1
|
Barmpagianni A, Karamanakos G, Anastasiou IA, Kountouri A, Lambadiari V, Liatis S. The relationship between residual insulin secretion and subclinical cardiovascular risk indices in young adults with type 1 diabetes. J Diabetes Complications 2025; 39:108946. [PMID: 39731973 DOI: 10.1016/j.jdiacomp.2024.108946] [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: 06/07/2024] [Revised: 09/25/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Patients with type 1 diabetes (DM1), even in the setting of adequate glycaemic control, have an excess risk for developing cardiovascular disease. Residual insulin secretion (RIS), measured by detectable C-peptide levels in patients with DM1, might protect against diabetes-related complications. This study aimed to examine the relationship between residual insulin secretion and prognostic markers of cardiovascular complications in patients with DM1. METHODS A total of 137 patients with DM1 were included in this analysis. They were of young age (<45 years), with an established diagnosis of over two years before the study entry and without a history of cardiovascular complications. All patients underwent complete clinical and laboratory evaluation. A c-peptide measurement of ≥0.05 ng/ml was used to identify the presence of RIS. Pulse wave velocity (PWV), cardiac autonomic function assessed both at rest, by total power of heart rate variability and dynamically, by the expiration to inspiration (e/i) index, albumin to creatinine ratio (ACR), and high sensitivity CRP (hs-CRP) were used as predictive biomarkers of cardiovascular complications. RESULTS Female participants represented 63.5% of the population [mean age: 29.7 (±8.1) years, mean HbA1c: 7.6% (±1.4), median diabetes duration:15 (10-21) years, median age at diabetes diagnosis: 13 (8-17) years]]. The median value of fasting c-peptide was 0.04 (0.03-0.05) ng/ml, and RIS was detected in 32 patients (23.4%). Patients with RIS had a shorter diabetes duration, an older age at diagnosis and a lower BMI, while no significant association was found between residual c-peptide and age or HbA1c. RIS was significantly associated with lower PWV values [8.1 m/s² (7-8.7) vs 9.2 m/s² (7.8-10.1), p <0,001], higher total power values [1124 Hz (600-3277) vs 577 Hz (207-2091), p <0,001], and higher E/I measurements [1.4 (1.2-1.5) vs. 1.3 (1.2-1.4), p=0.01]. No significant association was noted between RIS and either ACR or hs-CRP. In multivariable linear regression analysis, the association between RIS and lower PWV values remained significant (p= 0.007) regardless of age, sex, diabetes duration or age of diagnosis, blood pressure and BMI. Similarly, residual insulin secretion retained a significant independent association with total power (p= 0.032) and E/I (p=0.045). CONCLUSION In young patients with DM1, free of macrovascular complications, residual insulin secretion is independently associated with more favorable prognostic markers of subclinical atherosclerosis and cardiac autonomic function.
Collapse
Affiliation(s)
- Aikaterini Barmpagianni
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece.
| | - Georgios Karamanakos
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece
| | - Ioanna A Anastasiou
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece
| | - Aikaterini Kountouri
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece
| | - Vaia Lambadiari
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece
| | - Stavros Liatis
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece
| |
Collapse
|
2
|
Wu Z, Li L, Xu T, Hu Y, Peng X, Zhang Z, Yao X, Peng Q. Elucidating the multifaceted roles of GPR146 in non-specific orbital inflammation: a concerted analytical approach through the prisms of bioinformatics and machine learning. Front Med (Lausanne) 2024; 11:1309510. [PMID: 38903815 PMCID: PMC11188444 DOI: 10.3389/fmed.2024.1309510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 05/13/2024] [Indexed: 06/22/2024] Open
Abstract
Background Non-specific Orbital Inflammation (NSOI) is a chronic idiopathic condition marked by extensive polymorphic lymphoid infiltration in the orbital area. The integration of metabolic and immune pathways suggests potential therapeutic roles for C-peptide and G protein-coupled receptor 146 (GPR146) in diabetes and its sequelae. However, the specific mechanisms through which GPR146 modulates immune responses remain poorly understood. Furthermore, the utility of GPR146 as a diagnostic or prognostic marker for NSOI has not been conclusively demonstrated. Methods We adopted a comprehensive analytical strategy, merging differentially expressed genes (DEGs) from the Gene Expression Omnibus (GEO) datasets GSE58331 and GSE105149 with immune-related genes from the ImmPort database. Our methodology combined LASSO regression and support vector machine-recursive feature elimination (SVM-RFE) for feature selection, followed by Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) to explore gene sets co-expressed with GPR146, identifying a significant enrichment in immune-related pathways. The tumor microenvironment's immune composition was quantified using the CIBERSORT algorithm and the ESTIMATE method, which confirmed a positive correlation between GPR146 expression and immune cell infiltration. Validation of GPR146 expression was performed using the GSE58331 dataset. Results Analysis identified 113 DEGs associated with GPR146, with a significant subset showing distinct expression patterns. Using LASSO and SVM-RFE, we pinpointed 15 key hub genes. Functionally, these genes and GPR146 were predominantly linked to receptor ligand activity, immune receptor activity, and cytokine-mediated signaling. Specific immune cells, such as memory B cells, M2 macrophages, resting mast cells, monocytes, activated NK cells, plasma cells, and CD8+ T cells, were positively associated with GPR146 expression. In contrast, M0 macrophages, naive B cells, M1 macrophages, activated mast cells, activated memory CD4+ T cells, naive CD4+ T cells, and gamma delta T cells showed inverse correlations. Notably, our findings underscore the potential diagnostic relevance of GPR146 in distinguishing NSOI. Conclusion Our study elucidates the immunological signatures associated with GPR146 in the context of NSOI, highlighting its prognostic and diagnostic potential. These insights pave the way for GPR146 to be a novel biomarker for monitoring the progression of NSOI, providing a foundation for future therapeutic strategies targeting immune-metabolic pathways.
Collapse
Affiliation(s)
- Zixuan Wu
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Ling Li
- Dongying People’s Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, Shandong, China
| | - Tingting Xu
- Dongying People’s Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, Shandong, China
| | - Yi Hu
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Xin Peng
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Zheyuan Zhang
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Xiaolei Yao
- Department of Ophthalmology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Qinghua Peng
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
- Department of Ophthalmology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
3
|
Tharehalli U, Rimbert A. G protein-coupled receptor 146: new insights from genetics and model systems. Curr Opin Lipidol 2024; 35:162-169. [PMID: 38465903 DOI: 10.1097/mol.0000000000000929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
PURPOSE OF REVIEW Atherosclerotic cardiovascular diseases continue to be a significant global cause of death. Despite the availability of efficient treatments, there is an ongoing need for innovative strategies to lower lipid levels, especially for individuals experiencing refractory dyslipidemias or intolerable adverse effects. Based on human genetic findings and on mouse studies, the G protein-coupled receptor 146 (GPR146) emerges as a promising target against hypercholesterolemia and atherosclerosis. The present review aims at providing a thorough summary of the latest information acquired regarding GPR146, encompassing genetic evidence, functional insights, and its broader implications for cardiometabolic health. RECENT FINDINGS Human genetic studies uncovered associations between GPR146 variants, plasma lipid levels and metabolic parameters. Additionally, GPR146's influence extends beyond lipid regulation, impacting adipocyte differentiation, lipolysis, and inflammation pathways. Despite GPR146's orphan status, ongoing efforts to deorphanize it, suggest a potential ligand with downstream effects involving Gαi coupling. SUMMARY Here, we outline and deliberate on recent progress focused on: enhancing comprehension of the effects of inhibiting GPR146 in humans through genetic instruments, evaluating the extra-hepatic functions of GPR146, and discovering its natural ligand(s). Grasping these biological parameters and mechanisms is crucial in the exploration of GPR146 as a prospective therapeutic target.
Collapse
Affiliation(s)
- Umesh Tharehalli
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| |
Collapse
|
4
|
Dakroub A, Dbouk A, Asfour A, Nasser SA, El-Yazbi AF, Sahebkar A, Eid AA, Iratni R, Eid AH. C-peptide in diabetes: A player in a dual hormone disorder? J Cell Physiol 2024; 239:e31212. [PMID: 38308646 DOI: 10.1002/jcp.31212] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024]
Abstract
C-peptide, a byproduct of insulin synthesis believed to be biologically inert, is emerging as a multifunctional molecule. C-peptide serves an anti-inflammatory and anti-atherogenic role in type 1 diabetes mellitus (T1DM) and early T2DM. C-peptide protects endothelial cells by activating AMP-activated protein kinase α, thus suppressing the activity of NAD(P)H oxidase activity and reducing reactive oxygen species (ROS) generation. It also prevents apoptosis by regulating hyperglycemia-induced p53 upregulation and mitochondrial adaptor p66shc overactivation, as well as reducing caspase-3 activity and promoting expression of B-cell lymphoma-2. Additionally, C-peptide suppresses platelet-derived growth factor (PDGF)-beta receptor and p44/p42 mitogen-activated protein (MAP) kinase phosphorylation to inhibit vascular smooth muscle cells (VSMC) proliferation. It also diminishes leukocyte adhesion by virtue of its capacity to abolish nuclear factor kappa B (NF-kB) signaling, a major pro-inflammatory cascade. Consequently, it is envisaged that supplementation of C-peptide in T1DM might ameliorate or even prevent end-organ damage. In marked contrast, C-peptide increases monocyte recruitment and migration through phosphoinositide 3-kinase (PI-3 kinase)-mediated pathways, induces lipid accumulation via peroxisome proliferator-activated receptor γ upregulation, and stimulates VSMC proliferation and CD4+ lymphocyte migration through Src-kinase and PI-3K dependent pathways. Thus, it promotes atherosclerosis and microvascular damage in late T2DM. Indeed, C-peptide is now contemplated as a potential biomarker for insulin resistance in T2DM and linked to increased coronary artery disease risk. This shift in the understanding of the pathophysiology of diabetes from being a single hormone deficiency to a dual hormone disorder warrants a careful consideration of the role of C-peptide as a unique molecule with promising diagnostic, prognostic, and therapeutic applications.
Collapse
Affiliation(s)
- Ali Dakroub
- St. Francis Hospital and Heart Center, Roslyn, New York, USA
| | - Ali Dbouk
- Department of Medicine, Saint-Joseph University Medical School, Hotel-Dieu de France Hospital, Beirut, Lebanon
| | - Aref Asfour
- Leeds Teaching Hospitals NHS Trust, West Yorkshire, United Kingdom
| | | | - Ahmed F El-Yazbi
- Faculty of Pharmacy, Alamein International University (AIU), Alamein City, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, UAE
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
5
|
Zheng YS, Liu YL, Xu ZG, He C, Guo ZY. Is myeloid-derived growth factor a ligand of the sphingosine-1-phosphate receptor 2? Biochem Biophys Res Commun 2024; 706:149766. [PMID: 38484568 DOI: 10.1016/j.bbrc.2024.149766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 03/08/2024] [Indexed: 03/24/2024]
Abstract
Secretory myeloid-derived growth factor (MYDGF) exerts beneficial effects on organ repair, probably via a plasma membrane receptor; however, the identity of the expected receptor has remained elusive. In a recent study, MYDGF was reported as an agonist of the sphingosine-1-phosphate receptor 2 (S1PR2), an A-class G protein-coupled receptor that mediates the functions of the signaling lipid, sphingosine-1-phosphate (S1P). In the present study, we conducted living cell-based functional assays to test whether S1PR2 is a receptor for MYDGF. In the NanoLuc Binary Technology (NanoBiT)-based β-arrestin recruitment assay and the cAMP-response element (CRE)-controlled NanoLuc reporter assay, S1P could efficiently activate human S1PR2 overexpressed in human embryonic kidney (HEK) 293T cells; however, recombinant human MYDGF, overexpressed either from Escherichia coli or HEK293 cells, had no detectable effect. Thus, the results demonstrated that human MYDGF is not a ligand of human S1PR2. Considering the high conservation of MYDGF and S1PR2 in evolution, MYDGF is also probably not a ligand of S1PR2 in other vertebrates.
Collapse
Affiliation(s)
- Yong-Shan Zheng
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China; Shanghai Institute of Biological Products Co., Ltd., Shanghai, China
| | - Ya-Li Liu
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zeng-Guang Xu
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Cheng He
- Shanghai Institute of Biological Products Co., Ltd., Shanghai, China.
| | - Zhan-Yun Guo
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| |
Collapse
|
6
|
Scharf MM, Humphrys LJ, Berndt S, Di Pizio A, Lehmann J, Liebscher I, Nicoli A, Niv MY, Peri L, Schihada H, Schulte G. The dark sides of the GPCR tree - research progress on understudied GPCRs. Br J Pharmacol 2024. [PMID: 38339984 DOI: 10.1111/bph.16325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/24/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024] Open
Abstract
A large portion of the human GPCRome is still in the dark and understudied, consisting even of entire subfamilies of GPCRs such as odorant receptors, class A and C orphans, adhesion GPCRs, Frizzleds and taste receptors. However, it is undeniable that these GPCRs bring an untapped therapeutic potential that should be explored further. Open questions on these GPCRs span diverse topics such as deorphanisation, the development of tool compounds and tools for studying these GPCRs, as well as understanding basic signalling mechanisms. This review gives an overview of the current state of knowledge for each of the diverse subfamilies of understudied receptors regarding their physiological relevance, molecular mechanisms, endogenous ligands and pharmacological tools. Furthermore, it identifies some of the largest knowledge gaps that should be addressed in the foreseeable future and lists some general strategies that might be helpful in this process.
Collapse
Affiliation(s)
- Magdalena M Scharf
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Stockholm, Sweden
| | - Laura J Humphrys
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Sandra Berndt
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Antonella Di Pizio
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Life Science, Technical University of Munich, Freising, Germany
| | - Juliane Lehmann
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Alessandro Nicoli
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Life Science, Technical University of Munich, Freising, Germany
| | - Masha Y Niv
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Lior Peri
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Hannes Schihada
- Institute of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Gunnar Schulte
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Stockholm, Sweden
| |
Collapse
|
7
|
Kaczmarek I, Wower I, Ettig K, Kuhn CK, Kraft R, Landgraf K, Körner A, Schöneberg T, Horn S, Thor D. Identifying G protein-coupled receptors involved in adipose tissue function using the innovative RNA-seq database FATTLAS. iScience 2023; 26:107841. [PMID: 37766984 PMCID: PMC10520334 DOI: 10.1016/j.isci.2023.107841] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/26/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
G protein-coupled receptors (GPCRs) modulate the function of adipose tissue (AT) in general and of adipocytes, specifically. Although it is well-established that GPCRs are widely expressed in AT, their repertoire as well as their regulation and function in (patho)physiological conditions (e.g., obesity) is not fully resolved. Here, we established FATTLAS, an interactive public database, for improved access and analysis of RNA-seq data of mouse and human AT. After extracting the GPCRome of non-obese and obese individuals, highly expressed and differentially regulated GPCRs were identified. Exemplarily, we describe four receptors (GPR146, MRGPRF, FZD5, PTGER2) and analyzed their functions in a (pre)adipocyte cell model. Besides all receptors being involved in adipogenesis, MRGPRF is essential for adipocyte viability and regulates cAMP levels, while GPR146 modulates adipocyte lipolysis via constitutive activation of Gi proteins. Taken together, by implementing and using FATTLAS we describe four hitherto unrecognized GPCRs associated with AT function and adipogenesis.
Collapse
Affiliation(s)
- Isabell Kaczmarek
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Isabel Wower
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Katja Ettig
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Christina Katharina Kuhn
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Robert Kraft
- Carl Ludwig Institute for Physiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Kathrin Landgraf
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Antje Körner
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
- School of Medicine, University of Global Health Equity (UGHE), Kigali, Rwanda
| | - Susanne Horn
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, 45122 Essen, Germany
| | - Doreen Thor
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
8
|
Chen J, Huang Y, Liu C, Chi J, Wang Y, Xu L. The role of C-peptide in diabetes and its complications: an updated review. Front Endocrinol (Lausanne) 2023; 14:1256093. [PMID: 37745697 PMCID: PMC10512826 DOI: 10.3389/fendo.2023.1256093] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Worldwide, diabetes and its complications have seriously affected people's quality of life and become a serious public health problem. C-peptide is not only an indicator of pancreatic β-cell function, but also a biologically active peptide that can bind to cell membrane surface signaling molecules and activate downstream signaling pathways to play antioxidant, anti-apoptotic and inflammatory roles, or regulate cellular transcription through internalization. It is complex how C-peptide is related to diabetic complications. Both deficiencies and overproduction can lead to complications, but their mechanisms of action may be different. C-peptide replacement therapy has shown beneficial effects on diabetic complications in animal models when C-peptide is deficient, but results from clinical trials have been unsatisfactory. The complex pattern of the relationship between C-peptide and diabetic chronic complications has not yet been fully understood. Future basic and clinical studies of C-peptide replacement therapies will need to focus on baseline levels of C-peptide in addition to more attention also needs to be paid to post-treatment C-peptide levels to explore the optimal range of fasting C-peptide and postprandial C-peptide maintenance.
Collapse
Affiliation(s)
| | | | | | | | - Yangang Wang
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lili Xu
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
San Juan JA, Chakarawet K, He Z, Fernandez RL, Stevenson MJ, Harder NHO, Janisse SE, Wang LP, Britt RD, Heffern MC. Copper(II) Affects the Biochemical Behavior of Proinsulin C-peptide by Forming Ternary Complexes with Serum Albumin. J Am Chem Soc 2023. [PMID: 37486968 DOI: 10.1021/jacs.3c04599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Peptide hormones are essential signaling molecules with therapeutic importance. Identifying regulatory factors that drive their activity gives important insight into their mode of action and clinical development. In this work, we demonstrate the combined impact of Cu(II) and the serum protein albumin on the activity of C-peptide, a 31-mer peptide derived from the same prohormone as insulin. C-peptide exhibits beneficial effects, particularly in diabetic patients, but its clinical use has been hampered by a lack of mechanistic understanding. We show that Cu(II) mediates the formation of ternary complexes between albumin and C-peptide and that the resulting species depend on the order of addition. These ternary complexes notably alter peptide activity, showing differences from the peptide or Cu(II)/peptide complexes alone in redox protection as well as in cellular internalization of the peptide. In standard clinical immunoassays for measuring C-peptide levels, the complexes inflate the quantitation of the peptide, suggesting that such adducts may affect biomarker quantitation. Altogether, our work points to the potential relevance of Cu(II)-linked C-peptide/albumin complexes in the peptide's mechanism of action and application as a biomarker.
Collapse
Affiliation(s)
- Jessica A San Juan
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Khetpakorn Chakarawet
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Zhecheng He
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Rebeca L Fernandez
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Michael J Stevenson
- Department of Chemistry, University of San Francisco, 2130 Fulton Street, San Francisco, California 94117, United States
| | - Nathaniel H O Harder
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Samuel E Janisse
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Lee-Ping Wang
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - R David Britt
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Marie C Heffern
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California 95616, United States
| |
Collapse
|
10
|
Monsen VT, Attramadal H. Structural insights into regulation of CCN protein activities and functions. J Cell Commun Signal 2023:10.1007/s12079-023-00768-5. [PMID: 37245184 DOI: 10.1007/s12079-023-00768-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/07/2023] [Indexed: 05/29/2023] Open
Abstract
CCN proteins play important functions during development, in repair mechanisms following tissue injury, as well as in pathophysiologic mechanisms of metastasis of cancer. CCNs are secreted proteins that have a multimodular structure and are categorized as matricellular proteins. Although the prevailing view is that CCN proteins regulate biologic processes by interacting with a wide array of other proteins in the microenvironment of the extracellular matrix, the molecular mechanisms of action of CCN proteins are still poorly understood. Not dissuading the current view, however, the recent appreciation that these proteins are signaling proteins in their own right and may even be considered preproproteins controlled by endopeptidases to release a C-terminal bioactive peptide has opened new avenues of research. Also, the recent resolution of the crystal structure of two of the domains of CCN3 have provided new knowledge with implications for the entire CCN family. These resolved structures in combination with structural predictions based upon the AlphaFold artificial intelligence tool provide means to shed new light on CCN functions in context of the notable literature in the field. CCN proteins have emerged as important therapeutic targets in several disease conditions, and clinical trials are currently ongoing. Thus, a review that critically discusses structure - function relationship of CCN proteins, in particular as it relates to interactions with other proteins in the extracellular milieu and on the cell surface, as well as to cell signaling activities of these proteins, is very timely. Suggested mechanism for activation and inhibition of signaling by the CCN protein family (graphics generated with BioRender.com ).
Collapse
Affiliation(s)
- Vivi Talstad Monsen
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Håvard Attramadal
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
11
|
Franchini L, Orlandi C. Probing the orphan receptors: Tools and directions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 195:47-76. [PMID: 36707155 DOI: 10.1016/bs.pmbts.2022.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The endogenous ligands activating a large fraction of the G Protein Coupled Receptor (GPCR) family members have yet to be identified. These receptors are commonly labeled as orphans (oGPCRs), and because of the absence of available pharmacological tools they are currently understudied. Nonetheless, genome wide association studies, together with research using animal models identified many physiological functions regulated by oGPCRs. Similarly, mutations in some oGPCRs have been associated with rare genetic disorders or with an increased risk of developing pathologies. The once underestimated pharmacological potential of targeting oGPCRs is increasingly being exploited by the development of novel tools to understand their biology and by drug discovery endeavors aimed at identifying new modulators of their activity. Here, we summarize recent advancements in the field of oGPCRs and future directions.
Collapse
Affiliation(s)
- Luca Franchini
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, United States
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, United States.
| |
Collapse
|
12
|
Profiling of G-Protein Coupled Receptors in Adipose Tissue and Differentiating Adipocytes Offers a Translational Resource for Obesity/Metabolic Research. Cells 2023; 12:cells12030377. [PMID: 36766718 PMCID: PMC9913134 DOI: 10.3390/cells12030377] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/06/2023] [Accepted: 01/06/2023] [Indexed: 01/21/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are expressed essentially on all cells, facilitating cellular responses to external stimuli, and are involved in nearly every biological process. Several members of this family play significant roles in the regulation of adipogenesis and adipose metabolism. However, the expression and functional significance of a vast number of GPCRs in adipose tissue are unknown. We used a high-throughput RT-PCR panel to determine the expression of the entire repertoire of non-sensory GPCRs in mouse white, and brown adipose tissue and assess changes in their expression during adipogenic differentiation of murine adipocyte cell line, 3T3-L1. In addition, the expression of GPCRs in subcutaneous adipose tissues from lean, obese, and diabetic human subjects and in adipocytes isolated from regular chow and high-fat fed mice were evaluated by re-analyzing RNA-sequencing data. We detected a total of 292 and 271 GPCRs in mouse white and brown adipose tissue, respectively. There is a significant overlap in the expression of GPCRs between the two adipose tissue depots, but several GPCRs are specifically expressed in one of the two tissue types. Adipogenic differentiation of 3T3-L1 cells had a profound impact on the expression of several GPCRs. RNA sequencing of subcutaneous adipose from healthy human subjects detected 255 GPCRs and obesity significantly changed the expression of several GPCRs in adipose tissue. High-fat diet had a significant impact on adipocyte GPCR expression that was similar to human obesity. Finally, we report several highly expressed GPCRs with no known role in adipose biology whose expression was significantly altered during adipogenic differentiation, and/or in the diseased human subjects. These GPCRs could play an important role in adipose metabolism and serve as a valuable translational resource for obesity and metabolic research.
Collapse
|
13
|
Wensvoort G. Human C-peptide is a ligand of the elastin-receptor-complex and therewith central to human vascular remodelling and disease in metabolic syndrome. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
14
|
Rimbert A, Yeung MW, Dalila N, Thio CHL, Yu H, Loaiza N, Oldoni F, van der Graaf A, Wang S, Said MA, Blauw LL, Girardeau A, Bray L, Caillaud A, Bloks VW, Marrec M, Moulin P, Rensen PCN, van de Sluis B, Snieder H, Di Filippo M, van der Harst P, Tybjaerg-Hansen A, Zimmerman P, Cariou B, Kuivenhoven JA. Variants in the GPR146 Gene Are Associated With a Favorable Cardiometabolic Risk Profile. Arterioscler Thromb Vasc Biol 2022; 42:1262-1271. [PMID: 36047410 DOI: 10.1161/atvbaha.122.317514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND In mice, GPR146 (G-protein-coupled receptor 146) deficiency reduces plasma lipids and protects against atherosclerosis. Whether these findings translate to humans is unknown. METHODS Common and rare genetic variants in the GPR146 gene locus were used as research instruments in the UK Biobank. The Lifelines, The Copenhagen-City Heart Study, and a cohort of individuals with familial hypobetalipoproteinemia were used to find and study rare GPR146 variants. RESULTS In the UK Biobank, carriers of the common rs2362529-C allele present with lower low-density lipoprotein cholesterol, apo (apolipoprotein) B, high-density lipoprotein cholesterol, apoAI, CRP (C-reactive protein), and plasma liver enzymes compared with noncarriers. Carriers of the common rs1997243-G allele, associated with higher GPR146 expression, present with the exact opposite phenotype. The associations with plasma lipids of the above alleles are allele dose-dependent. Heterozygote carriers of a rare coding variant (p.Pro62Leu; n=2615), predicted to be damaging, show a stronger reductions in the above parameters compared with carriers of the common rs2362529-C allele. The p.Pro62Leu variant is furthermore shown to segregate with low low-density lipoprotein cholesterol in a family with familial hypobetalipoproteinemia. Compared with controls, carriers of the common rs2362529-C allele show a marginally reduced risk of coronary artery disease (P=0.03) concomitant with a small effect size on low-density lipoprotein cholesterol (average decrease of 2.24 mg/dL in homozygotes) of this variant. Finally, mendelian randomization analyses suggest a causal relationship between GPR146 gene expression and plasma lipid and liver enzyme levels. CONCLUSIONS This study shows that carriers of new genetic GPR146 variants have a beneficial cardiometabolic risk profile, but it remains to be shown whether genetic or pharmaceutical inhibition of GPR146 protects against atherosclerosis in humans.
Collapse
Affiliation(s)
- Antoine Rimbert
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, France (A.R., A.G., L.B., A.C., M.M., B.C.)
| | - Ming W Yeung
- Department of Cardiology (M.W.Y., S.W., M.S., P.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands.,Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, the Netherlands (M.W.Y., P.v.d.H.)
| | - Nawar Dalila
- Section for Molecular Genetics, Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (N.D., A.T.-H.)
| | - Chris H L Thio
- Department of Epidemiology (C.H.L.T., S.W., H.S.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Haojie Yu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (H.Y.).,Precision Medicine Research Programme, Yong Loo Lin School of Medicine, National University of Singapore (H.Y.)
| | - Natalia Loaiza
- Department of Pediatrics, Section Molecular Genetics (N.L., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Federico Oldoni
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas (F.O.)
| | - Adriaan van der Graaf
- Department of Genetics (A.v.d.G.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Siqi Wang
- Department of Cardiology (M.W.Y., S.W., M.S., P.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands.,Department of Epidemiology (C.H.L.T., S.W., H.S.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - M Abdullah Said
- Department of Cardiology (M.W.Y., S.W., M.S., P.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Lisanne L Blauw
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands (L.L.B., P.C.N.R.)
| | - Aurore Girardeau
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, France (A.R., A.G., L.B., A.C., M.M., B.C.)
| | - Lise Bray
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, France (A.R., A.G., L.B., A.C., M.M., B.C.)
| | - Amandine Caillaud
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, France (A.R., A.G., L.B., A.C., M.M., B.C.)
| | - Vincent W Bloks
- Sections of Molecular Metabolism and Nutrition, Department of Pediatrics (V.W.B.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Marie Marrec
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, France (A.R., A.G., L.B., A.C., M.M., B.C.)
| | - Philippe Moulin
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France (P.M., M.D.F.).,Hospices Civils de Lyon, Fédération d'endocrinologie, maladies métaboliques, diabète et nutrition, Hôpital Louis Pradel, Bron, France (P.M.)
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands (L.L.B., P.C.N.R.)
| | - Bart van de Sluis
- Department of Pediatrics, Section Molecular Genetics (N.L., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Harold Snieder
- Department of Epidemiology (C.H.L.T., S.W., H.S.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Mathilde Di Filippo
- CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France (P.M., M.D.F.).,Hospices Civils de Lyon, UF Dyslipidémies Service de Biochimie et de Biologie Moléculaire Grand Est, Bron, France (M.D.F.)
| | - Pim van der Harst
- Department of Cardiology (M.W.Y., S.W., M.S., P.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands.,Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, the Netherlands (M.W.Y., P.v.d.H.)
| | - Anne Tybjaerg-Hansen
- Section for Molecular Genetics, Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (N.D., A.T.-H.).,Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark (A.T.-H.).,The Copenhagen City Heart Study, Bispebjerg and Frederiksberg Hospital, Denmark (A.T.-H.)
| | | | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, France (A.R., A.G., L.B., A.C., M.M., B.C.)
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, Section Molecular Genetics (N.L., B.v.d.S., J.A.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| |
Collapse
|
15
|
Wilkins BP, Finch AM, Wang Y, Smith NJ. Orphan GPR146: an alternative therapeutic pathway to achieve cholesterol homeostasis? Trends Endocrinol Metab 2022; 33:481-492. [PMID: 35550855 DOI: 10.1016/j.tem.2022.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/07/2022] [Accepted: 04/10/2022] [Indexed: 11/27/2022]
Abstract
Atherosclerosis predisposes to myriad cardiovascular complications, including myocardial infarction and stroke. Statins have revolutionised cholesterol management but they do not work for all patients, particularly those with familial hypercholesterolaemia (FH). Genome-wide association studies have linked SNPs at orphan G protein-coupled receptor 146 (GPR146) to human atherosclerosis but how GPR146 influences serum cholesterol homeostasis was only recently described. Gpr146 deletion in mice reduces serum cholesterol and atherosclerotic plaque burden, confirming GPR146 as a potential therapeutic target for managing circulating cholesterol. Critically, this effect was independent of the low-density lipoprotein receptor. While still an orphan, the activation of GPR146 by serum suggests identification of its endogenous ligand is tantalisingly close. Herein, we discuss the evidence for GPR146 inhibition as a treatment for atherosclerosis.
Collapse
Affiliation(s)
- Brendan P Wilkins
- Orphan Receptor Pharmacology Laboratory, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia; Molecular Pharmacology Drug Design, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Angela M Finch
- Molecular Pharmacology Drug Design, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Yan Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Nicola J Smith
- Orphan Receptor Pharmacology Laboratory, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia; Molecular Pharmacology Drug Design, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
16
|
Andreassen SN, Toft-Bertelsen TL, Wardman JH, Villadsen R, MacAulay N. Transcriptional profiling of transport mechanisms and regulatory pathways in rat choroid plexus. Fluids Barriers CNS 2022; 19:44. [PMID: 35659263 PMCID: PMC9166438 DOI: 10.1186/s12987-022-00335-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dysregulation of brain fluid homeostasis associates with brain pathologies in which fluid accumulation leads to elevated intracranial pressure. Surgical intervention remains standard care, since specific and efficient pharmacological treatment options are limited for pathologies with disturbed brain fluid homeostasis. Such lack of therapeutic targets originates, in part, from the incomplete map of the molecular mechanisms underlying cerebrospinal fluid (CSF) secretion by the choroid plexus. METHODS The transcriptomic profile of rat choroid plexus was generated by RNA Sequencing (RNAseq) of whole tissue and epithelial cells captured by fluorescence-activated cell sorting (FACS), and compared to proximal tubules. The bioinformatic analysis comprised mapping to reference genome followed by filtering for type, location, and association with alias and protein function. The transporters and associated regulatory modules were arranged in discovery tables according to their transcriptional abundance and tied together in association network analysis. RESULTS The transcriptomic profile of choroid plexus displays high similarity between sex and species (human, rat, and mouse) and lesser similarity to another high-capacity fluid-transporting epithelium, the proximal tubules. The discovery tables provide lists of transport mechanisms that could participate in CSF secretion and suggest regulatory candidates. CONCLUSIONS With quantification of the transport protein transcript abundance in choroid plexus and their potentially linked regulatory modules, we envision a molecular tool to devise rational hypotheses regarding future delineation of choroidal transport proteins involved in CSF secretion and their regulation. Our vision is to obtain future pharmaceutical targets towards modulation of CSF production in pathologies involving disturbed brain water dynamics.
Collapse
Affiliation(s)
- Søren N Andreassen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Trine L Toft-Bertelsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Jonathan H Wardman
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - René Villadsen
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| |
Collapse
|
17
|
Webber T, Ronacher K, Conradie-Smit M, Kleynhans L. Interplay Between the Immune and Endocrine Systems in the Lung: Implications for TB Susceptibility. Front Immunol 2022; 13:829355. [PMID: 35273609 PMCID: PMC8901994 DOI: 10.3389/fimmu.2022.829355] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/02/2022] [Indexed: 12/25/2022] Open
Abstract
The role of the endocrine system on the immune response, especially in the lung, remains poorly understood. Hormones play a crucial role in the development, homeostasis, metabolism, and response to the environment of cells and tissues. Major infectious and metabolic diseases, such as tuberculosis and diabetes, continue to converge, necessitating the development of a clearer understanding of the immune and endocrine interactions that occur in the lung. Research in bacterial respiratory infections is at a critical point, where the limitations in identifying and developing antibiotics is becoming more profound. Hormone receptors on alveolar and immune cells may provide a plethora of targets for host-directed therapy. This review discusses the interactions between the immune and endocrine systems in the lung. We describe hormone receptors currently identified in the lungs, focusing on the effect hormones have on the pulmonary immune response. Altered endocrine responses in the lung affect the balance between pro- and anti-inflammatory immune responses and play a role in the response to infection in the lung. While some hormones, such as leptin, resistin and lipocalin-2 promote pro-inflammatory responses and immune cell infiltration, others including adiponectin and ghrelin reduce inflammation and promote anti-inflammatory cell responses. Furthermore, type 2 diabetes as a major endocrine disease presents with altered immune responses leading to susceptibility to lung infections, such as tuberculosis. A better understanding of these interactions will expand our knowledge of the mechanisms at play in susceptibility to infectious diseases and may reveal opportunities for the development of host-directed therapies.
Collapse
Affiliation(s)
- Tariq Webber
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Katharina Ronacher
- Translational Research Institute, Mater Research Institute - The University of Queensland, Brisbane, QLD, Australia
| | - Marli Conradie-Smit
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Léanie Kleynhans
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
18
|
Rossiter JL, Redlinger LJ, Kolar GR, Samson WK, Yosten GLC. The actions of C-peptide in HEK293 cells are dependent upon insulin and extracellular glucose concentrations. Peptides 2022; 150:170718. [PMID: 34954230 DOI: 10.1016/j.peptides.2021.170718] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 10/19/2022]
Abstract
Connecting peptide, or C-peptide, is a part of the insulin prohormone and is essential for the proper folding and processing of the mature insulin peptide. C-peptide is released from the same beta cell secretory granules as insulin in equimolar amounts. However, due to their relative stabilities in plasma, the two peptides are detected in the circulation at ratios of approximately 4:1 to 6:1 (C-peptide to insulin), depending on metabolic state. C-peptide binds specifically to human cell membranes and induces intracellular signaling cascades, likely through an interaction with the G protein coupled receptor, GPR146. C-peptide has been shown to exert protective effects against the vascular, renal, and ocular complications of diabetes. The effects of C-peptide appear to be dependent upon the presence of insulin and the absolute, extracellular concentration of glucose. In this study, we employed HEK293 cells to further examine the interactive effects of C-peptide, insulin, and glucose on cell signaling. We observed that C-peptide's cellular effects are dampened significantly when cells are exposed to physiologically relevant concentrations of both insulin and C-peptide. Likewise, the actions of C-peptide on cFos and GPR146 mRNA expressions were affected by changes in extracellular glucose concentration. In particular, C-peptide induced significant elevations in cFos expression in the setting of high (25 mmol) extracellular glucose concentration. These data indicate that future experimentation on the actions of C-peptide should control for the presence or absence of insulin and the concentration of glucose. Furthermore, these findings should be considered prior to the development of C-peptide-based therapeutics for the treatment of diabetes-associated complications.
Collapse
Affiliation(s)
- Jacqueline L Rossiter
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States
| | - Lauren J Redlinger
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States
| | - Grant R Kolar
- Department of Pathology, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States
| | - Willis K Samson
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States
| | - Gina L C Yosten
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Boulevard, Saint Louis, MO 63104, United States.
| |
Collapse
|
19
|
Abstract
Neuropeptides are a diverse class of signaling molecules in metazoans. They occur in all animals with a nervous system and also in neuron-less placozoans. However, their origin has remained unclear because no neuropeptide shows deep homology across lineages, and none have been found in sponges. Here, we identify two neuropeptide precursors, phoenixin (PNX) and nesfatin, with broad evolutionary conservation. By database searches, sequence alignments, and gene-structure comparisons, we show that both precursors are present in bilaterians, cnidarians, ctenophores, and sponges. We also found PNX and a secreted nesfatin precursor homolog in the choanoflagellate Salpingoeca rosetta. PNX, in particular, is highly conserved, including its cleavage sites, suggesting that prohormone processing occurs also in choanoflagellates. In addition, based on phyletic patterns and negative pharmacological assays, we question the originally proposed GPR-173 (SREB3) as a PNX receptor. Our findings revealed that secreted neuropeptide homologs derived from longer precursors have premetazoan origins and thus evolved before neurons.
Collapse
Affiliation(s)
| | - Daniel Thiel
- Living Systems Institute, University of Exeter, Stocker Road, Exeter, UK
| | - Gáspár Jékely
- Living Systems Institute, University of Exeter, Stocker Road, Exeter, UK
| |
Collapse
|
20
|
Zou Z, Zhuang J, Xia L, Li Y, Yin J, Mu Y. DCD-chip designed for the digital and ultraprecise quantification of copy number variation. Analyst 2022; 147:4371-4378. [DOI: 10.1039/d2an00982j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The progress of CNV detection in DCD-chip.
Collapse
Affiliation(s)
- Zheyu Zou
- Research Centre for Analytical Instrumentation, Institute of Cyber-Systems and Control, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou, P. R. China
- College of Life Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Jianjian Zhuang
- Department of Clinical pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, P. R. China
| | - Liping Xia
- Research Centre for Analytical Instrumentation, Institute of Cyber-Systems and Control, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou, P. R. China
- College of Life Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Ying Li
- Department of Public Health, School of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Juxin Yin
- School of information and Electrical Engineering, Zhejiang University City College, Hangzhou, Zhejiang Province, P. R. China
| | - Ying Mu
- Research Centre for Analytical Instrumentation, Institute of Cyber-Systems and Control, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou, P. R. China
| |
Collapse
|
21
|
Vejrazkova D, Vankova M, Lukasova P, Vcelak J, Bendlova B. Insights into the physiology of C-peptide. Physiol Res 2021; 69:S237-S243. [PMID: 33094622 DOI: 10.33549/physiolres.934519] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Current knowledge suggests a complex role of C-peptide in human physiology, but its mechanism of action is only partially understood. The effects of C-peptide appear to be variable depending on the target tissue, physiological environment, its combination with other bioactive molecules such as insulin, or depending on its concentration. It is apparent that C-peptide has therapeutic potential for the treatment of vascular and nervous damage caused by type 1 or late type 2 diabetes mellitus. The question remains whether the effect is mediated by the receptor, the existence of which is still uncertain, or whether an alternative non-receptor-mediated mechanism is responsible. The Institute of Endocrinology in Prague has been paying much attention to the issue of C-peptide and its metabolic effect since the 1980s. The RIA methodology of human C-peptide determination was introduced here and transferred to commercial production. By long-term monitoring of C-peptide oGTT-derived indices, the Institute has contributed to elucidating the pathophysiology of glucose tolerance disorders. This review summarizes the current knowledge of C-peptide physiology and highlights the contributions of the Institute of Endocrinology to this issue.
Collapse
Affiliation(s)
- D Vejrazkova
- Department of Molecular Endocrinology, Institute of Endocrinology, Prague, Czech Republic.
| | | | | | | | | |
Collapse
|
22
|
Samson WK, Salvemini D, Yosten GLC. Overcoming Stress, Hunger, and Pain: Cocaine- and Amphetamine-Regulated Transcript Peptide's Promise. Endocrinology 2021; 162:6287092. [PMID: 34043767 PMCID: PMC8210821 DOI: 10.1210/endocr/bqab108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Indexed: 01/17/2023]
Abstract
Cocaine- and amphetamine-regulated transcript encodes an eponymous peptide, CARTp, which exerts diverse pharmacologic actions in the central and peripheral nervous systems, as well as in several endocrine organs, including pancreas. Here we review those diverse actions, the physiological relevance of which had remained unestablished until recently. With the identification of a CARTp receptor, GPR160, the physiologic importance and therapeutic potential of CARTp or analogs are being revealed. Not only is the CARTp-GPR160 interaction essential for the circadian regulation of appetite and thirst but also for the transmission of nerve injury-induced pain. Molecular approaches now are uncovering additional physiologically relevant actions and the development of acute tissue-specific gene compromise approaches may reveal even more physiologically relevant actions of this pluripotent ligand/receptor pair.
Collapse
Affiliation(s)
- Willis K Samson
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience Saint Louis University School of Medicine, St Louis, MO 63104, USA
- Correspondence: Willis K. Samson, PhD DSc, Professor of Pharmacology and Physiology, Saint Louis University School of Medicine, Caroline Building, Room 2-207A, 1402 South Grand Boulevard, St Louis, MO 63104, USA.
| | - Daniela Salvemini
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - Gina L C Yosten
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience Saint Louis University School of Medicine, St Louis, MO 63104, USA
| |
Collapse
|
23
|
Moore MC, Warner SO, Dai Y, Sheanon N, Smith M, Farmer B, Cason RL, Cherrington AD, Winnick JJ. C-peptide enhances glucagon secretion in response to hyperinsulinemia under euglycemic and hypoglycemic conditions. JCI Insight 2021; 6:148997. [PMID: 34003799 PMCID: PMC8262495 DOI: 10.1172/jci.insight.148997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/12/2021] [Indexed: 12/17/2022] Open
Abstract
Several studies have associated the presence of residual insulin secretion capability (also referred to as being C-peptide positive) with lower risk of insulin-induced hypoglycemia in patients with type 1 diabetes (T1D), although the reason is unclear. We tested the hypothesis that C-peptide infusion would enhance glucagon secretion in response to hyperinsulinemia during euglycemic and hypoglycemic conditions in dogs (5 male/4 female). After a 2-hour basal period, an intravenous (IV) infusion of insulin was started, and dextrose was infused to maintain euglycemia for 2 hours. At the same time, an IV infusion of either saline (SAL) or C-peptide (CPEP) was started. After this euglycemic period, the insulin and SAL/CPEP infusions were continued for another 2 hours, but the glucose was allowed to fall to approximately 50 mg/dL. In response to euglycemic-hyperinsulinemia, glucagon secretion decreased in SAL but remained unchanged from the basal period in CPEP condition. During hypoglycemia, glucagon secretion in CPEP was 2 times higher than SAL, and this increased net hepatic glucose output and reduced the amount of exogenous glucose required to maintain glycemia. These data suggest that the presence of C-peptide during IV insulin infusion can preserve glucagon secretion during euglycemia and enhance it during hypoglycemia, which could explain why T1D patients with residual insulin secretion are less susceptible to hypoglycemia.
Collapse
Affiliation(s)
- Mary Courtney Moore
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Shana O. Warner
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yufei Dai
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Nicole Sheanon
- Department of Endocrinology, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Marta Smith
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Ben Farmer
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Rebecca L. Cason
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Alan D. Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jason J. Winnick
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
24
|
The Role of Peptide Hormones Discovered in the 21st Century in the Regulation of Adipose Tissue Functions. Genes (Basel) 2021; 12:genes12050756. [PMID: 34067710 PMCID: PMC8155905 DOI: 10.3390/genes12050756] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 12/14/2022] Open
Abstract
Peptide hormones play a prominent role in controlling energy homeostasis and metabolism. They have been implicated in controlling appetite, the function of the gastrointestinal and cardiovascular systems, energy expenditure, and reproduction. Furthermore, there is growing evidence indicating that peptide hormones and their receptors contribute to energy homeostasis regulation by interacting with white and brown adipose tissue. In this article, we review and discuss the literature addressing the role of selected peptide hormones discovered in the 21st century (adropin, apelin, elabela, irisin, kisspeptin, MOTS-c, phoenixin, spexin, and neuropeptides B and W) in controlling white and brown adipogenesis. Furthermore, we elaborate how these hormones control adipose tissue functions in vitro and in vivo.
Collapse
|
25
|
C-Peptide as a Therapy for Type 1 Diabetes Mellitus. Biomedicines 2021; 9:biomedicines9030270. [PMID: 33800470 PMCID: PMC8000702 DOI: 10.3390/biomedicines9030270] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus (DM) is a complex metabolic disease affecting one-third of the United States population. It is characterized by hyperglycemia, where the hormone insulin is either not produced sufficiently or where there is a resistance to insulin. Patients with Type 1 DM (T1DM), in which the insulin-producing beta cells are destroyed by autoimmune mechanisms, have a significantly increased risk of developing life-threatening cardiovascular complications, even when exogenous insulin is administered. In fact, due to various factors such as limited blood glucose measurements and timing of insulin administration, only 37% of T1DM adults achieve normoglycemia. Furthermore, T1DM patients do not produce C-peptide, a cleavage product from insulin processing. C-peptide has potential therapeutic effects in vitro and in vivo on many complications of T1DM, such as peripheral neuropathy, atherosclerosis, and inflammation. Thus, delivery of C-peptide in conjunction with insulin through a pump, pancreatic islet transplantation, or genetically engineered Sertoli cells (an immune privileged cell type) may ameliorate many of the cardiovascular and vascular complications afflicting T1DM patients.
Collapse
|
26
|
Yosten GLC, Haddock CJ, Harada CM, Almeida-Pereira G, Kolar GR, Stein LM, Hayes MR, Salvemini D, Samson WK. Past, present and future of cocaine- and amphetamine-regulated transcript peptide. Physiol Behav 2021; 235:113380. [PMID: 33705816 DOI: 10.1016/j.physbeh.2021.113380] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/22/2021] [Accepted: 03/01/2021] [Indexed: 01/02/2023]
Abstract
The existence of the peptide encoded by the cocaine- and amphetamine-regulated transcript (Cartpt) has been recognized since 1981, but it was not until 1995, that the gene encoding CART peptide (CART) was identified. With the availability of the predicted protein sequence of CART investigators were able to identify sites of peptide localization, which then led to numerous approaches attempting to clarify CART's multiple pharmacologic effects and even provide evidence of potential physiologic relevance. Although not without controversy, a picture emerged of the importance of CART in ingestive behaviors, reward behaviors and even pain sensation. Despite the wealth of data hinting at the significance of CART, in the absence of an identified receptor, the full potential for this peptide or its analogs to be developed into therapeutic agents remained unrealized. There was evidence favoring the action of CART via a G protein-coupled receptor (GPCR), but despite multiple attempts the identity of that receptor eluded investigators until recently. Now with the identification of the previously orphaned GPCR, GPR160, as a receptor for CART, focus on this pluripotent neuropeptide will in all likelihood experience a renaissance and the potential for the development of pharmcotherapies targeting GPR160 seems within reach.
Collapse
Affiliation(s)
- Gina L C Yosten
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA; Henry and Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Christopher J Haddock
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Caron M Harada
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA; Henry and Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Gislaine Almeida-Pereira
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Grant R Kolar
- Henry and Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO 63104, USA; Department of Pathology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Lauren M Stein
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Matthew R Hayes
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA; Henry and Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Willis K Samson
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA; Henry and Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| |
Collapse
|
27
|
Abid MSR, Mousavi S, Checco JW. Identifying Receptors for Neuropeptides and Peptide Hormones: Challenges and Recent Progress. ACS Chem Biol 2021; 16:251-263. [PMID: 33539706 DOI: 10.1021/acschembio.0c00950] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intercellular signaling events mediated by neuropeptides and peptide hormones represent important targets for both basic science and drug discovery. For many bioactive peptides, the protein receptors that transmit information across the receiving cell membrane are not known, severely limiting these signaling pathways as potential therapeutic targets. Identifying the receptor(s) for a given peptide of interest is complicated by several factors. Most notably, cell-cell signaling peptides are generated through dynamic biosynthetic pathways, can act on many different families of receptor proteins, and can participate in complex ligand-receptor interactions that extend beyond a simple one-to-one archetype. Here, we discuss recent methodological advances to identify signaling partners for bioactive peptides. Recent efforts have centered on methods to identify candidate receptors via transcript expression, methods to match peptide-receptor pairs through high throughput screening, and methods to capture direct ligand-receptor interactions using chemical probes. Future applications of the receptor identification approaches discussed here, as well as technical advancements to address their limitations, promise to lead to a greater understanding of how cells communicate to deliver complex physiologies. Importantly, such advancements will likely provide novel targets for the treatment of human diseases within the central nervous and endocrine systems.
Collapse
Affiliation(s)
- Md Shadman Ridwan Abid
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Somayeh Mousavi
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - James W. Checco
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| |
Collapse
|
28
|
Biological Activity of c-Peptide in Microvascular Complications of Type 1 Diabetes-Time for Translational Studies or Back to the Basics? Int J Mol Sci 2020; 21:ijms21249723. [PMID: 33419247 PMCID: PMC7766542 DOI: 10.3390/ijms21249723] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/15/2022] Open
Abstract
People with type 1 diabetes have an increased risk of developing microvascular complications, which have a negative impact on the quality of life and reduce life expectancy. Numerous studies in animals with experimental diabetes show that c-peptide supplementation exerts beneficial effects on diabetes-induced damage in peripheral nerves and kidneys. There is substantial evidence that c-peptide counteracts the detrimental changes caused by hyperglycemia at the cellular level, such as decreased activation of endothelial nitric oxide synthase and sodium potassium ATPase, and increase in formation of pro-inflammatory molecules mediated by nuclear factor kappa-light-chain-enhancer of activated B cells: cytokines, chemokines, cell adhesion molecules, vascular endothelial growth factor, and transforming growth factor beta. However, despite positive results from cell and animal studies, no successful c-peptide replacement therapies have been developed so far. Therefore, it is important to improve our understanding of the impact of c-peptide on the pathophysiology of microvascular complications to develop novel c-peptide-based treatments. This article aims to review current knowledge on the impact of c-peptide on diabetic neuro- and nephropathy and to evaluate its potential therapeutic role.
Collapse
|
29
|
Khaliq SA, Baek MO, Cho HJ, Chon SJ, Yoon MS. C-Peptide Inhibits Decidualization in Human Endometrial Stromal Cells via GSK3β-PP1. Front Cell Dev Biol 2020; 8:609551. [PMID: 33330513 PMCID: PMC7734312 DOI: 10.3389/fcell.2020.609551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/12/2020] [Indexed: 12/17/2022] Open
Abstract
Decidualization refers to the functional differentiation of endometrial stromal cells and plays a significant role in embryo implantation and pregnancy. C-peptide is excreted in equimolar concentrations as that of insulin during the metabolism of proinsulin in pancreatic beta-cells. High levels of C-peptide are correlated with hyperinsulinemia and polycystic ovarian syndrome, which show a defect in decidualization. However, the role of C-peptide in decidualization has not yet been studied. Here, we identified C-peptide as an endogenous antideciduogenic factor. This inhibitory function was confirmed by the reduced expression of decidual markers, including prolactin, insulin-like growth factor-binding protein-1, and Forkhead box protein O1 as well as by the fibroblastic morphological change in the presence of C-peptide. C-peptide also enhanced cellular senescence and decreased the proportion of apoptotic cells during decidualization. In addition, C-peptide potentiated the inhibitory effects of both insulin and palmitic acid in an AKT- and autophagy-independent manner, respectively. Furthermore, C-peptide augmented protein phosphatase 1 (PP1) activity, leading to a reduction in the inhibitory phosphorylation of glycogen synthase kinase (GSK)3β, which resulted in enhanced cellular senescence and decreased apoptosis during decidualization. Taken together, our findings suggest that C-peptide is an antideciduogenic factor acting via the regulation between PP1 and GSK3β in patients with hyperinsulinemia.
Collapse
Affiliation(s)
- Sana Abdul Khaliq
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea.,Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, South Korea
| | - Mi-Ock Baek
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea.,Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, South Korea
| | - Hye-Jeong Cho
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea
| | - Seung Joo Chon
- Department of Obstetrics and Gynecology, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon, South Korea
| | - Mee-Sup Yoon
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea.,Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, South Korea
| |
Collapse
|
30
|
A C-peptide complex with albumin and Zn 2+ increases measurable GLUT1 levels in membranes of human red blood cells. Sci Rep 2020; 10:17493. [PMID: 33060722 PMCID: PMC7566639 DOI: 10.1038/s41598-020-74527-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022] Open
Abstract
People with type 1 diabetes (T1D) require exogenous administration of insulin, which stimulates the translocation of the GLUT4 glucose transporter to cell membranes. However, most bloodstream cells contain GLUT1 and are not directly affected by insulin. Here, we report that C-peptide, the 31-amino acid peptide secreted in equal amounts with insulin in vivo, is part of a 3-component complex that affects red blood cell (RBC) membranes. Multiple techniques were used to demonstrate saturable and specific C-peptide binding to RBCs when delivered as part of a complex with albumin. Importantly, when the complex also included Zn2+, a significant increase in cell membrane GLUT1 was measured, thus providing a cellular effect similar to insulin, but on a transporter on which insulin has no effect.
Collapse
|
31
|
Biological activity versus physiological function of proinsulin C-peptide. Cell Mol Life Sci 2020; 78:1131-1138. [PMID: 32959070 PMCID: PMC7897624 DOI: 10.1007/s00018-020-03636-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/07/2020] [Accepted: 09/03/2020] [Indexed: 11/06/2022]
Abstract
Proinsulin C-peptide (C-peptide) has drawn much research attention. Even if the peptide has turned out not to be important in the treatment of diabetes, every phase of C-peptide research has changed our view on insulin and peptide hormone biology. The first phase revealed that peptide hormones can be subject to processing, and that their pro-forms may involve regulatory stages. The second phase revealed the possibility that one prohormone could harbor more than one activity, and that the additional activities should be taken into account in the development of hormone-based therapies. In the third phase, a combined view of the evolutionary patterns in hormone biology allowed an assessment of C-peptide´s role in physiology, and of how biological activities and physiological functions are shaped by evolutionary processes. In addition to this distinction, C-peptide research has produced further advances. For example, C-peptide fragments are successfully administered in immunotherapy of type I diabetes, and plasma C-peptide levels remain a standard for measurement of beta cell activity in patients. Even if the concept of C-peptide as a hormone is presently not supported, some of its bioactivities continue to influence our understanding of evolutionary changes of also other peptides.
Collapse
|
32
|
Derkach KV, Zorina II, Zakharova IO, Basova NE, Bakhtyukov AA, Shpakov AO. The Influence of Intranasally Administered
Insulin and
C-peptide on AMP-Activated Protein Kinase Activity, Mitochondrial
Dynamics and Apoptosis Markers in the Hypothalamus of Rats with
Streptozotocin-Induced Diabetes. J EVOL BIOCHEM PHYS+ 2020. [DOI: 10.1134/s0022093020030035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Zhang R, Wang J, Liu B, Wang W, Fan X, Zheng B, Yuan Q, Xue M, Xu F, Guo P, Chen Y. Differentially expressed lncRNAs, miRNAs and mRNAs with associated ceRNA networks in a mouse model of myocardial ischemia/reperfusion injury. Mol Med Rep 2020; 22:2487-2495. [PMID: 32705277 PMCID: PMC7411395 DOI: 10.3892/mmr.2020.11300] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 06/05/2020] [Indexed: 12/17/2022] Open
Abstract
Non‑coding RNAs, including long non‑coding RNAs (lncRNAs) and microRNAs (miRNAs/miRs), have significant regulatory effects on a number of biological processes in myocardial ischemia/reperfusion (I/R) injury, including cell differentiation, proliferation and apoptosis. In the present study, the expression levels of lncRNAs, miRNAs and mRNAs were evaluated in a mouse model of myocardial I/R injury. The potential functions of these differentially expressed genes were then analyzed via Gene Ontology and pathway analyses. Additionally, the interactions between lncRNA‑miRNA‑mRNA were predicted by constructing a competing endogenous RNA regulatory network. It was found that 14,366 lncRNAs, 151 miRNAs and 9,377 mRNAs were differentially expressed in mice hearts after I/R compared with the Sham group (fold change >2; P<0.05). The results indicated that these differentially expressed genes were involved in multiple molecular functions, including 'guanosine diphosphate binding', 'RNA polymerase II carboxy‑terminal domain kinase activity', 'TATA‑binding protein‑class protein binding', 'nicotinamide adenine dinucleotide binding' and 'protein phosphatase type 2A regulator activity'. The interactions between lncRNA‑miRNA‑mRNA, including five lncRNAs, 38 miRNAs and 196 mRNAs, were predicted, specifically Gm12040‑mmu‑miR‑125a‑5p‑decapping mRNA 1B, Rpl7l1‑ps1‑mmu‑miR‑124‑3p‑G protein‑coupled receptor 146, Gm11407‑mmu‑miR‑190a‑5p‑homeobox and leucine zipper encoding (HOMEZ), 1600029O15Rik‑mmu‑miR‑132‑3p‑HOMEZ and AK155692‑mmu‑miR‑1224‑3p‑activating transcription factor 6β. Collectively, these findings provided novel insights for future research on lncRNAs, miRNAs and mRNAs in myocardial I/R injury.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jiali Wang
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Baoshan Liu
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Wenjun Wang
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xinhui Fan
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Boyuan Zheng
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qiuhuan Yuan
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Mengyang Xue
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Feng Xu
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ping Guo
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yuguo Chen
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
34
|
Is GPR146 really the receptor for proinsulin C-peptide? Bioorg Med Chem Lett 2020; 30:127208. [DOI: 10.1016/j.bmcl.2020.127208] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 01/01/2023]
|
35
|
Luppi P, Drain N, To R, Stolz D, Wallace C, Watkins S, Drain P. Autocrine C-peptide protects INS1 β cells against palmitic acid-induced oxidative stress in peroxisomes by inducing catalase. Endocrinol Diabetes Metab 2020; 3:e00147. [PMID: 32704568 PMCID: PMC7375117 DOI: 10.1002/edm2.147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/26/2020] [Accepted: 05/02/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS C-peptide, produced by pancreatic β cells and co-secreted in the bloodstream with insulin, has antioxidant properties in glucose- and hydrogen peroxide (H2O2)-exposed INS1 β cells. Palmitic acid, the most physiologically abundant long-chain free fatty acid in humans, is metabolized in peroxisomes of β cells accumulating H2O2 that can lead to oxidative stress. Here, we tested the hypothesis that C-peptide protects β cells from palmitic acid-induced stress by lowering peroxisomal H2O2. MATERIALS AND METHODS We exposed INS1 β cells to palmitic acid and C-peptide in the setting of increasing glucose concentration and tested for changes in parameters of stress and death. To study the ability of C-peptide to lower peroxisomal H2O2, we engineered an INS1 β cell line stably expressing the peroxisomal-targeted H2O2 sensor HyPer, whose fluorescence increases with cellular H2O2. An INS1 β cell line stably expressing a live-cell fluorescent catalase reporter was used to detect changes in catalase gene expression. RESULTS C-peptide protects INS1 β cells from the combined effect of palmitic acid and glucose by reducing peroxisomal H2O2 to baseline levels and increasing expression of catalase. CONCLUSIONS In conditions of glucolipotoxicity, C-peptide increases catalase expression and reduces peroxisomal oxidative stress and death of INS1 β cells. Maintenance of C-peptide secretion is a pro-survival requisite for β cells in adverse conditions. Loss of C-peptide secretion would render β cells more vulnerable to stress and death leading to secretory dysfunction and diabetes.
Collapse
Affiliation(s)
- Patrizia Luppi
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Nicholas Drain
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Ramsey To
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Donna Stolz
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Callen Wallace
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Simon Watkins
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Peter Drain
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| |
Collapse
|
36
|
Poteryaeva ON, Usynin IF. [Molecular mechanisms of action and physiological effects of the proinsulin C-peptide (a systematic review)]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2020; 66:196-207. [PMID: 32588825 DOI: 10.18097/pbmc20206603196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The C-peptide is a fragment of proinsulin, the cleavage of which forms active insulin. In recent years, new information has appeared on the physiological effects of the C-peptide, indicating its positive effect on many organs and tissues, including the kidneys, nervous system, heart, vascular endothelium and blood microcirculation. Studies on experimental models of diabetes mellitus in animals, as well as clinical trials in patients with diabetes, have shown that the C-peptide has an important regulatory effect on the early stages of functional and structural disorders caused by this disease. The C-peptide exhibits its effects through binding to a specific receptor on the cell membrane and activation of downstream signaling pathways. Intracellular signaling involves G-proteins and Ca2+-dependent pathways, resulting in activation and increased expression of endothelial nitric oxide synthase, Na+/K+-ATPase and important transcription factors involved in apoptosis, anti-inflammatory and other intracellular defense mechanisms. This review gives an idea of the C-peptide as a bioactive endogenous peptide that has its own biological activity and therapeutic potential.
Collapse
Affiliation(s)
- O N Poteryaeva
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| | - I F Usynin
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| |
Collapse
|
37
|
Haddock CJ, Almeida-Pereira G, Stein LM, Yosten GLC, Samson WK. A novel regulator of thirst behavior: phoenixin. Am J Physiol Regul Integr Comp Physiol 2020; 318:R1027-R1035. [PMID: 32292064 PMCID: PMC7311681 DOI: 10.1152/ajpregu.00023.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/30/2020] [Accepted: 04/10/2020] [Indexed: 12/19/2022]
Abstract
There are examples of physiological conditions under which thirst is inappropriately exaggerated, and the mechanisms for these paradoxical ingestive behaviors remain unknown. We are interested in thirst mechanisms across the female life cycle and have identified a novel mechanism through which ingestive behavior may be activated. We discovered a previously unrecognized endogenous hypothalamic peptide, phoenixin (PNX), identified physiologically relevant actions of the peptide in brain and pituitary gland to control reproductive hormone secretion in female rodents, and in the process identified the previously orphaned G protein-coupled receptor Gpr173 to be a potential receptor for the peptide. Labeled PNX binding distribution in brain parallels areas known to be important in ingestive behaviors as well in areas where gonadal steroids feedback to control estrous cyclicity (Stein LM, Tullock CW, Mathews SK, Garcia-Galiano D, Elias CF, Samson WK, Yosten GLC, Am J Physiol Regul Integr Comp Physiol 311: R489-R496, 2016). We have demonstrated upregulation of Gpr173 during puberty, fluctuations across the estrous cycle, and, importantly, upregulation during the last third of gestation. It is during this hypervolemic, hyponatremic state that both vasopressin secretion and thirst are inappropriately elevated in humans. Here, we show that central administration of PNX stimulated water drinking in both males and females under ad libitum conditions, increased water drinking after overnight fluid deprivation, and increased both water and 1.5% NaCl ingestion under fed and hydrated conditions. Importantly, losartan pretreatment blocked the effect of PNX on water drinking, and knockdown of Gpr173 by use of short interfering RNA constructs significantly attenuated water drinking in response to overnight fluid deprivation. These actions, together with the stimulatory action of PNX on vasopressin secretion, suggest that this recently discovered neuropeptide may impact the recruitment of critically important neural circuits through which ingestive behaviors and endocrine mechanisms that maintain fluid and electrolyte homeostasis are regulated.
Collapse
Affiliation(s)
- Christopher J Haddock
- Department of Pharmacology and Physiology, Center for Neuroscience Research, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Gislaine Almeida-Pereira
- Department of Pharmacology and Physiology, Center for Neuroscience Research, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Lauren M Stein
- Department of Pharmacology and Physiology, Center for Neuroscience Research, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Gina L C Yosten
- Department of Pharmacology and Physiology, Center for Neuroscience Research, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Willis K Samson
- Department of Pharmacology and Physiology, Center for Neuroscience Research, Saint Louis University School of Medicine, St. Louis, Missouri
| |
Collapse
|
38
|
Yosten GL, Harada CM, Haddock C, Giancotti LA, Kolar GR, Patel R, Guo C, Chen Z, Zhang J, Doyle TM, Dickenson AH, Samson WK, Salvemini D. GPR160 de-orphanization reveals critical roles in neuropathic pain in rodents. J Clin Invest 2020; 130:2587-2592. [PMID: 31999650 PMCID: PMC7190928 DOI: 10.1172/jci133270] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/23/2020] [Indexed: 01/15/2023] Open
Abstract
Treating neuropathic pain is challenging and novel non-opioid-based medicines are needed. Using unbiased receptomics, transcriptomic analyses, immunofluorescence, and in situ hybridization, we found that the expression of the orphan GPCR Gpr160 and GPR160 increased in the rodent dorsal horn of the spinal cord following traumatic nerve injury. Genetic and immunopharmacological approaches demonstrated that GPR160 inhibition in the spinal cord prevented and reversed neuropathic pain in male and female rodents without altering normal pain response. GPR160 inhibition in the spinal cord attenuated sensory processing in the thalamus, a key relay in the sensory discriminative pathways of pain. We also identified cocaine- and amphetamine-regulated transcript peptide (CARTp) as a GPR160 ligand. Inhibiting endogenous CARTp signaling in spinal cord attenuated neuropathic pain, whereas exogenous intrathecal CARTp evoked painful hypersensitivity through GPR160-dependent ERK and cAMP response element-binding protein (CREB). Our findings de-orphanize GPR160, identify it as a determinant of neuropathic pain and potential therapeutic target, and provide insights into its signaling pathways. CARTp is involved in many diseases including depression and reward and addiction; de-orphanization of GPR160 is a major step forward understanding the role of CARTp signaling in health and disease.
Collapse
Affiliation(s)
- Gina L.C. Yosten
- Department of Pharmacology and Physiology
- Henry and Amelia Nasrallah Center for Neuroscience, and
| | - Caron M. Harada
- Department of Pharmacology and Physiology
- Henry and Amelia Nasrallah Center for Neuroscience, and
| | - Chris Haddock
- Department of Pharmacology and Physiology
- Henry and Amelia Nasrallah Center for Neuroscience, and
| | | | - Grant R. Kolar
- Henry and Amelia Nasrallah Center for Neuroscience, and
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Ryan Patel
- Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Chun Guo
- Department of Pharmacology and Physiology
| | - Zhoumou Chen
- Department of Pharmacology and Physiology
- Henry and Amelia Nasrallah Center for Neuroscience, and
| | - Jinsong Zhang
- Department of Pharmacology and Physiology
- Henry and Amelia Nasrallah Center for Neuroscience, and
| | - Timothy M. Doyle
- Department of Pharmacology and Physiology
- Henry and Amelia Nasrallah Center for Neuroscience, and
| | - Anthony H. Dickenson
- Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Willis K. Samson
- Department of Pharmacology and Physiology
- Henry and Amelia Nasrallah Center for Neuroscience, and
| | - Daniela Salvemini
- Department of Pharmacology and Physiology
- Henry and Amelia Nasrallah Center for Neuroscience, and
| |
Collapse
|
39
|
Wei P, Wang P, Li B, Gu H, Liu J, Wang Z. Divergence and Convergence of Cerebral Ischemia Pathways Profile Deciphers Differential Pure Additive and Synergistic Mechanisms. Front Pharmacol 2020; 11:80. [PMID: 32161541 PMCID: PMC7053362 DOI: 10.3389/fphar.2020.00080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/27/2020] [Indexed: 12/11/2022] Open
Abstract
Aim The variable mechanisms on additive and synergistic effects of jasminoidin (JA)-Baicalin (BA) combination and JA-ursodeoxycholic acid (UA) combination in treating cerebral ischemia are not completely understood. In this study, we explored the differential pure mechanisms of additive and synergistic effects based on pathway analysis that excluded ineffective interference. Methods The MCAO mice were divided into eight groups: sham, vehicle, BA, JA, UA, Concha Margaritifera (CM), BA-JA combination (BJ), and JA-UA combination (JU). The additive and synergistic effects of combination groups were identified by cerebral infarct volume calculation. The differentially expressed genes based on a microarray chip containing 16,463 oligoclones were uploaded to GeneGo MetaCore software for pathway analyses and function catalogue. The comparison of specific pathways and functions crosstalk between different groups were analyzed to reveal the underlying additive and synergistic pharmacological variations. Results Additive BJ and synergistic JU were more effective than monotherapies of BA, JA, and UA, while CM was ineffective. Compared with monotherapies, 43 pathways and six functions were found uniquely in BJ group, with 33 pathways and three functions in JU group. We found six overlapping pathways and six overlapping functions between BJ and JU groups, which mainly involved central nervous system development. Thirty-seven specific pathways and 10 functions were activated by additive BJ, which were mainly related to cell adhesion and G-protein signaling; and 27 specific pathways and three functions of synergistic JU were associated with regulation of metabolism, DNA damage, and translation. The overlapping and distinct pathways and functions may contribute to different additive and synergistic effects. Conclusion The divergence pathways of pure additive effect of BJ were mainly related to cell adhesion and G-protein signaling, while the pure synergistic mechanism of JU depended on metabolism, translation and DNA damage. Such a systematic analysis of pathways may provide an important paradigm to reveal the pharmacological mechanisms underlying drug combinations.
Collapse
Affiliation(s)
- Penglu Wei
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Pengqian Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bing Li
- Institute of Information on Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Gu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
40
|
Stevenson MJ, Farran IC, Uyeda KS, San Juan JA, Heffern MC. Analysis of Metal Effects on C-Peptide Structure and Internalization. Chembiochem 2019; 20:2447-2453. [PMID: 31074079 DOI: 10.1002/cbic.201900172] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Indexed: 01/17/2023]
Abstract
The connecting peptide (C-peptide) has received increased attention for its potential therapeutic effects in ameliorating illnesses such as kidney disease and diabetes. Although the mechanism of C-peptide signaling remains elusive, evidence supports its internalization and intracellular function. Emerging research is uncovering the diverse biological roles metals play in controlling and affecting the function of bioactive peptides. The work presented herein investigates interactions between C-peptide and first-row d-block transition metals, as well as their effects on C-peptide internalization into cells. Through spectroscopic techniques, it is demonstrated that CrIII , CuII , and ZnII bind to C-peptide with differing stoichiometries and biologically relevant affinities. In addition, metal binding elicits both subtle changes in secondary structure and inhibits adoption of an α-helical character in environments where the dielectric constants are reduced. This study shows how metal ions can modulate peptide hormone activity through subtle structural changes to disrupt cellular uptake.
Collapse
Affiliation(s)
- Michael J Stevenson
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Ian C Farran
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Kylie S Uyeda
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Jessica A San Juan
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Marie C Heffern
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA, 95616, USA
| |
Collapse
|
41
|
Martin JH, Aitken RJ, Bromfield EG, Cafe SL, Sutherland JM, Frost ER, Nixon B, Lord T. Investigation into the presence and functional significance of proinsulin C-peptide in the female germline†. Biol Reprod 2019; 100:1275-1289. [PMID: 30715203 DOI: 10.1093/biolre/ioz008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 12/10/2018] [Accepted: 01/28/2019] [Indexed: 08/02/2024] Open
Abstract
Diabetes is associated with poor oocyte quality and the dysregulation of ovarian function and is thus a leading contributor to the increasing prevalence of female reproductive pathologies. Accordingly, it is well-established that insulin fulfills a key role in the regulation of several facets of female reproduction. What remains less certain is whether proinsulin C-peptide, which has recently been implicated in cellular signaling cascades, holds a functional role in the female germline. In the present study, we examined the expression of insulin, C-peptide, and its purported receptor; GPR146, within the mouse ovary and oocyte. Our data establish the presence of abundant C-peptide within follicular fluid and raise the prospect that this bioactive peptide is internalized by oocytes in a G-protein coupled receptor-dependent manner. Further, our data reveal that internalized C-peptide undergoes pronounced subcellular relocalization from the ooplasm to the pronuclei postfertilization. The application of immunoprecipitation analysis and mass spectrometry identified breast cancer type 2 susceptibility protein (BRCA2), the meiotic resumption/DNA repair protein, as a primary binding partner for C-peptide within the oocyte. Collectively, these findings establish a novel accumulation profile for C-peptide in the female germline and provide the first evidence for an interaction between C-peptide and BRCA2. This interaction is particularly intriguing when considering the propensity for oocytes from diabetic women to experience aberrant meiotic resumption and perturbation of traditional DNA repair processes. This therefore provides a clear imperative for further investigation of the implications of dysregulated C-peptide production in these individuals.
Collapse
Affiliation(s)
- Jacinta H Martin
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The Hunter Medical Research Institute, New Lambton Heights and the University of Newcastle, Callaghan, Newcastle, Australia
| | - R John Aitken
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The Hunter Medical Research Institute, New Lambton Heights and the University of Newcastle, Callaghan, Newcastle, Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The Hunter Medical Research Institute, New Lambton Heights and the University of Newcastle, Callaghan, Newcastle, Australia
| | - Shenae L Cafe
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The Hunter Medical Research Institute, New Lambton Heights and the University of Newcastle, Callaghan, Newcastle, Australia
| | - Jessie M Sutherland
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The Hunter Medical Research Institute, New Lambton Heights and the University of Newcastle, Callaghan, Newcastle, Australia
| | - Emily R Frost
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The Hunter Medical Research Institute, New Lambton Heights and the University of Newcastle, Callaghan, Newcastle, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The Hunter Medical Research Institute, New Lambton Heights and the University of Newcastle, Callaghan, Newcastle, Australia
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The Hunter Medical Research Institute, New Lambton Heights and the University of Newcastle, Callaghan, Newcastle, Australia
- School of Molecular Biosciences, Centre for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
42
|
Stevenson MJ, Uyeda KS, Harder NHO, Heffern MC. Metal-dependent hormone function: the emerging interdisciplinary field of metalloendocrinology. Metallomics 2019; 11:85-110. [PMID: 30270362 PMCID: PMC10249669 DOI: 10.1039/c8mt00221e] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
For over 100 years, there has been an incredible amount of knowledge amassed concerning hormones in the endocrine system and their central role in human health. Hormones represent a diverse group of biomolecules that are released by glands, communicate signals to their target tissue, and are regulated by feedback loops to maintain organism health. Many disease states, such as diabetes and reproductive disorders, stem from misregulation or dysfunction of hormones. Increasing research is illuminating the intricate roles of metal ions in the endocrine system where they may act advantageously in concert with hormones or deleteriously catalyze hormone-associated disease states. As the critical role of metal ions in the endocrine system becomes more apparent, it is increasingly important to untangle the complex mechanisms underlying the connections between inorganic biochemistry and hormone function to understand and control endocrinological phenomena. This tutorial review harmonizes the interdisciplinary fields of endocrinology and inorganic chemistry in the newly-termed field of "metalloendocrinology". We describe examples linking metals to both normal and aberrant hormone function with a focus on highlighting insight to molecular mechanisms. Hormone activities related to both essential metal micronutrients, such as copper, iron, zinc, and calcium, and disruptive nonessential metals, such as lead and cadmium are discussed.
Collapse
Affiliation(s)
- Michael J Stevenson
- Department of Chemistry, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA.
| | | | | | | |
Collapse
|
43
|
Täubel J, Ferber G, Van Langenhoven L, Del Bianco T, Fernandes S, Djumanov D, Kanters JK, Graff C, Camm AJ. The Cardiovascular Effects of a Meal: J-T peak and T peak -T end Assessment and Further Insights Into the Physiological Effects. J Clin Pharmacol 2019; 59:799-810. [PMID: 30633366 PMCID: PMC6590239 DOI: 10.1002/jcph.1374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022]
Abstract
Meal intake leads to a significant and prolonged increase in cardiac output to supply the splanchnic vasculature. A meal is associated with sympathetic activation of the cardiovascular system, and food ingestion is correlated with an increase in heart rate, an increase in cardiac stroke volume, and QTc interval shortening for up to 7 hours. Given the complexity of the system, one or several of many mechanisms could explain this observation. The shortening of the QTc interval was correlated with a rise of C‐peptide following food ingestion, but the mechanisms by which C‐peptide may be involved in the modulation of cardiac repolarization are still unknown. This shortening of the myocardial action potential caused by the ingestion of food was further investigated in the present study by measuring the QRS, J‐Tpeak, and Tpeak‐Tend intervals in search of further clues to better understand the underlying mechanisms. A retrospective analysis was conducted based on data collected in a formal thorough QT/QTc study in which 32 subjects received a carbohydrate‐rich “continental” breakfast, moxifloxacin without food, and moxifloxacin with food. We assessed the effect of food on T‐wave morphology using validated algorithms for measurement of J‐Tpeak and Tpeak‐Tend intervals. Our findings demonstrate that a standardized meal significantly shortened J‐Tpeak for 4 hours after a meal and to a much lesser extent and shorter duration (up to 1 hour) prolonged the Tpeak‐Tend and QRS intervals. This suggests that the QTc shortening occurs mainly during phase 2 of the cardiac action potential. As there was no corresponding effect on Tpeak‐Tend beyond the first hour, we conclude that a meal does not interfere with the outward correcting potassium channels but possibly with Ca2+ currents. An effect on mainly Ca2+ aligns well with our understanding of physiology whereby an increase in stroke volume, as observed after a meal, is associated with changes in Ca2+ cycling in and out of the sarcoplasmic reticulum during cardiac myocyte contraction.
Collapse
Affiliation(s)
- Jörg Täubel
- Richmond Pharmacology Ltd., St George's University of London, Cranmer Terrace, London, UK.,Cardiovascular and Cell Sciences Research Institute, St George's University of London, London, UK
| | - Georg Ferber
- Statistik Georg Ferber GmbH, Cagliostrostrasse, Riehen, Switzerland
| | - Leen Van Langenhoven
- Richmond Pharmacology Ltd., St George's University of London, Cranmer Terrace, London, UK
| | - Teresa Del Bianco
- Richmond Pharmacology Ltd., St George's University of London, Cranmer Terrace, London, UK
| | - Sara Fernandes
- Richmond Pharmacology Ltd., St George's University of London, Cranmer Terrace, London, UK
| | - Dilshat Djumanov
- Richmond Pharmacology Ltd., St George's University of London, Cranmer Terrace, London, UK
| | - Jørgen K Kanters
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Graff
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - A John Camm
- Cardiovascular and Cell Sciences Research Institute, St George's University of London, London, UK
| |
Collapse
|
44
|
Derkach KV, Shpakova EA, Bondareva VM, Shpakov AO. The Effect of Intranasal Administration of Proinsulin C-peptide and Its C-terminal Fragment on Metabolic Parameters in Rats with Streptozotocin Diabetes. J EVOL BIOCHEM PHYS+ 2018. [DOI: 10.1134/s0022093018030092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Stein LM, Haddock CJ, Samson WK, Kolar GR, Yosten GLC. The phoenixins: From discovery of the hormone to identification of the receptor and potential physiologic actions. Peptides 2018; 106:45-48. [PMID: 29933026 PMCID: PMC6092957 DOI: 10.1016/j.peptides.2018.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/12/2018] [Accepted: 06/19/2018] [Indexed: 01/02/2023]
Abstract
Using a series of classical protein purification techniques, coupled with more modern molecular approaches, a family of neuropeptides, the Phoenixins, was identified to be produced in brain and heart, and to bind selectively in pituitary gland, ovary and brain. These same binding sites were revealed, using a novel receptor identification strategy, to express the orphan G protein-coupled receptor, GPR173, the expression of which was required for the actions of phoenixin both in vivo and in vitro. In fact, studies using small interfering RNA molecules to compromise GPR173 expression revealed the physiologic relevance of the initially reported pharmacologic actions of the peptides. Those include not only the reproductive actions of the peptides in brain and pituitary gland, but also a CNS site of action in the maintenance of fluid and electrolyte homeostasis. Additional pharmacologic actions of the phoenixins have been described and the race is on to establish the physiologic relevance of those actions as well as the therapeutic potential of phoenixin analogs.
Collapse
Affiliation(s)
- Lauren M Stein
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Christopher J Haddock
- Departments of Pharmacology and Physiology, Saint Louis University, St. Louis, MO, 63104, United States
| | - Willis K Samson
- Departments of Pharmacology and Physiology, Saint Louis University, St. Louis, MO, 63104, United States
| | - Grant R Kolar
- Department of Pathology, Saint Louis University, St. Louis, MO, 63104, United States
| | - Gina L C Yosten
- Departments of Pharmacology and Physiology, Saint Louis University, St. Louis, MO, 63104, United States.
| |
Collapse
|
46
|
Derkach KV, Bondareva VM, Shpakov AO. Coadministration of Intranasally Delivered Insulin and Proinsulin C-Peptide to Rats with Types 1 and 2 Diabetes Mellitus Restores Their Metabolic Parameters. ADVANCES IN GERONTOLOGY 2018. [DOI: 10.1134/s2079057018020030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
47
|
The G protein-coupled receptors deorphanization landscape. Biochem Pharmacol 2018; 153:62-74. [PMID: 29454621 DOI: 10.1016/j.bcp.2018.02.016] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) are usually highlighted as being both the largest family of membrane proteins and the most productive source of drug targets. However, most of the GPCRs are understudied and hence cannot be used immediately for innovative therapeutic strategies. Besides, there are still around 100 orphan receptors, with no described endogenous ligand and no clearly defined function. The race to discover new ligands for these elusive receptors seems to be less intense than before. Here, we present an update of the various strategies employed to assign a function to these receptors and to discover new ligands. We focus on the recent advances in the identification of endogenous ligands with a detailed description of newly deorphanized receptors. Replication being a key parameter in these endeavors, we also discuss the latest controversies about problematic ligand-receptor pairings. In this context, we propose several recommendations in order to strengthen the reporting of new ligand-receptor pairs.
Collapse
|
48
|
Halperin Kuhns VL, Pluznick JL. Novel differences in renal gene expression in a diet-induced obesity model. Am J Physiol Renal Physiol 2017; 314:F517-F530. [PMID: 29141937 DOI: 10.1152/ajprenal.00345.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Obesity is a significant risk factor for both chronic kidney disease and end-stage renal disease. To better understand disease development, we sought to identify novel genes differentially expressed early in disease progression. We first confirmed that mice fed a high-fat (HF) diet exhibit early signs of renal injury including hyperfiltration. We then performed RNA-Seq using renal cortex RNA from C57BL6/J male mice fed either HF or control (Ctrl) diet. We identified 1,134 genes differentially expressed in the cortex on HF vs. Ctrl, of which 31 genes were selected for follow-up analysis. This included the 9 most upregulated, the 11 most downregulated, and 11 genes of interest (primarily sensory receptors and G proteins). Quantitative (q)RT-PCR for these 31 genes was performed on additional male renal cortex and medulla samples, and 11 genes (including all 9 upregulated genes) were selected for further study based on qRT-PCR. We then examined expression of these 11 genes in Ctrl and HF male heart and liver samples, which demonstrated that these changes are relatively specific to the renal cortex. These 11 genes were also examined in female renal cortex, where we found that the expression changes seen in males on a HF diet are not replicated in females, even when the females are started on the diet sooner to match weight gain of the males. In sum, these data demonstrate that in a HF-diet model of early disease, novel transcriptional changes occur that are both sex specific and specific to the renal cortex.
Collapse
Affiliation(s)
| | - Jennifer L Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|
49
|
Huang H, Zhang N, Xiong Q, Chen R, Zhang C, Wang N, Wang L, Ren H, Liu M, Qian M, Du B. Elimination of GPR146-mediated antiviral function through IRF3/HES1-signalling pathway. Immunology 2017; 152:102-114. [PMID: 28464285 PMCID: PMC5543731 DOI: 10.1111/imm.12752] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 03/23/2017] [Accepted: 04/24/2017] [Indexed: 12/18/2022] Open
Abstract
As the most important host defence against viral infection, interferon (IFN) stimulates hundreds of antiviral genes (ISGs) that together establish an 'antiviral state'. However, the antiviral function of most ISGs in viral infection still need further exploration. Here, we demonstrated that the expression of G-protein-coupled receptor 146 (GPR146) is highly increased by both IFN-β and IFN-γ in a signal transducer and activator of transcription 1-dependent signalling pathway. Most importantly, overexpression of GPR146 protects the host cells from vesicular stomatitis virus and Newcastle disease virus infection but not from infection by herpes simplex virus. In contrast, the virus-induced IFN-β production changed little in Gpr146-knockout cells. Furthermore, the Gpr146-deficient mice showed similar susceptibility to wild-type mice with vesicular stomatitis virus infection. Interestingly, the expression of GPR146 in virus-infected cells was strikingly reduced and can partially explain why the viral infection was little influenced in Gpr146-knockout mice. Surprisingly, virus-activated IFN regulatory factor 3 (IRF3) signalling not only induces the expression of IFN but also represses GPR146 expression through HES1 (hairy and enhancer of split-1)-mediated transcriptional activity to establish a dynamic equilibrium between pro-viral and antiviral stages in host cells. Taken together, these data reveal the antiviral role of GPR146 in fighting viral infection although the GPR146-mediated protection is eliminated by IRF3/HES1-signalling, which suggests a potential therapeutic significance of both GPR146 and HES1 signalling in viral infection.
Collapse
MESH Headings
- Animals
- Chlorocebus aethiops
- Genotype
- HEK293 Cells
- Herpes Simplex/immunology
- Herpes Simplex/metabolism
- Herpes Simplex/prevention & control
- Herpes Simplex/virology
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/metabolism
- Host-Pathogen Interactions
- Humans
- Interferon Regulatory Factor-3/immunology
- Interferon Regulatory Factor-3/metabolism
- Interferon-beta/pharmacology
- Interferon-gamma/pharmacology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/virology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Newcastle Disease/immunology
- Newcastle Disease/metabolism
- Newcastle Disease/prevention & control
- Newcastle Disease/virology
- Newcastle disease virus/immunology
- Newcastle disease virus/metabolism
- Phenotype
- RAW 264.7 Cells
- RNA Interference
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/immunology
- Signal Transduction
- Transcription Factor HES-1/immunology
- Transcription Factor HES-1/metabolism
- Transfection
- Vero Cells
- Vesicular Stomatitis/immunology
- Vesicular Stomatitis/metabolism
- Vesicular Stomatitis/prevention & control
- Vesicular Stomatitis/virology
- Vesicular stomatitis Indiana virus/immunology
- Vesicular stomatitis Indiana virus/metabolism
- Virus Replication
Collapse
Affiliation(s)
- Hongjun Huang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Na Zhang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Qingqing Xiong
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Ruoyu Chen
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Chengfei Zhang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Ning Wang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Li Wang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Hua Ren
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Min Qian
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Bing Du
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghaiChina
| |
Collapse
|
50
|
Pinger CW, Entwistle KE, Bell TM, Liu Y, Spence DM. C-Peptide replacement therapy in type 1 diabetes: are we in the trough of disillusionment? MOLECULAR BIOSYSTEMS 2017; 13:1432-1437. [PMID: 28685788 PMCID: PMC5796410 DOI: 10.1039/c7mb00199a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Type 1 diabetes is associated with such complications as blindness, kidney failure, and nerve damage. Replacing C-peptide, a hormone normally co-secreted with insulin, has been shown to reduce diabetes-related complications. Interestingly, after nearly 30 years of positive research results, C-peptide is still not being co-administered with insulin to diabetic patients. The following review discusses the potential of C-peptide as an auxilliary replacement therapy and why it's not currently being used as a therapeutic.
Collapse
Affiliation(s)
- C W Pinger
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln, East Lansing, MI 48824, USA. and Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, MI 48824-6962, USA
| | - K E Entwistle
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln, East Lansing, MI 48824, USA.
| | - T M Bell
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln, East Lansing, MI 48824, USA. and Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, MI 48824-6962, USA
| | - Y Liu
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln, East Lansing, MI 48824, USA.
| | - D M Spence
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln, East Lansing, MI 48824, USA. and Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, MI 48824-6962, USA
| |
Collapse
|