1
|
Gujral P, Orozco-Alonso E, Saliba J, Yan X, Blank V. The NFE2L2 (NRF2) transcription factor controls genes involved in the oxidative stress response and inflammation in myometrial cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119985. [PMID: 40360022 DOI: 10.1016/j.bbamcr.2025.119985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025]
Abstract
The myometrium is the smooth muscle layer of the uterus, which mediates uterine contractions during labor. We treated PHM1-31 myometrial cells with the proinflammatory cytokine interleukin-1 beta (IL1B) and measured a significant increase in reactive oxygen species (ROS). We found that IL1B induces NFE2L2 (NRF2) transcription factor levels. We further showed that siRNA mediated knockdown of NFE2L2 results in a significant increase in ROS. Downregulation of NFE2L2 leads to a decrease of heme oxygenase-1 (HMOX1) and aldo-keto reductase family 1 member B (AKR1B) at the transcript and protein level both in the absence and presence of IL1B. NFE2L2 knockdown also results in reduced ferritin heavy chain 1 (FTH1) mRNA expression, but only upon IL1B exposure, while FTH1 protein is downregulated both under basal and IL1B treatment conditions. We confirmed that NFE2L2 directly binds to the regulatory regions of these targets. Previous reports have linked HMOX1 and FTH1 to the oxidative stress response, and AKR1B1 to prostaglandin synthesis. Our data demonstrate that NFE2L2 functions as a key regulator of inflammatory and oxidative stress signaling through the regulation of HMOX1, FTH1, and AKR1B1 expression in myometrial cells. While HMOX1 and FTH1 have established roles in oxidative stress responses, our findings identify AKR1B1 as a novel target of NFE2L2 in myometrial cells, suggesting a role for the transcription factor in prostaglandin metabolism. Thus, NFE2L2 links inflammation and the oxidative stress response to critical pathways that control myometrial cell function and parturition, highlighting their potential as therapeutic targets for treating infection-induced preterm labor.
Collapse
Affiliation(s)
- Palak Gujral
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada; Department of Medicine, McGill University, Montreal H4A 3J1, Canada
| | - Eduardo Orozco-Alonso
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada; Department of Medicine, McGill University, Montreal H4A 3J1, Canada
| | - James Saliba
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Xingyue Yan
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Volker Blank
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada; Department of Medicine, McGill University, Montreal H4A 3J1, Canada; Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
2
|
Szymulewska-Konopko K, Reszeć-Giełażyn J, Małeczek M. Ferritin as an Effective Prognostic Factor and Potential Cancer Biomarker. Curr Issues Mol Biol 2025; 47:60. [PMID: 39852175 PMCID: PMC11763953 DOI: 10.3390/cimb47010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Abstract
Ferritin is found in all cells of the body, serving as a reservoir of iron and protecting against damage to the molecules that make up cellular structures. It has emerged as a biomarker not only for iron-related disorders but also for inflammatory diseases and conditions in which inflammation plays a key role, including cancer, neurodegeneration, and infection. Oxidative stress, which can cause cellular damage, is induced by reactive oxygen species generated during the Fenton reaction, activating signaling pathways associated with tumor growth and proliferation. This review primarily emphasizes basic studies on the identification and function of ferritin, its essential role in iron metabolism, its involvement in inflammatory diseases, and its potential as an important prognostic factor and biomarker for cancer detection.
Collapse
Affiliation(s)
| | - Joanna Reszeć-Giełażyn
- Department of Medical Pathomorphology, Medical University of Bialystok, 15-089 Białystok, Poland; (K.S.-K.); (M.M.)
| | | |
Collapse
|
3
|
Mehta P, Samanta RJ, Wick K, Coll RC, Mawhinney T, McAleavey PG, Boyle AJ, Conlon J, Shankar-Hari M, Rogers A, Calfee CS, Matthay MA, Summers C, Chambers RC, McAuley DF, O'Kane CM. Elevated ferritin, mediated by IL-18 is associated with systemic inflammation and mortality in acute respiratory distress syndrome (ARDS). Thorax 2024; 79:227-235. [PMID: 38148147 DOI: 10.1136/thorax-2023-220292] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 11/03/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND Inflammatory subphenotypes have been identified in acute respiratory distress syndrome (ARDS). Hyperferritinaemia in sepsis is associated with hyperinflammation, worse clinical outcomes, and may predict benefit with immunomodulation. Our aim was to determine if raised ferritin identified a subphenotype in patients with ARDS. METHODS Baseline plasma ferritin concentrations were measured in patients with ARDS from two randomised controlled trials of simvastatin (Hydroxymethylglutaryl-CoA Reductase Inhibition with Simvastatin in Acute Lung Injury to Reduce Pulmonary Dysfunction-2 (HARP-2); discovery cohort, UK) and neuromuscular blockade (ROSE; validation cohort, USA). Results were analysed using a logistic regression model with restricted cubic splines, to determine the ferritin threshold associated with 28-day mortality. RESULTS Ferritin was measured in 511 patients from HARP-2 (95% of patients enrolled) and 847 patients (84% of patients enrolled) from ROSE. Ferritin was consistently associated with 28-day mortality in both studies and following a meta-analysis, a log-fold increase in ferritin was associated with an OR 1.71 (95% CI 1.01 to 2.90) for 28-day mortality. Patients with ferritin >1380 ng/mL (HARP-2 28%, ROSE 24%) had a significantly higher 28-day mortality and fewer ventilator-free days in both studies. Mediation analysis, including confounders (acute physiology and chronic health evaluation-II score and ARDS aetiology) demonstrated a statistically significant contribution of interleukin (IL)-18 as an intermediate pathway between ferritin and mortality. CONCLUSIONS Ferritin is a clinically useful biomarker in ARDS and is associated with worse patient outcomes. These results provide support for prospective interventional trials of immunomodulatory agents targeting IL-18 in this hyperferritinaemic subgroup of patients with ARDS.
Collapse
Affiliation(s)
- Puja Mehta
- Centre for inflammation and Tissue Repair (CITR), University College London Division of Medicine, London, UK
| | - Romit J Samanta
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Katherine Wick
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Rebecca C Coll
- Wellcome Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Thea Mawhinney
- Wellcome Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Patrick G McAleavey
- Wellcome Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Andrew J Boyle
- Wellcome Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
- Royal Victoria Hospital, Belfast Health and Social Care Trust, Belfast, UK
| | - John Conlon
- Wellcome Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Manu Shankar-Hari
- The Queen's Medical Research Institute, Edinburgh BioQuarter, Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
- Intensive Care Medicine, Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, UK
| | - Angela Rogers
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Carolyn S Calfee
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
- Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, California, USA
| | - Michael A Matthay
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
- Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, California, USA
| | | | | | - Daniel Francis McAuley
- Wellcome Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
- Royal Victoria Hospital, Belfast Health and Social Care Trust, Belfast, UK
| | - Cecilia M O'Kane
- Wellcome Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| |
Collapse
|
4
|
Shesh BP, Connor JR. A novel view of ferritin in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188917. [PMID: 37209958 PMCID: PMC10330744 DOI: 10.1016/j.bbcan.2023.188917] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/13/2023] [Accepted: 05/13/2023] [Indexed: 05/22/2023]
Abstract
Since its discovery more than 85 years ago, ferritin has principally been known as an iron storage protein. However, new roles, beyond iron storage, are being uncovered. Novel processes involving ferritin such as ferritinophagy and ferroptosis and as a cellular iron delivery protein not only expand our thinking on the range of contributions of this protein but present an opportunity to target these pathways in cancers. The key question we focus on within this review is whether ferritin modulation represents a useful approach for treating cancers. We discussed novel functions and processes of this protein in cancers. We are not limiting this review to cell intrinsic modulation of ferritin in cancers, but also focus on its utility in the trojan horse approach in cancer therapeutics. The novel functions of ferritin as discussed herein realize the multiple roles of ferritin in cell biology that can be probed for therapeutic opportunities and further research.
Collapse
Affiliation(s)
| | - James R Connor
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
5
|
Cavallazzi R, Bradley J, Chandler T, Furmanek S, Ramirez JA. Severity of Illness Scores and Biomarkers for Prognosis of Patients with Coronavirus Disease 2019. Semin Respir Crit Care Med 2023; 44:75-90. [PMID: 36646087 DOI: 10.1055/s-0042-1759567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The spectrum of disease severity and the insidiousness of clinical presentation make it difficult to recognize patients with coronavirus disease 2019 (COVID-19) at higher risk of worse outcomes or death when they are seen in the early phases of the disease. There are now well-established risk factors for worse outcomes in patients with COVID-19. These should be factored in when assessing the prognosis of these patients. However, a more precise prognostic assessment in an individual patient may warrant the use of predictive tools. In this manuscript, we conduct a literature review on the severity of illness scores and biomarkers for the prognosis of patients with COVID-19. Several COVID-19-specific scores have been developed since the onset of the pandemic. Some of them are promising and can be integrated into the assessment of these patients. We also found that the well-known pneumonia severity index (PSI) and CURB-65 (confusion, uremia, respiratory rate, BP, age ≥ 65 years) are good predictors of mortality in hospitalized patients with COVID-19. While neither the PSI nor the CURB-65 should be used for the triage of outpatient versus inpatient treatment, they can be integrated by a clinician into the assessment of disease severity and can be used in epidemiological studies to determine the severity of illness in patient populations. Biomarkers also provide valuable prognostic information and, importantly, may depict the main physiological derangements in severe disease. We, however, do not advocate the isolated use of severity of illness scores or biomarkers for decision-making in an individual patient. Instead, we suggest the use of these tools on a case-by-case basis with the goal of enhancing clinician judgment.
Collapse
Affiliation(s)
- Rodrigo Cavallazzi
- Division of Pulmonary, Critical Care Medicine, and Sleep Disorders, University of Louisville, Norton Healthcare, Louisville, Kentucky
| | - James Bradley
- Division of Pulmonary, Critical Care Medicine, and Sleep Disorders, University of Louisville, Norton Healthcare, Louisville, Kentucky
| | - Thomas Chandler
- Norton Infectious Diseases Institute, Norton Healthcare, Louisville, Kentucky
| | - Stephen Furmanek
- Norton Infectious Diseases Institute, Norton Healthcare, Louisville, Kentucky
| | - Julio A Ramirez
- Norton Infectious Diseases Institute, Norton Healthcare, Louisville, Kentucky
| |
Collapse
|
6
|
Ruscitti P, Di Cola I, Di Muzio C, Italiano N, Ursini F, Giacomelli R, Cipriani P. Expanding the spectrum of the hyperferritinemic syndrome, from pathogenic mechanisms to clinical observations, and therapeutic implications. Autoimmun Rev 2022; 21:103114. [PMID: 35595050 DOI: 10.1016/j.autrev.2022.103114] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/15/2022] [Indexed: 01/19/2023]
Abstract
From the introduction of hyperferritinemic syndrome concept, a growing body of evidence has suggested the role of ferritin as a pathogenic mediator and a relevant clinical feature in the management of patients with inflammatory diseases. From a pathogenic point of view, ferritin may directly stimulate the aberrant immune response by triggering the production of pro-inflammatory mediators in inducing a vicious pathogenic loop and contributing to the occurrence of cytokine storm syndrome. The latter has been recently defined as a clinical picture characterised by elevated circulating cytokine levels, acute systemic inflammatory symptoms, and secondary organ dysfunction beyond that which could be attributed to a normal response to a pathogen It is noteworthy that the occurrence of hyperferritinemia may be correlated with the development of the cytokine storm syndrome in the context of an inflammatory disease. In addition to adult onset Still's disease, macrophage activation syndrome, catastrophic anti-phospholipids syndrome, and septic shock, recent evidence has suggested this association between ferritin and life-threatening evolution in patients with systemic lupus erythematosus, with anti-MDA5 antibodies in the context of poly-dermatomyositis, with severe COVID-19, and with multisystem inflammatory syndrome. The possible underlying common inflammatory mechanisms, associated with hyperferritinemia, may led to the similar clinical picture observed in these patients. Furthermore, similar therapeutic strategies could be suggested inhibiting pro-inflammatory cytokines and improving long-term outcomes in these disorders. Thus, it could be possible to expand the spectrum of the hyperferritinemic syndrome to those diseases burdened by a dreadful clinical picture correlated with hyperferritinemia and the occurrence of the cytokine storm syndrome. In addition, the assessment of ferritin may provide useful information to the physicians in clinical practice to manage these patients. Therefore, ferritin may be considered a relevant clinical feature to be used as biomarker in dissecting the unmet needs in the management of these disorders. Novel evidence may thus support an expansion of the spectrum of the hyperferritinemic syndrome to these diseases burdened by a life-threatening clinical picture correlated with hyperferritinemia and the occurrence of the cytokine storm syndrome.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Claudia Di Muzio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Noemi Italiano
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesco Ursini
- Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome Campus Biomedico, Rome, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
7
|
Wen Y, Cheng M, Qin L, Xu W. TNFα-induced abnormal activation of TNFR/NF-κB/FTH1 in endometrium is involved in the pathogenesis of early spontaneous abortion. J Cell Mol Med 2022; 26:2947-2958. [PMID: 35441429 PMCID: PMC9097845 DOI: 10.1111/jcmm.17308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/13/2022] [Accepted: 03/21/2022] [Indexed: 11/30/2022] Open
Abstract
Early spontaneous abortion (ESA) is one of the most common complications during pregnancy and the inflammation condition in uterine environment such as long‐term exposure to high TNFα plays an essential role in the aetiology. Ferritin heavy chain (FTH1) is considered to be closely associated with inflammation and very important in normal pregnancy, yet the underlying mechanism of how TNFα induced abortion and its relationship with FTH1 remain elusive. In this study, we found that TNFα and FTH1 were positively expressed in decidual stromal cells and increased significantly in the ESA group compared with the normal pregnancy group (NP group). Besides, TNFα expression was positively correlated with FTH1 expression. Furthermore, in vitro cell model demonstrated that high TNFα could induce the abnormal signals of TNFR/NF‐κB/FTH1 and activate apoptosis both in human endometrium stromal cells (hESCs) and in local decidual tissues. Taken together, the present findings suggest that the excessive apoptosis in response to TNFα‐induced upregulation of FTH1 may be responsible for the occurrence of ESA, and thus provide a possible therapeutic target for the treatment of ESA.
Collapse
Affiliation(s)
- Yuting Wen
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Meng Cheng
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lang Qin
- The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Wenming Xu
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
SARS-CoV-2 Mediated Hyperferritinemia and Cardiac Arrest: Preliminary Insights. Drug Discov Today 2021; 26:1265-1274. [PMID: 33493677 PMCID: PMC7826001 DOI: 10.1016/j.drudis.2021.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/14/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023]
|
9
|
Iron in immune cell function and host defense. Semin Cell Dev Biol 2020; 115:27-36. [PMID: 33386235 DOI: 10.1016/j.semcdb.2020.12.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022]
Abstract
The control over iron availability is crucial under homeostatic conditions and even more in the case of an infection. This results from diverse properties of iron: first, iron is an important trace element for the host as well as for the pathogen for various cellular and metabolic processes, second, free iron catalyzes Fenton reaction and is therefore producing reactive oxygen species as a part of the host defense machinery, third, iron exhibits important effects on immune cell function and differentiation and fourth almost every immune activation in turn impacts on iron metabolism and spatio-temporal iron distribution. The central importance of iron in the host and microbe interplay and thus for the course of infections led to diverse strategies to restrict iron for invading pathogens. In this review, we focus on how iron restriction to the pathogen is a powerful innate immune defense mechanism of the host called "nutritional immunity". Important proteins in the iron-host-pathogen interplay will be discussed as well as the influence of iron on the efficacy of innate and adaptive immunity. Recently described processes like ferritinophagy and ferroptosis are further covered in respect to their impact on inflammation and infection control and how they impact on our understanding of the interaction of host and pathogen.
Collapse
|
10
|
Hypoferremia is Associated With Increased Hospitalization and Oxygen Demand in COVID-19 Patients. Hemasphere 2020; 4:e492. [PMID: 33205000 PMCID: PMC7665253 DOI: 10.1097/hs9.0000000000000492] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/04/2020] [Indexed: 01/18/2023] Open
Abstract
Iron metabolism might play a crucial role in cytokine release syndrome in COVID-19 patients. Therefore, we assessed iron metabolism markers in COVID-19 patients for their ability to predict disease severity. COVID-19 patients referred to the Heidelberg University Hospital were retrospectively analyzed. Patients were divided into outpatients (cohort A, n = 204), inpatients (cohort B, n = 81), and outpatients later admitted to hospital because of health deterioration (cohort C, n = 23). Iron metabolism parameters were severely altered in patients of cohort B and C compared to cohort A. In multivariate regression analysis including age, gender, CRP and iron-related parameters only serum iron and ferritin were significantly associated with hospitalization. ROC analysis revealed an AUC for serum iron of 0.894 and an iron concentration <6 μmol/l as the best cutoff-point predicting hospitalization with a sensitivity of 94.7% and a specificity of 67.9%. When stratifying inpatients in a low- and high oxygen demand group serum iron levels differed significantly between these two groups and showed a high negative correlation with the inflammatory parameters IL-6, procalcitonin, and CRP. Unexpectedly, serum iron levels poorly correlate with hepcidin. We conclude that measurement of serum iron can help predicting the severity of COVID-19. The differences in serum iron availability observed between the low and high oxygen demand group suggest that disturbed iron metabolism likely plays a causal role in the pathophysiology leading to lung injury.
Collapse
|
11
|
Ruscitti P, Di Benedetto P, Berardicurti O, Panzera N, Grazia N, Lizzi AR, Cipriani P, Shoenfeld Y, Giacomelli R. Pro-inflammatory properties of H-ferritin on human macrophages, ex vivo and in vitro observations. Sci Rep 2020; 10:12232. [PMID: 32699419 PMCID: PMC7376151 DOI: 10.1038/s41598-020-69031-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
Ferritin is an iron-binding molecule, which comprises 24 subunits, heavy (FeH) and light (FeL) subunits, suggested to have a pathogenic role by the 'hyperferritinemic syndrome'. In this work, we tested (1) FeH and FeL in bone marrow (BM) and sera in patients with macrophage activation syndrome (MAS); (2) pro-inflammatory effects of ferritin, FeL, and FeH on macrophages; (3) ability of FeH-stimulated macrophages to stimulate the proliferation of peripheral blood mononuclear cells (PBMCs); (4) production of mature IL-1β and IL-12p70 in extracellular compartments of FeH-stimulated macrophages. Immunofluorescence analysis and liquid chromatography mass spectrometry (LC-MS/MS) based proteomics were performed to identify FeL and FeH in BM and sera, respectively, in the same patients. Macrophages were stimulated with ferritin, FeH, and FeL to assess pro-inflammatory effects by RT-PCR and western blot. The proliferation of co-cultured PBMCs with FeH-stimulated macrophages was tested. Immunofluorescence showed an increased FeH expression in BMs, whereas LC-MS/MS identified that FeL was mainly represented in sera. FeH induced a significant increase of gene expressions of IL-1β, IL-6, IL-12, and TNF-α, more marked with FeH, which also stimulated NLRP3. FeH-stimulated macrophages enhanced the proliferation of PBMCs. The ELISA assays showed that mature form of IL-1β and IL-12p70 were increased, in extracellular compartments of FeH-stimulated macrophages. Our results showed FeH in BM biopsies of MAS patients, whereas, LC-MS/MS identified FeL in the sera. FeH showed pro-inflammatory effects on macrophages, stimulated NLRP3, and increased PBMCs proliferation.
Collapse
Affiliation(s)
- Piero Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy.
| | - Paola Di Benedetto
- Clinical Pathology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Onorina Berardicurti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Noemi Panzera
- Clinical Pathology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nicolò Grazia
- Clinical Pathology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Anna Rita Lizzi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paola Cipriani
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel HaShomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Saint Petersburg, Russia
| | - Roberto Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, Via dell'Ospedale, 67100, L'Aquila, Italy
| |
Collapse
|
12
|
Giemza-Stokłosa J, Islam MA, Kotyla PJ. Hyperferritinaemia: An Iron Sword of Autoimmunity. Curr Pharm Des 2020; 25:2909-2918. [PMID: 31686632 DOI: 10.2174/1381612825666190709202804] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 06/30/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Ferritin is a molecule that plays many roles being the storage for iron, signalling molecule, and modulator of the immune response. METHODS Different electronic databases were searched in a non-systematic way to find out the literature of interest. RESULTS The level of ferritin rises in many inflammatory conditions including autoimmune disorders. However, in four inflammatory diseases (i.e., adult-onset Still's diseases, macrophage activation syndrome, catastrophic antiphospholipid syndrome, and sepsis), high levels of ferritin are observed suggesting it as a remarkable biomarker and pathological involvement in these diseases. Acting as an acute phase reactant, ferritin is also involved in the cytokine-associated modulator of the immune response as well as a regulator of cytokine synthesis and release which are responsible for the inflammatory storm. CONCLUSION This review article presents updated information on the role of ferritin in inflammatory and autoimmune diseases with an emphasis on hyperferritinaemic syndrome.
Collapse
Affiliation(s)
| | - Md Asiful Islam
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Przemysław J Kotyla
- Department of Internal Medicine, Rheumatology and Clinical Immunology, Faculty in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| |
Collapse
|
13
|
Wang MY, Jia JC, Yang CD, Hu QY. Pathogenesis, disease course, and prognosis of adult-onset Still's disease: an update and review. Chin Med J (Engl) 2019; 132:2856-2864. [PMID: 31856058 PMCID: PMC6940076 DOI: 10.1097/cm9.0000000000000538] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Adult-onset Still's disease (AOSD) is a rare but clinically well-known polygenic systemic autoinflammatory disease. In this review, we aim to present frontiers in the pathogenesis, clinical features, diagnosis, biomarkers, disease course, prognosis, and treatment in AOSD. DATA SOURCES We retrieved information from the PubMed database up to July 2019, using various search terms and relevant words, including AOSD and Still's disease. STUDY SELECTION We included data from peer-reviewed journals. Both basic and clinical studies were selected. RESULTS Pathogenesis of AOSD involves genetic background, infectious triggers, and immunopathogenesis, mainly the activation of macrophages and neutrophils followed by a cytokine storm. Diagnosis and prognosis evaluation of AOSD is still challenging; therefore, there is an urgent need to identify better biomarkers. Biologic agents, including interleukin (IL)-1β, IL-6, and tumor necrosis factor-α antagonists in the treatment of AOSD, have good prospect. CONCLUSION This review highlights the advances in pathogenesis, potential biomarkers, disease course, and treatment in AOSD.
Collapse
Affiliation(s)
- Meng-Yan Wang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | | |
Collapse
|
14
|
Daher R, Lefebvre T, Puy H, Karim Z. Extrahepatic hepcidin production: The intriguing outcomes of recent years. World J Clin Cases 2019; 7:1926-1936. [PMID: 31423425 PMCID: PMC6695539 DOI: 10.12998/wjcc.v7.i15.1926] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/18/2019] [Accepted: 06/27/2019] [Indexed: 02/05/2023] Open
Abstract
Hepcidin is the hyposideremic hormone regulating iron metabolism. It is a defensin-like disulfide-bonded peptide with antimicrobial activity. The main site of hepcidin production is the liver where its synthesis is modulated by iron, inflammation and erythropoietic signaling. However, hepcidin locally produced in several peripheral organs seems to be an important actor for the maintenance of iron homeostasis in these organs. This review highlights the presence of peripheral hepcidin and its potential functions. Understanding the role of extrahepatic hepcidin could be of great physiological and therapeutic importance for several specific pathologies.
Collapse
Affiliation(s)
- Raêd Daher
- Université Paris Diderot, Bichat site, Paris 75018, France
- Inflammation Research Center (CRI), INSERM U1149/ERL CNRS 8252, Paris 75018, France
- Laboratory of Excellence, GR-Ex, Paris 75018, France
| | - Thibaud Lefebvre
- Université Paris Diderot, Bichat site, Paris 75018, France
- Inflammation Research Center (CRI), INSERM U1149/ERL CNRS 8252, Paris 75018, France
- Laboratory of Excellence, GR-Ex, Paris 75018, France
| | - Hervé Puy
- Université Paris Diderot, Bichat site, Paris 75018, France
- Inflammation Research Center (CRI), INSERM U1149/ERL CNRS 8252, Paris 75018, France
- Laboratory of Excellence, GR-Ex, Paris 75018, France
| | - Zoubida Karim
- Université Paris Diderot, Bichat site, Paris 75018, France
- Inflammation Research Center (CRI), INSERM U1149/ERL CNRS 8252, Paris 75018, France
- Laboratory of Excellence, GR-Ex, Paris 75018, France
| |
Collapse
|
15
|
Up-regulated ferritin in periodontitis promotes inflammatory cytokine expression in human periodontal ligament cells through transferrin receptor via ERK/P38 MAPK pathways. Clin Sci (Lond) 2019; 133:135-148. [PMID: 30552136 DOI: 10.1042/cs20180679] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/29/2018] [Accepted: 12/14/2018] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Ferritin, an iron-binding protein, is ubiquitous and highly conserved; it plays a crucial role in inflammation, which is the main symptom of periodontitis. Full-length cDNA library analyses have demonstrated abundant expression of ferritin in human periodontal ligament. The aims of the present study were to explore how ferritin is regulated by local inflammation, and to investigate its functions and mechanisms of action in the process of periodontitis. METHODS Human gingival tissues were collected from periodontitis patients and healthy individuals. Experimental periodontitis was induced by ligature of second molars in mice. The expression of ferritin light polypeptide (FTL) and ferritin heavy polypeptide (FTH) were assessed by immunohistochemistry. Meanwhile, after stimulating human periodontal ligament cells (HPDLCs) with P. gingivalis-lipopolysaccharide (LPS), interleukin (IL)-6, and tumor necrosis factor-α (TNF-α), the expression of FTH and FTL were measured. Then, IL-6 and IL-8 were measured after incubation with different concentrations of apoferritin (iron-free ferritin) and several intracellular signaling pathway inhibitors, or after knockdown of the transferrin receptor. RESULTS Both FTH and FTL were substantially higher in inflamed periodontal tissues than in healthy tissues. The location of the elevated expression correlated well with the extent of inflammatory infiltration. Moreover, expression of FTH and FTL were enhanced after stimulation with P. gingivalis-LPS, IL-6, TNF-α. Apoferritin induced the production of IL-6 and IL-8 in a dose-dependent manner partly through binding to the transferrin receptor and activating ERK/P38 signaling pathways in HPDLCs. CONCLUSIONS Ferritin is up-regulated by inflammation and exhibits cytokine-like activity in HPDLCs inducing a signaling cascade that promotes expression of pro-inflammatory cytokines associated with periodontitis.
Collapse
|
16
|
Ruscitti P, Giacomelli R. Pathogenesis of adult onset still’s disease: current understanding and new insights. Expert Rev Clin Immunol 2018; 14:965-976. [DOI: 10.1080/1744666x.2018.1533403] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Piero Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Roberto Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
17
|
Giacomelli R, Ruscitti P, Shoenfeld Y. A comprehensive review on adult onset Still's disease. J Autoimmun 2018; 93:24-36. [PMID: 30077425 DOI: 10.1016/j.jaut.2018.07.018] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 07/26/2018] [Accepted: 07/26/2018] [Indexed: 02/06/2023]
Abstract
Adult-onset Still's disease (AOSD) is a systemic inflammatory disorder of unknown etiology usually affecting young adults; spiking fever, arthritis and evanescent rash are commonly observed during the disease. Other frequently observed clinical features include sore throat, hepatomegaly, splenomegaly, lymphadenopathy and serositis. Furthermore, AOSD patients may experience different life-threating complications. Macrophage activation syndrome (MAS) has been reported up to 15% of AOSD patients and it is considered to be the most severe complication of the disease being characterised by high mortality rate. During AOSD, laboratory tests reflect the systemic inflammatory process showing high levels of erythrocyte sedimentation rate and C-reactive protein. In addition, the ferritin levels are typically higher than those observed in other autoimmune, inflammatory, infectious, or neoplastic diseases. Analysing AOSD disease course, 3 different clinical patterns of AOSD have been identified: i. monocyclic pattern, characterised by a systemic single episode; ii. polycyclic pattern, characterised by multiple, ≤ 1 year lasting, flares, alternating with remissions; iii. chronic pattern, related to a persistently active disease with associated polyarthritis. At present, AOSD therapeutic strategy is aimed at targeting pro-inflammatory signs and symptoms, preventing organ damage and life-threating complications and minimising adverse effects of treatment. However, the treatment of AOSD remains largely empirical, lacking controlled clinical trials. High dosages of corticosteroids are usually the first line therapy when the systemic symptoms predominate. Despite this treatment, a large percentage of patients experiences several flares with an evolution toward the chronic disease course and up to 16% of patients die during the follow up, due to AOSD-related complications. On these bases, in the last years, biological agents have been successfully used in refractory cases. Finally, multiple recent lines of evidence have suggested new insights in AOSD pathogenesis unmasking further therapeutic targets. In fact, small molecules, used in experimental MAS models, might represent new therapeutic options.
Collapse
Affiliation(s)
- Roberto Giacomelli
- Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Piero Ruscitti
- Rheumatology Section, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
18
|
Kernan KF, Carcillo JA. Hyperferritinemia and inflammation. Int Immunol 2017; 29:401-409. [PMID: 28541437 PMCID: PMC5890889 DOI: 10.1093/intimm/dxx031] [Citation(s) in RCA: 382] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/23/2017] [Indexed: 12/18/2022] Open
Abstract
Understanding of ferritin biology has traditionally centered on its role in iron storage and homeostasis, with low ferritin levels indicative of deficiency and high levels indicative of primary or secondary hemochromatosis. However, further work has shown that iron, redox biology and inflammation are inexorably linked. During infection, increased ferritin levels represent an important host defense mechanism that deprives bacterial growth of iron and protects immune cell function. It may also be protective, limiting the production of free radicals and mediating immunomodulation. Additionally, hyperferritinemia is a key acute-phase reactants, used by clinicians as an indication for therapeutic intervention, aimed at controlling inflammation in high-risk patients. One school of thought maintains that hyperferritinemia is an 'innocent bystander' biomarker of uncontrolled inflammation that can be used to gauge effectiveness of intervention. Other schools of thought maintain that ferritin induction could be a protective negative regulatory loop. Others maintain that ferritin is a key mediator of immune dysregulation, especially in extreme hyperferritinemia, via direct immune-suppressive and pro-inflammatory effects. There is a clear need for further investigation of the role of ferritin in uncontrolled inflammatory conditions both as a biomarker and mediator of disease because its occurrence identifies patients with high mortality risk and its resolution predicts their improved survival.
Collapse
Affiliation(s)
- Kate F Kernan
- Department of Critical Care Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | - Joseph A Carcillo
- Department of Critical Care Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| |
Collapse
|
19
|
Ruscitti P, Cipriani P, Di Benedetto P, Liakouli V, Berardicurti O, Carubbi F, Ciccia F, Guggino G, Triolo G, Giacomelli R. H-ferritin and proinflammatory cytokines are increased in the bone marrow of patients affected by macrophage activation syndrome. Clin Exp Immunol 2017; 191:220-228. [PMID: 28960260 DOI: 10.1111/cei.13057] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2017] [Indexed: 12/12/2022] Open
Abstract
Macrophage activation syndrome (MAS) is hyperinflammatory life-threatening syndrome, associated typically with high levels of serum ferritin. This is an iron storage protein including heavy (H) and light (L) subunits, categorized on their molecular weight. The H-/L subunits ratio may be different in tissues, depending on the specific tissue and pathophysiological status. In this study, we analysed the bone marrow (BM) biopsies of adult MAS patients to assess the presence of: (i) H-ferritin and L-ferritin; (ii) CD68+ /H-ferritin+ and CD68+ /L-ferritin+ ; and (iii) interleukin (IL)-1β, tumour necrosis factor (TNF) and interferon (IFN)-γ. We also explored possible correlations of these results with clinical data. H-ferritin, IL-1β, TNF and IFN-γ were increased significantly in MAS. Furthermore, an increased number of CD68+ /H-ferritin+ cells and an infiltrate of cells co-expressing H-ferritin and IL-12, suggesting an infiltrate of M1 macrophages, were observed. H-ferritin levels and CD68+ /H-ferritin+ cells were correlated with haematological involvement of the disease, serum ferritin and C-reactive protein. L-ferritin and CD68+ /L-ferritin+ cells did not correlate with these parameters. In conclusion, during MAS, H-ferritin, CD68+ /H-ferritin+ cells and proinflammatory cytokines were increased significantly in the BM inflammatory infiltrate, pointing out a possible vicious pathogenic loop. To date, H-ferritin and CD68+ /H-ferritin+ were associated significantly with haematological involvement of the disease, suggesting biomarkers assessing severity of clinical picture.
Collapse
Affiliation(s)
- P Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - P Cipriani
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - P Di Benedetto
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - V Liakouli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - O Berardicurti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - F Carubbi
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - F Ciccia
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - G Guggino
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - G Triolo
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - R Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
20
|
Ruscitti P, Cipriani P, Di Benedetto P, Liakouli V, Carubbi F, Berardicurti O, Ciccia F, Guggino G, Triolo G, Giacomelli R. Advances in immunopathogenesis of macrophage activation syndrome during rheumatic inflammatory diseases: toward new therapeutic targets? Expert Rev Clin Immunol 2017; 13:1041-1047. [PMID: 28837367 DOI: 10.1080/1744666x.2017.1372194] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Macrophage activation syndrome (MAS) is a severe, hyperinflammatory life-threatening syndrome, generally complicating different rheumatic diseases. Despite the severity of the disease, little is known about the pathogenic mechanisms and, thus, possible targeted therapies in the management of these patients. Areas covered: In this review, we aimed to update the current pathogenic knowledge of MAS, during rheumatic diseases, focusing mainly on immunologic abnormalities and on new possible therapeutic strategies. Expert commentary: The difficult pathogenic scenario of MAS, in which genetic defects, predisposing diseases, and triggers are mixed together with the high mortality rate, make it difficult to manage these patients. Although most efforts have been focused on investigating the disease in children, in recent years, several studies are trying to elucidate the possible pathogenic mechanism in adult MAS patients. In this context, genetic and immunological studies might lead to advances in the knowledge of pathogenic mechanisms and possible new therapeutic targets. In the future, the results of ongoing clinical trials are awaited in order to improve the management and, thus, the survival of these patients.
Collapse
Affiliation(s)
- Piero Ruscitti
- a Division of Rheumatology , University of L'Aquila , L'Aquila , Italy
| | - Paola Cipriani
- a Division of Rheumatology , University of L'Aquila , L'Aquila , Italy
| | | | - Vasiliky Liakouli
- a Division of Rheumatology , University of L'Aquila , L'Aquila , Italy
| | - Francesco Carubbi
- a Division of Rheumatology , University of L'Aquila , L'Aquila , Italy
| | | | - Francesco Ciccia
- b Division of Rheumatology , University of Palermo , Palermo , Italy
| | - Giuliana Guggino
- b Division of Rheumatology , University of Palermo , Palermo , Italy
| | - Giovanni Triolo
- b Division of Rheumatology , University of Palermo , Palermo , Italy
| | | |
Collapse
|
21
|
Ruscitti P, Cipriani P, Ciccia F, Masedu F, Liakouli V, Carubbi F, Berardicurti O, Guggino G, Di Benedetto P, Di Bartolomeo S, Valenti M, Triolo G, Giacomelli R. Prognostic factors of macrophage activation syndrome, at the time of diagnosis, in adult patients affected by autoimmune disease: Analysis of 41 cases collected in 2 rheumatologic centers. Autoimmun Rev 2016; 16:16-21. [PMID: 27664384 DOI: 10.1016/j.autrev.2016.09.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 07/29/2016] [Indexed: 12/21/2022]
Abstract
Macrophage activation syndrome (MAS) is a rare, life-threatening disease in which early diagnosis and aggressive therapeutic strategy may improve the outcome. Due to its rarity, epidemiologic data are still lacking. Hyperferritinemia is frequently associated with MAS and might modulate the cytokine storm, which is involved in the development of multiple organ failure. In this paper, we investigated clinical data, treatments, and outcome of a homogeneous cohort of 41 adult MAS patients, complicating autoimmune rheumatic diseases. MAS-related death occurred in 17 patients (42.5%) during the follow-up, and older age and increased serum ferritin levels, at the time of diagnosis, were significantly associated with mortality. In conclusion, adult MAS is associated with high mortality rate. Some clinical features at diagnosis may be predictive of MAS-associated death.
Collapse
Affiliation(s)
- Piero Ruscitti
- Division of Rheumatology, University of L'Aquila, L'Aquila, Italy
| | - Paola Cipriani
- Division of Rheumatology, University of L'Aquila, L'Aquila, Italy
| | | | - Francesco Masedu
- Division of Medical Statistic Unit, University of L'Aquila, L'Aquila, Italy
| | | | | | | | | | | | | | - Marco Valenti
- Division of Medical Statistic Unit, University of L'Aquila, L'Aquila, Italy
| | - Giovanni Triolo
- Division of Rheumatology, University of Palermo, Palermo, Italy
| | | |
Collapse
|
22
|
Ruscitti P, Cipriani P, Ciccia F, Di Benedetto P, Liakouli V, Berardicurti O, Carubbi F, Guggino G, Di Bartolomeo S, Triolo G, Giacomelli R. H-ferritin and CD68(+) /H-ferritin(+) monocytes/macrophages are increased in the skin of adult-onset Still's disease patients and correlate with the multi-visceral involvement of the disease. Clin Exp Immunol 2016; 186:30-8. [PMID: 27317930 DOI: 10.1111/cei.12826] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2016] [Indexed: 12/28/2022] Open
Abstract
Adult-onset Still's disease (AOSD) patients may show an evanescent salmon-pink erythema appearing during febrile attacks and reducing without fever. Some patients may experience this eruption for many weeks. During AOSD, exceptionally high serum levels of ferritin may be observed; it is an iron storage protein composed of 24 subunits, heavy (H) subunits and light (L) subunits. The ferritin enriched in L subunits (L-ferritin) and the ferritin enriched in H subunits (H-ferritin) may be observed in different tissues. In this work, we aimed to investigate the skin expression of both H-and L-ferritin and the number of macrophages expressing these molecules from AOSD patients with persistent cutaneous lesions. We observed an increased expression of H-ferritin in the skin, associated with an infiltrate in the biopsies obtained from persistent cutaneous lesions of AOSD patients. Furthermore, a positive correlation between H-ferritin skin levels as well as the number of CD68(+) /H-ferritin(+) cells and the multi-visceral involvement of the disease was observed. Our data showed an increased expression of H-ferritin in the skin of AOSD patients, associated with a strong infiltrate of CD68(+) /H-ferritin(+) cells. Furthermore, a correlation between the levels of H-ferritin as well as of the number of CD68(+) /H-ferritin(+) cells and the multi-visceral involvement of the disease was observed.
Collapse
Affiliation(s)
- P Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - P Cipriani
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - F Ciccia
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - P Di Benedetto
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - V Liakouli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - O Berardicurti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - F Carubbi
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - G Guggino
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - S Di Bartolomeo
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - G Triolo
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - R Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
23
|
Lu CH, Allen K, Oei F, Leoni E, Kuhle J, Tree T, Fratta P, Sharma N, Sidle K, Howard R, Orrell R, Fish M, Greensmith L, Pearce N, Gallo V, Malaspina A. Systemic inflammatory response and neuromuscular involvement in amyotrophic lateral sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2016; 3:e244. [PMID: 27308305 PMCID: PMC4897985 DOI: 10.1212/nxi.0000000000000244] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/14/2016] [Indexed: 11/20/2022]
Abstract
Objective: To evaluate the combined blood expression of neuromuscular and inflammatory biomarkers as predictors of disease progression and prognosis in amyotrophic lateral sclerosis (ALS). Methods: Logistic regression adjusted for markers of the systemic inflammatory state and principal component analysis were carried out on plasma levels of creatine kinase (CK), ferritin, and 11 cytokines measured in 95 patients with ALS and 88 healthy controls. Levels of circulating biomarkers were used to study survival by Cox regression analysis and correlated with disease progression and neurofilament light chain (NfL) levels available from a previous study. Cytokines expression was also tested in blood samples longitudinally collected for up to 4 years from 59 patients with ALS. Results: Significantly higher levels of CK, ferritin, tumor necrosis factor (TNF)–α, and interleukin (IL)–1β, IL-2, IL-8, IL-12p70, IL-4, IL-5, IL-10, and IL-13 and lower levels of interferon (IFN)–γ were found in plasma samples from patients with ALS compared to controls. IL-6, TNF-α, and IFN-γ were the most highly regulated markers when all explanatory variables were jointly analyzed. High ferritin and IL-2 levels were predictors of poor survival. IL-5 levels were positively correlated with CK, as was TNF-α with NfL. IL-6 was strongly associated with CRP levels and was the only marker showing increasing expression towards end-stage disease in the longitudinal analysis. Conclusions: Neuromuscular pathology in ALS involves the systemic regulation of inflammatory markers mostly active on T-cell immune responses. Disease stratification based on the prognostic value of circulating inflammatory markers could improve clinical trials design in ALS.
Collapse
Affiliation(s)
- Ching-Hua Lu
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Kezia Allen
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Felicia Oei
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Emanuela Leoni
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Jens Kuhle
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Timothy Tree
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Pietro Fratta
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Nikhil Sharma
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Katie Sidle
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Robin Howard
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Richard Orrell
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Mark Fish
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Linda Greensmith
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Neil Pearce
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Valentina Gallo
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| | - Andrea Malaspina
- Centre for Neuroscience & Trauma (C.L., F.O., J.K., A.M.) and Centre of Primary Care and Public Health (V.G.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.L., P.F., L.G.), MRC Centre for Neuromuscular Diseases (P.F., L.G.), MRC Unit for Lifelong Health and Ageing (N.S.), Department of Molecular Neuroscience (K.S.), and Department of Clinical Neuroscience (R.O.), UCL Institute of Neurology; Basildon and Thurrock University Hospitals (K.A., A.M.), NHS Foundation Trust, Basildon; William Harvey Hospital (F.O.), Kent; Proteome Sciences plc (E.L.), South Wing Laboratory, Institute of Psychiatry, UK; Neurology (J.K.), University Hospital Basel, Switzerland; Department of Immunobiology (T.T.), King's College London; National Hospital for Neurology and Neurosurgery (N.S., R.H., R.O.), London; Musgrove Park Hospital (M.F.), Taunton; and Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, UK
| |
Collapse
|
24
|
Belaidi AA, Bush AI. Iron neurochemistry in Alzheimer's disease and Parkinson's disease: targets for therapeutics. J Neurochem 2016; 139 Suppl 1:179-197. [DOI: 10.1111/jnc.13425] [Citation(s) in RCA: 324] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/24/2015] [Accepted: 11/03/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Abdel A. Belaidi
- The Florey Institute for Neuroscience and Mental Health; The University of Melbourne; Parkville Vic. Australia
| | - Ashley I. Bush
- The Florey Institute for Neuroscience and Mental Health; The University of Melbourne; Parkville Vic. Australia
| |
Collapse
|
25
|
Ruscitti P, Ciccia F, Cipriani P, Guggino G, Di Benedetto P, Rizzo A, Liakouli V, Berardicurti O, Carubbi F, Triolo G, Giacomelli R. The CD68(+)/H-ferritin(+) cells colonize the lymph nodes of the patients with adult onset Still's disease and are associated with increased extracellular level of H-ferritin in the same tissue: correlation with disease severity and implication for pathogenesis. Clin Exp Immunol 2015; 183:397-404. [PMID: 26540556 DOI: 10.1111/cei.12738] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2015] [Indexed: 12/11/2022] Open
Abstract
In this work, we aimed to evaluate the levels of ferritin enriched in H subunits (H-ferritin) and ferritin enriched in L subunits (L-ferritin) and the cells expressing these two molecules in the lymph node (LN) biopsies obtained from adult-onset Still's disease (AOSD) patients, and the possible correlation among these data and the severity of the disease. Ten patients with AOSD underwent LN biopsy. All the samples were stained by immunofluorescence. A statistical analysis was performed to estimate the possible correlation among both H-ferritin and L-ferritin tissue expression and the clinical picture of the disease. Furthermore, the same analysis was performed to evaluate the possible correlation among the number of CD68(+)/H-ferritin(+) or CD68(+)/L-ferritin(+) cells and the clinical picture. Immunofluorescence analysis demonstrated an increased tissue H-ferritin expression in the LNs of AOSD patients. This increased expression correlated with the severity of the disease. An increased number of CD68 macrophages expressing H-ferritin was observed in the LN samples of our patients. Furthermore, we observed that the number of CD68(+)/H-ferritin(+) cells correlated significantly with the severity of the clinical picture. Our data showed an imbalance between the levels of H- and L-ferritin in LNs of AOSD patients and the evidence of an increased number of CD68(+)/H-ferritin(+) cells in the same organs. Furthermore, a correlation among both the tissue H-ferritin levels and the CD68(+)/H-ferritin(+) cells and the clinical picture was observed.
Collapse
Affiliation(s)
- P Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - F Ciccia
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - P Cipriani
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - G Guggino
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - P Di Benedetto
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - A Rizzo
- General Pathology, Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - V Liakouli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - O Berardicurti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - F Carubbi
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | - G Triolo
- Division of Rheumatology, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - R Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, School of Medicine, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
26
|
Induction of Interleukin-1β by Human Immunodeficiency Virus-1 Viral Proteins Leads to Increased Levels of Neuronal Ferritin Heavy Chain, Synaptic Injury, and Deficits in Flexible Attention. J Neurosci 2015. [PMID: 26203149 DOI: 10.1523/jneurosci.4403-14.2015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Synaptodendritic pruning and alterations in neurotransmission are the main underlying causes of HIV-associated neurocognitive disorders (HAND). Our studies in humans and nonhuman primates indicated that the protein ferritin heavy chain (FHC) is a critical player in neuronal changes and ensuing cognitive deficit observed in these patients. Here we focus on the effect of HIV proteins and inflammatory cytokines implicated in HAND on neuronal FHC levels, dendritic changes, and neurocognitive behavior. In two well characterized models of HAND (HIV transgenic and gp120-treated rats), we report reductions in spine density and dendritic branches in prefrontal cortex pyramidal neurons compared with age-matched controls. FHC brain levels are elevated in these animals, which also show deficits in reversal learning. Moreover, IL-1β, TNF-α, and HIV gp120 upregulate FHC in rat cortical neurons. However, although the inflammatory cytokines directly altered neuronal FHC, gp120 only caused significant FHC upregulation in neuronal/glial cocultures, suggesting that glia are necessary for sustained elevation of neuronal FHC by the viral protein. Although the envelope protein induced secretion of IL-1β and TNF-α in cocultures, TNF-α blockade did not affect gp120-mediated induction of FHC. Conversely, studies with an IL-1β neutralizing antibody or specific IL-1 receptor antagonist revealed the primary involvement of IL-1β in gp120-induced FHC changes. Furthermore, silencing of neuronal FHC abrogates the effect of gp120 on spines, and spine density correlates negatively with FHC levels or cognitive deficit. These results demonstrate that viral and host components of HIV infection increase brain expression of FHC, leading to cellular and functional changes, and point to IL-1β-targeted strategies for prevention of these alterations. Significance statement: This work demonstrates the key role of the cytokine IL-1β in the regulation of a novel intracellular mediator [i.e., the protein ferritin heavy chain (FHC)] of HIV-induced dendritic damage and the resulting neurocognitive impairment. This is also the first study that systematically investigates dendritic damage in layer II/III prefrontal cortex neurons of two different non-infectious models of HIV-associated neurocognitive disorders (HAND) and reveals a precise correlation of these structural changes with specific biochemical and functional alterations also reported in HIV patients. Overall, these data suggest that targeting the IL-1β-dependent FHC increase may represent a valid strategy for neuroprotective adjuvant therapies in HAND.
Collapse
|
27
|
Hatcher HC, Tesfay L, Torti SV, Torti FM. Cytoprotective Effect of Ferritin H in Renal Ischemia Reperfusion Injury. PLoS One 2015; 10:e0138505. [PMID: 26379029 PMCID: PMC4574775 DOI: 10.1371/journal.pone.0138505] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/31/2015] [Indexed: 01/08/2023] Open
Abstract
Oxidative stress is a major contributor to kidney injury following ischemia reperfusion. Ferritin, a highly conserved iron-binding protein, is a key protein in the maintenance of cellular iron homeostasis and protection from oxidative stress. Ferritin mitigates oxidant stress by sequestering iron and preventing its participation in reactions that generate reactive oxygen species. Ferritin is composed of two subunit types, ferritin H and ferritin L. Using an in vivo model that enables conditional tissue-specific doxycycline-inducible expression of ferritin H in the mouse kidney, we tested the hypothesis that an increased level of H-rich ferritin is renoprotective in ischemic acute renal failure. Prior to induction of ischemia, doxycycline increased ferritin H in the kidneys of the transgenic mice nearly 6.5-fold. Following reperfusion for 24 hours, induction of neutrophil gelatinous-associated lipocalin (NGAL, a urine marker of renal dysfunction) was reduced in the ferritin H overexpressers compared to controls. Histopathologic examination following ischemia reperfusion revealed that ferritin H overexpression increased intact nuclei in renal tubules, reduced the frequency of tubular profiles with luminal cast materials, and reduced activated caspase-3 in the kidney. In addition, generation of 4-hydroxy 2-nonenal protein adducts, a measurement of oxidant stress, was decreased in ischemia-reperfused kidneys of ferritin H overexpressers. These studies demonstrate that ferritin H can inhibit apoptotic cell death, enhance tubular epithelial viability, and preserve renal function by limiting oxidative stress following ischemia reperfusion injury.
Collapse
Affiliation(s)
- Heather C. Hatcher
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Lia Tesfay
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Suzy V. Torti
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
- Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Frank M. Torti
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
- Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
28
|
Serum ferritin levels predict histological severity in patients with nonalcoholic fatty liver disease in India. Indian J Gastroenterol 2015; 34:200-8. [PMID: 26108652 DOI: 10.1007/s12664-015-0572-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/13/2015] [Indexed: 02/04/2023]
Abstract
AIM The aims of the study were to determine the levels of serum ferritin which predict fibrosis in Indian patients with nonalcoholic fatty liver disease (NAFLD) and to establish correlation between Fibroscan values and serum ferritin levels. METHODS The clinical, biochemical, radiologic, and histological findings of consecutive adult NAFLD patients accessed at a tertiary care center over a 3-year period were analyzed. Those with concurrent liver diseases were excluded. Fifty-five of 250 NAFLD patients with fatty liver on ultrasound and raised enzymes (>40 IU/L) underwent liver biopsy. Patients were stratified into two groups based on their histological stage steatosis (with or without inflammation) but no fibrosis and nonalcoholic steatohepatitis (NASH) with fibrosis/cirrhosis. Serum ferritin levels were measured at the same time as getting liver biopsy. Fibroscan was carried out in each of these patients. These were compared with 50 age- and sex-matched controls with normal ultrasound, liver enzymes, and no history of alcohol. Student's t test was used as the test for significance. RESULTS Fifty-five NAFLD patients diagnosed on ultrasound and with raised enzymes underwent biopsy. Steatosis (with or without inflammation, but no fibrosis/ballooning) was seen in 35 patients, fibrosis/ballooning in 14 patients, and cirrhosis in 6 patients. Mean ferritin levels in groups with nonalcoholic fatty liver (NAFL) and nonalcoholic steatohepatitis (NASH) were 39.4 and 72.7 ng/mL, respectively (p < 0.001). The mean ferritin levels in NAFLD and controls were 51.2 and 35.2 ng/mL, respectively (p < 0.05). The area under the curve (AUC) of serum ferritin at value 48.0 ng/mL was 0.779. The coefficient of correlation between Fibroscan and serum ferritin levels was 0.9864 while that with alanine transaminase and aspartate aminotransferase was 0.69. Serum ferritin at the cutoff of 48 ng/mL differentiated significantly patients with fibrosis and higher Fibroscan levels. CONCLUSION Serum ferritin was low in Indian individuals, and levels even within apparently normal range indicated fibrosis and cirrhosis. A cutoff level of 48.0 IU/mL distinguished fibrosis in NAFLD. Fibroscan correlated well with serum ferritin levels.
Collapse
|
29
|
Increased level of H-ferritin and its imbalance with L-ferritin, in bone marrow and liver of patients with adult onset Still's disease, developing macrophage activation syndrome, correlate with the severity of the disease. Autoimmun Rev 2015; 14:429-37. [PMID: 25599955 DOI: 10.1016/j.autrev.2015.01.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 01/12/2015] [Indexed: 02/04/2023]
Abstract
In this paper, we aimed to evaluate the levels of ferritin enriched in H subunits (H-ferritin) and ferritin enriched in L subunits (L-ferritin) and the cells expressing these 2 molecules, in the bone marrow (BM) and liver biopsies obtained from adult onset Still's disease (AOSD) patients who developed macrophage activation syndrome (MAS), and correlating these data with the severity of the disease. Twenty-one patients with MAS-associated AOSD underwent BM biopsy and among them, 9 patients with hepatomegaly and elevated liver enzymes underwent liver biopsy. All the samples were stained by both immunohistochemistry and immunofluorescence. A statistical analysis was performed to estimate the possible correlation among both H-ferritin and L-ferritin tissue expression and the clinical picture of the disease. Furthermore, the same analysis was performed to evaluate the possible correlation among the number of CD68/H-ferritin or CD68/L-ferritin positive cells and the clinical picture. Both immunohistochemical and immunofluorescence analysis demonstrated an increased tissue H-ferritin expression, in the BM and liver samples of our patients. This increased expression correlated with the severity of the disease. An inflammatory infiltrate, enriched in CD68 macrophages, expressing H-ferritin was observed in both the BM and the liver samples of our patients. Furthermore, we observed, that this increased number of CD68/H-ferritin positive cells significantly correlated with the severity of clinical picture and this specific BM infiltrate correlated with the mortality rate, reported in our cohort. Our data showed an imbalance between the levels of H- and L-ferritin in different organs of patients with MAS-associated AOSD and the evidence of a strong infiltrate of CD68/H-ferritin positive cells in the same organs. Furthermore, a strong correlation among both the tissue H-ferritin and the CD68/H-ferritin positive cells and the clinical picture was observed.
Collapse
|
30
|
Molecular cloning and expression analysis of ferritin, heavy polypeptide 1 gene from duck (Anas platyrhynchos). Mol Biol Rep 2014; 41:6233-40. [PMID: 24981929 DOI: 10.1007/s11033-014-3503-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 06/18/2014] [Indexed: 01/14/2023]
Abstract
H-ferritin is a core subunit of the iron storage protein ferritin, and is related to the pathogenesis of malignant diseases. A differential expressed sequence tag of the ferritin, heavy polypeptide 1 gene (FTH1) was obtained from our previously constructed suppression subtractive cDNA library from 3-day-old ducklings challenged with duck hepatitis virus type I (DHV-1). The expression and function of FTH1 in immune defense against infection remains largely unknown in ducks. In this study, the full-length duFTH1 cDNA was obtained using reverse transcription-polymerase chain reaction (RT-PCR) and rapid amplification of cDNA ends. It consisted of 153 basepairs (bp) 5'untranslated region (UTR), 183 bp 3'UTR, and 546 bp open reading frame that encodes a single protein of 181 amino acid residues. duFTH1 shares high similarity with FTH1 genes from other vertebrates. The amino acid sequence possesses the conserved domain of typical ferritin H subunits, including seven metal ligands in the ferroxidase center, one iron binding region signature, and a potential bio-mineralization residue (Thy(29)). Moreover, in agreement with a previously reported ferritin H subunit, we identified an iron response element in the 5'UTR. RT-PCR analyses revealed duFTH1 mRNA is widely expressed in various tissues. Real-time quantitative polymerase chain reaction analyses suggested that duFTH1 mRNA is significantly up-regulated in the liver after DHV-1 injection or polyriboinosinic polyribocytidylic acid (polyI:C) treatment, reaching a peak 4 h post-infection, and dropping progressively and returning to normal after 24 h. Our findings suggest that duFTH1 functions as an iron chelating protein subunit in duck and contributes to the innate immune responses against viral infections.
Collapse
|
31
|
Aiba Y, Yamashita M, Katakura Y, Furukawa Y, Matsumoto SE, Tomimatsu K, Teruya K, Shirahata S. Identification of Genes Involved in the Suppression of Antibody Production from Human Peripheral Blood Lymphocytes. Biosci Biotechnol Biochem 2014; 70:966-70. [PMID: 16636465 DOI: 10.1271/bbb.70.966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Pretreatment with L-leucyl-L-leucine methyl ester (LLME) is a prerequisite for peripheral blood mononuclear cells (PBMCs) to produce antigen-specific antibodies when sensitized with an antigen. Little information, however, is available regarding the mechanisms involved in LLME-induced augmentation of antibody production from PBMCs that are antigen sensitized. In the present study, we attempted to identify the genes involved in the suppression of antibody production from PBMCs that was not treated with LLME, but sensitized with an antigen. Using subtractive screening, we obtained 63 independent genes, including 17 EST genes, that are specific for LLME-nontreated PBMC. Among these genes, the expression of heavy chain ferritin (H-ferritin), CC chemokine ligand 18 (CCL18), and matrix metalloproteinase 12 (MMP12) were augmented in LLME-nontreated PBMCs, suggesting that inflammatory factors might be involved in the suppression of antibody production in LLME-nontreated PBMCs.
Collapse
Affiliation(s)
- Yoshihiro Aiba
- Graduate School of Systems Life Sciences, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Ren C, Chen T, Jiang X, Wang Y, Hu C. Identification and functional characterization of a novel ferritin subunit from the tropical sea cucumber, Stichopus monotuberculatus. FISH & SHELLFISH IMMUNOLOGY 2014; 38:265-274. [PMID: 24698995 DOI: 10.1016/j.fsi.2014.03.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 03/12/2014] [Accepted: 03/15/2014] [Indexed: 06/03/2023]
Abstract
Ferritin is one of the major non-harm iron storage proteins that found in most cell types of animals, plants and microorganisms. In this study, a ferritin subunit named StmFer was identified from the sea cucumber (Stichopus monotuberculatus) and characterized functionally. The full-length cDNA of StmFer is 1184 bp in size with a 5'-untranslated region (UTR) of 131 bp, a 3'-UTR of 531 bp and an open reading frame of 522 bp that encoding a protein of 173 amino acids with a deduced molecular weight of 19.95 kDa. StmFer possesses both the ferroxidase center of vertebrate ferritin heavy subunit and iron nucleation sites of vertebrate ferritin light subunit. For the gene structure, StmFer contains only three exons separated by two introns. Higher levels of mRNA expression were noticed in intestine and coelomocyte of S. monotuberculatus by northern blot analysis. In in vitro experiments performed in coelomocytes, transcriptional expression of StmFer showed the strongest response to polyriboinosinic polyribocytidylic acid [Poly (I:C)] (9.08 fold up-regulation), followed by lipopolysaccharides (LPS), ferrous chloride (FeCl2) and inactivated bacteria (Vibrio alginolyticus) (7.84, 7.41 and 4.90 fold up-regulation, respectively) after 3 h post-challenge. In addition, the anti-oxidation activity and iron binding capacity of recombinant ferritin protein were demonstrated in this study. As a whole, our study suggested that the ferritin from sea cucumber may play critical roles not only in the cellular and organismic iron homeostasis, but also in the innate immune defense.
Collapse
Affiliation(s)
- Chunhua Ren
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB); Key Laboratory of Applied Marine Biology of Guangdong Province and Chinese Academy of Sciences (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China.
| | - Ting Chen
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB); Key Laboratory of Applied Marine Biology of Guangdong Province and Chinese Academy of Sciences (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China.
| | - Xiao Jiang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB); Key Laboratory of Applied Marine Biology of Guangdong Province and Chinese Academy of Sciences (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China.
| | - Yanhong Wang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB); Key Laboratory of Applied Marine Biology of Guangdong Province and Chinese Academy of Sciences (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China.
| | - Chaoqun Hu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB); Key Laboratory of Applied Marine Biology of Guangdong Province and Chinese Academy of Sciences (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China.
| |
Collapse
|
33
|
Abstract
SIGNIFICANCE Inflammation and immunity can be associated with varying degrees of heme release from hemoproteins, eventually leading to cellular and tissue iron (Fe) overload, oxidative stress, and tissue damage. Presumably, these deleterious effects contribute to the pathogenesis of systemic infections. RECENT ADVANCES Heme release from hemoglobin sensitizes parenchyma cells to undergo programmed cell death in response to proinflammatory cytokines, such as tumor necrosis factor. This cytotoxic effect is driven by a mechanism involving intracellular accumulation of free radicals, which sustain the activation of the c-Jun N-terminal kinase (JNK) signaling transduction pathway. While heme catabolism by heme oxygenase-1 (HO-1) prevents programmed cell death, this cytoprotective effect requires the co-expression of ferritin H (heart/heavy) chain (FTH), which controls the pro-oxidant effect of labile Fe released from the protoporphyrin IX ring of heme. This antioxidant effect of FTH restrains JNK activation, whereas JNK activation inhibits FTH expression, a cross talk that controls metabolic adaptation to cellular Fe overload associated with systemic infections. CRITICAL ISSUES AND FUTURE DIRECTIONS Identification and characterization of the mechanisms via which FTH provides metabolic adaptation to tissue Fe overload should provide valuable information to our current understanding of the pathogenesis of systemic infections as well as other immune-mediated inflammatory diseases.
Collapse
|
34
|
Singh N, Haldar S, Tripathi AK, Horback K, Wong J, Sharma D, Beserra A, Suda S, Anbalagan C, Dev S, Mukhopadhyay CK, Singh A. Brain iron homeostasis: from molecular mechanisms to clinical significance and therapeutic opportunities. Antioxid Redox Signal 2014; 20:1324-63. [PMID: 23815406 PMCID: PMC3935772 DOI: 10.1089/ars.2012.4931] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Iron has emerged as a significant cause of neurotoxicity in several neurodegenerative conditions, including Alzheimer's disease (AD), Parkinson's disease (PD), sporadic Creutzfeldt-Jakob disease (sCJD), and others. In some cases, the underlying cause of iron mis-metabolism is known, while in others, our understanding is, at best, incomplete. Recent evidence implicating key proteins involved in the pathogenesis of AD, PD, and sCJD in cellular iron metabolism suggests that imbalance of brain iron homeostasis associated with these disorders is a direct consequence of disease pathogenesis. A complete understanding of the molecular events leading to this phenotype is lacking partly because of the complex regulation of iron homeostasis within the brain. Since systemic organs and the brain share several iron regulatory mechanisms and iron-modulating proteins, dysfunction of a specific pathway or selective absence of iron-modulating protein(s) in systemic organs has provided important insights into the maintenance of iron homeostasis within the brain. Here, we review recent information on the regulation of iron uptake and utilization in systemic organs and within the complex environment of the brain, with particular emphasis on the underlying mechanisms leading to brain iron mis-metabolism in specific neurodegenerative conditions. Mouse models that have been instrumental in understanding systemic and brain disorders associated with iron mis-metabolism are also described, followed by current therapeutic strategies which are aimed at restoring brain iron homeostasis in different neurodegenerative conditions. We conclude by highlighting important gaps in our understanding of brain iron metabolism and mis-metabolism, particularly in the context of neurodegenerative disorders.
Collapse
Affiliation(s)
- Neena Singh
- 1 Department of Pathology, Case Western Reserve University , Cleveland, Ohio
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Pitcher J, Abt A, Myers J, Han R, Snyder M, Graziano A, Festa L, Kutzler M, Garcia F, Gao WJ, Fischer-Smith T, Rappaport J, Meucci O. Neuronal ferritin heavy chain and drug abuse affect HIV-associated cognitive dysfunction. J Clin Invest 2014; 124:656-69. [PMID: 24401274 DOI: 10.1172/jci70090] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 10/24/2013] [Indexed: 11/17/2022] Open
Abstract
Interaction of the chemokine CXCL12 with its receptor CXCR4 promotes neuronal function and survival during embryonic development and throughout adulthood. Previous studies indicated that μ-opioid agonists specifically elevate neuronal levels of the protein ferritin heavy chain (FHC), which negatively regulates CXCR4 signaling and affects the neuroprotective function of the CXCL12/CXCR4 axis. Here, we determined that CXCL12/CXCR4 activity increased dendritic spine density, and also examined FHC expression and CXCR4 status in opiate abusers and patients with HIV-associated neurocognitive disorders (HAND), which is typically exacerbated by illicit drug use. Drug abusers and HIV patients with HAND had increased levels of FHC, which correlated with reduced CXCR4 activation, within cortical neurons. We confirmed these findings in a nonhuman primate model of SIV infection with morphine administration. Transfection of a CXCR4-expressing human cell line with an iron-deficient FHC mutant confirmed that increased FHC expression deregulated CXCR4 signaling and that this function of FHC was independent of iron binding. Furthermore, examination of morphine-treated rodents and isolated neurons expressing FHC shRNA revealed that FHC contributed to morphine-induced dendritic spine loss. Together, these data implicate FHC-dependent deregulation of CXCL12/CXCR4 as a contributing factor to cognitive dysfunction in neuroAIDS.
Collapse
|
36
|
Abstract
Iron is an essential nutrient that is tightly regulated. A principal function of the liver is the regulation of iron homeostasis. The liver senses changes in systemic iron requirements and can regulate iron concentrations in a robust and rapid manner. The last 10 years have led to the discovery of several regulatory mechanisms in the liver that control the production of iron regulatory genes, storage capacity, and iron mobilization. Dysregulation of these functions leads to an imbalance of iron, which is the primary cause of iron-related disorders. Anemia and iron overload are two of the most prevalent disorders worldwide and affect over a billion people. Several mutations in liver-derived genes have been identified, demonstrating the central role of the liver in iron homeostasis. During conditions of excess iron, the liver increases iron storage and protects other tissues, namely, the heart and pancreas from iron-induced cellular damage. However, a chronic increase in liver iron stores results in excess reactive oxygen species production and liver injury. Excess liver iron is one of the major mechanisms leading to increased steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Erik R Anderson
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
37
|
Alkhateeb AA, Connor JR. The significance of ferritin in cancer: anti-oxidation, inflammation and tumorigenesis. Biochim Biophys Acta Rev Cancer 2013; 1836:245-54. [PMID: 23891969 DOI: 10.1016/j.bbcan.2013.07.002] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 07/09/2013] [Accepted: 07/18/2013] [Indexed: 12/16/2022]
Abstract
The iron storage protein ferritin has been continuously studied for over 70years and its function as the primary iron storage protein in cells is well established. Although the intracellular functions of ferritin are for the most part well-characterized, the significance of serum (extracellular) ferritin in human biology is poorly understood. Recently, several lines of evidence have demonstrated that ferritin is a multi-functional protein with possible roles in proliferation, angiogenesis, immunosuppression, and iron delivery. In the context of cancer, ferritin is detected at higher levels in the sera of many cancer patients, and the higher levels correlate with aggressive disease and poor clinical outcome. Furthermore, ferritin is highly expressed in tumor-associated macrophages which have been recently recognized as having critical roles in tumor progression and therapy resistance. These characteristics suggest ferritin could be an attractive target for cancer therapy because its down-regulation could disrupt the supportive tumor microenvironment, kill cancer cells, and increase sensitivity to chemotherapy. In this review, we provide an overview of the current knowledge on the function and regulation of ferritin. Moreover, we examine the literature on ferritin's contributions to tumor progression and therapy resistance, in addition to its therapeutic potential.
Collapse
Affiliation(s)
- Ahmed A Alkhateeb
- Department of Neurosurgery, The Pennsylvania State University Hershey Medical Center, Hershey, PA, USA
| | | |
Collapse
|
38
|
Abstract
ABSTRACT Iron is an essential transition metal for mammalian cellular and tissue viability. It is critical to supplying oxygen through heme, the mitochondrial respiratory chain, and enzymes such as ribonucleotide reductase. Mammalian organisms have evolved with the means of regulating the metabolism of iron, because if left unregulated, the resulting excess amounts of iron may induce chronic toxicities affecting multiple organ systems. Several homeostatic mechanisms exist to control the amount of intestinal dietary iron uptake, cellular iron uptake, distribution, and export. Within these processes, numerous molecular participants have been identified because of advancements in basic cell biology and efforts in disease-based research of iron storage abnormalities. For example, dietary iron uptake across the intestinal duodenal mucosa is mediated by an intramembrane divalent metal transporter 1 (DMT1), and cellular iron efflux involves ferroportin, the only known iron exporter. In addition to duodenal enterocytes, ferroportin is present in other cell types, and exports iron into plasma. Ferroportin was recently discovered to be regulated by the expression of the circulating hormone hepcidin, a small peptide synthesized in hepatocytes. These recent studies on the role of hepcidin in the regulation of dietary, cellular, and extracellular iron have led to a better understanding of the pathways by which iron balance in humans is influenced, especially its involvement in human genetic diseases of iron overload. Other important molecular pathways include iron binding to transferrin in the bloodstream for cellular delivery through the plasma membrane transferrin receptor (TfR1). In the cytosol, iron regulatory proteins 1 and 2 (IRP1 and IRP2) play a prominent role in sensing the presence of iron in order to posttranscriptionally regulate the expression of TfR1 and ferritin, two important participants in iron metabolism. From a toxicological standpoint, posttranscriptional regulation of these genes aids in the sequestration, control, and hence prevention of cytotoxic effects from free-floating nontransferrin-bound iron. Given the importance of dietary iron in normal physiology, its potential to induce chronic toxicity, and recent discoveries in the regulation of human iron metabolism by hepcidin, this review will address the regulatory mechanisms of normal iron metabolism in mammals with emphasis on dietary exposure. It is the goal of this review that this information may provide in a concise format our current understanding of major pathways and mechanisms involved in mammalian iron metabolism, which is a basis for control of iron toxicity. Such a discussion is intended to facilitate the identification of deficiencies so that future metabolic or toxicological studies may be appropriately focused. A better knowledge of iron metabolism from normal to pathophysiological conditions will ultimately broaden the spectrum of the usefulness of this information in biomedical and toxicological sciences for improving and protecting human health.
Collapse
Affiliation(s)
- Luis G Valerio
- U.S. Food and Drug Administration, Center for Food Safety and Applied Nutrition,Office of Food Additive Safety, Division of Biotechnology and GRAS Notice Review, College Park, MD, 20470, USA
| |
Collapse
|
39
|
Zhu B, Huang L, Huang HQ. Cloning analysis of ferritin and the cisplatin-subunit for cancer cell apoptosis in Aplysia juliana hepatopancreas. Comp Biochem Physiol C Toxicol Pharmacol 2012; 156:95-103. [PMID: 22579997 DOI: 10.1016/j.cbpc.2012.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 04/03/2012] [Accepted: 04/25/2012] [Indexed: 11/27/2022]
Abstract
Ferritin, an iron storage protein, plays a key role in iron metabolism in vivo. Here, we have cloned an inducible ferritin cDNA with 519 bp within the open reading frame fragment from the hepatopancreas of Aplysia juliana (AJ). The subunit sequence of the ferritin was predicted to be a polypeptide of 172 amino acids with a molecular mass of 19.8291kDa and an isoelectric point of 5.01. The cDNA sequence of hepatopancreas ferritin in AJ was constructed into a pET-32a system for expressing its relative protein efficiently in E. coli strain BL21, under isopropyl-β-d-thiogalactoside induction. The recombinant ferritin, which was further purified on a Ni-NTA resin column and digested with enterokinase, was detected as a single subunit of approximately 20 kDa mass using both SDS-PAGE and mass spectrometry. The secondary structure and phosphorylation sites of the deduced amino acids were predicted using both ExPASy proteomic tools and the NetPhos 2.0 server, and the subunit space structure of the recombinant AJ ferritin (rAjFer) was built using a molecular operating environment software system. The result of in-gel digestion and identification using MALDI-TOF MS/MS showed that the recombinant protein was AjFer. ICP-MS results indicated that the rAjFer subunit could directly bind to cisplatin[cis-Diaminedichloroplatinum(CDDP)], giving approximately 17.6 CDDP/ferritin subunits and forming a novel CDDP-subunit. This suggests that a nanometer CDDP core-ferritin was constructed, which could be developed as a new anti-cancer drug. The flow cytometry results indicated that CDDP-rAjFer could induce Hela cell apoptosis. Results of the real-time PCR and Western blotting showed that the expression of AjFer mRNA was up-regulated in AJ under Cd(2+) stress. The recombinant AjFer protein should prove to be useful for further study of the structure and function of ferritin in Aplysia.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antineoplastic Agents/pharmacology
- Aplysia/drug effects
- Aplysia/genetics
- Aplysia/metabolism
- Apoptosis
- Base Sequence
- Cadmium/pharmacology
- Cell Proliferation/drug effects
- Cisplatin/metabolism
- Cisplatin/pharmacology
- Cloning, Molecular
- DNA, Complementary/genetics
- DNA, Complementary/metabolism
- Drug Screening Assays, Antitumor
- Electrophoresis, Polyacrylamide Gel
- Escherichia coli/genetics
- Escherichia coli/metabolism
- Ferritins/classification
- Ferritins/genetics
- Ferritins/metabolism
- Ferritins/pharmacology
- Flow Cytometry
- Gene Expression Regulation
- HeLa Cells
- Hepatopancreas/drug effects
- Hepatopancreas/metabolism
- Humans
- Isoelectric Point
- Isopropyl Thiogalactoside/metabolism
- Molecular Sequence Data
- Molecular Weight
- Open Reading Frames
- Phosphorylation
- Phylogeny
- Protein Binding
- Protein Structure, Secondary
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Recombinant Fusion Proteins/pharmacology
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
Collapse
Affiliation(s)
- Bo Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, China
| | | | | |
Collapse
|
40
|
Ferritin in adult-onset still's disease: just a useful innocent bystander? Int J Inflam 2012; 2012:298405. [PMID: 22536541 PMCID: PMC3321299 DOI: 10.1155/2012/298405] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 01/16/2012] [Indexed: 12/11/2022] Open
Abstract
Background. Adult-Onset Still's Disease (AOSD) is an immune-mediated systemic disease with quotidian-spiking fever, rash, and inflammatory arthritis. Hyperferritinemia is a prominent feature, often used for screening. Methods. The key terms “ferritin” and “hyperferritinemia” were used to search PubMed and Medline and were cross-referenced with “Still's Disease.” Results. Hyperferritinemia, although nonspecific, is particularly prevalent in AOSD. While most clinicians associate ferritin with iron metabolism, this is mostly true for the H isoform and not for the L isoform that tends to increase dramatically in hyperferritenemia. In these situations, hyperferritinemia is not associated with iron metabolism and may even mask an underlying iron deficiency. We review, in systematic fashion, the current basic science and clinical literature regarding the regulation of ferritin and its use in the diagnosis and management of AOSD. Conclusion. Serum hyperferritinemia in AOSD has been described for 2 decades, although its mechanism has not yet been completely elucidated. Regulation by proinflammatory cytokines such as interleukin (IL)-1b, IL-6, IL-18, MCSF, and INF-α provides a link to the disease pathogenesis and may explain rapid resolution of hyperferritinemia after targeted treatment and inhibition of key cytokines.
Collapse
|
41
|
Li J, Li L, Zhang S, Li J, Zhang G. Three ferritin subunits involved in immune defense from bay scallop Argopecten irradians. FISH & SHELLFISH IMMUNOLOGY 2012; 32:368-372. [PMID: 22146701 DOI: 10.1016/j.fsi.2011.10.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Revised: 10/24/2011] [Accepted: 10/25/2011] [Indexed: 05/31/2023]
Abstract
Ferritin is a ubiquitous protein that plays an important role in iron storage and iron-withholding strategy of innate immunity. In this study, three genes encoding different ferritin subunits were cloned from bay scallop Argopecten irradians (AiFer1, AiFer2 and AiFer3) by rapid amplification of cDNA ends (RACE) approaches based on the known ESTs. The open reading frames of the three ferritins are of 516 bp, 522 bp and 519 bp, encoding 171,173 and 172 amino acids, respectively. All the AiFers contain a putative Iron Regulatory Element (IRE) in their 5'-untranslated regions. The deduced amino acid sequences of AiFers possess both the ferroxidase center of mammalian H ferritin and the iron nucleation site of mammalian L ferritin. Gene structure study revealed two distinct structured genes encoding a ferritin subunit (AiFer3). Quantitative real-time PCR analysis indicated the significant up-regulation of AiFers in hemocytes after challenged with Listonella anguillarum, though the magnitudes of AiFer1 and AiFer2 were much higher than that of AiFer3. Taken together, these results suggest that AiFers are likely to play roles in both iron storage and innate immune defense against microbial infections.
Collapse
Affiliation(s)
- Juan Li
- Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Rd., Qingdao 266071, China
| | | | | | | | | |
Collapse
|
42
|
Kowdley KV, Belt P, Wilson LA, Yeh MM, Neuschwander-Tetri BA, Chalasani N, Sanyal AJ, Nelson JE. Serum ferritin is an independent predictor of histologic severity and advanced fibrosis in patients with nonalcoholic fatty liver disease. Hepatology 2012; 55:77-85. [PMID: 21953442 PMCID: PMC3245347 DOI: 10.1002/hep.24706] [Citation(s) in RCA: 394] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 09/26/2011] [Indexed: 12/11/2022]
Abstract
UNLABELLED Serum ferritin (SF) levels are commonly elevated in patients with nonalcoholic fatty liver disease (NAFLD) because of systemic inflammation, increased iron stores, or both. The aim of this study was to examine the relationship between elevated SF and NAFLD severity. Demographic, clinical, histologic, laboratory, and anthropometric data were analyzed in 628 adult patients with NAFLD (age, ≥ 18 years) with biopsy-proven NAFLD and an SF measurement within 6 months of their liver biopsy. A threshold SF >1.5 × upper limit of normal (ULN) (i.e., >300 ng/mL in women and >450 ng/mL in men) was significantly associated with male sex, elevated serum alanine aminotransferase, aspartate aminotransferase, iron, transferrin-iron saturation, iron stain grade, and decreased platelets (P < 0.01). Histologic features of NAFLD were more severe among patients with SF >1.5 × ULN, including steatosis, fibrosis, hepatocellular ballooning, and diagnosis of NASH (P < 0.026). On multiple regression analysis, SF >1.5 × ULN was independently associated with advanced hepatic fibrosis (odds ratio [OR], 1.66; 95% confidence interval [CI], 1.05-2.62; P = 0.028) and increased NAFLD Activity Score (NAS) (OR, 1.99; 95% CI, 1.06-3.75; P = 0.033). CONCLUSIONS A SF >1.5 × ULN is associated with hepatic iron deposition, a diagnosis of NASH, and worsened histologic activity and is an independent predictor of advanced hepatic fibrosis among patients with NAFLD. Furthermore, elevated SF is independently associated with higher NAS, even among patients without hepatic iron deposition. We conclude that SF is useful to identify NAFLD patients at risk for NASH and advanced fibrosis.
Collapse
Affiliation(s)
- Kris V. Kowdley
- Center for Liver Disease, Digestive Disease Institute, Virginia Mason Medical Center, Seattle, WA,Benaroya Research Institute at Virginia Mason Medical Center, Seattle, WA
| | - Patricia Belt
- Johns Hopkins University School of Public Health, Baltimore, MD
| | - Laura A. Wilson
- Johns Hopkins University School of Public Health, Baltimore, MD
| | | | | | | | | | - James E. Nelson
- Benaroya Research Institute at Virginia Mason Medical Center, Seattle, WA
| | | |
Collapse
|
43
|
Zhu B, Lin Q, Ke CH, Huang HQ. Single subunit type of ferritin from visceral mass of Saccostrea cucullata: cloning, expression and cisplatin-subunit analysis. FISH & SHELLFISH IMMUNOLOGY 2011; 31:453-461. [PMID: 21729755 DOI: 10.1016/j.fsi.2011.06.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 06/13/2011] [Accepted: 06/20/2011] [Indexed: 05/31/2023]
Abstract
Ferritin, the iron storage protein, plays a key role in iron metabolism. Here, we have cloned an inducible ferritin cDNA with 516 bp within the open reading frame fragment from the visceral mass of Saccostrea cucullata. The subunit sequence of the ferritin was predicted to be a polypeptide of 171 amino acids with a molecular weight (MW) of 19.9182 kDa and an isoelectric point of 5.24. The cDNA sequence of S. cucullata ferritin was constructed into a pET-32a expression system for expressing its relative protein efficiently in the Escherichia coli BL21 strain under isopropyl-β-D-thiogalactoside (IPTG) induction. The recombinant ferritin, which was further purified on a Ni-NTA resin column and digested with enterokinase, was detected as a single subunit of approximately MW 20 kDa using both SDS-PAGE and mass spectrometry. S. cucullata ferritin (ScFer) showed 98% identity with Crassostrea gigas ferritin at the amino acid level. The secondary structure and phosphorylation sites of deduced amino acids were predicted with ExPASy proteomics tools and the NetPhos 2.0 server, respectively, and the subunit space structure of recombinant S. cucullata ferritin (rScFer) was built using the molecular operating environmental software system. The results of both in-gel digestion and identification using MALDI-TOF MS/MS showed that the recombinant protein was ScFer. ICP-MS indicated that rScFer subunit can directly bind to cisplatin[cis-Diaminedichloroplatinum(CDDP)], giving approximately 22.9 CDDP/ferritin subunit for forming a novel complex of CDDP-subunit, which suggests that it constructs a nanometer CDDP core-ferritin for developing a new drug of anti-cancer. The results of both the real-time PCR and Western blotting showed that the expression of ScFer mRNA was up-regulated in the oyster under the stress of Cd(2+). In addition, the expression increment of ScFer mRNA under bacterial challenge indicated that ferritin participated in the immune response of S. cucullata. The recombinant ScFer should prove to be useful for further study of the structure and function of ferritin in S. cucullata.
Collapse
Affiliation(s)
- Bo Zhu
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | | | | | | |
Collapse
|
44
|
Regulation of neuronal ferritin heavy chain, a new player in opiate-induced chemokine dysfunction. J Neuroimmune Pharmacol 2011; 6:466-76. [PMID: 21465240 DOI: 10.1007/s11481-011-9278-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Accepted: 03/21/2011] [Indexed: 12/19/2022]
Abstract
The heavy chain subunit of ferritin (FHC), a ubiquitous protein best known for its iron-sequestering activity as part of the ferritin complex, has recently been described as a novel inhibitor of signaling through the chemokine receptor CXCR4. Levels of FHC as well as its effects on CXCR4 activation increase in cortical neurons exposed to mu-opioid receptor agonists such as morphine, an effect likely specific to neurons. Major actions of CXCR4 signaling in the mature brain include a promotion of neurogenesis, activation of pro-survival signals, and modulation of excitotoxic pathways; thus, FHC up-regulation may contribute to the neuronal dysfunction often associated with opiate drug abuse. This review summarizes our knowledge of neuronal CXCR4 function, its regulation by opiates and the role of FHC in this process, and known mechanisms controlling FHC production. We speculate on the mechanism involved in FHC regulation by opiates and offer FHC as a new target in opioid-induced neuropathology.
Collapse
|
45
|
The many faces of the octahedral ferritin protein. Biometals 2011; 24:489-500. [PMID: 21267633 DOI: 10.1007/s10534-011-9415-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Accepted: 01/13/2011] [Indexed: 12/14/2022]
Abstract
Iron is an essential trace nutrient required for the active sites of many enzymes, electron transfer and oxygen transport proteins. In contrast, to its important biological roles, iron is a catalyst for reactive oxygen species (ROS). Organisms must acquire iron but must protect against oxidative damage. Biology has evolved siderophores, hormones, membrane transporters, and iron transport and storage proteins to acquire sufficient iron but maintain iron levels at safe concentrations that prevent iron from catalyzing the formation of ROS. Ferritin is an important hub for iron metabolism because it sequesters iron during times of iron excess and releases iron during iron paucity. Ferritin is expressed in response to oxidative stress and is secreted into the extracellular matrix and into the serum. The iron sequestering ability of ferritin is believed to be the source of the anti-oxidant properties of ferritin. In fact, ferritin has been used as a biomarker for disease because it is synthesized in response to oxidative damage and inflammation. The function of serum ferritin is poorly understood, however serum ferritin concentrations seem to correlate with total iron stores. Under certain conditions, ferritin is also associated with pro-oxidant activity. The source of this switch from anti-oxidant to pro-oxidant has not been established but may be associated with unregulated iron release from ferritin. Recent reports demonstrate that ferritin is involved in other aspects of biology such as cell activation, development, immunity and angiogenesis. This review examines ferritin expression and secretion in correlation with anti-oxidant activity and with respect to these new functions. In addition, conditions that lead to pro-oxidant conditions are considered.
Collapse
|
46
|
Hu YH, Zheng WJ, Sun L. Identification and molecular analysis of a ferritin subunit from red drum (Sciaenops ocellatus). FISH & SHELLFISH IMMUNOLOGY 2010; 28:678-686. [PMID: 20064620 DOI: 10.1016/j.fsi.2010.01.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2009] [Revised: 12/23/2009] [Accepted: 01/03/2010] [Indexed: 05/28/2023]
Abstract
Ferritin is a conserved iron binding protein existing ubiquitously in prokaryotes and eukaryotes. In this study, the gene encoding a ferritin M subunit homologue (SoFer1) was cloned from red drum (Sciaenops ocellatus) and analyzed at expression and functional levels. The open reading frame of SoFer1 is 531 bp and preceded by a 5'-untranslated region that contains a putative Iron Regulatory Element (IRE) preserved in many ferritins. The deduced amino acid sequence of SoFer1 possesses both the ferroxidase center of mammalian H ferritin and the iron nucleation site of mammalian L ferritin. Expression of SoFer1 was tissue specific and responded positively to experimental challenges with Gram-positive and Gram-negative fish pathogens. Treatment of red drum liver cells with iron, copper, and oxidant significantly upregulated the expression of SoFer1 in time-dependent manners. To further examine the potential role of SoFer1 in antioxidation, red drum liver cells transfected transiently with SoFer1 were prepared. Compared to control cells, SoFer1 transfectants exhibited reduced production of reactive oxygen species following H(2)O(2) challenge. Finally, to examine the iron binding potential of SoFer1, SoFer1 was expressed in and purified from Escherichia coli as a recombinant protein. Iron-chelating analysis showed that purified recombinant SoFer1 was capable of iron binding. Taken together, these results suggest that SoFer1 is likely to be a functional ferritin involved in iron sequestration, host immune defence against bacterial infection, and antioxidation.
Collapse
Affiliation(s)
- Yong-hua Hu
- Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, PR China
| | | | | |
Collapse
|
47
|
Weiss G. Iron metabolism in the anemia of chronic disease. Biochim Biophys Acta Gen Subj 2009; 1790:682-93. [DOI: 10.1016/j.bbagen.2008.08.006] [Citation(s) in RCA: 192] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2008] [Revised: 07/27/2008] [Accepted: 08/14/2008] [Indexed: 02/08/2023]
|
48
|
Jeong SY, Rathore KI, Schulz K, Ponka P, Arosio P, David S. Dysregulation of iron homeostasis in the CNS contributes to disease progression in a mouse model of amyotrophic lateral sclerosis. J Neurosci 2009; 29:610-9. [PMID: 19158288 PMCID: PMC6665152 DOI: 10.1523/jneurosci.5443-08.2009] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 12/07/2008] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS), characterized by degeneration of spinal motor neurons, consists of sporadic and familial forms. One cause of familial ALS is missense mutations in the superoxide dismutase 1 (SOD1) gene. Iron accumulation occurs in the CNS of both forms of ALS; however, its contribution to the pathogenesis of ALS is not known. We examined the role of iron in a transgenic mouse line overexpressing the human SOD1(G37R) mutant. We show that multiple mechanisms may underlie the iron accumulation in neurons and glia in SOD1(G37R) transgenic mice. These include dysregulation of proteins involved in iron influx and sensing of intracellular iron; iron accumulation in ventral motor neurons secondary to blockage of anterograde axonal transport; and increased mitochondrial iron load in neurons and glia. We also show that treatment of SOD1(G37R) mice with an iron chelator extends life span by 5 weeks, accompanied by increased survival of spinal motor neurons and improved locomotor function. These data suggest that iron chelator therapy might be useful for the treatment of ALS.
Collapse
Affiliation(s)
- Suh Young Jeong
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada H3G 1A4
| | - Khizr I. Rathore
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada H3G 1A4
| | - Katrin Schulz
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada H3G 1A4
| | - Prem Ponka
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada H3T 1E2, and
| | - Paolo Arosio
- Dipartimento Materno Infantile e Tecnologie Biomediche, Università di Brescia, 25123 Brescia, Italy
| | - Samuel David
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada H3G 1A4
| |
Collapse
|
49
|
Banerjee A, Russell WK, Jayaraman A, Ramaiah SK. Identification of proteins to predict the molecular basis for the observed gender susceptibility in a rat model of alcoholic steatohepatitis by 2-D gel proteomics. Proteomics 2009; 8:4327-37. [PMID: 18924223 DOI: 10.1002/pmic.200700368] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Females are reported to be highly susceptible to alcoholic steatohepatitis (ASH) compared to the males. Although a variety of mechanisms have been proposed to explain this higher sensitivity of females, the precise mechanism is not well understood. The objective of this study was to identify changes in global protein expression in liver tissues of male and female rats with pathologically evident ASH by 2-DE (dimensional electrophoresis). ASH was induced in the SD (Sprague-Dawley) rats by feeding ethanol (EtOH) containing Lieber-DeCarli diet for 6 wk followed by a single injection of lipopolysaccharide (LPS, 10 mg/kg, i.p.). Higher liver injury in females in the ASH group as compared to the males was confirmed by HE stained liver sections. As identified by 2-DE, 22 protein-spots were differentially expressed in the females in the ASH group as compared to the males. Following identification of these proteins by MALDI-MS, they were mainly categorized into metabolism and oxidative stress-related proteins. The expression pattern of a few of these oxidative stress-related proteins like Ferritin Heavy chain (Ferritin-H chain), ER stress protein 60 (ER 60) and Heat-shock protein-60 (HSP 60) were verified by Western blotting. To conclude, the current study has identified a set of proteins that highlights potential novel mechanisms associated with higher liver injury noted in the female rat ASH model.
Collapse
Affiliation(s)
- Atrayee Banerjee
- Department of Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4467, USA
| | | | | | | |
Collapse
|
50
|
Xue Y, Yun D, Esmon A, Zou P, Zuo S, Yu Y, He F, Yang P, Chen X. Proteomic dissection of agonist-specific TLR-mediated inflammatory responses on macrophages at subcellular resolution. J Proteome Res 2008; 7:3180-93. [PMID: 18572962 DOI: 10.1021/pr800021a] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Upon stimulation by distinct bacterial/viral products/agonists, APCs including macrophages tend to express particular TLR molecules to coordinate the signaling that ultimately target at chromatin and mediate the activity of downstream transcriptional factors in regulating characteristic sets of gene expression for innate immune response. To investigate largely unknown regulatory mechanism underlying agonist-specific TLR-mediated innate immune responses, at subcellular resolution, we first analyzed Pam3CSK4-induced proteome changes in living macrophages and identified the differentially expressed proteins in the cytosol and chromatin-associated fractions, respectively, by using AACT/SILAC-based quantitative proteomic approach. In the cytosol fraction, we found that the proteins with notable Pam3CSK4-induced expression changes were primarily involved in post-translational events, energy metabolism, protein transporting, and apoptosis. Among them, a ubiquitous and highly conserved iron-binding protein, Ferritin, was further characterized as a modulator for the expression of a TLR2-specific cytokine IL-10 in murine macrophage cells by using small-interfering RNA (siRNA). Interestingly, we simultaneously identified multiple apoptosis-related proteins showing opposite trend in their regulated expressions, which clearly indicated the existence of systems regulation in differentially modulating the signal for the cross-road balance between protecting cell from apoptosis and the apoptosis of infected cells. For those regulated proteins identified in the nuclear fraction, we integrated bioinformatics to find the interactions of certain chromatin-associated proteins, which suggested their interconnected involvements in proteasome-ubiquitin pathway, DNA replication, and post-translational activity upon Pam3CSK4 stimulation. Certain regulated proteins in our quantitative proteomic data set showed the similar trend of up-regulation in both Pam3CSK4- and LPS-stimulated macrophages (Nature 2007, 447, 972), suggesting their belonging to the recently identified class of pro-inflammatory genes. The regulatory discrepancy between both data sets for other set of genes indicated their agonist-specific nature in innate immune responses.
Collapse
Affiliation(s)
- Yan Xue
- Department of Chemistry and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|