.01), collagen III (p < 0.01), and α-smooth muscle actin (α-SMA) (p < 0.01). Additionally, oxidative stress-associated biomarkers such as myeloperoxidase (MPO) (p < 0.01) and oxidized low-density lipoprotein (ox-LDL) (p < 0.01), inflammation-associated biomarkers, including interleukin-1 beta (IL-1β) (p < 0.01), interleukin-6 (IL-6) (p < 0.01), interleukin-8 (IL-8) (p < 0.01), and tumor necrosis factor α (TNFα) (p < 0.01), and apoptosis-associated biomarkers like cleaved caspase-3 (p < 0.01) and BCL2-associated X (BAX) (p < 0.01) were also upregulated in HFpEF+siGDF15 group. CONCLUSIONS Our research indicates that GDF15 preserves cardiac function by inhibiting myocardial fibrosis, reducing myocardial cell oxidative stress, alleviating cardiac inflammation, and suppressing myocardial cell apoptosis.
Collapse
Affiliation(s)
- Xuyang Meng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 100730 Beijing, China
| | - Yi Li
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 100730 Beijing, China
| | - Lingbing Meng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China
| | - Chenguang Yang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 100730 Beijing, China
| | - ChenXi Xia
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China
- Peking University Fifth School of Clinical Medicine, 100730 Beijing, China
| | - Xiang Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 100730 Beijing, China
| | - Fang Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 100730 Beijing, China
| |
Collapse
|
4
|
Sato R, da Fonseca GWP, das Neves W, von Haehling S. Mechanisms and pharmacotherapy of cancer cachexia-associated anorexia. Pharmacol Res Perspect 2025; 13:e70031. [PMID: 39776294 PMCID: PMC11707257 DOI: 10.1002/prp2.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 09/17/2024] [Accepted: 10/16/2024] [Indexed: 01/30/2025] Open
Abstract
Cachexia is a multifactorial metabolic syndrome characterized by weight and skeletal muscle loss caused by underlying illnesses such as cancer, heart failure, and renal failure. Inflammation, insulin resistance, increased muscle protein degradation, decreased food intake, and anorexia are the primary pathophysiological drivers of cachexia. Cachexia causes physical deterioration and functional impairment, loss of quality of life, lower response to active treatment, and ultimately morbidity and mortality, while the difficulties in tackling cachexia in its advanced phases and the heterogeneity of the syndrome among patients require an individualized and multidisciplinary approach from an early stage. Specifically, strategies combining nutritional and exercise interventions as well as pharmacotherapy that directly affect the pathogenesis of cachexia, such as anti-inflammatory, metabolism-improving, and appetite-stimulating agents, have been proposed, but none of which have demonstrated sufficient evidence to date. Nevertheless, several agents have recently emerged, including anamorelin, a ghrelin receptor agonist, growth differentiation factor 15 neutralization therapy, and melanocortin receptor antagonist, as candidates for ameliorating anorexia associated with cancer cachexia. Therefore, in this review, we outline cancer cachexia-associated anorexia and its pharmacotherapy, including corticosteroids, progesterone analogs, cannabinoids, anti-psychotics, and thalidomide which have been previously explored for their efficacy, in addition to the aforementioned novel agents, along with their mechanisms.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Cardiology and PneumologyUniversity of Göttingen Medical CenterGöttingenGermany
- DZHK (German Center for Cardiovascular Research), Partner Site Lower SaxonyGermany
| | - Guilherme Wesley Peixoto da Fonseca
- Heart Institute (InCor)University of São Paulo Medical SchoolSão PauloSão PauloBrazil
- School of Physical Education and SportUniversity of São PauloSão PauloBrazil
| | - Willian das Neves
- Department of Anatomy, Institute of Biomedical SciencesUniversity of Sao PauloSao PauloBrazil
| | - Stephan von Haehling
- Department of Cardiology and PneumologyUniversity of Göttingen Medical CenterGöttingenGermany
- DZHK (German Center for Cardiovascular Research), Partner Site Lower SaxonyGermany
| |
Collapse
|
5
|
Sabapathy V, Price A, Cheru NT, Venkatadri R, Dogan M, Costlow G, Mohammad S, Sharma R. ST2 + T-Regulatory Cells in Renal Inflammation and Fibrosis after Ischemic Kidney Injury. J Am Soc Nephrol 2025; 36:73-86. [PMID: 39186386 PMCID: PMC11706559 DOI: 10.1681/asn.0000000000000471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024] Open
Abstract
Key Points IL-33/ST2 alarmin pathway regulates inflammation, fibrosis, and resolution of ischemia-reperfusion injury of kidneys. ST2 regulates the transcriptome of T-regulatory cells related to suppressive and reparative functions. The secretome of ST2+ T-regulatory cells regulates hypoxic injury in an amphiregulin-dependent manner. Background Inflammation is a major cause of kidney injury. IL-1 family cytokine IL-33 is released from damaged cells and modulates the immune response through its receptor ST2 expressed on many cell types, including regulatory T cells (Tregs). Although a proinflammatory role of IL-33 has been proposed, exogenous IL-33 expanded Tregs and suppressed renal inflammation. However, the contribution of endogenous IL-33/ST2 for the role of Tregs in the resolution of kidney injury has not been investigated. Methods We used murine renal ischemia-reperfusion injury and kidney organoids (KDOs) to delineate the role of the ST2 and amphiregulin (AREG) specifically in Tregs using targeted deletion. Bulk and single-cell RNA sequencing were performed on flow-sorted Tregs from spleen and CD4 T cells from postischemic kidneys, respectively. The protective role of ST2-sufficient Tregs was analyzed using a novel coculture system of syngeneic KDOs and Tregs under hypoxic conditions. Results Bulk RNA sequencing of splenic and single-cell RNA sequencing of kidney CD4 T cells showed that ST2+ Tregs are enriched for genes related to Treg proliferation and function. Genes for reparative factors, such as Areg , were also enriched in ST2+ Tregs. Treg-specific deletion of ST2 or AREG exacerbated kidney injury and fibrosis in the unilateral ischemia-reperfusion injury model. In coculture studies, wild-type but not ST2-deficient Tregs preserved hypoxia-induced loss of kidney organoid viability, which was restored by AREG supplementation. Conclusions Our study identified the role of the IL-33/ST2 pathway in Tregs for resolution of kidney injury. The transcriptome of ST2+ Tregs was enriched for reparative factors including Areg . Lack of ST2 or AREG in Tregs worsened kidney injury. Tregs protected KDOs from hypoxia in a ST2- and AREG-dependent manner.
Collapse
Affiliation(s)
- Vikram Sabapathy
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| | - Airi Price
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Department of International Health, Georgetown University, Washington, DC
| | - Nardos Tesfaye Cheru
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut
| | - Rajkumar Venkatadri
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Immunology Research Unit, GlaxoSmithKline (GSK), Collegeville, Pennsylvania
| | - Murat Dogan
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Department of Transplant Surgery, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Gabrielle Costlow
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| | - Saleh Mohammad
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| | - Rahul Sharma
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| |
Collapse
|
6
|
Hong D. Identification of Ferroptosis-Associated Genes in Primary Open-Angle Glaucoma through Bioinformatics Analysis. Crit Rev Eukaryot Gene Expr 2025; 35:15-26. [PMID: 40228223 DOI: 10.1615/critreveukaryotgeneexpr.2025057767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
This study aims to examine ferroptosis-associated genes in primary open-angle glaucoma (POAG) and offer new insights into the underlying disease mechanisms and potential therapeutic approaches. Differentially expressed genes (DEGs) between the POAG and control groups were identified using bioinformatics analysis and subsequently intersected with a ferroptosis gene set to isolate ferroptosis-related DEGs (Ferr DEGs). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted to examine their biological functions. Core genes were identified through protein-protein interaction (PPI) network and Friends analysis. The diagnostic potential of core Ferr DEGs was assessed using receiver operating characteristic (ROC) curve analysis, while immune cell infiltration was examined using the CIBERSORT algorithm. Additionally, Spearman correlation analysis was used to examine the relationships between the identified genes and immune cell populations. A total of 25 Ferr DEGs were identified, with DDIT4, GDF15, NAMPT, HBA1, and IGFBP7 recognized as key core genes. ROC analysis demonstrated that these genes exhibited high diagnostic accuracy, with an AUC > 0.7. Additionally, the infiltration levels of memory B cells and macrophage_M2 were significantly elevated in POAG tissues compared to the control group. Notably, the core genes revealed significant correlations with various immune cell types. Our findings underscore the involvement of ferroptosis-related genes in POAG pathogenesis and highlight their potential as diagnostic biomarkers and therapeutic targets. Future research should focus on validating these findings in clinical settings and exploring the therapeutic modulation of ferroptosis in POAG management.
Collapse
|
7
|
Zhou H, Xiao J, Cheng Q, Wang W, Peng H, Lin X, Chen J, Wang X. Metformin inhibits migration and epithelial-to-mesenchymal transition in non-small cell lung cancer cells through AMPK-mediated GDF15 induction. Eur J Pharmacol 2024; 985:177127. [PMID: 39528101 DOI: 10.1016/j.ejphar.2024.177127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/06/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
The growth differentiation factor 15 (GDF15) may serve as a biomarker of metformin, which mediates the bodyweight lowering effect of metformin. However, whether GDF15 also serves as a molecular target of metformin to inhibit carcinogenesis remains largely unknown. This study examined the role and molecular mechanisms of GDF15 in the anticancer effects of metformin in non-small cell lung cancer (NSCLC) cells, which has never been reported before. We found that metformin significantly inhibited the migration of NSCLC A549 and NCI-H460 cells and reduced the expression of epithelial-to-mesenchymal transition (EMT)-related molecules, including neuro-cadherin (N-cadherin), matrix metalloproteinase 2 (MMP2), and the zinc finger transcription factor Snail, but increased epithelial cadherin (E-cadherin) expression. Furthermore, metformin increased GDF15 and its upstream transcription factors activated transcription factor 4 (ATF4) and C/EBP-homologous protein (CHOP) expressions and increased AMP-activated protein kinase (AMPK) phosphorylation in NSCLC cells. GDF15 siRNA partially reverses the inhibitory effect of metformin on NSCLC cell migration. Moreover, metformin-induced increases in GDF15, CHOP, and ATF4 expression and the inhibition of migration were partially reversed by treatment with Compound C, a specific AMPK inhibitor. Meanwhile, metformin significantly inhibited NCI-H460 xenograft tumor growth in nude mice, increased GDF15 expression, and regulated EMT- and migration-related protein expression in xenograft tumors. In conclusion, our results provide novel insights into revealing that GDF15 can serve as a potential molecular target of metformin owing to its anti-cancer effect in NSCLC, which is mediated by AMPK activation.
Collapse
Affiliation(s)
- Hongyu Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Jun Xiao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Qi Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Wen Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - He Peng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Xiaojian Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Jiajun Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Xingya Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China.
| |
Collapse
|
8
|
Wang J, Fendler NL, Shukla A, Wu SY, Challa A, Lee J, Joachimiak LA, Minna JD, Chiang CM, Vos SM, D'Orso I. ARF alters PAF1 complex integrity to selectively repress oncogenic transcription programs upon p53 loss. Mol Cell 2024; 84:4538-4557.e12. [PMID: 39532099 PMCID: PMC12001331 DOI: 10.1016/j.molcel.2024.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 09/03/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
The polymerase associated factor 1 (PAF1) complex (PAF1c) promotes RNA polymerase II (RNA Pol II) transcription at the elongation step; however, how PAF1c transcription activity is selectively regulated during cell fate transitions remains poorly understood. Here, we reveal that the alternative reading frame (ARF) tumor suppressor operates at two levels to restrain PAF1c-dependent oncogenic transcriptional programs upon p53 loss in mouse cells. First, ARF assembles into homo-oligomers to bind the PAF1 subunit to promote PAF1c disassembly, consequently dampening PAF1c interaction with RNA Pol II and PAF1c-dependent transcription. Second, ARF targets the RUNX family transcription factor 1 (RUNX1) to selectively tune gene transcription. Consistently, ARF loss triggers RUNX1- and PAF1c-dependent transcriptional activation of pro-growth ligands (growth differentiation factor/bone morphogenetic protein [GDF/BMP]), promoting a cell-intrinsic GDF/BMP-Smad1/5 axis that aberrantly induce cell growth. Notably, pharmacologic inactivation of GDF/BMP signaling and genetic perturbation of RUNX1 significantly attenuate cell proliferation mediated by dual p53 and ARF loss, offering therapeutic utility. Our data underscore the significance of selective ARF-mediated tumor-suppressive functions through a universal transcriptional regulator.
Collapse
Affiliation(s)
- Jinli Wang
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nikole L Fendler
- Massachusetts Institute of Technology, Department of Biology, Cambridge, MA, USA
| | - Ashutosh Shukla
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shwu-Yuan Wu
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ashwini Challa
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeon Lee
- Lydia Hill Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lukasz A Joachimiak
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John D Minna
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Hamon Center for Therapeutic Oncology Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cheng-Ming Chiang
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Seychelle M Vos
- Massachusetts Institute of Technology, Department of Biology, Cambridge, MA, USA; Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Iván D'Orso
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
9
|
Wang B, Zhang J, Ma R, Bai M, Song Y, Liang G. Diagnostic value of serum GDF-15 in patients with pseudomyxoma peritonei. Clin Biochem 2024; 133-134:110827. [PMID: 39304173 DOI: 10.1016/j.clinbiochem.2024.110827] [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: 07/05/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND AND AIMS Pseudomyxoma peritonei (PMP) is a rare malignancy that lacks a highly sensitive and specific biomarker for its diagnosis. Identifying reliable serum markers is crucial for improving the diagnostic accuracy and management of PMP. This study aims to explore the diagnostic value of serum growth differentiation factor 15 (GDF-15) in patients with PMP. MATERIAL AND METHODS We carried on a 1:1 matched case-control study. 44 patients with PMP hospitalized in Aerospace Center Hospital were recruited as cases, and 44 sex- and age-matched apparently healthy participants were selected as controls. The serum GDF-15 concentrations were tested using an ELISA method. The diagnostic value of GDF-15 in PMP patients was assessed by receiver operating characteristic (ROC) curve analysis. RESULTS The median serum GDF-15 level in PMP patients was 1192.77 (843.03-1879.06) pg/mL, notably higher than that in healthy controls [533.27 (410.46-641.47) pg/mL] (P<0.001). The area under the curve (AUC) of serum GDF-15 for PMP diagnosis was 0.907, the optimal diagnostic threshold value was 644.58 pg/mL, the sensitivity was 93.18 %, and the specificity was 77.27 %. The AUC of GDF-15 combined with carbohydrate antigen 125 (CA125) was larger than that of GDF-15 alone (P=0.027), and the sensitivity and specificity achieved 86.36 % and 95.45 %. GDF-15 levels showed a significant correlation with age (P=0.042), with younger PMP patients exhibiting notably lower concentrations of GDF-15 compared to older patients. CONCLUSION Serum GDF-15 could become a new marker for the PMP diagnosis. The combination of GDF-15 and CA125 demonstrated superior diagnostic performance for PMP compared to GDF-15 alone, achieving a sensitivity of 86.36% and a specificity of 95.45%.
Collapse
Affiliation(s)
- Bing Wang
- Department of Clinical Laboratory of Aerospace Center Hospital, Beijing 100049, China
| | - Jie Zhang
- Department of Clinical Laboratory of Aerospace Center Hospital, Beijing 100049, China
| | - Ruiqing Ma
- Department of Myxoma, Aerospace Center Hospital, Beijing 100049, China
| | - Mingjian Bai
- Department of Clinical Laboratory of Aerospace Center Hospital, Beijing 100049, China
| | - Yan Song
- Department of Clinical Laboratory of Aerospace Center Hospital, Beijing 100049, China
| | - Guowei Liang
- Department of Clinical Laboratory of Aerospace Center Hospital, Beijing 100049, China.
| |
Collapse
|
10
|
Zhang J, Sun J, Li J, Xia H. Targeting the GDF15 Signalling for Obesity Treatment: Recent Advances and Emerging Challenges. J Cell Mol Med 2024; 28:e70251. [PMID: 39700016 DOI: 10.1111/jcmm.70251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/21/2024] Open
Abstract
The growth differentiation factor 15 (GDF15)-glial cell-derived neurotrophic factor family receptor alpha-like (GFRAL) pathway plays a crucial role in the regulation of metabolism, appetite and body weight control. Obesity is an increasingly prevalent chronic disease worldwide, necessitating effective treatment strategies. Recent preclinical and clinical studies have highlighted that targeting the GDF15-GFRAL signalling pathway is a promising approach for treating obesity, particularly because it has minimal impact on skeletal muscle mass, which is essential to preserve during weight loss. Given its distinctive mechanisms, the GDF15-GFRAL axis represents an attractive target for addressing various metabolic disorders, especially obesity. In this review, we will explore how the GDF15-GFRAL axis is regulated, its distribution in the body and its role in the regulation of metabolism, appetite and obesity. Additionally, we will discuss recent advances and potential challenges in targeting the GDF15-GFRAL axis for obesity treatment.
Collapse
Affiliation(s)
- Jincheng Zhang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and National Clinical Research Center for Geriatrics and Laboratory of Molecular Targeted Therapy in Oncology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Physical Education and Sports, Sichuan University, Chengdu, China
- Research Institute of Molecular Exercise Science, Hungarian University of Sports Science, Budapest, Hungary
| | - Jingquan Sun
- School of Physical Education and Sports, Sichuan University, Chengdu, China
| | - Jielang Li
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and National Clinical Research Center for Geriatrics and Laboratory of Molecular Targeted Therapy in Oncology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hongwei Xia
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and National Clinical Research Center for Geriatrics and Laboratory of Molecular Targeted Therapy in Oncology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Xue Y, Zhang Y, Su Y, Zhao J, Yu D, Jo Y, Joo J, Lee HJ, Ryu D, Wei S. The implicated role of GDF15 in gastrointestinal cancer. Eur J Clin Invest 2024; 54:e14290. [PMID: 39044314 DOI: 10.1111/eci.14290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/03/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Growth differentiation factor 15 (GDF15), a stress-responsive cytokine from transforming growth factor superfamily, is highly expressed in mammalian tissues, including pancreas, stomach and intestine under pathological conditions. In particular, elevated levels of GDF15 might play an important role in the development and progression of various gastrointestinal cancers (GCs), suggesting its potential as a promising target for disease prediction and treatment. METHODS In this review, systematic reviews addressing the role of GDF15 in GCs were updated, along with the latest clinical trials focussing on the GDF15-associated digestive malignancies. RESULTS The multiple cellular pathways through which GDF15 is involved in the regulation of physiological and pathological conditions were first summarized. Then, GDF15 was also established as a valuable clinical index, functioning as a predictive marker in diverse GCs. Notably, latest clinical treatments targeting GDF15 were also highlighted, demonstrating its promising potential in mitigating and curing digestive malignancies. CONCLUSIONS This review unveils the pivotal roles of GDF15 and its potential as a promising target in the pathogenesis of GCs, which may provide insightful directions for future investigations.
Collapse
Affiliation(s)
- Yingqi Xue
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Yan Zhang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Yale Su
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jiangqi Zhao
- Department of Dermatology, The Second Hospital of Jilin University, Changchun, China
| | - Daoquan Yu
- Department of Hepatological Surgery, Shuangliao Center Hospital, Shuangliao, China
| | - Yunju Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Jongkil Joo
- Department of Obstetrics and Gynecology, Pusan National University Hospital, Busan, Korea
| | - Hyun Joo Lee
- Department of Obstetrics and Gynecology, Pusan National University Hospital, Busan, Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Shibo Wei
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
12
|
Streeter J, Persaud L, Gao J, Manika D, Fairman W, García-Peña LM, Marti A, Manika C, Gaddi S, Schickling B, Pereira RO, Abel ED. ATF4-dependent and independent mitokine secretion from OPA1 deficient skeletal muscle in mice is sexually dimorphic. Front Endocrinol (Lausanne) 2024; 15:1325286. [PMID: 39381436 PMCID: PMC11458430 DOI: 10.3389/fendo.2024.1325286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 08/30/2024] [Indexed: 10/10/2024] Open
Abstract
Introduction Reducing Optic Atrophy 1 (OPA1) expression in skeletal muscle in male mice induces Activation Transcription Factor 4 (ATF4) and the integrated stress response (ISR). Additionally, skeletal muscle secretion of Fibroblast Growth Factor 21 (FGF21) is increased, which mediates metabolic adaptations including resistance to diet-induced obesity (DIO) and glucose intolerance in these mice. Although FGF21 induction in this model can be reversed with pharmacological attenuation of ER stress, it remains to be determined if ATF4 is responsible for FGF21 induction and its metabolic benefits in this model. Methods We generated mice with homozygous floxed Opa1 and Atf4 alleles and a tamoxifen-inducible Cre transgene controlled by the human skeletal actin promoter to enable simultaneous depletion of OPA1 and ATF4 in skeletal muscle (mAO DKO). Mice were fed high fat (HFD) or control diet and evaluated for ISR activation, body mass, fat mass, glucose tolerance, insulin tolerance and circulating concentrations of FGF21 and growth differentiation factor 15 (GDF15). Results In mAO DKO mice, ATF4 induction is absent. Other indices of ISR activation, including XBP1s, ATF6, and CHOP were induced in mAO DKO males, but not in mOPA1 or mAO DKO females. Resistance to diet-induced obesity was not reversed in mAO DKO mice of both sexes. Circulating FGF21 and GDF15 illustrated sexually dimorphic patterns. Loss of OPA1 in skeletal muscle increases circulating FGF21 in mOPA1 males, but not in mOPA1 females. Additional loss of ATF4 decreased circulating FGF21 in mAO DKO male mice, but increased circulating FGF21 in female mAO DKO mice. Conversely, circulating GDF15 was increased in mAO DKO males and mOPA1 females, but not in mAO DKO females. Conclusion Sex differences exist in the transcriptional outputs of the ISR following OPA deletion in skeletal muscle. Deletion of ATF4 in male and female OPA1 KO mice does not reverse the resistance to DIO. Induction of circulating FGF21 is ATF4 dependent in males, whereas induction of circulating GDF15 is ATF4 dependent in females. Elevated GDF15 in males and FGF21 in females could reflect activation by other transcriptional outputs of the ISR, that maintain mitokine-dependent metabolic protection in an ATF4-independent manner.
Collapse
Affiliation(s)
- Jennifer Streeter
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Luis Persaud
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Jason Gao
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Deeraj Manika
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Will Fairman
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Luis Miguel García-Peña
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Alex Marti
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Chethan Manika
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Shreya Gaddi
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Brandon Schickling
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Renata O. Pereira
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - E. Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
13
|
Enzer NA, Chiles J, Mason S, Shirahata T, Castro V, Regan E, Choi B, Yuan NF, Diaz AA, Washko GR, McDonald ML, Estépar RSJ, Ash SY. Proteomics and machine learning in the prediction and explanation of low pectoralis muscle area. Sci Rep 2024; 14:17981. [PMID: 39097658 PMCID: PMC11297919 DOI: 10.1038/s41598-024-68447-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024] Open
Abstract
Low muscle mass is associated with numerous adverse outcomes independent of other associated comorbid diseases. We aimed to predict and understand an individual's risk for developing low muscle mass using proteomics and machine learning. We identified eight biomarkers associated with low pectoralis muscle area (PMA). We built three random forest classification models that used either clinical measures, feature selected biomarkers, or both to predict development of low PMA. The area under the receiver operating characteristic curve for each model was: clinical-only = 0.646, biomarker-only = 0.740, and combined = 0.744. We displayed the heterogenetic nature of an individual's risk for developing low PMA and identified two distinct subtypes of participants who developed low PMA. While additional validation is required, our methods for identifying and understanding individual and group risk for low muscle mass could be used to enable developments in the personalized prevention of low muscle mass.
Collapse
Affiliation(s)
- Nicholas A Enzer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Joe Chiles
- Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Stefanie Mason
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Toru Shirahata
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan
| | - Victor Castro
- Boston University School of Medicine, Boston, MA, USA
| | - Elizabeth Regan
- COPDGene Study Consortium, Denver, CO, USA
- Division of Rheumatology, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Bina Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Nancy F Yuan
- Department of Biomedical Informatics, University of California at San Diego, San Diego, CA, USA
| | - Alejandro A Diaz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Merry-Lynn McDonald
- Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Raúl San José Estépar
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Samuel Y Ash
- Department of Critical Care Medicine, South Shore Hospital, 55 Fogg Road, South Weymouth, MA, 02190, USA.
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
14
|
Mohammedi K, Hess S, McQueen M, Pigeyre M, Lee SF, Pare G, Gerstein HC. Determinants of serious health outcome-free status in middle-aged and older people with dysglycaemia: Exploratory analysis of the ORIGIN trial. Diabetes Obes Metab 2024; 26:3272-3280. [PMID: 38747213 DOI: 10.1111/dom.15654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 07/10/2024]
Abstract
AIM To assess clinical and biochemical measurements that can identify people with dysglycaemia (i.e. diabetes or pre-diabetes) who remain free of serious outcomes during follow-up. MATERIALS AND METHODS We conducted exploratory analyses using data from the Outcomes Reduction with an Initial Glargine Intervention (ORIGIN) study to identify independent determinants of outcome-free status in 12 537 middle-aged and older adults with prediabetes and early type 2 diabetes from 40 countries. Serious outcome-free status was defined as the absence of major cardiovascular outcomes, kidney or retinal outcomes, peripheral artery disease, dementia, cancer, any hospitalization, or death during follow-up. RESULTS In total, 3328 (26.6%) participants remained free of serious outcomes during a median follow-up of 6.2 years (IQR 5.8, 6.7). Independent clinical determinants of outcome-free status included younger age, female sex, non-White ethnicity, shorter diabetes duration, absence of previous cardiovascular disease, current or former smokers, higher grip strength, Mini-Mental State Examination score, and ankle-brachial index, lower body mass index and kidney disease index, and non-use of renin-angiotensin system drugs and beta-blockers. In a subset of 8401 people with baseline measurements of 238 biomarkers, growth differentiation factor 15, kidney injury molecule-1, N-terminal pro-brain natriuretic peptide, uromodulin, C-reactive protein, factor VII and ferritin were independent determinants. The combination of clinical determinants and biomarkers best identified participants who remained outcome-free (C-statistics 0.71, 95% confidence interval 0.70-0.73; net reclassification improvement 0.55, 95% confidence interval 0.48-0.58). CONCLUSIONS A set of routinely measured clinical characteristics and seven protein biomarkers identify middle-aged and older people with prediabetes or early type 2 diabetes as least likely to experience serious outcomes during follow-up.
Collapse
Affiliation(s)
- Kamel Mohammedi
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Canada
- Université de Bordeaux, INSERM, BMC, U1034, Avenue de Magellan, Pessac, France
| | - Sibylle Hess
- Sanofi, Global Medical Diabetes, Frankfurt, Germany
| | - Matthew McQueen
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Canada
| | - Marie Pigeyre
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Canada
| | - Shun Fu Lee
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Canada
| | - Guillaume Pare
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Canada
| | - Hertzel C Gerstein
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Canada
| |
Collapse
|
15
|
Greer CE, Chew-Harris J, Adamson PD, Pemberton CJ, Pickering JW, Pilbrow AP, Frampton CM, Troughton RW, Doughty RN, Richards AM. Convalescent Growth Differentiation Factor-15 and Long-Term Outcomes after an Acute Coronary Syndrome. J Appl Lab Med 2024; 9:672-683. [PMID: 38635817 DOI: 10.1093/jalm/jfae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/01/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Growth differentiation factor-15 (GDF-15) has been shown to be associated with adverse clinical outcomes in patients after an acute coronary syndrome when measured soon after an event. Although dynamic in the acute phase after myocardial injury, GDF-15 has been shown to remain stable during convalescence. In this study, we aimed to assess the value of GDF-15 as a long-term prognostic marker for clinical outcomes when measured in the convalescent phase following an acute coronary syndrome. METHODS GDF-15 concentrations were measured in 1945 patients who were recruited between 2002 and 2009 to the Coronary Disease Cohort Study. For this analysis, follow-up was curtailed at 10 years and association of GDF-15 with all-cause death, cardiovascular death, recurrent myocardial infarction, and heart failure hospitalizations were assessed with multivariate Cox proportional hazard regression analysis. RESULTS After 10 years of follow-up, there were 648 deaths (348 from cardiovascular causes), 500 admissions for myocardial infarction, and 436 for heart failure. Four-month convalescent GDF-15 demonstrated a robust independent association with all endpoints, which remained after adjustment for Global Registry of Acute Coronary Events score and other convalescent biomarkers. When compared to the lowest quartile of GDF-15 concentrations, those in the highest quartile had a 3-fold increased risk of all-cause death. CONCLUSIONS Convalescent plasma GDF-15 is a strong and independent predictor of 10-year all-cause death, cardiovascular death, recurrent myocardial infarction, and heart failure admission following an acute coronary syndrome. AUSTRALIAN NEW ZEALAND CLINICAL TRIALS REGISTRY TRIAL ID ACTRN12605000431628.
Collapse
Affiliation(s)
- Charlotte E Greer
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Janice Chew-Harris
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Philip D Adamson
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Chris J Pemberton
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - John W Pickering
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Anna P Pilbrow
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Chris M Frampton
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Richard W Troughton
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Robert N Doughty
- Greenlane Cardiovascular Service, Te Toka Tumai Auckland Hospital, Auckland, New Zealand
- Heart Health Research Group, Department of Medicine, University of Auckland, Auckland, New Zealand
| | - A Mark Richards
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
16
|
Lerma-Treviño C, Hernández-Cadena L, Acosta-Montes JO, Hernández-Montes G, Alvarado-Cruz I, Romieu I, Barraza-Villarreal A. Prenatal Arsenic Exposure on DNA Methylation of C18ORF8 and ADAMTS9 Genes of Newborns from the POSGRAD Birth Cohort Study. TOXICS 2024; 12:476. [PMID: 39058128 PMCID: PMC11280544 DOI: 10.3390/toxics12070476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/08/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024]
Abstract
Exposure to arsenic (As) is a public health problem associated with cancer (skin and colon) and it has been reported that epigenetic changes may be a potential mechanism of As carcinogenesis. It is pertinent to evaluate this process in genes that have been associated with cancer, such as ADAMTS9 and C18ORF8. Gestation and delivery data were obtained from the POSGRAD study. Exposure to As was measured in urine during pregnancy. Gene methylation was performed by sodium bisulfite sequencing; 26 CpG sites for the C18ORF8 gene and 21 for ADAMTS9 were analyzed. These sites are located on the CpG islands near the start of transcription. Sociodemographic characteristics were obtained by a questionnaire. The statistical analysis was performed using multiple linear regression models adjusted for potential confounders. Newborns with an As exposure above 49.4 μg g-1 showed a decrease of 0.21% on the methylation rate in the sites CpG15, CpG19, and CpG21 of the C18ORF8 gene (adjusted ß = -0.21, p-value = 0.02). No statistically significant association was found between prenatal exposure to As and methylation of the ADAMTS9 gene. Prenatal exposure to As was associated with decreased DNA methylation at the CpG15, CpG19, and CpG21 sites of the C18ORF8 gene. These sites can provide information to elucidate epigenetic mechanisms associated with prenatal exposure to As and cancer.
Collapse
Affiliation(s)
- Carolina Lerma-Treviño
- Centro de Investigación en Nutrición y Salud, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico;
| | - Leticia Hernández-Cadena
- Dirección de Salud Ambiental, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (L.H.-C.); (I.R.)
| | | | - Georgina Hernández-Montes
- CIC-UNAM-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico;
| | - Isabel Alvarado-Cruz
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA;
| | - Isabelle Romieu
- Dirección de Salud Ambiental, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (L.H.-C.); (I.R.)
| | - Albino Barraza-Villarreal
- Dirección de Salud Ambiental, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (L.H.-C.); (I.R.)
| |
Collapse
|
17
|
Du YN, Zhao JW. GDF15: Immunomodulatory Role in Hepatocellular Carcinoma Pathogenesis and Therapeutic Implications. J Hepatocell Carcinoma 2024; 11:1171-1183. [PMID: 38911292 PMCID: PMC11193986 DOI: 10.2147/jhc.s471239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/07/2024] [Indexed: 06/25/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths globally and the sixth most common cancer worldwide. Evidence shows that growth differentiation factor 15 (GDF15) contributes to hepatocarcinogenesis through various mechanisms. This paper reviews the latest insights into the role of GDF15 in the development of HCC, its role in the immune microenvironment of HCC, and its molecular mechanisms in metabolic dysfunction associated steatohepatitis (MASH) and metabolic associated fatty liver disease (MAFLD)-related HCC. Additionally, as a serum biomarker for HCC, diagnostic and prognostic value of GDF15 for HCC is summarized. The article elaborates on the immunological effects of GDF15, elucidating its effects on hepatic stellate cells (HSCs), liver fibrosis, as well as its role in HCC metastasis and tumor angiogenesis, and its interactions with anticancer drugs. Based on the impact of GDF15 on the immune response in HCC, future research should identify its signaling pathways, affected immune cells, and tumor microenvironment interactions. Clinical studies correlating GDF15 levels with patient outcomes can aid personalized treatment. Additionally, exploring GDF15-targeted therapies with immunotherapies could improve anti-tumor responses and patient outcomes.
Collapse
Affiliation(s)
- Yi-Ning Du
- Department of Medical Sciences, Li Ka-shing School of Medicine, University of Hong Kong, Hong Kong, People’s Republic of China
| | - Jin-Wei Zhao
- Department of Hepatopancreatobiliary Surgery, Second Hospital of Jilin University, Jilin University, Changchun, Jilin Province, People’s Republic of China
| |
Collapse
|
18
|
Sato R, Vatic M, Peixoto da Fonseca GW, Anker SD, von Haehling S. Biological basis and treatment of frailty and sarcopenia. Cardiovasc Res 2024:cvae073. [PMID: 38828887 DOI: 10.1093/cvr/cvae073] [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: 07/27/2022] [Revised: 11/23/2022] [Accepted: 12/20/2022] [Indexed: 06/05/2024] Open
Abstract
In an ageing society, the importance of maintaining healthy life expectancy has been emphasized. As a result of age-related decline in functional reserve, frailty is a state of increased vulnerability and susceptibility to adverse health outcomes with a serious impact on healthy life expectancy. The decline in skeletal muscle mass and function, also known as sarcopenia, is key in the development of physical frailty. Both frailty and sarcopenia are highly prevalent in patients not only with advanced age but also in patients with illnesses that exacerbate their progression like heart failure (HF), cancer, or dementia, with the prevalence of frailty and sarcopenia in HF patients reaching up to 50-75% and 19.5-47.3%, respectively, resulting in 1.5-3 times higher 1-year mortality. The biological mechanisms of frailty and sarcopenia are multifactorial, complex, and not yet fully elucidated, ranging from DNA damage, proteostasis impairment, and epigenetic changes to mitochondrial dysfunction, cellular senescence, and environmental factors, many of which are further linked to cardiac disease. Currently, there is no gold standard for the treatment of frailty and sarcopenia, however, growing evidence supports that a combination of exercise training and nutritional supplement improves skeletal muscle function and frailty, with a variety of other therapies being devised based on the underlying pathophysiology. In this review, we address the involvement of frailty and sarcopenia in cardiac disease and describe the latest insights into their biological mechanisms as well as the potential for intervention through exercise, diet, and specific therapies.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Mirela Vatic
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Guilherme Wesley Peixoto da Fonseca
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Stefan D Anker
- Department of Cardiology (CVK) of German Heart Center Charité; German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| |
Collapse
|
19
|
Lasaad S, Crambert G. GDF15, an Emerging Player in Renal Physiology and Pathophysiology. Int J Mol Sci 2024; 25:5956. [PMID: 38892145 PMCID: PMC11172470 DOI: 10.3390/ijms25115956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
These last years, the growth factor GDF15 has emerged as a key element in many different biological processes. It has been established as being produced in response to many pathological states and is now referred to as a stress-related hormone. Regarding kidney functions, GDF15 has been involved in different pathologies such as chronic kidney disease, diabetic nephropathy, renal cancer, and so on. Interestingly, recent studies also revealed a role of GDF15 in the renal homeostatic mechanisms allowing to maintain constant, as far as possible, the plasma parameters such as pH and K+ values. In this review, we recapitulate the role of GDF15 in physiological and pathological context by focusing our interest on its renal effect.
Collapse
Affiliation(s)
- Samia Lasaad
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Gilles Crambert
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Scientifique (INSERM), Sorbonne Université, Université Paris Cité, Laboratoire de Physiologie Rénale et Tubulopathies, F-75006 Paris, France
- Unité Métabolisme et Physiologie Rénale, Centre National de la Recherche Scientifique (CNRS) EMR 8228, F-75006 Paris, France
| |
Collapse
|
20
|
Ono R, Takayama S, Abe M, Arita R, Abe T, Ishii T. Growth Differentiation Factor-15 Is Considered a Predictive Biomarker of Long COVID in Non-hospitalized Patients. Cureus 2024; 16:e59433. [PMID: 38826986 PMCID: PMC11140824 DOI: 10.7759/cureus.59433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2024] [Indexed: 06/04/2024] Open
Abstract
Mitochondrial dysfunction is associated with various diseases. Mitochondria plays a regulatory role during infection. The association between mitokines and subsequent COVID progression has not been previously studied. The retrospective cohort study aimed to investigate the potential of serum mitokines as long COVID biomarkers in non-hospitalized patients. Patients with confirmed SARS-CoV-2 infection and blood test reports between January 2021 and April 2023 were included. Patients were categorized into two groups, the recovered and long COVID groups, based on fatigue, decline in focus, and pain. Serum levels of growth differentiation factor 15 (GDF-15) and fibroblast growth factor-21 (FGF-21), which are affected by mitochondrial function, along with inflammatory and vascular endothelium markers, were measured using enzyme-linked immunosorbent assays (ELISA). A receiver operating characteristic curve was used to screen the biomarkers. The threshold value of GDF-15 in the acute phase was 965 pg/mL (sensitivity: 71.4%, specificity: 83.3%), indicating that GDF-15 may be associated with the presence of symptoms three months post onset. No association with inflammatory markers and vascular structures was observed. Therefore, elevated GDF-15 levels in the acute phase may act as a predictive biomarker of long COVID.
Collapse
Affiliation(s)
- Rie Ono
- Department of Kampo Medicine, Tohoku University Hospital, Sendai, JPN
- Department of Education and Support for Regional Medicine, Tohoku University Hospital, Sendai, JPN
- Department of Anesthesiology and Perioperative Medicine, Tohoku University Hospital, Sendai, JPN
| | - Shin Takayama
- Department of Kampo Medicine, Tohoku University Hospital, Sendai, JPN
- Department of Education and Support for Regional Medicine, Tohoku University Hospital, Sendai, JPN
- Department of Kampo and Integrative Medicine, Tohoku University Graduate School of Medicine, Sendai, JPN
| | - Michiaki Abe
- Department of Education and Support for Regional Medicine, Tohoku University Hospital, Sendai, JPN
| | - Ryutaro Arita
- Department of Kampo Medicine, Tohoku University Hospital, Sendai, JPN
- Department of Education and Support for Regional Medicine, Tohoku University Hospital, Sendai, JPN
- Department of Kampo and Integrative Medicine, Tohoku University Graduate School of Medicine, Sendai, JPN
| | - Takaaki Abe
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, JPN
- Department of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, JPN
| | - Tadashi Ishii
- Department of Education and Support for Regional Medicine, Tohoku University Hospital, Sendai, JPN
- Department of Kampo and Integrative Medicine, Tohoku University Graduate School of Medicine, Sendai, JPN
| |
Collapse
|
21
|
Gallucci G, Turazza FM, Inno A, Canale ML, Silvestris N, Farì R, Navazio A, Pinto C, Tarantini L. Atherosclerosis and the Bidirectional Relationship between Cancer and Cardiovascular Disease: From Bench to Bedside-Part 1. Int J Mol Sci 2024; 25:4232. [PMID: 38673815 PMCID: PMC11049833 DOI: 10.3390/ijms25084232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Atherosclerosis, a complex metabolic-immune disease characterized by chronic inflammation driven by the buildup of lipid-rich plaques within arterial walls, has emerged as a pivotal factor in the intricate interplay between cancer and cardiovascular disease. This bidirectional relationship, marked by shared risk factors and pathophysiological mechanisms, underscores the need for a comprehensive understanding of how these two formidable health challenges intersect and influence each other. Cancer and its treatments can contribute to the progression of atherosclerosis, while atherosclerosis, with its inflammatory microenvironment, can exert profound effects on cancer development and outcomes. Both cancer and cardiovascular disease involve intricate interactions between general and personal exposomes. In this review, we aim to summarize the state of the art of translational data and try to show how oncologic studies on cardiotoxicity can broaden our knowledge of crucial pathways in cardiovascular biology and exert a positive impact on precision cardiology and cardio-oncology.
Collapse
Affiliation(s)
| | - Fabio Maria Turazza
- Struttura Complessa di Cardiologia, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy;
| | - Alessandro Inno
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy;
| | - Maria Laura Canale
- Division of Cardiology, Azienda USL Toscana Nord-Ovest, Versilia Hospital, 55041 Lido di Camaiore, Italy;
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology “G.Barresi”, University of Messina, 98100 Messina, Italy;
| | - Roberto Farì
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Alessandro Navazio
- Cardiologia Ospedaliera, Department of Specialized Medicine, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy;
| | - Carmine Pinto
- Provincial Medical Oncology, Department of Oncology and Advanced Technologies, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy;
| | - Luigi Tarantini
- Cardiologia Ospedaliera, Department of Specialized Medicine, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy;
| |
Collapse
|
22
|
Abel TR, Kosarek NN, Parvizi R, Jarnagin H, Torres GM, Bhandari R, Huang M, Toledo DM, Smith A, Popovich D, Mariani MP, Yang H, Wood T, Garlick J, Pioli PA, Whitfield ML. Single-cell epigenomic dysregulation of Systemic Sclerosis fibroblasts via CREB1/EGR1 axis in self-assembled human skin equivalents. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586316. [PMID: 38585776 PMCID: PMC10996484 DOI: 10.1101/2024.03.22.586316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by skin fibrosis, internal organ involvement and vascular dropout. We previously developed and phenotypically characterized an in vitro 3D skin-like tissue model of SSc, and now analyze the transcriptomic (scRNA-seq) and epigenetic (scATAC-seq) characteristics of this model at single-cell resolution. SSc 3D skin-like tissues were fabricated using autologous fibroblasts, macrophages, and plasma from SSc patients or healthy control (HC) donors. SSc tissues displayed increased dermal thickness and contractility, as well as increased α-SMA staining. Single-cell transcriptomic and epigenomic analyses identified keratinocytes, macrophages, and five populations of fibroblasts (labeled FB1 - 5). Notably, FB1 APOE-expressing fibroblasts were 12-fold enriched in SSc tissues and were characterized by high EGR1 motif accessibility. Pseudotime analysis suggests that FB1 fibroblasts differentiate from a TGF-β1-responsive fibroblast population and ligand-receptor analysis indicates that the FB1 fibroblasts are active in macrophage crosstalk via soluble ligands including FGF2 and APP. These findings provide characterization of the 3D skin-like model at single cell resolution and establish that it recapitulates subsets of fibroblasts and macrophage phenotypes observed in skin biopsies.
Collapse
|
23
|
Yeyeodu S, Hanafi D, Webb K, Laurie NA, Kimbro KS. Population-enriched innate immune variants may identify candidate gene targets at the intersection of cancer and cardio-metabolic disease. Front Endocrinol (Lausanne) 2024; 14:1286979. [PMID: 38577257 PMCID: PMC10991756 DOI: 10.3389/fendo.2023.1286979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/07/2023] [Indexed: 04/06/2024] Open
Abstract
Both cancer and cardio-metabolic disease disparities exist among specific populations in the US. For example, African Americans experience the highest rates of breast and prostate cancer mortality and the highest incidence of obesity. Native and Hispanic Americans experience the highest rates of liver cancer mortality. At the same time, Pacific Islanders have the highest death rate attributed to type 2 diabetes (T2D), and Asian Americans experience the highest incidence of non-alcoholic fatty liver disease (NAFLD) and cancers induced by infectious agents. Notably, the pathologic progression of both cancer and cardio-metabolic diseases involves innate immunity and mechanisms of inflammation. Innate immunity in individuals is established through genetic inheritance and external stimuli to respond to environmental threats and stresses such as pathogen exposure. Further, individual genomes contain characteristic genetic markers associated with one or more geographic ancestries (ethnic groups), including protective innate immune genetic programming optimized for survival in their corresponding ancestral environment(s). This perspective explores evidence related to our working hypothesis that genetic variations in innate immune genes, particularly those that are commonly found but unevenly distributed between populations, are associated with disparities between populations in both cancer and cardio-metabolic diseases. Identifying conventional and unconventional innate immune genes that fit this profile may provide critical insights into the underlying mechanisms that connect these two families of complex diseases and offer novel targets for precision-based treatment of cancer and/or cardio-metabolic disease.
Collapse
Affiliation(s)
- Susan Yeyeodu
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
- Charles River Discovery Services, Morrisville, NC, United States
| | - Donia Hanafi
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
| | - Kenisha Webb
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Nikia A. Laurie
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
| | - K. Sean Kimbro
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
24
|
Enzer NA, Chiles J, Mason S, Shirahata T, Castro V, Regan E, Choi B, Yuan NF, Diaz AA, Washko GR, McDonald ML, Estépar RSJ, Ash SY. Proteomics and Machine Learning in the Prediction and Explanation of Low Pectoralis Muscle Area. RESEARCH SQUARE 2024:rs.3.rs-3957125. [PMID: 38496412 PMCID: PMC10942559 DOI: 10.21203/rs.3.rs-3957125/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Low muscle mass is associated with numerous adverse outcomes independent of other associated comorbid diseases. We aimed to predict and understand an individual's risk for developing low muscle mass using proteomics and machine learning. We identified 8 biomarkers associated with low pectoralis muscle area (PMA). We built 3 random forest classification models that used either clinical measures, feature selected biomarkers, or both to predict development of low PMA. The area under the receiver operating characteristic curve for each model was: clinical-only = 0.646, biomarker-only = 0.740, and combined = 0.744. We displayed the heterogenetic nature of an individual's risk for developing low PMA and identified 2 distinct subtypes of participants who developed low PMA. While additional validation is required, our methods for identifying and understanding individual and group risk for low muscle mass could be used to enable developments in the personalized prevention of low muscle mass.
Collapse
|
25
|
Başpinar O, Elibol A, Koçer D, Tokmak TT, Doğan S, Dizdar OS. Evaluation of the relationship between atherosclerosis and Helicobacter pylori infection with measurement of growth differentiation factor 15 and atherosclerosis indicators in adults with no comorbidity. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:51-59. [PMID: 37858436 DOI: 10.1016/j.arteri.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/30/2023] [Accepted: 09/19/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND The aim of this study was to investigate presence of subclinical atherosclerosis by measuring carotid intima-media thickness (CIMT) in patients with Helicobacter pylori (HP) and to assess effects of HP on atherosclerosis by evaluating markers of atherosclerosis and blood growth differentiation factor (GDF-15) levels. MATERIALS AND METHODS This cross-sectional study included 59 patients without comorbid disease who had HP and 30 healthy controls without HP in upper endoscopic biopsy. In order to assess atherosclerosis, the CIMT measurement was performed by sonography. Serum GDF-15 level was measured by ELISA method. In all patients, atherosclerosis markers were recorded. Atherogenic indices were calculated, including Castelli risk index I and II (TG/HDL-c and LDL-c/HDL-c, respectively), plasma atherogenic index (PAI; log TG/HDL-c), non-HDL-c (TH-HDL-c) and atherogenic coefficient (AC; non-HDL-HDL-c). RESULTS The GDF-15 level and CIMT were significantly higher in HP-positive group when compared to HP-negative group (p≤0.001). There was a significant correlation between serum GDF-15 level and CIMT (r=0.445; p≤0.001). There was no correlation between other atherosclerosis markers and serum GDF-15 level or CIMT. The bacterial intensity on endoscopic specimen was only correlated with CIMT (p<0.001). Vitamin B12 and D levels were comparable among groups. CONCLUSION This study suggested that there was a correlation between GDF-15 level and subclinical atherosclerosis development in patients with HP. However, GDF-15 level, which was found to be elevated while atherogenic indices were normal, can be an earlier marker for subclinical atherosclerosis.
Collapse
Affiliation(s)
- Osman Başpinar
- Department of Internal Medicine, Kayseri City Training and Research Hospital, Kayseri, Turkey
| | - Ayça Elibol
- Department of Internal Medicine, Kayseri City Training and Research Hospital, Kayseri, Turkey
| | - Derya Koçer
- Department of Medical Biochemistry, Kayseri City Training and Research Hospital, Kayseri, Turkey
| | - Turgut Tursem Tokmak
- Department of Radiology, Kayseri City Training and Research Hospital, Kayseri, Turkey
| | - Serkan Doğan
- Department of Gastroenterology, Kayseri City Training and Research Hospital, Kayseri, Turkey
| | - Oğuzhan Sıtkı Dizdar
- Department of Internal Medicine, Kayseri City Training and Research Hospital, Kayseri, Turkey.
| |
Collapse
|
26
|
Shibasaki I, Otani N, Ouchi M, Fukuda T, Matsuoka T, Hirota S, Yokoyama S, Kanazawa Y, Kato T, Shimizu R, Tezuka M, Takei Y, Tsuchiya G, Saito S, Konishi T, Ogata K, Toyoda S, Fukuda H, Nakajima T. Utility of growth differentiation factor-15 as a predictor of cardiovascular surgery outcomes: Current research and future directions. J Cardiol 2024; 83:211-218. [PMID: 37648079 DOI: 10.1016/j.jjcc.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/16/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
In a world increasingly confronted by cardiovascular diseases (CVDs) and an aging population, accurate risk assessment prior to cardiac surgery is critical. Although effective, traditional risk calculators such as the Japan SCORE, Society of Thoracic Surgeons score, and EuroSCORE II may not completely capture contemporary risks, particularly due to emerging factors such as frailty and sarcopenia. These calculators often focus on regional and ethnic specificity and rely heavily on evaluations based on age and underlying diseases. Growth differentiation factor-15 (GDF-15) is a stress-responsive cytokine that has been identified as a potential biomarker for sarcopenia and a tool for future cardiac risk assessment. Preoperative plasma GDF-15 levels have been associated with preoperative, intraoperative, and postoperative factors and short- and long-term mortality rates in patients undergoing cardiac surgery. Increased plasma GDF-15 levels have prognostic significance, having been correlated with the use of cardiopulmonary bypass during surgery, amount of bleeding, postoperative acute kidney injury, and intensive care unit stay duration. Notably, the inclusion of preoperative levels of GDF-15 in risk stratification models enhances their predictive value, especially when compared with those of the N-terminal prohormone of brain natriuretic peptide, which does not lead to reclassification. Thus, this review examines traditional risk assessments for cardiac surgery and the role of the novel biomarker GDF-15. This study acknowledges that the relationship between patient outcomes and elevated GDF-15 levels is not limited to CVDs or cardiac surgery but can be associated with variable diseases, including diabetes and cancer. Moreover, the normal range of GDF-15 is not well defined. Given its promise for improving patient care and outcomes in cardiovascular surgery, future research should explore the potential of GDF-15 as a biomarker for postoperative outcomes and target therapeutic intervention.
Collapse
Affiliation(s)
- Ikuko Shibasaki
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan.
| | - Naoyuki Otani
- Department of Cardiology, Dokkyo Medical University, Nikko Medical Center, Nikko, Japan
| | - Motoshi Ouchi
- Department of Pharmacology and Toxicology, Dokkyo Medical University, School of Medicine, Mibu, Japan; Department of Health Promotion in Nursing and Midwifery, Innovative Nursing for Life Course, Chiba University Graduate School of Nursing, Chiba, Japan
| | - Taira Fukuda
- Department of Liberal Arts and Human Development, Kanagawa University of Human Services, Yokosuka, Japan
| | - Taiki Matsuoka
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Shotaro Hirota
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Shohei Yokoyama
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Yuta Kanazawa
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Takashi Kato
- Department of Cardiovascular Surgery, Maebashi Red Cross Hospital, Maebashi, Japan
| | - Riha Shimizu
- Department of Cardiovascular Surgery, Maebashi Red Cross Hospital, Maebashi, Japan
| | - Masahiro Tezuka
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Yusuke Takei
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Go Tsuchiya
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Shunsuke Saito
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Taisuke Konishi
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Koji Ogata
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Hirotsugu Fukuda
- Department of Cardiac and Vascular Surgery, Dokkyo Medical University, School of Medicine, Mibu, Japan
| | - Toshiaki Nakajima
- Department of Cardiovascular Medicine, Dokkyo Medical University, School of Medicine, Mibu, Japan
| |
Collapse
|
27
|
Wang SF, Chang YL, Liu TY, Huang KH, Fang WL, Li AFY, Yeh TS, Hung GY, Lee HC. Mitochondrial dysfunction decreases cisplatin sensitivity in gastric cancer cells through upregulation of integrated stress response and mitokine GDF15. FEBS J 2024; 291:1131-1150. [PMID: 37935441 DOI: 10.1111/febs.16992] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/18/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Gastric neoplasm is a high-mortality cancer worldwide. Chemoresistance is the obstacle against gastric cancer treatment. Mitochondrial dysfunction has been observed to promote malignant progression. However, the underlying mechanism is still unclear. The mitokine growth differentiation factor 15 (GDF15) is a significant biomarker for mitochondrial disorder and is activated by the integrated stress response (ISR) pathway. The serum level of GDF15 was found to be correlated with the poor prognosis of gastric cancer patients. In this study, we found that high GDF15 protein expression might increase disease recurrence in adjuvant chemotherapy-treated gastric cancer patients. Moreover, treatment with mitochondrial inhibitors, especially oligomycin (a complex V inhibitor) and salubrinal (an ISR activator), respectively, was found to upregulate GDF15 and enhance cisplatin insensitivity of human gastric cancer cells. Mechanistically, it was found that the activating transcription factor 4-C/EBP homologous protein pathway has a crucial function in the heightened manifestation of GDF15. In addition, reactive oxygen species-activated general control nonderepressible 2 mediates the oligomycin-induced ISR, and upregulates GDF15. The GDF15-glial cell-derived neurotrophic factor family receptor a-like-ISR-cystine/glutamate transporter-enhanced glutathione production was found to be involved in cisplatin resistance. These results suggest that mitochondrial dysfunction might enhance cisplatin insensitivity through GDF15 upregulation, and targeting mitokine GDF15-ISR regulation might be a strategy against cisplatin resistance of gastric cancer.
Collapse
Affiliation(s)
- Sheng-Fan Wang
- Department of Pharmacy, Taipei Veterans General Hospital, Taiwan
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taiwan
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yuh-Lih Chang
- Department of Pharmacy, Taipei Veterans General Hospital, Taiwan
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Pharmacy, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Yu Liu
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kuo-Hung Huang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan
- Department of Surgery, Gastric Cancer Medical Center, Taipei Veterans General Hospital, Taiwan
| | - Wen-Liang Fang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan
- Department of Surgery, Gastric Cancer Medical Center, Taipei Veterans General Hospital, Taiwan
| | - Anna Fen-Yau Li
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Anatomical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Tien-Shun Yeh
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Giun-Yi Hung
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Taipei Veterans General Hospital, Taiwan
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Pharmacy, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
28
|
Zhao F, Fan Z, Jia R, Liu Q, Wang M, Sui J, Liu H. Mesenchymal Stem Cells Accelerate Recovery of Acetic Acid-Induced Chronic Gastric Ulcer by Regulating Ekt/Akt/TRIM29 Axis. Stem Cells Int 2024; 2024:6202123. [PMID: 38213743 PMCID: PMC10781525 DOI: 10.1155/2024/6202123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/08/2023] [Accepted: 12/07/2023] [Indexed: 01/13/2024] Open
Abstract
Chronic gastric ulcer (CGU), a prevalent digestive disease, has a high incidence and is seriously harmful to human health. Mesenchymal stem cells (MSCs) have been proven to have beneficial therapeutic effects in many human diseases. Here, a CGU model induced by acetic acid in mice was used to evaluate the repair effects and potential mechanism of human umbilical cord-derived MSCs (hUC-MSCs) and hUC-MSCs derived conditioned medium (hUC-MSC-CM). We found that hUC-MSCs and hUC-MSC-CM treatment significantly repaired morphological characteristics of CGU, improved proliferation and decreased apoptosis of gastric cells, and promoted the generation of new blood vessels in granulation tissues. In addition, we could detect the homing of MSCs in gastric tissue, and MSCs may differentiate into Lgr5-positive cells. As well as this, in vitro experiments showed that hUC-MSC-CM could promote cell proliferation, stimulate cell cycle progression, and reduce the incidence of apoptosis. The transcriptome of cells and the iTRAQ proteome of gastric tissues suggest that MSCs may play a therapeutic role by increasing the expression of TRIM29. Additionally, it was found that knocking down TRIM29 significantly decreased the ameliorative effects of hUC-MSC-CM on cell apoptosis. As a result of further molecular experiments, it was found that TRIM29 is capable of phosphorylating Erk/Akt in specific cell type. As a whole, it appears that hUC-MSCs can be an effective therapeutic approach for promoting gastric ulcer healing and may exert therapeutic effects in the form of paracrine and differentiation into gastric cells.
Collapse
Affiliation(s)
- Feiyue Zhao
- Handan Pharmaceutical Co. Ltd., Handan, Hebei Province, China
- Key Laboratory of Chinese Medicine for Gastric Medicine, Handan, Hebei Province, China
| | - Zhibin Fan
- Handan Pharmaceutical Co. Ltd., Handan, Hebei Province, China
| | - Ruikang Jia
- Handan Pharmaceutical Co. Ltd., Handan, Hebei Province, China
| | - Qichao Liu
- Handan Pharmaceutical Co. Ltd., Handan, Hebei Province, China
| | - Menglei Wang
- Key Laboratory of Chinese Medicine for Gastric Medicine, Handan, Hebei Province, China
| | - Jianliang Sui
- School of Life Science and Food Engineering, Hebei University of Engineering, Handan, Hebei Province, China
| | - Huiyun Liu
- Handan Pharmaceutical Co. Ltd., Handan, Hebei Province, China
- Key Laboratory of Chinese Medicine for Gastric Medicine, Handan, Hebei Province, China
| |
Collapse
|
29
|
Shin HE, Won CW, Kim M. Development of multiple biomarker panels for prediction of sarcopenia in community-dwelling older adults. Arch Gerontol Geriatr 2023; 115:105115. [PMID: 37422966 DOI: 10.1016/j.archger.2023.105115] [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: 05/01/2023] [Revised: 06/22/2023] [Accepted: 06/29/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND It is required to consider multiple biomarkers simultaneously to predict sarcopenia and to understand its complex pathological mechanisms. This study aimed to develop multiple biomarker panels for predicting sarcopenia in older adults and to further examine its association with the incidence of sarcopenia. METHODS A total of 1,021 older adults were selected from the Korean Frailty and Aging Cohort Study. Sarcopenia was defined by the Asian Working Group for Sarcopenia 2019 criteria. Among the 14 biomarker candidates at baseline, eight biomarkers that could optimally detect individuals with sarcopenia were selected to develop a multi-biomarker risk score (range from 0 to 10). The utility of developed multi-biomarker risk score in discriminating sarcopenia was investigated using receiver operating characteristic (ROC) analysis. RESULTS The multi-biomarker risk score had an area under the ROC curve (AUC) of 0.71 with an optimal cut-off of 1.76 score, which was significantly higher than all single biomarkers with AUC of <0.7 (all, p<0.01). During the two-year follow-up, the incidence of sarcopenia was 11.1%. Continuous multi-biomarker risk score was positively associated with incidence of sarcopenia after adjusting confounders (odds ratio [OR]=1.63; 95% confidence interval [CI]=1.23-2.17). Participants with a high risk score had higher odds of sarcopenia than those with a low risk score (OR=1.82; 95% CI=1.04-3.19). CONCLUSIONS Multi-biomarker risk score, which was a combination of eight biomarkers with different pathophysiologies, better discriminated the presence of sarcopenia than a single biomarker, and it could further predict the incidence of sarcopenia over two years in older adults.
Collapse
Affiliation(s)
- Hyung Eun Shin
- Department of Biomedical Science and Technology, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Chang Won Won
- Elderly Frailty Research Center, Department of Family Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Korea.
| | - Miji Kim
- Department of Biomedical Science and Technology, College of Medicine, East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Korea.
| |
Collapse
|
30
|
Kobayashi S, Yamazaki H, Imamura T, Fujioka H, Kakeshita K, Koike T, Kinugawa K. Implication of serum growth differentiation factor-15 level in patients with renal diseases. Int Urol Nephrol 2023; 55:2935-2941. [PMID: 37010737 PMCID: PMC10069349 DOI: 10.1007/s11255-023-03580-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 03/26/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND The synthesis of growth differentiation factor-15 (GDF-15) is induced by inflammation, hypoxia, and oxidative stress and is receiving great interest as a predictive biomarker for cardiovascular disease. However, its detailed impact on patients with renal disease remains uncertain. METHODS Patients who underwent renal biopsy for evaluation of renal disease between 2012 and 2017 in our institute were prospectively included. Serum GDF-15 levels were measured and its association with baseline characteristics and its impact on the 3-year composites of renal prognosis (composites of > 1.5 folds of serum creatinine and renal replacement therapy) were investigated. RESULTS A total of 110 patients (64 [42, 73] years old, 61 men) were included. The median serum GDF-15 level at baseline was 1885 (998, 3496) pg/mL. A higher serum GDF-15 level was associated with comorbidities including diabetes mellitus, anemia, renal impairment, and pathologic features including crescent formation, hyaline degeneration, and interstitial fibrosis (p < 0.05 for all). Serum GDF-15 level was a significant predictor of 3-year composite renal outcomes with an odds ratio per 100 pg/mL of 1.072 (95% confidence interval 1.001-1.103, p = 0.036) after adjustment for potential confounders. CONCLUSIONS Serum GDF-15 levels were associated with several renal pathological features and renal prognosis in patients with renal diseases.
Collapse
Affiliation(s)
- Shiori Kobayashi
- The Second Department of Internal Medicine, Toyama University, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Hidenori Yamazaki
- The Second Department of Internal Medicine, Toyama University, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Teruhiko Imamura
- The Second Department of Internal Medicine, Toyama University, 2630 Sugitani, Toyama, 930-0194, Japan.
| | - Hayato Fujioka
- The Second Department of Internal Medicine, Toyama University, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Kota Kakeshita
- The Second Department of Internal Medicine, Toyama University, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Tsutomu Koike
- The Second Department of Internal Medicine, Toyama University, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Koichiro Kinugawa
- The Second Department of Internal Medicine, Toyama University, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
31
|
Patil K, Naigaonkar A, Hinduja I, Mukherjee S. Transcriptomic profile of GLCs of PCOS women highlights metabolic dysregulation as a plausible contributor to PCOS pathophysiology. Reprod Biol 2023; 23:100787. [PMID: 37467532 DOI: 10.1016/j.repbio.2023.100787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/22/2023] [Accepted: 07/08/2023] [Indexed: 07/21/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a complex heterogeneous disorder with reproductive and metabolic consequences whose aetiology is still elusive. To understand the cellular mechanisms that potentially govern follicular defect in women with PCOS, we performed transcriptomic profiles of granulosa-lutein cells (GLCs) by RNA-Seq analysis. We found differential expression of 876 genes in GLCs between PCOS and controls that belonged to various processes such as cell cycle, extracellular matrix organization, angiogenesis, oxidative stress, metabolism, etc. that support folliculogenesis, oocyte development, and maturation. The cross-talk between oocyte and GLCs is a fundamental cornerstone in determining oocyte quality and highly interlinked pathways of metabolism and redox homeostasis may influence this. We found several genes involved in the metabolism of carbohydrates, nucleotides, cholesterol, and lipids were dysregulated, which may impair the supply of metabolites to the growing oocyte, affecting oocyte development and competence. Additionally, high metabolic activity during folliculogenesis may augment oxidative damage to cells and macromolecules if not counter-balanced. We observed dysregulation of redox homeostasis and AGE-RAGE signalling in the follicular environment. Among the validated genes, prokineticin-1 and growth differentiation factor-15 were found to be negatively regulated, while, S100, calcium-binding protein A9 and angiomotin-like-2 were positively regulated in GLCs of women with PCOS. Comparing our data with previously published relevant transcriptomic studies showed metabolic, cytokine-cytokine receptor interaction, IL-17, and chemokine signalling pathways were most commonly affected in PCOS. Overall, this data can provide insights into mechanisms contributing to PCOS pathophysiology and can be explored as potential indicators for oocyte/embryo quality in IVF settings.
Collapse
Affiliation(s)
- Krutika Patil
- Department of Molecular Endocrinology, National Institute for Research in Reproductive and Child Health, Indian Council of Medical Research, J.M. Street, Parel, Mumbai 400012, India
| | - Aalaap Naigaonkar
- Department of Molecular Endocrinology, National Institute for Research in Reproductive and Child Health, Indian Council of Medical Research, J.M. Street, Parel, Mumbai 400012, India
| | - Indira Hinduja
- P. D. Hinduja National Hospital and Medical Research Centre, Mahim, Mumbai 400016, India
| | - Srabani Mukherjee
- Department of Molecular Endocrinology, National Institute for Research in Reproductive and Child Health, Indian Council of Medical Research, J.M. Street, Parel, Mumbai 400012, India.
| |
Collapse
|
32
|
Sluka M, Hutyra M, Nykl R, Ostransky J, Furst T, Petrova P, Precek J, Hudec S, Taborsky M. Risk stratification using growth differentiation factor 15 in patients undergoing transcatheter aortic valve implantation. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2023; 167:263-271. [PMID: 35416185 DOI: 10.5507/bp.2022.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/24/2022] [Indexed: 11/23/2022] Open
Abstract
AIMS Growth differentiation factor 15 (GDF15) shows potential predictive value in various cardiac conditions. We investigated relationships between GDF15 and clinical or procedural outcomes in patients with severe aortic stenosis undergoing transcatheter aortic valve implantation (TAVI) in order to propose clinically useful predictive risk stratification model. METHODS This prospective single-center registry enrolled 88 consecutive patients with severe symptomatic aortic stenosis treated with TAVI. Clinical parameters were collected and biomarkers including GDF-15 were measured within 24 h before TAVI. All relevant clinical outcomes according to the Valve Academic Research Consortium-2 were collected over the follow-up period. RESULTS The cohort included 52.3% of females. The mean age of study participants was 81 years; the mean Society of Thoracic Surgeons (STS) score and logistic EuroSCORE were 3.6% and 15.4%, respectively. The mortality over the entire follow-up period was 10.2%; no death was observed within the first 30 days following TAVI. Univariate analysis showed significant associations between GDF15 and mortality (P=0.0006), bleeding (P=0.0416) and acute kidney injury (P=0.0399). A standard multivariate logistic regression model showed GDF-15 as the only significant predictor of mortality (P=0.003); the odds ratio corresponding to an increase in GDF15 of 1000 pg/mL was 1.22. However, incremental predictive value was not observed when the STS score was combined with GDF15 in this predictive model. CONCLUSIONS Based on our observations, preprocedural elevated GDF15 levels are associated with increased mortality and demonstrate their additional value in predicting adverse clinical outcomes in a TAVI population.
Collapse
Affiliation(s)
- Martin Sluka
- Department of Internal Medicine I - Cardiology, University Hospital Olomouc, I. P. Pavlova 6, 779 00, Czech Republic
| | - Martin Hutyra
- Department of Internal Medicine I - Cardiology, University Hospital Olomouc, I. P. Pavlova 6, 779 00, Czech Republic
| | - Radomir Nykl
- Department of Internal Medicine I - Cardiology, University Hospital Olomouc, I. P. Pavlova 6, 779 00, Czech Republic
| | - Jiri Ostransky
- Department of Internal Medicine I - Cardiology, University Hospital Olomouc, I. P. Pavlova 6, 779 00, Czech Republic
| | - Tomas Furst
- Department of Mathematical Analysis and Applications of Mathematics, Faculty of Science, Palacky University Olomouc, Czech Republic
| | - Pavla Petrova
- Department of Clinical Biochemistry, University Hospital Olomouc, I. P. Pavlova 6, 779 00, Czech Republic
| | - Jan Precek
- Department of Internal Medicine I - Cardiology, University Hospital Olomouc, I. P. Pavlova 6, 779 00, Czech Republic
| | - Stepan Hudec
- Department of Internal Medicine I - Cardiology, University Hospital Olomouc, I. P. Pavlova 6, 779 00, Czech Republic
| | - Milos Taborsky
- Department of Internal Medicine I - Cardiology, University Hospital Olomouc, I. P. Pavlova 6, 779 00, Czech Republic
| |
Collapse
|
33
|
Ast T, Wang H, Marutani E, Nagashima F, Malhotra R, Ichinose F, Mootha VK. Continuous, but not intermittent, regimens of hypoxia prevent and reverse ataxia in a murine model of Friedreich's ataxia. Hum Mol Genet 2023; 32:2600-2610. [PMID: 37260376 PMCID: PMC10407700 DOI: 10.1093/hmg/ddad091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 05/08/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023] Open
Abstract
Friedreich's ataxia (FA) is a devastating, multi-systemic neurodegenerative disease affecting thousands of people worldwide. We previously reported that oxygen is a key environmental variable that can modify FA pathogenesis. In particular, we showed that chronic, continuous normobaric hypoxia (11% FIO2) prevents ataxia and neurological disease in a murine model of FA, although it did not improve cardiovascular pathology or lifespan. Here, we report the pre-clinical evaluation of seven 'hypoxia-inspired' regimens in the shFxn mouse model of FA, with the long-term goal of designing a safe, practical and effective regimen for clinical translation. We report three chief results. First, a daily, intermittent hypoxia regimen (16 h 11% O2/8 h 21% O2) conferred no benefit and was in fact harmful, resulting in elevated cardiac stress and accelerated mortality. The detrimental effect of this regimen is likely owing to transient tissue hyperoxia that results when daily exposure to 21% O2 combines with chronic polycythemia, as we could blunt this toxicity by pharmacologically inhibiting polycythemia. Second, we report that more mild regimens of chronic hypoxia (17% O2) confer a modest benefit by delaying the onset of ataxia. Third, excitingly, we show that initiating chronic, continuous 11% O2 breathing once advanced neurological disease has already started can rapidly reverse ataxia. Our studies showcase both the promise and limitations of candidate hypoxia-inspired regimens for FA and underscore the need for additional pre-clinical optimization before future translation into humans.
Collapse
Affiliation(s)
- Tslil Ast
- Broad Institute, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Hong Wang
- Broad Institute, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Eizo Marutani
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Fumiaki Nagashima
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Rajeev Malhotra
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Fumito Ichinose
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Vamsi K Mootha
- Broad Institute, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
34
|
Sawalha K, Norgard NB, Drees BM, López-Candales A. Growth Differentiation Factor 15 (GDF-15), a New Biomarker in Heart Failure Management. Curr Heart Fail Rep 2023; 20:287-299. [PMID: 37289373 DOI: 10.1007/s11897-023-00610-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
The emergence of biomarkers across medicine's subspecialties continues to evolve. In essence, a biomarker is a biological observation that clearly substitutes a clinical endpoint or intermediate outcome not only are more difficult to observe but also, biomarkers are easier, less expensive and could be measured over shorter periods. In general, biomarkers are versatile and not only used for disease screening and diagnosis but, most importantly, for disease characterization, monitoring, and determination of prognosis as well as individualized therapeutic responses. Obviously, heart failure (HF) is no exception to the use of biomarkers. Currently, natriuretic peptides are the most used biomarkers for both diagnosis and prognostication, while their role in the monitoring of treatment is still debatable. Although several other new biomarkers are currently under investigation regarding diagnosis and determination of prognosis, none of them are specific for HF, and none are recommended for routine clinical use at present. However, among these emerging biomarkers, we would like to highlight the potential for growth differentiation factor (GDF)-15 as a plausible new biomarker that could be helpful in providing prognostic information regarding HF morbidity and mortality.
Collapse
Affiliation(s)
- Khalid Sawalha
- Cardiometabolic Medicine Fellowship, University of Missouri-Kansas City, Kansas City, MO, USA.
- Section of Cardiovascular Medicine, University Health, Truman Medical Center, University of Missouri-Kansas City, 2301 Holmes Street, Kansas City, MO, 64108, USA.
| | - Nicholas B Norgard
- Department of Medicine, University Health Truman Medical Center, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Betty M Drees
- Department(s) of Internal Medicine, Biomedical and Health Informatics, Section of Endocrinology, UMKC School of Medicine, Kansas City, MO, USA
| | - Angel López-Candales
- Section of Cardiovascular Medicine, University Health, Truman Medical Center, University of Missouri-Kansas City, 2301 Holmes Street, Kansas City, MO, 64108, USA
| |
Collapse
|
35
|
Jigoranu RA, Roca M, Costache AD, Mitu O, Oancea AF, Miftode RS, Haba MȘC, Botnariu EG, Maștaleru A, Gavril RS, Trandabat BA, Chirica SI, Haba RM, Leon MM, Costache II, Mitu F. Novel Biomarkers for Atherosclerotic Disease: Advances in Cardiovascular Risk Assessment. Life (Basel) 2023; 13:1639. [PMID: 37629496 PMCID: PMC10455542 DOI: 10.3390/life13081639] [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: 06/11/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Atherosclerosis is a significant health concern with a growing incidence worldwide. It is directly linked to an increased cardiovascular risk and to major adverse cardiovascular events, such as acute coronary syndromes. In this review, we try to assess the potential diagnostic role of biomarkers in the early identification of patients susceptible to the development of atherosclerosis and other adverse cardiovascular events. We have collected publications concerning already established parameters, such as low-density lipoprotein cholesterol (LDL-C), as well as newer markers, e.g., apolipoprotein B (apoB) and the ratio between apoB and apoA. Additionally, given the inflammatory nature of the development of atherosclerosis, high-sensitivity c-reactive protein (hs-CRP) or interleukin-6 (IL-6) are also discussed. Additionally, newer publications on other emerging components linked to atherosclerosis were considered in the context of patient evaluation. Apart from the already in-use markers (e.g., LDL-C), emerging research highlights the potential of newer molecules in optimizing the diagnosis of atherosclerotic disease in earlier stages. After further studies, they might be fully implemented in the screening protocols.
Collapse
Affiliation(s)
- Raul-Alexandru Jigoranu
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Mihai Roca
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Alexandru-Dan Costache
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ovidiu Mitu
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Alexandru-Florinel Oancea
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Radu-Stefan Miftode
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Mihai Ștefan Cristian Haba
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Eosefina Gina Botnariu
- Department of Internal Medicine II, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania;
- Department of Diabetes, Nutrition and Metabolic Diseases, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Alexandra Maștaleru
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Radu-Sebastian Gavril
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Bogdan-Andrei Trandabat
- Department of Surgery II, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania;
- Department of Orthopedics and Trauma, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Sabina Ioana Chirica
- Faculty of General Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (S.I.C.); (R.M.H.)
| | - Raluca Maria Haba
- Faculty of General Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (S.I.C.); (R.M.H.)
| | - Maria Magdalena Leon
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Irina-Iuliana Costache
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Florin Mitu
- Department of Medical Specialties I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (R.-A.J.); (O.M.); (A.-F.O.); (R.-S.M.); (M.Ș.C.H.); (A.M.); (R.-S.G.); (M.M.L.); (I.-I.C.); (F.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
- Romanian Academy of Medical Sciences, 030167 Bucharest, Romania
- Romanian Academy of Scientists, 050045 Bucharest, Romania
| |
Collapse
|
36
|
Zhou Z, Liu H, Ju H, Chen H, Jin H, Sun M. Circulating GDF-15 in relation to the progression and prognosis of chronic kidney disease: A systematic review and dose-response meta-analysis. Eur J Intern Med 2023; 110:77-85. [PMID: 36740468 DOI: 10.1016/j.ejim.2023.01.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/26/2022] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) typically exhibit circulating growth differentiation factor-15 (GDF-15) at high levels. This meta-analysis aimed to evaluate the potential value of GDF-15 in predicting CKD progression and prognosis. Furthermore, when sufficient information was provided, the dose-response correlation was studied. METHODS Studies were searched in Web of Science, Embase, and PubMed from inception until November 2022. By using random- or fixed-effects models, the pooled effect size was estimated in accordance with heterogeneity in existing research. RESULTS This study covered 14 studies from 12 articles with 7813 subjects participating in the research. CKD patients in the top GDF-15 tertile had notably higher risks of CKD progression (HR 2.60, 95% CI 2.06-3.27), all-cause mortality (HR 2.05, 95% CI 1.44-2.92), cardiovascular mortality (HR 2.82, 95% CI 1.85-4.30), and cardiovascular events (HR 2.74, 95% CI 2.21-3.40), as compared to CKD patients in the bottom tertile. In the dose-response study, the risks for CKD progression, all-cause death, cardiovascular death, and cardiovascular events were increased by 31% (HR 1.31, 95% CI 1.06-1.61), 44% (HR 1.44, 95% CI 1.08-1.92), 67% (HR 1.67, 95% CI 1.37-2.03), and 55% (HR 1.55, 95% CI 1.31-1.83), respectively, with per 1 ng/mL increase in GDF-15. The positive linear correlations between GDF-15 and CKD progression and prognosis in a certain GDF-15 concentration range of approximately 0-3 ng/mL were indicated by the dose-response curve. CONCLUSIONS Circulating GDF-15 independently predicted CKD progression and worse prognosis; however, the predicted correlations may fall into a specific range of GDF-15 concentrations.
Collapse
Affiliation(s)
- Zhongwei Zhou
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Jiangsu 224001, China
| | - Hongli Liu
- Department of Clinical Laboratory, Nantong Tumor Hospital, Tumor Hospital Affiliated to Nantong University, Jiangsu 226361, China
| | - Huixiang Ju
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Jiangsu 224001, China
| | - Hongmei Chen
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Jiangsu 224001, China
| | - Hao Jin
- Department of Blood Transfusion, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Jiangsu 224001, China.
| | - Mingzhong Sun
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Jiangsu 224001, China.
| |
Collapse
|
37
|
Joo M, Kim D, Lee MW, Lee HJ, Kim JM. GDF15 Promotes Cell Growth, Migration, and Invasion in Gastric Cancer by Inducing STAT3 Activation. Int J Mol Sci 2023; 24:2925. [PMID: 36769245 PMCID: PMC9917887 DOI: 10.3390/ijms24032925] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Growth differentiation factor 15 (GDF15) has been reported to play an important role in cancer and is secreted and involved in the progression of various cancers, including ovarian cancer, prostate cancer, and thyroid cancer. Nevertheless, the functional mechanism of GDF15 in gastric cancer is still unclear. Immunohistochemical staining was performed to estimate the expression of GDF15 in 178 gastric cancer tissues. The biological role and action mechanism of GDF15 were investigated by examining the effect of GDF15 knockdown in AGS and SNU216 gastric cancer cells. Here, we report that the high expression of GDF15 was associated with invasion depth (p = 0.002), nodal involvement (p = 0.003), stage III/IV (p = 0.01), lymphatic invasion (p = 0.05), and tumor size (p = 0.049), which are related to poor survival in gastric cancer patients. GDF15 knockdown induced G0/G1 cell cycle arrest and remarkably inhibited cell proliferation and reduced cell motility, migration, and invasion compared to the control. GDF15 knockdown inhibited the epithelial-mesenchymal transition by regulating the STAT3 phosphorylation signaling pathways. Taken together, our results indicate that GDF15 expression is associated with aggressive gastric cancer by promoting STAT3 phosphorylation, suggesting that the GDF15-STAT3 signaling axis is a potential therapeutic target against gastric cancer progression.
Collapse
Affiliation(s)
- Mina Joo
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Donghyun Kim
- Department of Pathology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Myung-Won Lee
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Hyo Jin Lee
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jin-Man Kim
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Pathology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
38
|
Imran M, Sachdeva G, Menon S, Das D, Davuluri S, Acharya K, Chaudhari U. Therapeutic metformin concentrations positively regulate proliferation in endometrial epithelial cells via mTOR activation and augmented mitochondrial strength. Can J Physiol Pharmacol 2023; 101:52-64. [PMID: 36322951 DOI: 10.1139/cjpp-2022-0307] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Metformin, an antidiabetic drug, has recently been repositioned in the treatment of several nondiabetic disorders, including reproductive disorders such as polycystic ovarian syndrome, where it improves endometrial functions. In vitro studies employing supratherapeutic concentrations (5-20 mmol/L) of metformin have reported antiproliferative effects on endometrial epithelial and stromal cells. However, animal and human studies have revealed that therapeutic serum concentrations of metformin range between 20 and 70 µmol/L. In the present study, the effect of therapeutic concentrations of metformin was studied on endometrial epithelial cells (EECs). Therapeutic concentrations of metformin induced proliferation in Ishikawa and HEC-1A cells. The proliferation of EECs was found to be mammalian target of rapamycin (mTOR) dependent. Interestingly, therapeutic metformin concentrations were not able to activate the classical AMP-activated protein kinase (AMPK) signaling. On the contrary, supratherapeutic metformin concentration (10 mmol/L) inhibited mTOR and activated AMPK signaling. Microarray analysis of metformin-treated HEC-1A cells revealed dose-dependent differential effects on biological pathways associated with translation, ribosomal RNA processing, mitochondrial translation, and cell proliferation. Therapeutic concentrations of metformin upregulated mitochondrial number as demonstrated by increased MitoTracker™ Red staining and enhanced succinate dehydrogenase expression; however, higher concentration (10 mmol/L) abrogated the same. Our results suggest that therapeutic concentrations of metformin augment mitochondrial strength and induce mTOR-dependent endometrial cell proliferation.
Collapse
Affiliation(s)
- M Imran
- Cell Physiology and Pathology Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Geetanjali Sachdeva
- Cell Physiology and Pathology Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Shyla Menon
- Stem Cell Biology Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Dhanjit Das
- Stem Cell Biology Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | | | - Kshitish Acharya
- Shodhaka Life Sciences Pvt. Ltd., Bengaluru, Karnataka, India.,Institute of Bioinformatics and Applied Biotechnology (IBAB), Bengaluru, Karnataka, India
| | - Uddhav Chaudhari
- Cell Physiology and Pathology Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| |
Collapse
|
39
|
Lu YC, Liu SL, Zhang YS, Liang F, Zhu XY, Xiao Y, Wang J, Ding C, Banerjee S, Yin JY, Ma QP. Association between growth differentiation factor 15 levels and gestational diabetes mellitus: A combined analysis. Front Endocrinol (Lausanne) 2023; 14:1084896. [PMID: 36742413 PMCID: PMC9895392 DOI: 10.3389/fendo.2023.1084896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE Gestational diabetes mellitus (GDM) is a common glucose metabolism disease occurs in pregnancy that affects both maternal and neonatal health. Recently, increasing studies have attached importance to the relationship between growth differentiation factor 15 (GDF-15) and GDM, but the results were inconclusive. Therefore, we conducted a meta-analysis to examine the association between GDF-15 and GDM. MATERIALS AND METHODS A systematical search was performed in Gene Expression Omnibus (GEO), PubMed and Google Scholar till Oct 27, 2022. We first calculated the mean and standard deviation of GDF-15 expression levels from the included eligible datasets and articles. Then, a meta-analysis was conducted to depict the difference in GDF-15 mRNA or GDF-15 protein expression between case and control groups by using conservative random effect model. Moreover, the potential publication bias was checked with the aid of Begg's test and Egger's test. Finally, sensitivity analyses were performed by changing the inclusion criteria. RESULTS In summary, 12 GEO datasets and 5 articles were enrolled in our study, including 789 GDM patients and 1202 non-GDM pregnant women. It was found that the expression levels of GDF-15 mRNA and GDF-15 protein in late pregnancy were significantly higher in GDM patients compared with non-GDM pregnant women, with the standard mean difference (SMD) and 95% confidence interval (95% CI) of 0.48 (0.14, 0.83) and 0.82 (0.32-1.33), respectively. Meanwhile, a slightly weakened association between GDF-15 protein levels and GDM was also observed in the middle pregnancy, with SMD (95% CI) of 0.53 (0.04-1.02). CONCLUSION In all, our results suggested that the expression levels of GDF-15 were significantly higher in GDM patients compared with non-GDM pregnant women, especially in the late pregnancy.
Collapse
Affiliation(s)
- Yi-Cheng Lu
- Taicang Affiliated Hospital of Soochow University, The First People’s Hospital of TaiCang, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Song-Liang Liu
- Taicang Affiliated Hospital of Soochow University, The First People’s Hospital of TaiCang, Soochow University, Suzhou, China
| | - Yu-Shan Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Fei Liang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Xiao-Yan Zhu
- Suzhou Center for Disease Prevention and Control, Suzhou, China
- Department of Sample Application and Management, Institute of Suzhou Biobank, Suzhou, China
| | - Yue Xiao
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jing Wang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Cong Ding
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Sudipta Banerjee
- Department of Endocrinology and Metabolism, Institute of Post-Graduate Medical Education and Research and Seth Sukhlal Karnani Memorial Hospital (IPGME & R and SSKM Hospital), Kolkata, India
| | - Jie-Yun Yin
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Suzhou, China
- *Correspondence: Jie-Yun Yin, ; Qiu-Ping Ma,
| | - Qiu-Ping Ma
- Taicang Affiliated Hospital of Soochow University, The First People’s Hospital of TaiCang, Soochow University, Suzhou, China
- *Correspondence: Jie-Yun Yin, ; Qiu-Ping Ma,
| |
Collapse
|
40
|
Xie B, Murali A, Vandevender AM, Chen J, Silva AG, Bello FM, Chuan B, Bahudhanapati H, Sipula I, Dedousis N, Shah FA, O'Donnell CP, Alder JK, Jurczak MJ. Hepatocyte-derived GDF15 suppresses feeding and improves insulin sensitivity in obese mice. iScience 2022; 25:105569. [PMID: 36465107 PMCID: PMC9708916 DOI: 10.1016/j.isci.2022.105569] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/15/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2022] Open
Abstract
Growth differentiation factor 15 (GDF15) is a stress-induced secreted protein whose circulating levels are increased in the context of obesity. Recombinant GDF15 reduces body weight and improves glycemia in obese models, which is largely attributed to the central action of GDF15 to suppress feeding and reduce body weight. Despite these advances in knowledge, the tissue-specific sites of GDF15 production during obesity are unknown, and the effects of modulating circulating GDF15 levels on insulin sensitivity have not been evaluated directly. Here, we demonstrate that hepatocyte Gdf15 expression is sufficient for changes in circulating levels of GDF15 during obesity and that restoring Gdf15 expression specifically in hepatocytes of Gdf15 knockout mice results in marked improvements in hyperinsulinemia, hepatic insulin sensitivity, and to a lesser extent peripheral insulin sensitivity. These data support that liver hepatocytes are the primary source of circulating GDF15 in obesity.
Collapse
Affiliation(s)
- Bingxian Xie
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anjana Murali
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amber M Vandevender
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeffrey Chen
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Agustin Gil Silva
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fiona M Bello
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Byron Chuan
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Harinath Bahudhanapati
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ian Sipula
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nikolaos Dedousis
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Faraaz A Shah
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher P O'Donnell
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan K Alder
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael J Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
41
|
Liao J, Gan Y, Peng M, Giri M, Yang S, Gu L, Li A, Xiao R, He C, Li Y, Bai Y, Xu L, Guo S. GDF15 alleviates the progression of benign tracheobronchial stenosis by inhibiting epithelial-mesenchymal transition and inactivating fibroblasts. Exp Cell Res 2022; 421:113410. [PMID: 36336027 DOI: 10.1016/j.yexcr.2022.113410] [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: 06/17/2022] [Revised: 09/23/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Benign tracheobronchial stenosis (BTS) is a fatal and incurable disease. Epithelial repair and matrix reconstruction play an important role in the wound repair process. If the interstitial context is not restored and stabilized in time, it can lead to pathological fibrosis. Here we attempted to identify cytokines that are involved in promoting wound repair. Growth differentiation factor 15 (GDF15) is a cytokine secreted by tracheal epithelial cells, which is indispensable for the growth of epithelial cells and inhibits the overgrowth of fibroblasts. GDF15 can counteract transforming growth factor-β (TGFβ1) stimulation of epithelial-mesenchymal transition (EMT) in tracheal epithelial cells and inhibit fibroblast activation via the TGFβ1-SMAD2/3 pathway. In a rat model of tracheal stenosis, GDF15 supplementation alleviated the degree of tracheal stenosis. These results suggest that GDF15 prevents fibroblast hyperactivation and promotes epithelial repair in injured trachea. GDF15 may be a potential therapy to improve benign tracheobronchial stenosis.
Collapse
Affiliation(s)
- Jiaxin Liao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiling Gan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Mingyu Peng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Mohan Giri
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Shu Yang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lei Gu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Anmao Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Rui Xiao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chunyan He
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yishi Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yang Bai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Li Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Shuliang Guo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
42
|
Liu X, Pan S, Xanthakis V, Vasan RS, Psaty BM, Austin TR, Newman AB, Sanders JL, Wu C, Tracy RP, Gerszten RE, Odden MC. Plasma proteomic signature of decline in gait speed and grip strength. Aging Cell 2022; 21:e13736. [PMID: 36333824 PMCID: PMC9741503 DOI: 10.1111/acel.13736] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/27/2022] [Accepted: 10/21/2022] [Indexed: 11/08/2022] Open
Abstract
The biological mechanisms underlying decline in physical function with age remain unclear. We examined the plasma proteomic profile associated with longitudinal changes in physical function measured by gait speed and grip strength in community-dwelling adults. We applied an aptamer-based platform to assay 1154 plasma proteins on 2854 participants (60% women, aged 76 years) in the Cardiovascular Health Study (CHS) in 1992-1993 and 1130 participants (55% women, aged 54 years) in the Framingham Offspring Study (FOS) in 1991-1995. Gait speed and grip strength were measured annually for 7 years in CHS and at cycles 7 (1998-2001) and 8 (2005-2008) in FOS. The associations of individual protein levels (log-transformed and standardized) with longitudinal changes in gait speed and grip strength in two populations were examined separately by linear mixed-effects models. Meta-analyses were implemented using random-effects models and corrected for multiple testing. We found that plasma levels of 14 and 18 proteins were associated with changes in gait speed and grip strength, respectively (corrected p < 0.05). The proteins most strongly associated with gait speed decline were GDF-15 (Meta-analytic p = 1.58 × 10-15 ), pleiotrophin (1.23 × 10-9 ), and TIMP-1 (5.97 × 10-8 ). For grip strength decline, the strongest associations were for carbonic anhydrase III (1.09 × 10-7 ), CDON (2.38 × 10-7 ), and SMOC1 (7.47 × 10-7 ). Several statistically significant proteins are involved in the inflammatory responses or antagonism of activin by follistatin pathway. These novel proteomic biomarkers and pathways should be further explored as future mechanisms and targets for age-related functional decline.
Collapse
Affiliation(s)
- Xiaojuan Liu
- Department of Epidemiology and Population HealthStanford University School of MedicineStanfordCaliforniaUSA
| | - Stephanie Pan
- Framingham Heart Study and Section of Preventive Medicine and EpidemiologyBoston University School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Vanessa Xanthakis
- Framingham Heart Study and Section of Preventive Medicine and EpidemiologyBoston University School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Ramachandran S. Vasan
- Framingham Heart Study and Section of Preventive Medicine and EpidemiologyBoston University School of MedicineBostonMassachusettsUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
- Section of Cardiovascular Medicine, Department of MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Systems and Population HealthUniversity of WashingtonSeattleWashingtonUSA
| | - Thomas R. Austin
- Department of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Anne B. Newman
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - Chenkai Wu
- Global Health Research CenterDuke Kunshan UniversityKunshanChina
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, The Robert Larner M.D. College of MedicineUniversity of VermontBurlingtonVermontUSA
- Department of Biochemistry, The Robert Larner M.D. College of MedicineUniversity of VermontBurlingtonVermontUSA
| | - Robert E. Gerszten
- Division of Cardiovascular MedicineBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Michelle C. Odden
- Department of Epidemiology and Population HealthStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
43
|
Tang W, Zhang Y, Cui S, Yi F. The Growth Factors: Potential Biomarkers and Therapeutic Targets in Kidney Diseases. KIDNEY DISEASES (BASEL, SWITZERLAND) 2022; 8:368-380. [PMID: 36466071 PMCID: PMC9710479 DOI: 10.1159/000526208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/17/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Kidney diseases are a prevalent health problem worldwide. Although substantial progress has been made in understanding the pathophysiology of kidney disease, currently there is no satisfactory clinical treatment available to prevent or treat kidney disease. Therefore, strategies to establish early diagnosis, identify the key molecules, and develop novel therapeutic interventions to slow the progression of kidney diseases and reduce their complications are encouraged. SUMMARY The growth factors play a crucial role in the development of kidney diseases. The altered levels of growth factors are usually detected in circulation and urine in the disease course. A growing body of studies has suggested that growth factors, receptors, and related regulators are promising biomarkers for the diagnosis and/or prognosis and potential therapeutic targets for the treatment of kidney diseases. In this review, we summarize recent advances in the potential applications of growth factors for diagnostic biomarkers and therapeutic targets in kidney diseases and highlight their performances in clinical trials. KEY MESSAGES Most diagnostic and therapeutic strategies targeting growth factors are still far from clinical implementation. The better understanding of growth factor-regulated pathophysiology and the progress of new intervention approaches are expected to facilitate the clinical translation of growth factor-based diagnosis and therapy of kidney diseases.
Collapse
Affiliation(s)
- Wei Tang
- Department of Pharmacology, The Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yufeng Zhang
- Department of Pharmacology, The Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Sijia Cui
- Department of Pharmacology, The Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Fan Yi
- Department of Pharmacology, The Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
44
|
Abstract
Single-pass transmembrane receptors (SPTMRs) represent a diverse group of integral membrane proteins that are involved in many essential cellular processes, including signal transduction, cell adhesion, and transmembrane transport of materials. Dysregulation of the SPTMRs is linked with many human diseases. Despite extensive efforts in past decades, the mechanisms of action of the SPTMRs remain incompletely understood. One major hurdle is the lack of structures of the full-length SPTMRs in different functional states. Such structural information is difficult to obtain by traditional structural biology methods such as X-ray crystallography and nuclear magnetic resonance (NMR). The recent rapid development of single-particle cryo-electron microscopy (cryo-EM) has led to an exponential surge in the number of high-resolution structures of integral membrane proteins, including SPTMRs. Cryo-EM structures of SPTMRs solved in the past few years have tremendously improved our understanding of how SPTMRs function. In this review, we will highlight these progresses in the structural studies of SPTMRs by single-particle cryo-EM, analyze important structural details of each protein involved, and discuss their implications on the underlying mechanisms. Finally, we also briefly discuss remaining challenges and exciting opportunities in the field.
Collapse
Affiliation(s)
- Kai Cai
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
| | - Xuewu Zhang
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Departments of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Corresponding Author: Xuewu Zhang, Department of pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Xiao-chen Bai
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Departments of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Corresponding Author: Xiao-chen Bai, Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| |
Collapse
|
45
|
BMP2 as a promising anticancer approach: functions and molecular mechanisms. Invest New Drugs 2022; 40:1322-1332. [PMID: 36040572 DOI: 10.1007/s10637-022-01298-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/22/2022] [Indexed: 10/14/2022]
Abstract
Bone morphogenetic protein 2 (BMP2), a pluripotent factor, is a member of the transforming growth factor-beta (TGF-β) superfamily and is implicated in embryonic development and postnatal homeostasis in tissues and organs. Experimental research in the contexts of physiology and pathology has indicated that BMP2 can induce macrophages to differentiate into osteoclasts and accelerate the osteolytic mechanism, aggravating cancer cell bone metastasis. Emerging studies have stressed the potent regulatory effect of BMP2 in cancer cell differentiation, proliferation, survival, and apoptosis. Complicated signaling networks involving multiple regulatory proteins imply the significant biological functions of BMP2 in cancer. In this review, we comprehensively summarized and discussed the current evidence related to the modulation of BMP2 in tumorigenesis and development, including evidence related to the roles and molecular mechanisms of BMP2 in regulating cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT), cancer angiogenesis and the tumor microenvironment (TME). All these findings suggest that BMP2 may be an effective therapeutic target for cancer and a new marker for assessing treatment efficacy.
Collapse
|
46
|
Chen YC, Wu CT, Chen JH, Tsai CF, Wu CY, Chang PC, Yeh WL. Diltiazem inhibits breast cancer metastasis via mediating growth differentiation factor 15 and epithelial-mesenchymal transition. Oncogenesis 2022; 11:48. [PMID: 35963873 PMCID: PMC9376069 DOI: 10.1038/s41389-022-00423-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 12/22/2022] Open
Abstract
Migration and metastasis commonly happen to triple-negative breast cancer (TNBC) patients with advanced diseases. In many studies, it has been suggested that epithelial-mesenchymal transition (EMT) is one of the key mechanisms triggering cancer metastasis. Accumulating evidence has proven that calcium channel blockers mediate cell motility. Therefore, we attempt to investigate the effects of diltiazem, which has been selected from several FDA-approved clinical calcium channel blockers, on EMT in TNBC. By using both mouse and human TNBC cell lines, we found that diltiazem decreases colony formation and cell migration in breast cancer cells. The expression of epithelial markers such as E-cadherin and ZO-1 were increased dose-dependently by diltiazem, while mesenchymal markers such as Snail and Twist were decreased. In addition, we found that the expression of growth differentiation factor-15 (GDF-15) was also increased by diltiazem. Administering recombinant GDF-15 also reverses EMT, inhibits colony formation and migration in breast cancer cells. Moreover, treatment with diltiazem in tumor-bearing mice also decreases cancer metastasis and nodule formation, with more GDF-15 expression in diltiazem-treated mice than saline-treated mice, respectively. These findings suggest that diltiazem regulates EMT and cell motility through elevating GDF-15 expression in breast cancers in vitro and in vivo.
Collapse
Affiliation(s)
- Yen-Chang Chen
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan
| | - Chen-Teng Wu
- Department of Surgery, China Medical University Hospital, No. 2, Yude Road, Taichung, 404332, Taiwan
| | - Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 88, Sec. 1, Fengxing Road, Taichung, 427213, Taiwan
| | - Cheng-Fang Tsai
- Department of Medical Laboratory Science and Biotechnology, Asia University, No.500 Lioufeng Road, Taichung, 413305, Taiwan
| | - Chen-Yun Wu
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan
| | - Pei-Chun Chang
- Department of Bioinformatics and Medical Engineering, Asia University, No.500 Lioufeng Road, Taichung, 413305, Taiwan
| | - Wei-Lan Yeh
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan.
- Department of Biochemistry, School of Medicine, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan.
| |
Collapse
|
47
|
Lemmelä S, Wigmore EM, Benner C, Havulinna AS, Ong RMY, Kempf T, Wollert KC, Blankenberg S, Zeller T, Peters JE, Salomaa V, Fritsch M, March R, Palotie A, Daly M, Butterworth AS, Kinnunen M, Paul DS, Matakidou A. Integrated analyses of growth differentiation factor-15 concentration and cardiometabolic diseases in humans. eLife 2022; 11:e76272. [PMID: 35916366 PMCID: PMC9391041 DOI: 10.7554/elife.76272] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 08/01/2022] [Indexed: 02/02/2023] Open
Abstract
Growth differentiation factor-15 (GDF15) is a stress response cytokine that is elevated in several cardiometabolic diseases and has attracted interest as a potential therapeutic target. To further explore the association of GDF15 with human disease, we conducted a broad study into the phenotypic and genetic correlates of GDF15 concentration in up to 14,099 individuals. Assessment of 772 traits across 6610 participants in FINRISK identified associations of GDF15 concentration with a range of phenotypes including all-cause mortality, cardiometabolic disease, respiratory diseases and psychiatric disorders, as well as inflammatory markers. A meta-analysis of genome-wide association studies (GWAS) of GDF15 concentration across three different assay platforms (n=14,099) confirmed significant heterogeneity due to a common missense variant (rs1058587; p.H202D) in GDF15, potentially due to epitope-binding artefacts. After conditioning on rs1058587, statistical fine mapping identified four independent putative causal signals at the locus. Mendelian randomisation (MR) analysis found evidence of a causal relationship between GDF15 concentration and high-density lipoprotein (HDL) but not body mass index (BMI). Using reverse MR, we identified a potential causal association of BMI on GDF15 (IVW pFDR = 0.0040). Taken together, our data derived from human population cohorts do not support a role for moderately elevated GDF15 concentrations as a causal factor in human cardiometabolic disease but support its role as a biomarker of metabolic stress.
Collapse
Affiliation(s)
- Susanna Lemmelä
- Institute for Molecular Medicine Finland, University of HelsinkiHelsinkiFinland
| | | | - Christian Benner
- Institute for Molecular Medicine Finland, University of HelsinkiHelsinkiFinland
| | - Aki S Havulinna
- Institute for Molecular Medicine Finland, University of HelsinkiHelsinkiFinland
- Finnish Institute for Health and WelfareHelsinkiFinland
| | - Rachel MY Ong
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of CambridgeCambridgeUnited Kingdom
| | - Tibor Kempf
- Department of Cardiology and Angiology, Hannover Medical SchoolHannoverGermany
| | - Kai C Wollert
- Department of Cardiology and Angiology, Hannover Medical SchoolHannoverGermany
| | - Stefan Blankenberg
- Clinic of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg-EppendorfHamburgGermany
- Population Health Research Department, University Heart and Vascular Center, University Medical Center Hamburg-EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
| | - Tanja Zeller
- Clinic of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg-EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- University Center of Cardiovascular Science, University Medical Center Hamburg-EppendorfHamburgGermany
| | - James E Peters
- Department of Immunology and Inflammation, Imperial College LondonLondonUnited Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of CambridgeCambridgeUnited Kingdom
| | | | - Maria Fritsch
- Bioscience Renal, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Ruth March
- Precision Medicine, Oncology R&D, AstraZenecaCambridgeUnited Kingdom
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of HelsinkiHelsinkiFinland
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General HospitalBostonUnited States
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Mark Daly
- Institute for Molecular Medicine Finland, University of HelsinkiHelsinkiFinland
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General HospitalBostonUnited States
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of CambridgeCambridgeUnited Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of CambridgeCambridgeUnited Kingdom
- British Heart Foundation Centre of Research Excellence, University of CambridgeCambridgeUnited Kingdom
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of CambridgeCambridgeUnited Kingdom
| | - Mervi Kinnunen
- Institute for Molecular Medicine Finland, University of HelsinkiHelsinkiFinland
| | - Dirk S Paul
- Centre for Genomics Research, AstraZenecaCambridgeUnited Kingdom
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of CambridgeCambridgeUnited Kingdom
- British Heart Foundation Centre of Research Excellence, University of CambridgeCambridgeUnited Kingdom
| | - Athena Matakidou
- Centre for Genomics Research, AstraZenecaCambridgeUnited Kingdom
| |
Collapse
|
48
|
Selle A, Benamar M. [Regulatory T cells promote lung inflammation through Notch4-dependent reprogramming]. Med Sci (Paris) 2022; 38:648-650. [PMID: 36094233 DOI: 10.1051/medsci/2022096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Amandine Selle
- Division of immunology, Boston Children's Hospital, Boston, Massachusetts, États-Unis. - Department of pediatrics, Harvard Medical School, Boston, Massachusetts, États-Unis
| | - Mehdi Benamar
- Division of immunology, Boston Children's Hospital, Boston, Massachusetts, États-Unis. - Department of pediatrics, Harvard Medical School, Boston, Massachusetts, États-Unis
| |
Collapse
|
49
|
Wang Y, Chen J, Chen C, Peng H, Lin X, Zhao Q, Chen S, Wang X. Growth differentiation factor-15 overexpression promotes cell proliferation and predicts poor prognosis in cerebral lower-grade gliomas correlated with hypoxia and glycolysis signature. Life Sci 2022; 302:120645. [PMID: 35588865 DOI: 10.1016/j.lfs.2022.120645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/15/2022] [Accepted: 05/12/2022] [Indexed: 12/30/2022]
Abstract
AIMS Growth differentiation factor-15 (GDF15) plays complex and controversial roles in cancer. In this study, the prognostic value and the exact biological function of GDF15 in cerebral lower-grade gliomas (LGGs) and its potential molecular targets were examined. MAIN METHODS Wilcoxon signed-rank test and logistic regression were applied to analyze associations between GDF15 expression and clinical characteristics using the Cancer Genome Atlas (TCGA) database. Overall survival was analyzed using Kaplan-Meier and Cox analyses. Gene set enrichment analysis (GSEA) and the hypoxia risk model was conducted to identify the potential molecular mechanisms underlying the effects of GDF15 on LGGs tumorigenesis. The biological function of GDF15 was examined using gain- and loss-of-function experiments, and a recombinant hGDF15 protein in LGG SW1783 cells in vitro. KEY FINDINGS We found that higher GDF15 expression is associated with poor clinical features in LGG patients, and an independent risk factor for overall survival among LGG patients. GSEA results showed that the poor prognostic role of GDF15 in LGGs is related to hypoxia and glycolysis signatures, which was further validated using the hypoxia risk model. Furthermore, GDF15 overexpression facilitated cell proliferation, while GDF15 siRNA inhibits cell proliferation in LGG SW1783 cells. In addition, GDF15 was upregulated upon CoCl2 treatment which induces hypoxia, correlating with the upregulation of the expressions of HIF-1α and glycolysis-related key genes in SW1783 cells. SIGNIFICANCE GDF15 may promote LGG tumorigenesis that is associated with the hypoxia and glycolysis pathways, and thus could serve as a promising molecular target for LGG prevention and therapy.
Collapse
Affiliation(s)
- Ying Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jiajun Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Chaojie Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - He Peng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Xiaojian Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Qian Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Shengjia Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Xingya Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China.
| |
Collapse
|
50
|
Hou CP, Tsui KH, Chen ST, Chang KS, Sung HC, Hsu SY, Lin YH, Feng TH, Juang HH. The Upregulation of Caffeic Acid Phenethyl Ester on Growth Differentiation Factor 15 Inhibits Transforming Growth Factor β/Smad Signaling in Bladder Carcinoma Cells. Biomedicines 2022; 10:biomedicines10071625. [PMID: 35884930 PMCID: PMC9312961 DOI: 10.3390/biomedicines10071625] [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: 06/14/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Growth differentiation factor 15 (GDF15) is known as a TGFβ-like cytokine acting on the TGFβ receptor to modulate target genes. GDF15 is regarded as a tumor suppressor gene in the human bladder and the caffeic acid phenethyl ester (CAPE) induces GDF15 expression to inhibit the tumor growth in vitro and in vivo. However, the interactions among GDF15, CAPE, and TGFβ/Smads signaling in the human bladder carcinoma cells remain unexplored. Results revealed that TGFβ downregulated the expression of GDF15 via the activation of Smad 2/3 and Smad 1/5. Induction of GDF15 on its downstream genes, NDRG1 and maspin, is dependent on the TGFβ/Smad pathways. Moreover, TGFβ blocked the CAPE-inducing expressions of GDF15, maspin, and NDRG1. Pretreatment of TGF receptor kinase inhibitor not only blocked the activation of TGFβ but also attenuated the activation of GDF15 on the expressions of maspin and NDRG1. The CAPE treatment attenuated the activation of TGFβ on cell proliferation and invasion. Our findings indicate that TGFβ downregulated the expressions of GDF15, maspin, and NDRG1 via TGFβ/Smad signaling. Whereas, CAPE acts as an antagonist on TGFβ/Smad signaling to block the effect of TGFβ on the GDF15 expression and cell proliferation and invasion in bladder carcinoma cells.
Collapse
Affiliation(s)
- Chen-Pang Hou
- Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan;
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan;
- Department of Healthcare Management, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan
| | - Ke-Hung Tsui
- Department of Urology, Shuang Ho Hospital, New Taipei City 235041, Taiwan;
- TMU Research Center of Urology and Kindey, Department of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Syue-Ting Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Kang-Shuo Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
| | - Hsin-Ching Sung
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Shu-Yuan Hsu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan;
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan;
| | - Horng-Heng Juang
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan;
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Correspondence: ; Tel.: +886-3-2118800; Fax: +886-3-2118112
| |
Collapse
|