1
|
Lee S, Kim YJ, Jeong S, Rho KN, Noh JE, Kim H, Cho H, Choi YD, Yang DH, Hwang EC, Bae WK, Yun SJ, Yun JS, Park C, Oh I, Cho J. Ex vivo STAT3 phosphorylation in circulating immune cells: a novel biomarker for early cancer diagnosis and response to anti-PD-1 therapy. Cancer Commun (Lond) 2025; 45:58-62. [PMID: 39568320 PMCID: PMC11758149 DOI: 10.1002/cac2.12631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024] Open
Affiliation(s)
- Sung‐Woo Lee
- Department of Microbiology and ImmunologyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- Medical Research Center for Combinatorial Tumor ImmunotherapyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- National Immunotherapy Innovation CenterChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
| | - Young Ju Kim
- Department of Microbiology and ImmunologyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- Medical Research Center for Combinatorial Tumor ImmunotherapyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- National Immunotherapy Innovation CenterChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- BioMedical Sciences Graduate ProgramChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
| | - Saei Jeong
- Department of Microbiology and ImmunologyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- Medical Research Center for Combinatorial Tumor ImmunotherapyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- National Immunotherapy Innovation CenterChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- BioMedical Sciences Graduate ProgramChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
| | - Kyung Na Rho
- Department of Microbiology and ImmunologyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- Medical Research Center for Combinatorial Tumor ImmunotherapyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- National Immunotherapy Innovation CenterChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- BioMedical Sciences Graduate ProgramChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
| | - Jeong Eun Noh
- Department of Microbiology and ImmunologyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- Medical Research Center for Combinatorial Tumor ImmunotherapyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- National Immunotherapy Innovation CenterChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- BioMedical Sciences Graduate ProgramChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
| | | | - Hyun‐Ju Cho
- Department of Internal MedicineChonnam National University Medical SchoolHwasun HospitalHwasunupJeollanamdoRepublic of Korea
| | - Yoo Duk Choi
- Department of PathologyChonnam National University Medical SchoolHwasun HospitalHwasunupJeollanamdoRepublic of Korea
| | - Deok Hwan Yang
- Department of Internal MedicineChonnam National University Medical SchoolHwasun HospitalHwasunupJeollanamdoRepublic of Korea
| | - Eu Chang Hwang
- Department of UrologyChonnam National University Medical SchoolHwasun HospitalHwasunupJeollanamdoRepublic of Korea
| | - Woo Kyun Bae
- Department of Internal MedicineChonnam National University Medical SchoolHwasun HospitalHwasunupJeollanamdoRepublic of Korea
| | - Sook Jung Yun
- Department of DermatologyChonnam National University Medical SchoolHwasun HospitalHwasunupJeollanamdoRepublic of Korea
| | - Ju Sik Yun
- Department of Thoracic and Cardiovascular SurgeryChonnam National University Medical SchoolHwasun HospitalHwasunupJeollanamdoRepublic of Korea
| | - Cheol‐Kyu Park
- Department of Internal MedicineChonnam National University Medical SchoolHwasun HospitalHwasunupJeollanamdoRepublic of Korea
| | - In‐Jae Oh
- Department of Internal MedicineChonnam National University Medical SchoolHwasun HospitalHwasunupJeollanamdoRepublic of Korea
| | - Jae‐Ho Cho
- Department of Microbiology and ImmunologyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- Medical Research Center for Combinatorial Tumor ImmunotherapyChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- National Immunotherapy Innovation CenterChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
- BioMedical Sciences Graduate ProgramChonnam National University Medical SchoolHwasunupJeollanamdoRepublic of Korea
| |
Collapse
|
2
|
Sarma U, Ripka L, Anyaegbunam UA, Legewie S. Modeling Cellular Signaling Variability Based on Single-Cell Data: The TGFβ-SMAD Signaling Pathway. Methods Mol Biol 2023; 2634:215-251. [PMID: 37074581 DOI: 10.1007/978-1-0716-3008-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Nongenetic heterogeneity is key to cellular decisions, as even genetically identical cells respond in very different ways to the same external stimulus, e.g., during cell differentiation or therapeutic treatment of disease. Strong heterogeneity is typically already observed at the level of signaling pathways that are the first sensors of external inputs and transmit information to the nucleus where decisions are made. Since heterogeneity arises from random fluctuations of cellular components, mathematical models are required to fully describe the phenomenon and to understand the dynamics of heterogeneous cell populations. Here, we review the experimental and theoretical literature on cellular signaling heterogeneity, with special focus on the TGFβ/SMAD signaling pathway.
Collapse
Affiliation(s)
- Uddipan Sarma
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Lorenz Ripka
- Institute of Molecular Biology (IMB), Mainz, Germany
- Department of Systems Biology, Institute for Biomedical Genetics, University of Stuttgart, Stuttgart, Germany
| | - Uchenna Alex Anyaegbunam
- Institute of Molecular Biology (IMB), Mainz, Germany
- Department of Systems Biology, Institute for Biomedical Genetics, University of Stuttgart, Stuttgart, Germany
| | - Stefan Legewie
- Institute of Molecular Biology (IMB), Mainz, Germany.
- Department of Systems Biology, Institute for Biomedical Genetics, University of Stuttgart, Stuttgart, Germany.
- Stuttgart Research Center for Systems Biology, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
3
|
Serna-Duque JA, Cuesta A, Esteban MÁ. Massive gene expansion of hepcidin, a host defense peptide, in gilthead seabream (Sparus aurata). FISH & SHELLFISH IMMUNOLOGY 2022; 124:563-571. [PMID: 35489593 DOI: 10.1016/j.fsi.2022.04.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 03/09/2022] [Accepted: 04/21/2022] [Indexed: 06/14/2023]
Abstract
Host defense peptides (HDP) are among the most ancient immune molecules in animals and clearly reflect an ancestral evolutionary history involving pathogen-host interactions. Hepcidins are a very widespread family of HDPs among vertebrates and are especially diverse in teleosts. We have investigated the identification of new hepcidins in gilthead seabream (Sparus aurata), a fish farmed in the Mediterranean. Targeted gene predictions supported with expressed sequence tags (ESTs) derived from Hidden Markov Models were used to find the hamp genes in the seabream genome. The results revealed a massively clustered hamp duplication on chromosome 17. In fact, the seabream genome contains the largest number of hepcidin copies described in any vertebrate. The evolutionary history of hepcidins in seabream, and vertebrates generally, clearly indicates high adaptation in teleosts and novel subgroups within hepcidin type II. Furthermore, basal hepcidin gene expression analysis indicates specific-tissue expression profiles, while the presence and distribution of transcription factor binding sites (TFBS) in hamp promoters as well as their transcription profile upon bacterial challenge indicates different immune roles depending on the type of hepcidin and tissue. This massive duplication of HDP genes in a bony fish could point to a far more specific and adaptive innate immune system than assumed in the classic concept of immunity in mammals. Hence, a new world of knowledge regarding hepcidins in fish and vertebrates is being initiated.
Collapse
Affiliation(s)
- Jhon A Serna-Duque
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus of International Excellence, Campus Mare Nostrum, University of Murcia, 30100, Murcia, Spain
| | - Alberto Cuesta
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus of International Excellence, Campus Mare Nostrum, University of Murcia, 30100, Murcia, Spain
| | - M Ángeles Esteban
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus of International Excellence, Campus Mare Nostrum, University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
4
|
Hoffmann A, de Souza LV, Seifert M, von Raffay L, Haschka D, Grubwieser P, Grander M, Mitterstiller AM, Nairz M, Poli M, Weiss G. Pharmacological Targeting of BMP6-SMAD Mediated Hepcidin Expression Does Not Improve the Outcome of Systemic Infections With Intra-Or Extracellular Gram-Negative Bacteria in Mice. Front Cell Infect Microbiol 2021; 11:705087. [PMID: 34368018 PMCID: PMC8342937 DOI: 10.3389/fcimb.2021.705087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
Introduction Hepcidin is the systemic master regulator of iron metabolism as it degrades the cellular iron exporter ferroportin. In bacterial infections, hepcidin is upregulated to limit circulating iron for pathogens, thereby increasing iron retention in macrophages. This mechanism withholds iron from extracellular bacteria but could be of disadvantage in infections with intracellular bacteria. We aimed to understand the role of hepcidin in infections with intra- or extracellular bacteria using different hepcidin inhibitors. Methods For the experiments LDN-193189 and oversulfated heparins were used, which interact with the BMP6-SMAD pathway thereby inhibiting hepcidin expression. We infected male C57BL/6N mice with either the intracellular bacterium Salmonella Typhimurium or the extracellular bacterium Escherichia coli and treated these mice with the different hepcidin inhibitors. Results Both inhibitors effectively reduced hepcidin levels in vitro under steady state conditions and upon stimulation with the inflammatory signals interleukin-6 or lipopolysaccharide. The inhibitors also reduced hepcidin levels and increased circulating iron concentration in uninfected mice. However, both compounds failed to decrease liver- and circulating hepcidin levels in infected mice and did not affect ferroportin expression in the spleen or impact on serum iron levels. Accordingly, both BMP-SMAD signaling inhibitors did not influence bacterial numbers in different organs in the course of E.coli or S.Tm sepsis. Conclusion These data indicate that targeting the BMP receptor or the BMP-SMAD pathway is not sufficient to suppress hepcidin expression in the course of infection with both intra- or extracellular bacteria. This suggests that upon pharmacological inhibition of the central SMAD-BMP pathways during infection, other signaling cascades are compensatorily induced to ensure sufficient hepcidin formation and iron restriction to circulating microbes.
Collapse
Affiliation(s)
- Alexander Hoffmann
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Lara Valente de Souza
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Laura von Raffay
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Philipp Grubwieser
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Manuel Grander
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna-Maria Mitterstiller
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Maura Poli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
5
|
Shibabaw T, Teferi B, Molla MD, Ayelign B. Inflammation Mediated Hepcidin-Ferroportin Pathway and Its Therapeutic Window in Breast Cancer. BREAST CANCER-TARGETS AND THERAPY 2020; 12:165-180. [PMID: 33116818 PMCID: PMC7585830 DOI: 10.2147/bctt.s276404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
Abstract
Experimental and clinical data strongly support that iron is an essential element which plays a big role in cancer biology. Thus, hepcidin (Hp) and ferroportin (Fpn) are molecules that regulate and maintain the metabolism of iron. A peptide hormone hepcidin limits recycled and stored iron fluxes in macrophage and hepatic hepatocyte, respectively, to the blood stream by promoting degradation of the only iron exporter, Fpn, in the target cells. Moreover, the inflammatory microenvironment of breast cancer and altered hepcidin/ferroportin pathway is intimately linked. Breast cancer exhibits an iron seeking phenotype that is accomplished by tumor-associated macrophage (TAM). Because macrophages contribute to breast cancer growth and progression, this review will discuss TAM with an emphasis on describing how TAM (M2Ф phenotypic) interacts with their surrounding microenvironment and results in dysregulated Hp/Fpn and pathologic accumulation of iron as a hallmark of its malignant condition. Moreover, the underlying stroma or tumor microenvironment releases significant inflammatory cytokines like IL-6 and bone morphogenetic proteins like BMP-2 and 6 leading in aberrant Hp/Fpn pathways in breast cancer. Inflammation is primarily associated with the high intracellular iron levels, deregulated hepcidin/ferroportin pathway, and its upstream signaling in breast cancer. Subsequently, scholars have been reported that reducing iron level and manipulating the signaling molecules involved in iron metabolism can be used as a promising strategy of tumor chemotherapy. Here, we review the key molecular aspects of iron metabolism and its regulatory mechanisms of the hepcidin/ferroportin pathways and its current therapeutic strategies in breast cancer.
Collapse
Affiliation(s)
- Tewodros Shibabaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Banchamlak Teferi
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Meseret Derbew Molla
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
6
|
Cheng Z, Yan M, Lu Y, Pan XT. Expression of serum BMP6 and hepcidin in cancer-related anemia. ACTA ACUST UNITED AC 2020; 25:134-138. [PMID: 32153255 DOI: 10.1080/16078454.2020.1738098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Objective: To study the function of human bone morphogenetic protein-6 (BMP6) and hepcidin in cancer-related anemia.Methods: The levels of Hemoglobin (Hb), serum C-reactive protein (CRP), BMP6, hepcidin and ferritin (SF) were measured in 115 patients with solid tumors, who were divided into elevated CRP group and normal CRP group, and further divided into anemia subgroup and non-anemia subgroup according to the CRP level.Results: The prevalence of anemia was 53.9% in all patients. In the elevated CRP group, the levels of CRP, hepcidin and SF of the anemia subgroup were higher than the non-anemia subgroup (P < 0.05); the BMP6 levels had no difference between the subgroups (P > 0.05). In the normal CRP group, the BMP6 level in the anemia subgroup was higher than the non-anemia subgroup (P < 0.01); the CRP, hepcidin and SF levels showed no difference between the subgroups (P > 0.05). Among the 115 patients, CRP and SF were both positively correlated with hepcidin (P < 0.05), and CRP and hepcidin were negatively correlated with Hb (P < 0.05), while SF had no correlation with Hb (P > 0.05); BMP6 was negatively correlated with Hb (P < 0.05) while had no correlation with CRP, hepcidin and SF (P < 0.05).Conclusions: The occurrence of anemia in patients with elevated CRP is associated with hepcidin over-expression, while in patients with normal CRP is associated with BMP6 over-expression. We speculate BMP6 and hepcidin likely play different roles in the occurrence of cancer-related anemia.
Collapse
Affiliation(s)
- Zhen Cheng
- Department of Hematology, Taicang Hospital of Soochow University, Taicang, People's Republic of China
| | - Min Yan
- Department of Hematology, Taicang Hospital of Soochow University, Taicang, People's Republic of China
| | - Ye Lu
- Department of Hematology, Taicang Hospital of Soochow University, Taicang, People's Republic of China
| | - Xiang-Tao Pan
- Department of Hematology, Taicang Hospital of Soochow University, Taicang, People's Republic of China
| |
Collapse
|
7
|
Schirm S, Scholz M. A biomathematical model of human erythropoiesis and iron metabolism. Sci Rep 2020; 10:8602. [PMID: 32451387 PMCID: PMC7248076 DOI: 10.1038/s41598-020-65313-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 04/23/2020] [Indexed: 11/09/2022] Open
Abstract
Anaemia therapy or perisurgical support of erythropoiesis often require both, EPO and iron medication. However, excessive iron medication can result in iron overload and it is challenging to control haemoglobin levels in a desired range. To support this task, we develop a biomathematical model to simulate EPO- and iron medication in humans. We combine our previously established model of human erythropoiesis including comprehensive pharmacokinetic models of EPO applications with a newly developed model of iron metabolism including iron supplementation. Equations were derived by translating known biological mechanisms into ordinary differential equations. Qualitative model behaviour is studied in detail considering a variety of interventions such as bleeding, iron malnutrition and medication. The model can explain time courses of erythrocytes, reticulocytes, haemoglobin, haematocrit, red blood cells, EPO, serum iron, ferritin, transferrin saturation, and transferrin under a variety of scenarios including EPO and iron application into healthy volunteers or chemotherapy patients. Unknown model parameters were determined by fitting the predictions of the model to time series data from literature. We demonstrate how the model can be used to make predictions of untested therapy options such as cytotoxic chemotherapy supported by iron and EPO. Following our ultimate goal of establishing a model of anaemia treatment in chronic kidney disease, we aim at translating our model to this pathological condition in the near future.
Collapse
Affiliation(s)
- Sibylle Schirm
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
8
|
Enculescu M, Braun S, Thonta Setty S, Busch A, Zarnack K, König J, Legewie S. Exon Definition Facilitates Reliable Control of Alternative Splicing in the RON Proto-Oncogene. Biophys J 2020; 118:2027-2041. [PMID: 32336349 DOI: 10.1016/j.bpj.2020.02.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 01/01/2023] Open
Abstract
Alternative splicing is a key step in eukaryotic gene expression that allows for the production of multiple transcript and protein isoforms from the same gene. Even though splicing is perturbed in many diseases, we currently lack insights into regulatory mechanisms promoting its precision and efficiency. We analyze high-throughput mutagenesis data obtained for an alternatively spliced exon in the proto-oncogene RON and determine the functional units that control this splicing event. Using mathematical modeling of distinct splicing mechanisms, we show that alternative splicing is based in RON on a so-called "exon definition" mechanism. Here, the recognition of the adjacent exons by the spliceosome is required for removal of an intron. We use our model to analyze the differences between the exon and intron definition scenarios and find that exon definition prevents the accumulation of deleterious, partially spliced retention products during alternative splicing regulation. Furthermore, it modularizes splicing control, as multiple regulatory inputs are integrated into a common net input, irrespective of the location and nature of the corresponding cis-regulatory elements in the pre-messenger RNA. Our analysis suggests that exon definition promotes robust and reliable splicing outcomes in RON splicing.
Collapse
Affiliation(s)
| | - Simon Braun
- Institute of Molecular Biology, Mainz, Germany
| | - Samarth Thonta Setty
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Anke Busch
- Institute of Molecular Biology, Mainz, Germany
| | - Kathi Zarnack
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | | |
Collapse
|
9
|
Sheetz M, Barrington P, Callies S, Berg PH, McColm J, Marbury T, Decker B, Dyas GL, Truhlar SME, Benschop R, Leung D, Berg J, Witcher DR. Targeting the hepcidin-ferroportin pathway in anaemia of chronic kidney disease. Br J Clin Pharmacol 2019; 85:935-948. [PMID: 30677788 DOI: 10.1111/bcp.13877] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 01/04/2019] [Accepted: 01/13/2019] [Indexed: 12/15/2022] Open
Abstract
AIMS Erythropoiesis-stimulating agents used to treat anaemia in patients with chronic kidney disease (CKD) have been associated with cardiovascular adverse events. Hepcidin production, controlled by bone morphogenic protein 6 (BMP6), regulates iron homeostasis via interactions with the iron transporter, ferroportin. High hepcidin levels are thought to contribute to increased iron sequestration and subsequent anaemia in CKD patients. To investigate alternative therapies to erythropoiesis-stimulating agents for CKD patients, monoclonal antibodies, LY3113593 and LY2928057, targeting BMP6 and ferroportin respectively, were tested in CKD patients. METHODS Preclinical in vitro/vivo data and clinical data in healthy subjects and CKD patients were used to illustrate the translation of pharmacological properties of LY3113593 and LY2928057, highlighting the novelty of targeting these nodes within the hepcidin-ferroportin pathway. RESULTS LY2928057 bound ferroportin and blocked interactions with hepcidin, allowing iron efflux, leading to increased serum iron and transferrin saturation levels and increased hepcidin in monkeys and humans. In CKD patients, LY2928057 led to slower haemoglobin decline and reduction in ferritin (compared to placebo). Serum iron increase was (mean [90% confidence interval]) 1.98 [1.46-2.68] and 1.36 [1.22-1.51] fold-relative to baseline following LY2928057 600 mg and LY311593 150 mg respectively in CKD patients. LY3113593 specifically blocked BMP6 binding to its receptor and produced increases in iron and transferrin saturation and decreases in hepcidin preclinically and clinically. In CKD patients, LY3113593 produced an increase in haemoglobin and reduction in ferritin (compared to placebo). CONCLUSION LY3113593 and LY2928057 pharmacological effects (serum iron and ferritin) were translated from preclinical-to-clinical development. Such interventions may lead to new CKD anaemia treatments.
Collapse
Affiliation(s)
| | | | | | - Paul H Berg
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | | | - Brian Decker
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | | | | - Jolene Berg
- DaVita Clinical Research, Minneapolis, Minnesota, USA
| | | |
Collapse
|
10
|
Katsarou MS, Papasavva M, Latsi R, Drakoulis N. Hemochromatosis: Hereditary hemochromatosis and HFE gene. VITAMINS AND HORMONES 2019; 110:201-222. [PMID: 30798813 DOI: 10.1016/bs.vh.2019.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hereditary Hemochromatosis (HH) is an autosomal recessive genetic disease, characterized by an excessively increased absorption of dietary iron. Excess iron can be accumulated because of the lack of an effective excretory mechanism leading to toxic effects. HH is one of the most common genetic disorders in individuals of European descent. Genetic polymorphisms of the HFE gene (rs1800562, rs1799945 and rs1800730) also affect the normal activity of another protein, hepcidin, a negative regulator of iron homeostasis. If left untreated, hereditary hemochromatosis can lead to morbidity and eventually death. Clinical onset hereditary hemochromatosis symptoms occur more frequently in adult men than women, as the monthly loss of iron due to menstruation in women slows down accumulation and the symptoms usually start appearing after menopause. Therapeutic phlebotomy is the primary form of treatment for this disease so far, combined with the use of chelating agents. Orthotopic liver transplantation (OTL) is performed in patients with advanced cirrhosis. In order to prevent the progression of iron accumulation, an early detection may be achieved by genotypic check of the frequent mutations of the HFE. Consequently, initiation of treatment may take place before the development of clinical symptoms, particularly cirrhosis, contributing significantly in achieving normal life expectancy. Therefore, genotypic check is vital in order to prevent the development of this type of hemochromatosis.
Collapse
Affiliation(s)
- Martha-Spyridoula Katsarou
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Papasavva
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Rozana Latsi
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Drakoulis
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
11
|
Enculescu M, Metzendorf C, Sparla R, Hahnel M, Bode J, Muckenthaler MU, Legewie S. Modelling Systemic Iron Regulation during Dietary Iron Overload and Acute Inflammation: Role of Hepcidin-Independent Mechanisms. PLoS Comput Biol 2017; 13:e1005322. [PMID: 28068331 PMCID: PMC5261815 DOI: 10.1371/journal.pcbi.1005322] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 01/24/2017] [Accepted: 12/19/2016] [Indexed: 01/01/2023] Open
Abstract
Systemic iron levels must be maintained in physiological concentrations to prevent diseases associated with iron deficiency or iron overload. A key role in this process plays ferroportin, the only known mammalian transmembrane iron exporter, which releases iron from duodenal enterocytes, hepatocytes, or iron-recycling macrophages into the blood stream. Ferroportin expression is tightly controlled by transcriptional and post-transcriptional mechanisms in response to hypoxia, iron deficiency, heme iron and inflammatory cues by cell-autonomous and systemic mechanisms. At the systemic level, the iron-regulatory hormone hepcidin is released from the liver in response to these cues, binds to ferroportin and triggers its degradation. The relative importance of individual ferroportin control mechanisms and their interplay at the systemic level is incompletely understood. Here, we built a mathematical model of systemic iron regulation. It incorporates the dynamics of organ iron pools as well as regulation by the hepcidin/ferroportin system. We calibrated and validated the model with time-resolved measurements of iron responses in mice challenged with dietary iron overload and/or inflammation. The model demonstrates that inflammation mainly reduces the amount of iron in the blood stream by reducing intracellular ferroportin transcription, and not by hepcidin-dependent ferroportin protein destabilization. In contrast, ferroportin regulation by hepcidin is the predominant mechanism of iron homeostasis in response to changing iron diets for a big range of dietary iron contents. The model further reveals that additional homeostasis mechanisms must be taken into account at very high dietary iron levels, including the saturation of intestinal uptake of nutritional iron and the uptake of circulating, non-transferrin-bound iron, into liver. Taken together, our model quantitatively describes systemic iron metabolism and generated experimentally testable predictions for additional ferroportin-independent homeostasis mechanisms. The importance of iron in many physiological processes relies on its ability to participate in reduction-oxidation reactions. This property also leads to potential toxicity if concentrations of free iron are not properly managed by cells and tissues. Multicellular organisms therefore evolved intricate regulatory mechanisms to control systemic iron levels. A central regulatory mechanism is the binding of the hormone hepcidin to the iron exporter ferroportin, which controls the major fluxes of iron into blood plasma. Here, we present a mathematical model that is fitted and validated against experimental data to simulate the iron content in different organs following dietary changes and/or inflammatory states, or genetic perturbation of the hepcidin/ferroportin regulatory system. We find that hepcidin mediated ferroportin control is essential, but not sufficient to quantitatively explain several of our experimental findings. Thus, further regulatory mechanisms had to be included in the model to reproduce reduced serum iron levels in response to inflammation, the preferential accumulation of iron in the liver in the case of iron overload, or the maintenance of physiological serum iron concentrations if dietary iron levels are very high. We conclude that hepcidin-independent mechanisms play an important role in maintaining systemic iron homeostasis.
Collapse
Affiliation(s)
| | - Christoph Metzendorf
- Pediatric Oncology, Hematology & Immunology, University Hospital Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit, Heidelberg University, Heidelberg, Germany
| | - Richard Sparla
- Molecular Medicine Partnership Unit, Heidelberg University, Heidelberg, Germany
| | - Maximilian Hahnel
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Johannes Bode
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Martina U Muckenthaler
- Pediatric Oncology, Hematology & Immunology, University Hospital Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit, Heidelberg University, Heidelberg, Germany
| | | |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Anemia is prevalent in patients with infections and other inflammatory conditions. Induction of the iron regulatory hormone hepcidin has been implicated in the pathogenesis of anemia of inflammation. This review outlines recent discoveries in understanding how hepcidin and its receptor ferroportin are regulated, how they contribute to anemia of inflammation, and how this knowledge may help guide new diagnostic and therapeutic strategies for this disease. RECENT FINDINGS IL-6 is a primary driver for hepcidin induction in many models of anemia of inflammation, but the SMAD1/5/8 pathway also contributes, likely via Activin B and SMAD-STAT3 interactions at the hepcidin promoter. Hepcidin has an important functional role in many, but not all forms of anemia of inflammation, although hepcidin-independent mechanisms also contribute. In certain populations, hepcidin assays may help target therapy with iron or erythropoiesis-stimulating agents to patients who may benefit most. New therapies targeting the hepcidin-ferroportin axis have shown efficacy in preclinical and early clinical studies. SUMMARY Recent studies confirm an important role for the hepcidin-ferroportin axis in the development of anemia of inflammation, but also highlight the diverse and complex pathogenesis of this disorder depending on the underlying disease. Hepcidin-based diagnostic and therapeutic strategies offer promise to improve anemia treatment, but more work is needed in this area.
Collapse
|
13
|
Combined treatment of 3-hydroxypyridine-4-one derivatives and green tea extract to induce hepcidin expression in iron-overloaded β-thalassemic mice. Asian Pac J Trop Biomed 2015. [DOI: 10.1016/j.apjtb.2015.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
14
|
Miseta A, Nagy J, Nagy T, Poór VS, Fekete Z, Sipos K. Hepcidin and its potential clinical utility. Cell Biol Int 2015; 39:1191-202. [PMID: 26109250 DOI: 10.1002/cbin.10505] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/12/2015] [Indexed: 02/06/2023]
Abstract
A number of pathophysiological conditions are related to iron metabolism disturbances. Some of them are well known, others are newly discovered or special. Hepcidin is a newly identified iron metabolism regulating hormone, which could be a promising biomarker for many disorders. In this review, we provide background information about mammalian iron metabolism, cellular iron trafficking, and the regulation of expression of hepcidin. Beside these molecular biological processes, we summarize the methods that have been used to determine blood and urine hepcidin levels and present those pathological conditions (cancer, inflammation, neurological disorders) when hepcidin measurement may have clinical relevance.
Collapse
Affiliation(s)
- Attila Miseta
- Department of Laboratory Medicine, Faculty of Medical Sciences, University of Pécs, 7624 Ifjusag Street 13. Pecs, Hungary
| | - Judit Nagy
- Department of Anaesthesiology and Intensive Care, Faculty of Medical Sciences, University of Pécs, 7624 Ifjusag Street 13. Pecs, Hungary
| | - Tamas Nagy
- Department of Laboratory Medicine, Faculty of Medical Sciences, University of Pécs, 7624 Ifjusag Street 13. Pecs, Hungary
| | - Viktor Soma Poór
- Department of Forensic Medicine, Faculty of Medical Sciences, University of Pécs, 7624 Szigeti Street 12. Pecs, Hungary
| | - Zsuzsanna Fekete
- Department of Medical Biology, Faculty of Medical Sciences, University of Pécs, 7624 Szigeti Street 12. Pecs, Hungary
| | - Katalin Sipos
- Department of Pharmaceutical Biology, Faculty of Medical Sciences, University of Pécs, 7624 Rokus Street 2. Pecs, Hungary
| |
Collapse
|
15
|
Drasdo D, Bode J, Dahmen U, Dirsch O, Dooley S, Gebhardt R, Ghallab A, Godoy P, Häussinger D, Hammad S, Hoehme S, Holzhütter HG, Klingmüller U, Kuepfer L, Timmer J, Zerial M, Hengstler JG. The virtual liver: state of the art and future perspectives. Arch Toxicol 2015; 88:2071-5. [PMID: 25331938 DOI: 10.1007/s00204-014-1384-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Dirk Drasdo
- Institut National de Recherche en Informatique et en Automatique (INRIA), Domaine de Voluceau - Rocquencourt, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Dittrich A, Hessenkemper W, Schaper F. Systems biology of IL-6, IL-12 family cytokines. Cytokine Growth Factor Rev 2015; 26:595-602. [PMID: 26187858 DOI: 10.1016/j.cytogfr.2015.07.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/01/2015] [Indexed: 10/23/2022]
Abstract
Interleukin-6-type cytokines play important roles in the communication between cells of multicellular organisms. They are involved in the regulation of complex cellular processes such as proliferation and differentiation and act as key player during inflammation and immune response. A major challenge is to understand how these complex non-linear processes are connected and regulated. Systems biology approaches are used to tackle this challenge in an iterative process of quantitative experimental and mathematical analyses. Here we review quantitative experimental studies and systems biology approaches dealing with the function of Interleukin-6-type cytokines in physiological and pathophysiological conditions. These approaches cover the analyses of signal transduction on a cellular level up to pharmacokinetic and pharmacodynamic studies on a whole organism level.
Collapse
Affiliation(s)
- Anna Dittrich
- Institute of Biology, Otto-von-Guericke-University, Universitätsplatz 2, 39106 Magdeburg, Germany.
| | - Wiebke Hessenkemper
- Institute of Biology, Otto-von-Guericke-University, Universitätsplatz 2, 39106 Magdeburg, Germany.
| | - Fred Schaper
- Institute of Biology, Otto-von-Guericke-University, Universitätsplatz 2, 39106 Magdeburg, Germany.
| |
Collapse
|
17
|
Schulthess P, Löffler A, Vetter S, Kreft L, Schwarz M, Braeuning A, Blüthgen N. Signal integration by the CYP1A1 promoter--a quantitative study. Nucleic Acids Res 2015; 43:5318-30. [PMID: 25934798 PMCID: PMC4477655 DOI: 10.1093/nar/gkv423] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 04/17/2015] [Indexed: 01/23/2023] Open
Abstract
Genes involved in detoxification of foreign compounds exhibit complex spatiotemporal expression patterns in liver. Cytochrome P450 1A1 (CYP1A1), for example, is restricted to the pericentral region of liver lobules in response to the interplay between aryl hydrocarbon receptor (AhR) and Wnt/β-catenin signaling pathways. However, the mechanisms by which the two pathways orchestrate gene expression are still poorly understood. With the help of 29 mutant constructs of the human CYP1A1 promoter and a mathematical model that combines Wnt/β-catenin and AhR signaling with the statistical mechanics of the promoter, we systematically quantified the regulatory influence of different transcription factor binding sites on gene induction within the promoter. The model unveils how different binding sites cooperate and how they establish the promoter logic; it quantitatively predicts two-dimensional stimulus-response curves. Furthermore, it shows that crosstalk between Wnt/β-catenin and AhR signaling is crucial to understand the complex zonated expression patterns found in liver lobules. This study exemplifies how statistical mechanical modeling together with combinatorial reporter assays has the capacity to disentangle the promoter logic that establishes physiological gene expression patterns.
Collapse
Affiliation(s)
- Pascal Schulthess
- Institute for Pathology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany Integrative Research Institute for the Life Sciences and Institute for Theoretical Biology, Humboldt University of Berlin, Philippstr. 13, 10115 Berlin, Germany
| | - Alexandra Löffler
- Institute for Experimental and Clinical Pharmacology and Toxicology, Department of Toxicology, University of Tübingen, Wilhelmstraße 56, 72074 Tübingen, Germany
| | - Silvia Vetter
- Institute for Experimental and Clinical Pharmacology and Toxicology, Department of Toxicology, University of Tübingen, Wilhelmstraße 56, 72074 Tübingen, Germany
| | - Luisa Kreft
- Institute for Experimental and Clinical Pharmacology and Toxicology, Department of Toxicology, University of Tübingen, Wilhelmstraße 56, 72074 Tübingen, Germany
| | - Michael Schwarz
- Institute for Experimental and Clinical Pharmacology and Toxicology, Department of Toxicology, University of Tübingen, Wilhelmstraße 56, 72074 Tübingen, Germany
| | - Albert Braeuning
- Department of Food Safety, Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany
| | - Nils Blüthgen
- Institute for Pathology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany Integrative Research Institute for the Life Sciences and Institute for Theoretical Biology, Humboldt University of Berlin, Philippstr. 13, 10115 Berlin, Germany
| |
Collapse
|
18
|
D'Alessandro LA, Hoehme S, Henney A, Drasdo D, Klingmüller U. Unraveling liver complexity from molecular to organ level: challenges and perspectives. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 117:78-86. [PMID: 25433231 DOI: 10.1016/j.pbiomolbio.2014.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/28/2014] [Accepted: 11/19/2014] [Indexed: 12/13/2022]
Abstract
Biological responses are determined by information processing at multiple and highly interconnected scales. Within a tissue the individual cells respond to extracellular stimuli by regulating intracellular signaling pathways that in turn determine cell fate decisions and influence the behavior of neighboring cells. As a consequence the cellular responses critically impact tissue composition and architecture. Understanding the regulation of these mechanisms at different scales is key to unravel the emergent properties of biological systems. In this perspective, a multidisciplinary approach combining experimental data with mathematical modeling is introduced. We report the approach applied within the Virtual Liver Network to analyze processes that regulate liver functions from single cell responses to the organ level using a number of examples. By facilitating interdisciplinary collaborations, the Virtual Liver Network studies liver regeneration and inflammatory processes as well as liver metabolic functions at multiple scales, and thus provides a suitable example to identify challenges and point out potential future application of multi-scale systems biology.
Collapse
Affiliation(s)
- L A D'Alessandro
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, 69120 Heidelberg, Germany
| | - S Hoehme
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, Germany
| | - A Henney
- Obsidian Biomedical Consulting Ltd., Macclesfield, UK; The German Virtual Liver Network, University of Heidelberg, 69120 Heidelberg, Germany
| | - D Drasdo
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, Germany; Institut National de Recherche en Informatique et en Automatique (INRIA), Domaine de Voluceau, 78150 Rocquencourt, France; University Pierre and Marie Curie and CNRS UMR 7598, LJLL, F-75005 Paris, France; CNRS, 7598 Paris, France
| | - U Klingmüller
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, 69120 Heidelberg, Germany.
| |
Collapse
|
19
|
Novel loci affecting iron homeostasis and their effects in individuals at risk for hemochromatosis. Nat Commun 2014; 5:4926. [PMID: 25352340 PMCID: PMC4215164 DOI: 10.1038/ncomms5926] [Citation(s) in RCA: 205] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 08/06/2014] [Indexed: 12/13/2022] Open
Abstract
Variation in body iron is associated with or causes diseases, including anaemia and iron overload. Here, we analyse genetic association data on biochemical markers of iron status from 11 European-population studies, with replication in eight additional cohorts (total up to 48,972 subjects). We find 11 genome-wide-significant (P<5 × 10(-8)) loci, some including known iron-related genes (HFE, SLC40A1, TF, TFR2, TFRC, TMPRSS6) and others novel (ABO, ARNTL, FADS2, NAT2, TEX14). SNPs at ARNTL, TF, and TFR2 affect iron markers in HFE C282Y homozygotes at risk for hemochromatosis. There is substantial overlap between our iron loci and loci affecting erythrocyte and lipid phenotypes. These results will facilitate investigation of the roles of iron in disease.
Collapse
|
20
|
A systems model of phosphorylation for inflammatory signaling events. PLoS One 2014; 9:e110913. [PMID: 25333362 PMCID: PMC4205014 DOI: 10.1371/journal.pone.0110913] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 09/19/2014] [Indexed: 12/24/2022] Open
Abstract
Phosphorylation is a fundamental biochemical reaction that modulates protein activity in cells. While a single phosphorylation event is relatively easy to understand, multisite phosphorylation requires systems approaches for deeper elucidation of the underlying molecular mechanisms. In this paper we develop a mechanistic model for single- and multi-site phosphorylation. The proposed model is compared with previously reported studies. We compare the predictions of our model with experiments published in the literature in the context of inflammatory signaling events in order to provide a mechanistic description of the multisite phosphorylation-mediated regulation of Signal Transducer and Activator of Transcription 3 (STAT3) and Interferon Regulatory Factor 5 (IRF-5) proteins. The presented model makes crucial predictions for transcription factor phosphorylation events in the immune system. The model proposes potential mechanisms for T cell phenotype switching and production of cytokines. This study also provides a generic framework for the better understanding of a large number of multisite phosphorylation-regulated biochemical circuits.
Collapse
|