1
|
Parveen S, Fatma M, Mir SS, Dermime S, Uddin S. JAK-STAT Signaling in Autoimmunity and Cancer. Immunotargets Ther 2025; 14:523-554. [PMID: 40376194 PMCID: PMC12080488 DOI: 10.2147/itt.s485670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025] Open
Abstract
The JAK-STAT pathway is an essential cell survival signaling that regulates gene expressions related to inflammation, immunity and cancer. Cytokine receptors, signal transducer and activator of transcription (STAT) proteins, and Janus kinases (JAKs) are the critical component of this signaling cascade. When JAKs are stimulated by cytokines, STAT phosphorylation, dimerization, and nuclear translocation occur, which eventually impacts gene transcription. Dysregulation of JAK-STAT signaling is linked with various autoimmune diseases, including rheumatoid arthritis, psoriasis, and inflammatory bowel disease. This pathway is constitutively activated in human malignancies and leads to tumor cell survival, proliferation, and immune evasion. Oncogenic mutations in the JAK and STAT genes have been found in solid tumors, leukemia, and lymphoma. Targeting the JAK-STAT pathway is a viable and promising therapeutic strategy for the treatment of autoimmune diseases and cancers.
Collapse
Affiliation(s)
- Sana Parveen
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, India
- Molecular Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary Research-4 (ICEIR-4) Integral University, Lucknow, India
| | - Mariyam Fatma
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, India
- Molecular Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary Research-4 (ICEIR-4) Integral University, Lucknow, India
| | - Snober Shabnam Mir
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, India
- Molecular Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary Research-4 (ICEIR-4) Integral University, Lucknow, India
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, 3050, Qatar
- College of Health Sciences, Qatar University, Doha, Qatar
| | - Shahab Uddin
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, India
- Translational Research Institute & Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
2
|
Peng A, Li J, Xing J, Yao Y, Niu X, Zhang K. The function of nicotinamide phosphoribosyl transferase (NAMPT) and its role in diseases. Front Mol Biosci 2024; 11:1480617. [PMID: 39513038 PMCID: PMC11540786 DOI: 10.3389/fmolb.2024.1480617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/11/2024] [Indexed: 11/15/2024] Open
Abstract
Nicotinamide phosphoribosyl transferase (NAMPT) is a rate-limiting enzyme in the mammalian nicotinamide adenine dinucleotide (NAD) salvage pathway, and plays a vital role in the regulation of cell metabolic activity, reprogramming, aging and apoptosis. NAMPT synthesizes nicotinamide mononucleotide (NMN) through enzymatic action, which is a key protein involved in host defense mechanism and plays an important role in metabolic homeostasis and cell survival. NAMPT is involved in NAD metabolism and maintains intracellular NAD levels. Sirtuins (SIRTs) are a family of nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylases (HDACs), the members are capable of sensing cellular NAD+ levels. NAMPT-NAD and SIRT constitute a powerful anti-stress defense system. In this paper, the structure, biological function and correlation with diseases of NAMPT are introduced, aiming to provide new ideas for the targeted therapy of related diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Center Hospital, Taiyuan, China
| |
Collapse
|
3
|
Welz L, Harris DM, Kim NM, Alsaadi AI, Wu Q, Oumari M, Taubenheim J, Volk V, Credido G, Koncina E, Mukherjee PK, Tran F, Sievers LK, Pavlidis P, Powell N, Rieder F, Letellier E, Waschina S, Kaleta C, Feuerhake F, Verstockt B, McReynolds MR, Rosenstiel P, Schreiber S, Aden K. A metabolic constraint in the kynurenine pathway drives mucosal inflammation in IBD. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.08.24311598. [PMID: 39211892 PMCID: PMC11361206 DOI: 10.1101/2024.08.08.24311598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Inflammatory bowel disease (IBD) is associated with perturbed metabolism of the essential amino acid tryptophan (Trp). Whether increased degradation of Trp directly fuels mucosal inflammation or acts as a compensatory attempt to restore cellular energy levels via de-novo nicotinamide adenine dinucleotide (NAD + ) synthesis is not understood. Employing a systems medicine approach on longitudinal IBD therapy intervention cohorts and targeted screening in preclinical IBD models, we discover that steady increases in Trp levels upon therapy success coincide with a rewiring of metabolic processes within the kynurenine pathway (KP). In detail, we identify that Trp catabolism in IBD is metabolically constrained at the level of quinolinate phosphorybosyltransferase (QPRT), leading to accumulation of quinolinic acid (Quin) and a decrease of NAD + . We further demonstrate that Trp degradation along the KP occurs locally in the inflamed intestinal mucosa and critically depends on janus kinase / signal transducers and activators of transcription (JAK/STAT) signalling. Subsequently, knockdown of QPRT in-vitro induces NAD + depletion and a pro-inflammatory state, which can largely be rescued by bypassing QPRT via other NAD + precursors. We hence propose a model of impaired de-novo NAD + synthesis from Trp in IBD. These findings point towards the replenishment of NAD + precursors as a novel therapeutic pathway in IBD.
Collapse
|
4
|
He F, Yu J, Ma S, Zhao W, Zhang M, Wang J, Zhang C, Wu J, Zhu L. γδT Cells Induce the Inflammatory Response of Human Fibroblast-Like Synoviocytes Directly or by Stimulating B Cells to Activate IL-17/STAT3 Signaling Pathway. Int Arch Allergy Immunol 2024; 185:1154-1165. [PMID: 38991517 DOI: 10.1159/000539703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) combined with hashimoto thyroiditis (HT) is an important cause of various fatal comorbidities of RA. There is no precise conclusion about the cause of this disease. METHODS Peripheral blood and synovial tissue were collected from healthy participants, patients with RA, and patients with both RA and HT. Immunofluorescence staining and Pearson correlation analysis were used to detect the levels of γδTCR and the correlation between IL-17 and p-STAT3, respectively. ELISA, chemiluminescence assays, qRT-PCR and Western blot were performed to detect the levels of IgG, IgM, IFN-γ, IL-1β, TNF-α, Tg-Ab, Tpo-Ab, IL-17, IL-2, p-SATA3, and STAT3, respectively. RESULTS There was increased proportion of γδT cells, IL-17, and p-STAT3 levels in RA and HT patients. IL-17 was positively correlated with p-STAT3. γδT cells significantly promoted the expression of IgG, Tg-Ab, Tpo-Ab, and IL-17. When γδT and human fibroblast-like synoviocytes (FLSs) were co-cultured, the levels of IL-2, IFN-γ, IL-1β, TNF-α, and IL-17 were increased, and the IL-17/STAT3 signaling pathway was activated. When IL-17-silenced γδT cells and STAT3-silenced FLSs were co-cultured, the levels of IL-1β and TNF-α in FLSs were significantly decreased. Furthermore, when STAT3-silenced FLSs were added to the co-culture medium of B cells and γδT cells, the levels of IL-1β and TNF-α were also decreased significantly. CONCLUSION γδT cells induced RA directly or by stimulating B cells to activate STAT3 through IL-17.
Collapse
Affiliation(s)
- Fang He
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Juan Yu
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Sha Ma
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Weiqing Zhao
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Mingxing Zhang
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Juan Wang
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Chunpan Zhang
- Department of Pain, The First People's Hospital of Yunnan Province, Kunming, China
| | - Jiangping Wu
- Department of Pain, The First People's Hospital of Yunnan Province, Kunming, China
| | - Lixuan Zhu
- Department of Pain, The First People's Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
5
|
Valle-Mendiola A, Rocha-Zavaleta L, Maldonado-Lagunas V, Morelos-Laguna D, Gutiérrez-Hoya A, Weiss-Steider B, Soto-Cruz I. STAT5 Is Necessary for the Metabolic Switch Induced by IL-2 in Cervical Cancer Cell Line SiHa. Int J Mol Sci 2024; 25:6835. [PMID: 38999946 PMCID: PMC11241652 DOI: 10.3390/ijms25136835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/07/2024] [Accepted: 06/10/2024] [Indexed: 07/14/2024] Open
Abstract
The tumor cells reprogram their metabolism to cover their high bioenergetic demands for maintaining uncontrolled growth. This response can be mediated by cytokines such as IL-2, which binds to its receptor and activates the JAK/STAT pathway. Some reports show a correlation between the JAK/STAT pathway and cellular metabolism, since the constitutive activation of STAT proteins promotes glycolysis through the transcriptional activation of genes related to energetic metabolism. However, the role of STAT proteins in the metabolic switch induced by cytokines in cervical cancer remains poorly understood. In this study, we analyzed the effect of IL-2 on the metabolic switch and the role of STAT5 in this response. Our results show that IL-2 induces cervical cancer cell proliferation and the tyrosine phosphorylation of STAT5. Also, it induces an increase in lactate secretion and the ratio of NAD+/NADH, which suggest a metabolic reprogramming of their metabolism. When STAT5 was silenced, the lactate secretion and the NAD+/NADH ratio decreased. Also, the expression of HIF1α and GLUT1 decreased. These results indicate that STAT5 regulates IL-2-induced cell proliferation and the metabolic shift to aerobic glycolysis by regulating genes related to energy metabolism. Our results suggest that STAT proteins modulate the metabolic switch in cervical cancer cells to attend to their high demand of energy required for cell growth and proliferation.
Collapse
Affiliation(s)
- Arturo Valle-Mendiola
- Laboratorio de Oncología Molecular, Unidad de Investigación en Diferenciación Celular y Cáncer, FES Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de Mayo s/n Col. Ejército de Oriente, Mexico City 09230, Mexico; (A.V.-M.); (D.M.-L.); (A.G.-H.); (B.W.-S.)
| | - Leticia Rocha-Zavaleta
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Vilma Maldonado-Lagunas
- Laboratorio de Epigenética, Instituto Nacional de Medicina Genómica (INMEGEN), Periférico Sur no. 4809, Col. Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico;
| | - Diego Morelos-Laguna
- Laboratorio de Oncología Molecular, Unidad de Investigación en Diferenciación Celular y Cáncer, FES Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de Mayo s/n Col. Ejército de Oriente, Mexico City 09230, Mexico; (A.V.-M.); (D.M.-L.); (A.G.-H.); (B.W.-S.)
| | - Adriana Gutiérrez-Hoya
- Laboratorio de Oncología Molecular, Unidad de Investigación en Diferenciación Celular y Cáncer, FES Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de Mayo s/n Col. Ejército de Oriente, Mexico City 09230, Mexico; (A.V.-M.); (D.M.-L.); (A.G.-H.); (B.W.-S.)
- Cátedra CONAHCYT, FES Zaragoza, Universidad Nacional Autónoma de México, Mexico City 68020, Mexico
| | - Benny Weiss-Steider
- Laboratorio de Oncología Molecular, Unidad de Investigación en Diferenciación Celular y Cáncer, FES Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de Mayo s/n Col. Ejército de Oriente, Mexico City 09230, Mexico; (A.V.-M.); (D.M.-L.); (A.G.-H.); (B.W.-S.)
| | - Isabel Soto-Cruz
- Laboratorio de Oncología Molecular, Unidad de Investigación en Diferenciación Celular y Cáncer, FES Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de Mayo s/n Col. Ejército de Oriente, Mexico City 09230, Mexico; (A.V.-M.); (D.M.-L.); (A.G.-H.); (B.W.-S.)
| |
Collapse
|
6
|
Zhang Q, Basappa J, Wang HY, Nunez-Cruz S, Lobello C, Wang S, Liu X, Chekol S, Guo L, Ziober A, Nejati R, Shestov A, Feldman M, Glickson JD, Turner SD, Blair IA, Van Dang C, Wasik MA. Chimeric kinase ALK induces expression of NAMPT and selectively depends on this metabolic enzyme to sustain its own oncogenic function. Leukemia 2023; 37:2436-2447. [PMID: 37773266 PMCID: PMC11152057 DOI: 10.1038/s41375-023-02038-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 08/31/2023] [Accepted: 09/13/2023] [Indexed: 10/01/2023]
Abstract
As we show in this study, NAMPT, the key rate-limiting enzyme in the salvage pathway, one of the three known pathways involved in NAD synthesis, is selectively over-expressed in anaplastic T-cell lymphoma carrying oncogenic kinase NPM1::ALK (ALK + ALCL). NPM1::ALK induces expression of the NAMPT-encoding gene with STAT3 acting as transcriptional activator of the gene. Inhibition of NAMPT affects ALK + ALCL cells expression of numerous genes, many from the cell-signaling, metabolic, and apoptotic pathways. NAMPT inhibition also functionally impairs the key metabolic and signaling pathways, strikingly including enzymatic activity and, hence, oncogenic function of NPM1::ALK itself. Consequently, NAMPT inhibition induces cell death in vitro and suppresses ALK + ALCL tumor growth in vivo. These results indicate that NAMPT is a novel therapeutic target in ALK + ALCL and, possibly, other similar malignancies. Targeting metabolic pathways selectively activated by oncogenic kinases to which malignant cells become "addicted" may become a novel therapeutic approach to cancer, alternative or, more likely, complementary to direct inhibition of the kinase enzymatic domain. This potential therapy to simultaneously inhibit and metabolically "starve" oncogenic kinases may not only lead to higher response rates but also delay, or even prevent, development of drug resistance, frequently seen when kinase inhibitors are used as single agents.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Johnvesly Basappa
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Hong Y Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Selene Nunez-Cruz
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cosimo Lobello
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Shengchun Wang
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Xiaobin Liu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Seble Chekol
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lili Guo
- Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy Ziober
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Reza Nejati
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alex Shestov
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Feldman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jerry D Glickson
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ian A Blair
- Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Chi Van Dang
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Wistar Institute, Philadelphia, PA, USA
| | - Mariusz A Wasik
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Bilski J, Schramm-Luc A, Szczepanik M, Mazur-Biały AI, Bonior J, Luc K, Zawojska K, Szklarczyk J. Adipokines in Rheumatoid Arthritis: Emerging Biomarkers and Therapeutic Targets. Biomedicines 2023; 11:2998. [PMID: 38001998 PMCID: PMC10669400 DOI: 10.3390/biomedicines11112998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease manifested by joint involvement, extra-articular manifestations, and general symptoms. Adipose tissue, previously perceived as an inert energy storage organ, has been recognised as a significant contributor to RA pathophysiology. Adipokines modulate immune responses, inflammation, and metabolic pathways in RA. Although most adipokines have a pro-inflammatory and aggravating effect on RA, some could counteract this pathological process. The coexistence of RA and sarcopenic obesity (SO) has gained attention due to its impact on disease severity and outcomes. Sarcopenic obesity further contributes to the inflammatory milieu and metabolic disturbances. Recent research has highlighted the intricate crosstalk between adipose tissue and skeletal muscle, suggesting potential interactions between these tissues in RA. This review summarizes the roles of adipokines in RA, particularly in inflammation, immune modulation, and joint destruction. In addition, it explores the emerging role of adipomyokines, specifically irisin and myostatin, in the pathogenesis of RA and their potential as therapeutic targets. We discuss the therapeutic implications of targeting adipokines and adipomyokines in RA management and highlight the challenges and future directions for research in this field.
Collapse
Affiliation(s)
- Jan Bilski
- Department of Biomechanics and Kinesiology, Chair of Biomedical Sciences, Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, 31-008 Krakow, Poland; (A.I.M.-B.); (K.Z.)
| | - Agata Schramm-Luc
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Krakow, Poland; (A.S.-L.); (K.L.)
| | - Marian Szczepanik
- Chair of Biomedical Sciences, Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, 31-034 Krakow, Poland;
| | - Agnieszka Irena Mazur-Biały
- Department of Biomechanics and Kinesiology, Chair of Biomedical Sciences, Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, 31-008 Krakow, Poland; (A.I.M.-B.); (K.Z.)
| | - Joanna Bonior
- Department of Medical Physiology, Chair of Biomedical Sciences, Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, 31-126 Krakow, Poland; (J.B.); (J.S.)
| | - Kevin Luc
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Krakow, Poland; (A.S.-L.); (K.L.)
| | - Klaudia Zawojska
- Department of Biomechanics and Kinesiology, Chair of Biomedical Sciences, Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, 31-008 Krakow, Poland; (A.I.M.-B.); (K.Z.)
| | - Joanna Szklarczyk
- Department of Medical Physiology, Chair of Biomedical Sciences, Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, 31-126 Krakow, Poland; (J.B.); (J.S.)
| |
Collapse
|
8
|
Semerena E, Nencioni A, Masternak K. Extracellular nicotinamide phosphoribosyltransferase: role in disease pathophysiology and as a biomarker. Front Immunol 2023; 14:1268756. [PMID: 37915565 PMCID: PMC10616597 DOI: 10.3389/fimmu.2023.1268756] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/03/2023] [Indexed: 11/03/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) plays a central role in mammalian cell metabolism by contributing to nicotinamide adenine dinucleotide biosynthesis. However, NAMPT activity is not limited to the intracellular compartment, as once secreted, the protein accomplishes diverse functions in the extracellular space. Extracellular NAMPT (eNAMPT, also called visfatin or pre-B-cell colony enhancing factor) has been shown to possess adipocytokine, pro-inflammatory, and pro-angiogenic activities. Numerous studies have reported the association between elevated levels of circulating eNAMPT and various inflammatory and metabolic disorders such as obesity, diabetes, atherosclerosis, arthritis, inflammatory bowel disease, lung injury and cancer. In this review, we summarize the current state of knowledge on eNAMPT biology, proposed roles in disease pathogenesis, and its potential as a disease biomarker. We also briefly discuss the emerging therapeutic approaches for eNAMPT inhibition.
Collapse
Affiliation(s)
- Elise Semerena
- Light Chain Bioscience - Novimmune SA, Plan-les-Ouates, Switzerland
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | | |
Collapse
|
9
|
Hanlon MM, McGarry T, Marzaioli V, Amaechi S, Song Q, Nagpal S, Veale DJ, Fearon U. Rheumatoid arthritis macrophages are primed for inflammation and display bioenergetic and functional alterations. Rheumatology (Oxford) 2023; 62:2611-2620. [PMID: 36398893 PMCID: PMC10321118 DOI: 10.1093/rheumatology/keac640] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/28/2022] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVES Myeloid cells with a monocyte/macrophage phenotype are present in large numbers in the RA joint, significantly contributing to disease; however, distinct macrophage functions have yet to be elucidated. This study investigates the metabolic activity of infiltrating polarized macrophages and their impact on pro-inflammatory responses in RA. METHODS CD14+ monocytes from RA and healthy control (HC) bloods were isolated and examined ex vivo or following differentiation into 'M1/M2' macrophages. Inflammatory responses and metabolic analysis ± specific inhibitors were quantified by RT-PCR, western blot, Seahorse XFe technology, phagocytosis assays and transmission electron microscopy along with RNA-sequencing (RNA-seq) transcriptomic analysis. RESULTS Circulating RA monocytes are hyper-inflammatory upon stimulation, with significantly higher expression of key cytokines compared with HC (P < 0.05) a phenotype which is maintained upon differentiation into mature ex vivo polarized macrophages. This induction in pro-inflammatory mechanisms is paralleled by cellular bioenergetic changes. RA macrophages are highly metabolic, with a robust boost in both oxidative phosphorylation and glycolysis in RA along with altered mitochondrial morphology compared with HC. RNA-seq analysis revealed divergent transcriptional variance between pro- and anti-inflammatory RA macrophages, revealing a role for STAT3 and NAMPT in driving macrophage activation states. STAT3 and NAMPT inhibition results in significant decrease in pro-inflammatory gene expression observed in RA macrophages. Interestingly, NAMPT inhibition specifically restores macrophage phagocytic function and results in reciprocal STAT3 inhibition, linking these two signalling pathways. CONCLUSION This study demonstrates a unique inflammatory and metabolic phenotype of RA monocyte-derived macrophages and identifies a key role for NAMPT and STAT3 signalling in regulating this phenotype.
Collapse
Affiliation(s)
- Megan M Hanlon
- Molecular Rheumatology Research Group, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | - Success Amaechi
- Molecular Rheumatology Research Group, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Qingxuan Song
- Immunology and Discovery Sciences, Janssen Research & Development, Philadelphia, PA, USA
| | - Sunil Nagpal
- Immunology and Discovery Sciences, Janssen Research & Development, Philadelphia, PA, USA
| | - Douglas J Veale
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Ursula Fearon
- Correspondence to: Ursula Fearon, Molecular Rheumatology Research Group, School of Medicine, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin D02 R590, Dublin, Ireland. E-mail:
| |
Collapse
|
10
|
Bhayana S, Zhao Y, Merchant M, Cummins T, Dougherty JA, Kamigaki Y, Pathmasiri W, McRitchie S, Mariani LH, Sumner S, Klein JB, Li L, Smoyer WE. Multiomics Analysis of Plasma Proteomics and Metabolomics of Steroid Resistance in Childhood Nephrotic Syndrome Using a "Patient-Specific" Approach. Kidney Int Rep 2023; 8:1239-1254. [PMID: 37284673 PMCID: PMC10239920 DOI: 10.1016/j.ekir.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/20/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction Nephrotic syndrome (NS) occurs commonly in children with glomerular disease and glucocorticoids (GCs) are the mainstay treatment. Steroid resistant NS (SRNS) develops in 15% to 20% of children, increasing the risk of chronic kidney disease compared to steroid sensitive NS (SSNS). NS pathogenesis is unclear in most children, and no biomarkers exist that predict the development of pediatric SRNS. Methods We studied a unique patient cohort with plasma specimens collected before GC treatment, yielding a disease-only sample not confounded by steroid-induced gene expression changes (SSNS n = 8; SRNS n = 7). A novel "patient-specific" bioinformatic approach merged paired pretreatment and posttreatment proteomic and metabolomic data and identified candidate SRNS biomarkers and altered molecular pathways in SRNS versus SSNS. Results Joint pathway analyses revealed perturbations in nicotinate or nicotinamide and butanoate metabolic pathways in patients with SRNS. Patients with SSNS had perturbations of lysine degradation, mucin type O-glycan biosynthesis, and glycolysis or gluconeogenesis pathways. Molecular analyses revealed frequent alteration of molecules within these pathways that had not been observed by separate proteomic and metabolomic studies. We observed upregulation of NAMPT, NMNAT1, and SETMAR in patients with SRNS, in contrast to upregulation of ALDH1B1, ACAT1, AASS, ENPP1, and pyruvate in patients with SSNS. Pyruvate regulation was the change seen in our previous analysis; all other targets were novel. Immunoblotting confirmed increased NAMPT expression in SRNS and increased ALDH1B1 and ACAT1 expression in SSNS, following GC treatment. Conclusion These studies confirmed that a novel "patient-specific" bioinformatic approach can integrate disparate omics datasets and identify candidate SRNS biomarkers not observed by separate proteomic or metabolomic analysis.
Collapse
Affiliation(s)
- Sagar Bhayana
- Center for Clinical and Translational Research, Nationwide Children’s Hospital; Columbus, Ohio, USA
| | - Yue Zhao
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Michael Merchant
- Department of Medicine, Division of Nephrology and Hypertension, University of Louisville; Louisville, Kentucky, USA
| | - Timothy Cummins
- Department of Medicine, Division of Nephrology and Hypertension, University of Louisville; Louisville, Kentucky, USA
| | - Julie A. Dougherty
- Center for Clinical and Translational Research, Nationwide Children’s Hospital; Columbus, Ohio, USA
| | - Yu Kamigaki
- Center for Clinical and Translational Research, Nationwide Children’s Hospital; Columbus, Ohio, USA
| | - Wimal Pathmasiri
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill; Kannapolis, North Carolina, USA
| | - Susan McRitchie
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill; Kannapolis, North Carolina, USA
| | - Laura H. Mariani
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Susan Sumner
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill; Kannapolis, North Carolina, USA
| | - Jon B. Klein
- Department of Medicine, Division of Nephrology and Hypertension, University of Louisville; Louisville, Kentucky, USA
- Robley Rex VA Medical Center, Louisville, Kentucky, USA
| | - Lang Li
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - William E. Smoyer
- Center for Clinical and Translational Research, Nationwide Children’s Hospital; Columbus, Ohio, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
11
|
Gatsiou A, Tual-Chalot S, Napoli M, Ortega-Gomez A, Regen T, Badolia R, Cesarini V, Garcia-Gonzalez C, Chevre R, Ciliberti G, Silvestre-Roig C, Martini M, Hoffmann J, Hamouche R, Visker JR, Diakos N, Wietelmann A, Silvestris DA, Georgiopoulos G, Moshfegh A, Schneider A, Chen W, Guenther S, Backs J, Kwak S, Selzman CH, Stamatelopoulos K, Rose-John S, Trautwein C, Spyridopoulos I, Braun T, Waisman A, Gallo A, Drakos SG, Dimmeler S, Sperandio M, Soehnlein O, Stellos K. The RNA editor ADAR2 promotes immune cell trafficking by enhancing endothelial responses to interleukin-6 during sterile inflammation. Immunity 2023; 56:979-997.e11. [PMID: 37100060 DOI: 10.1016/j.immuni.2023.03.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 01/02/2023] [Accepted: 03/30/2023] [Indexed: 04/28/2023]
Abstract
Immune cell trafficking constitutes a fundamental component of immunological response to tissue injury, but the contribution of intrinsic RNA nucleotide modifications to this response remains elusive. We report that RNA editor ADAR2 exerts a tissue- and stress-specific regulation of endothelial responses to interleukin-6 (IL-6), which tightly controls leukocyte trafficking in IL-6-inflamed and ischemic tissues. Genetic ablation of ADAR2 from vascular endothelial cells diminished myeloid cell rolling and adhesion on vascular walls and reduced immune cell infiltration within ischemic tissues. ADAR2 was required in the endothelium for the expression of the IL-6 receptor subunit, IL-6 signal transducer (IL6ST; gp130), and subsequently, for IL-6 trans-signaling responses. ADAR2-induced adenosine-to-inosine RNA editing suppressed the Drosha-dependent primary microRNA processing, thereby overwriting the default endothelial transcriptional program to safeguard gp130 expression. This work demonstrates a role for ADAR2 epitranscriptional activity as a checkpoint in IL-6 trans-signaling and immune cell trafficking to sites of tissue injury.
Collapse
Affiliation(s)
- Aikaterini Gatsiou
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; RNA Metabolism and Vascular Inflammation Laboratory, Institute of Cardiovascular Regeneration and Department of Cardiology, JW Goethe University Frankfurt, Frankfurt am Main, Germany.
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Matteo Napoli
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Almudena Ortega-Gomez
- Institute for Cardiovascular Prevention (IPEK), LMU Munich Hospital, Munich, Germany
| | - Tommy Regen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Rachit Badolia
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Valeriana Cesarini
- Department of Pediatric Hematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Raphael Chevre
- Institute for Cardiovascular Prevention (IPEK), LMU Munich Hospital, Munich, Germany; Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation, WWU Muenster, Muenster, Germany
| | - Giorgia Ciliberti
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
| | - Carlos Silvestre-Roig
- Institute for Cardiovascular Prevention (IPEK), LMU Munich Hospital, Munich, Germany; Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation, WWU Muenster, Muenster, Germany
| | - Maurizio Martini
- Fondazione Policlinico Universitario "A. Gemelli," IRCCS, UOC Anatomia Patologica, Rome, Italy; Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Jedrzej Hoffmann
- Department of Cardiology, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Rana Hamouche
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Joseph R Visker
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Nikolaos Diakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Astrid Wietelmann
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Domenico Alessandro Silvestris
- Department of Pediatric Hematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece; Translational Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ali Moshfegh
- Kancera AB, Stockholm, Sweden; Department of Oncology and Pathology at Karolinska Institutet, Stockholm, Sweden
| | - Andre Schneider
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Wei Chen
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China; Medi-X Institute, SUSTech Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Stefan Guenther
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Johannes Backs
- Institute of Experimental Cardiology, University Hospital Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Heidelberg and Mannheim, Germany
| | - Shin Kwak
- Department of Molecular Neuropathogenesis, Tokyo Medical University, Tokyo, Japan
| | - Craig H Selzman
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA; Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece; Translational Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Ioakim Spyridopoulos
- Translational Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; Department of Cardiology, Freeman Hospital, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Thomas Braun
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Angela Gallo
- Department of Pediatric Hematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Stavros G Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA; Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, JW Goethe University Frankfurt, Frankfurt am Main, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Frankfurt Partner Site, Germany
| | - Markus Sperandio
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), LMU Munich Hospital, Munich, Germany; Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation, WWU Muenster, Muenster, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany; Department of Physiology and Pharmacology (FyFa), Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; RNA Metabolism and Vascular Inflammation Laboratory, Institute of Cardiovascular Regeneration and Department of Cardiology, JW Goethe University Frankfurt, Frankfurt am Main, Germany; Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Heidelberg and Mannheim, Germany; Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany.
| |
Collapse
|
12
|
Helman T, Braidy N. Importance of NAD+ Anabolism in Metabolic, Cardiovascular and Neurodegenerative Disorders. Drugs Aging 2023; 40:33-48. [PMID: 36510042 DOI: 10.1007/s40266-022-00989-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2022] [Indexed: 12/14/2022]
Abstract
The role of nicotinamide adenine dinucleotide (NAD+) in ageing has emerged as a critical factor in understanding links to a wide range of chronic diseases. Depletion of NAD+, a central redox cofactor and substrate of numerous metabolic enzymes, has been detected in many major age-related diseases. However, the mechanisms behind age-associated NAD+ decline remains poorly understood. Despite limited conclusive evidence, supplements aimed at increasing NAD+ levels are becoming increasingly popular. This review provides renewed insights regarding the clinical utility and benefits of NAD+ precursors, namely nicotinamide (NAM), nicotinic acid (NA), nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN), in attenuating NAD+ decline and phenotypic characterization of age-related disorders, including metabolic, cardiovascular and neurodegenerative diseases. While it is anticipated that NAD+ precursors can play beneficial protective roles in several conditions, they vary in their ability to promote NAD+ anabolism with differing adverse effects. Careful evaluation of the role of NAD+, whether friend or foe in ageing, should be considered.
Collapse
Affiliation(s)
- Tessa Helman
- Centre for Healthy Brain Ageing, School of Psychiatry, NPI, Euroa Centre, Prince of Wales Hospital, University of New South Wales, Barker Street, Randwick, Sydney, NSW, 2031, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, NPI, Euroa Centre, Prince of Wales Hospital, University of New South Wales, Barker Street, Randwick, Sydney, NSW, 2031, Australia.
| |
Collapse
|
13
|
Hong Z, Tie Q, Zhang L. Targeted inhibition of the GRK2/HIF-1α pathway is an effective strategy to alleviate synovial hypoxia and inflammation. Int Immunopharmacol 2022; 113:109271. [PMID: 36461590 DOI: 10.1016/j.intimp.2022.109271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/11/2022]
Abstract
G-protein coupled receptor (GPCR) kinases (GRKs) and hypoxia-inducible factor-1α (HIF-1α) play key roles in rheumatoid arthritis (RA). Several studies have demonstrated that HIF-1α expression is positively regulated by GRK2, suggesting its posttranscriptional effects on HIF-1α. In this study, we review the role of HIF-1α and GRK2 in RA pathophysiology, focusing on their proinflammatory roles in immune cells and fibroblast-like synoviocytes (FLS).We then introduce several drugs that inhibit GRK2 and HIF-1α, and briefly outline their molecular mechanisms. We conclude by presenting gaps in knowledge and our prospects for the pharmacological potential of targeting these proteins and the relevant downstream signaling pathways.Future research is warranted and paramount for untangling these novel and promising roles for GRK2 and HIF-1α in RA.
Collapse
Affiliation(s)
- Zhongyang Hong
- Department of Pharmacy, Affiliated the Jianhu People's Hospital, Yancheng 224700, China.
| | - Qingsong Tie
- Department of Pharmacy, Affiliated the Jianhu People's Hospital, Yancheng 224700, China.
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
14
|
Hong SM, Lee AY, Hwang SM, Ha YJ, Kim MJ, Min S, Hwang W, Yoon G, Kwon SM, Woo HG, Kim HH, Jeong WI, Shen HM, Im SH, Lee D, Kim YS. NAMPT mitigates colitis severity by supporting redox-sensitive activation of phagocytosis in inflammatory macrophages. Redox Biol 2022; 50:102237. [PMID: 35063804 PMCID: PMC8784331 DOI: 10.1016/j.redox.2022.102237] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 12/02/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme in the nicotinamide adenine dinucleotide (NAD+) salvage pathway and plays a crucial role in the maintenance of the NAD+ pool during inflammation. Considering that macrophages are essential for tissue homeostasis and inflammation, we sought to examine the functional impact of NAMPT in inflammatory macrophages, particularly in the context of inflammatory bowel disease (IBD). In this study, we show that mice with NAMPT deletion within the myeloid compartment (Namptf/fLysMCre+/-, Nampt mKO) have more pronounced colitis with lower survival rates, as well as numerous uncleared apoptotic corpses within the mucosal layer. Nampt-deficient macrophages exhibit reduced phagocytic activity due to insufficient NAD+ abundance, which is required to produce NADPH for the oxidative burst. Nicotinamide mononucleotide (NMN) treatment rescues NADPH levels in Nampt mKO macrophages and sustains superoxide generation via NADPH oxidase. Consequently, Nampt mKO mice fail to clear dead cells during tissue repair, leading to substantially prolonged chronic colitis. Moreover, systemic administration of NMN, to supply NAD+, effectively suppresses the disease severity of DSS-induced colitis. Collectively, our findings suggest that activation of the NAMPT-dependent NAD+ biosynthetic pathway, via NMN administration, is a potential therapeutic strategy for managing inflammatory diseases.
Collapse
Affiliation(s)
- Sun Mi Hong
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - A-Yeon Lee
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Sung-Min Hwang
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Yu-Jin Ha
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Moo-Jin Kim
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Seongki Min
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Won Hwang
- MSBIOTECH. LTD, Chungbuk, 27672, Republic of Korea
| | - Gyesoon Yoon
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - So Mee Kwon
- Department of Physiology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Hyun Goo Woo
- Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Physiology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Hee-Hoon Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Han-Ming Shen
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea; ImmunoBiome, Bio Open Innovation Center, Pohang, 37673, Republic of Korea
| | - Dakeun Lee
- Department of Pathology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea.
| | - You-Sun Kim
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea.
| |
Collapse
|
15
|
Millrine D, Jenkins RH, Hughes STO, Jones SA. Making sense of IL-6 signalling cues in pathophysiology. FEBS Lett 2022; 596:567-588. [PMID: 34618359 PMCID: PMC9673051 DOI: 10.1002/1873-3468.14201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022]
Abstract
Unravelling the molecular mechanisms that account for functional pleiotropy is a major challenge for researchers in cytokine biology. Cytokine-receptor cross-reactivity and shared signalling pathways are considered primary drivers of cytokine pleiotropy. However, reports epitomized by studies of Jak-STAT cytokine signalling identify interesting biochemical and epigenetic determinants of transcription factor regulation that affect the delivery of signal-dependent cytokine responses. Here, a regulatory interplay between STAT transcription factors and their convergence to specific genomic enhancers support the fine-tuning of cytokine responses controlling host immunity, functional identity, and tissue homeostasis and repair. In this review, we provide an overview of the signalling networks that shape the way cells sense and interpret cytokine cues. With an emphasis on the biology of interleukin-6, we highlight the importance of these mechanisms to both physiological processes and pathophysiological outcomes.
Collapse
Affiliation(s)
- David Millrine
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
- Present address:
Medical Research Council Protein Phosphorylation and Ubiquitylation UnitSir James Black CentreSchool of Life SciencesUniversity of Dundee3rd FloorDundeeUK
| | - Robert H. Jenkins
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| | - Stuart T. O. Hughes
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| | - Simon A. Jones
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| |
Collapse
|
16
|
Funk-Hilsdorf TC, Behrens F, Grune J, Simmons S. Dysregulated Immunity in Pulmonary Hypertension: From Companion to Composer. Front Physiol 2022; 13:819145. [PMID: 35250621 PMCID: PMC8891568 DOI: 10.3389/fphys.2022.819145] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/20/2022] [Indexed: 12/26/2022] Open
Abstract
Pulmonary hypertension (PH) represents a grave condition associated with high morbidity and mortality, emphasizing a desperate need for innovative and targeted therapeutic strategies. Cumulative evidence suggests that inflammation and dysregulated immunity interdependently affect maladaptive organ perfusion and congestion as hemodynamic hallmarks of the pathophysiology of PH. The role of altered cellular and humoral immunity in PH gains increasing attention, especially in pulmonary arterial hypertension (PAH), revealing novel mechanistic insights into the underlying immunopathology. Whether these immunophysiological aspects display a universal character and also hold true for other types of PH (e.g., PH associated with left heart disease, PH-LHD), or whether there are unique immunological signatures depending on the underlying cause of disease are points of consideration and discussion. Inflammatory mediators and cellular immune circuits connect the local inflammatory landscape in the lung and heart through inter-organ communication, involving, e.g., the complement system, sphingosine-1-phosphate (S1P), cytokines and subsets of, e.g., monocytes, macrophages, natural killer (NK) cells, dendritic cells (DCs), and T- and B-lymphocytes with distinct and organ-specific pro- and anti-inflammatory functions in homeostasis and disease. Perivascular macrophage expansion and monocyte recruitment have been proposed as key pathogenic drivers of vascular remodeling, the principal pathological mechanism in PAH, pinpointing toward future directions of anti-inflammatory therapeutic strategies. Moreover, different B- and T-effector cells as well as DCs may play an important role in the pathophysiology of PH as an imbalance of T-helper-17-cells (TH17) activated by monocyte-derived DCs, a potentially protective role of regulatory T-cells (Treg) and autoantibody-producing plasma cells occur in diverse PH animal models and human PH. This article highlights novel aspects of the innate and adaptive immunity and their interaction as disease mediators of PH and its specific subtypes, noticeable inflammatory mediators and summarizes therapeutic targets and strategies arising thereby.
Collapse
Affiliation(s)
- Teresa C. Funk-Hilsdorf
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Felix Behrens
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Jana Grune
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Szandor Simmons
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- *Correspondence: Szandor Simmons,
| |
Collapse
|
17
|
Campanelli R, Massa M, Rosti V, Barosi G. New Markers of Disease Progression in Myelofibrosis. Cancers (Basel) 2021; 13:5324. [PMID: 34771488 PMCID: PMC8582535 DOI: 10.3390/cancers13215324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/30/2022] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm due to the clonal proliferation of a hematopoietic stem cell. The vast majority of patients harbor a somatic gain of function mutation either of JAK2 or MPL or CALR genes in their hematopoietic cells, resulting in the activation of the JAK/STAT pathway. Patients display variable clinical and laboratoristic features, including anemia, thrombocytopenia, splenomegaly, thrombotic complications, systemic symptoms, and curtailed survival due to infections, thrombo-hemorrhagic events, or progression to leukemic transformation. New drugs have been developed in the last decade for the treatment of PMF-associated symptoms; however, the only curative option is currently represented by allogeneic hematopoietic cell transplantation, which can only be offered to a small percentage of patients. Disease prognosis is based at diagnosis on the classical International Prognostic Scoring System (IPSS) and Dynamic-IPSS (during disease course), which comprehend clinical parameters; recently, new prognostic scoring systems, including genetic and molecular parameters, have been proposed as meaningful tools for a better patient stratification. Moreover, new biological markers predicting clinical evolution and patient survival have been associated with the disease. This review summarizes basic concepts of PMF pathogenesis, clinics, and therapy, focusing on classical prognostic scoring systems and new biological markers of the disease.
Collapse
Affiliation(s)
- Rita Campanelli
- Center for the Study of Myelofibrosis, General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (V.R.); (G.B.)
| | - Margherita Massa
- General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy;
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (V.R.); (G.B.)
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy; (V.R.); (G.B.)
| |
Collapse
|
18
|
Umar MI, Hassan W, Murtaza G, Buabeid M, Arafa E, Irfan HM, Asmawi MZ, Huang X. The Adipokine Component in the Molecular Regulation of Cancer Cell Survival, Proliferation and Metastasis. Pathol Oncol Res 2021; 27:1609828. [PMID: 34588926 PMCID: PMC8473628 DOI: 10.3389/pore.2021.1609828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/09/2021] [Indexed: 12/22/2022]
Abstract
A hormonal imbalance may disrupt the rigorously monitored cellular microenvironment by hampering the natural homeostatic mechanisms. The most common example of such hormonal glitch could be seen in obesity where the uprise in adipokine levels is in virtue of the expanding bulk of adipose tissue. Such aberrant endocrine signaling disrupts the regulation of cellular fate, rendering the cells to live in a tumor supportive microenvironment. Previously, it was believed that the adipokines support cancer proliferation and metastasis with no direct involvement in neoplastic transformations and tumorigenesis. However, the recent studies have reported discrete mechanisms that establish the direct involvement of adipokine signaling in tumorigenesis. Moreover, the individual adipokine profile of the patients has never been considered in the prognosis and staging of the disease. Hence, the present manuscript has focused on the reported extensive mechanisms that culminate the basis of poor prognosis and diminished survival rate in obese cancer patients.
Collapse
Affiliation(s)
| | - Waseem Hassan
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Manal Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Elshaimaa Arafa
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | | | - Mohd Zaini Asmawi
- School of Pharmaceutical Sciences, University of Science Malaysia, Pulau Pinang, Malaysia
| | - Xianju Huang
- College of Pharmacy, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
19
|
Coutinho A, Reddy N, Chatterjee A, Khan MI. The Role of Visfatin (Adipocytokine) Biomarker in Oral Health and Diseases among Nonobese Indian Population: A Proteomic Assay. Glob Med Genet 2021; 8:104-108. [PMID: 34430962 PMCID: PMC8378923 DOI: 10.1055/s-0041-1728690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Visfatin is an adipocytokine and a potential biomarker encoded by the nicotinamide phosphoribosyltransferase gene. It belongs to the nicotinic acid phosphoribosyltransferase family and involved in various metabolic processes and aging. The aim of this study was to evaluate the role of visfatin biomarker in oral diseases like periodontitis. A total of 60 patients (20–50 years) were included in this study, and they were divided into three groups. Group I consisted of 20 subjects with healthy periodontium, group II consisted of 20 subjects with generalized moderate gingivitis, and group III consisted of 20 subjects with generalized periodontitis. The clinical periodontal parameters, including plaque index, gingival index, probing pocket depth, and clinical attachment levels, were recorded, and saliva samples were collected. Salivary visfatin concentrations were assessed using standard enzyme-linked immunosorbent assay. The results of the study showed that the visfatin concentrations were higher in patients with gingivitis and periodontitis compared with those of healthy individuals. Visfatin was found highest in group III (38.22 ± 3.38 ng/mL) followed by group II (26.66 ± 2.24 ng/mL) and the group I (25.60 ± 2.19 ng/mL). Thus, salivary visfatin is a potential inflammatory biomarker and acts as a mediator in the pathogenesis of periodontal disease and, might serve as a diagnostic and therapeutic biomarker in oral diseases like periodontitis.
Collapse
Affiliation(s)
- Amita Coutinho
- Department of Periodontics, The Oxford Dental College, Bangalore, Karnataka, India
| | - Neethu Reddy
- Department of Periodontics, The Oxford Dental College, Bangalore, Karnataka, India
| | - Anirban Chatterjee
- Department of Periodontics, The Oxford Dental College, Bangalore, Karnataka, India
| | - Mahamad Irfanulla Khan
- Department of Orthodontics and Dentofacial Orthopedics, The Oxford Dental College, Bangalore, Karnataka, India
| |
Collapse
|
20
|
Enhanced NAMPT-Mediated NAD Salvage Pathway Contributes to Psoriasis Pathogenesis by Amplifying Epithelial Auto-Inflammatory Circuits. Int J Mol Sci 2021; 22:ijms22136860. [PMID: 34202251 PMCID: PMC8267663 DOI: 10.3390/ijms22136860] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/21/2021] [Indexed: 01/06/2023] Open
Abstract
Dysregulated cross-talk between immune cells and epithelial compartments is responsible for the onset and amplification of pathogenic auto-inflammatory circuits occurring in psoriasis. NAMPT-mediated NAD salvage pathway has been recently described as an immunometabolic route having inflammatory function in several disorders, including arthritis and inflammatory bowel diseases. To date, the role of NAD salvage pathway has not been explored in the skin of patients affected by psoriasis. Here, we show that NAD content is enhanced in lesional skin of psoriatic patients and is associated to high NAMPT transcriptional levels. The latter are drastically reduced in psoriatic skin following treatment with the anti-IL-17A biologics secukinumab. We provide evidence that NAMPT-mediated NAD+ metabolism fuels the immune responses executed by resident skin cells in psoriatic skin. In particular, intracellular NAMPT, strongly induced by Th1/Th17-cytokines, acts on keratinocytes by inducing hyper-proliferation and impairing their terminal differentiation. Furthermore, NAMPT-mediated NAD+ boosting synergizes with psoriasis-related cytokines in the upregulation of inflammatory chemokines important for neutrophil and Th1/Th17 cell recruitment. In addition, extracellular NAMPT, abundantly released by keratinocytes and dermal fibroblasts, acts in a paracrine manner on endothelial cells by inducing their proliferation and migration, as well as the expression of ICAM-1 membrane molecule and chemokines important for leukocyte recruitment into inflamed skin. In conclusion, our results showed that NAMPT-mediated NAD salvage pathway contributes to psoriasis pathogenic processes by amplifying epithelial auto-inflammatory responses in psoriasis.
Collapse
|
21
|
Davies R, Williams J, Sime K, Jin HS, Thompson C, Jordan L, Lang D, Halcox JP, Ellins E, Jones GW, Jones SA, Rose-John S, Williams A, Choy E. The role of interleukin-6 trans-signalling on cardiovascular dysfunction in inflammatory arthritis. Rheumatology (Oxford) 2021; 60:2852-2861. [PMID: 33313793 PMCID: PMC8213430 DOI: 10.1093/rheumatology/keaa725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/19/2020] [Indexed: 12/27/2022] Open
Abstract
Objectives Cardiovascular (CV) mortality in RA patients is 50% higher than in the general population. There is increasing recognition that systemic inflammation is a major driver of this. IL-6 is implicated in cardiovascular disease (CVD) in the general population but its role in CVD in RA is undefined. Of the two modes of IL-6 signalling, trans-signalling is pro-inflammatory whereas classical signalling is linked with inflammation resolution. This study examines the role of IL-6 trans-signalling in CVD in a mouse model and patients with RA. Methods Myography determined the effect of IL-6 trans-signalling blockade, using sgp130Fc, on aortic constriction in murine collagen-induced arthritis. Serum CCL2 and sVCAM-1 as soluble biomarkers of sIL-6R trans-signalling were investigated in a human cross-sectional study. An observational longitudinal study investigated the association between these biomarkers and progression of subclinical atherosclerosis in early RA by measuring carotid intima-media thickness (CIMT). Results sgp130Fc reduced arthritis severity, serum CCL2 and sVCAM-1 and restored vascular function in collagen-induced arthritis (CIA). In established RA, sVCAM-1 correlated with the 28-joint DAS (DAS28) and CV risk. In early RA, baseline DAS28 was associated with CIMT change at 6 months. CIMT ‘rapid progressors’ at 12 months had higher baseline sVCAM-1, haemoglobin A1c, cholesterol:high-density lipoprotein cholesterol ratio and LDL cholesterol. Conclusions IL-6 trans-signalling plays a pivotal role in vascular dysfunction in CIA. In early RA, sVCAM-1 was associated with progression of subclinical atherosclerosis. Inflammation from RA onset in CVD-susceptible individuals may accelerate atherosclerosis. IL-6 trans-signalling blockade may be beneficial to RA patients and perhaps for atherosclerosis in the general population.
Collapse
Affiliation(s)
- Ruth Davies
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Jessica Williams
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Katie Sime
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Hyun-Sun Jin
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | | | - Lauren Jordan
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Derek Lang
- Division of Medical Education, Cardiff University School of Medicine, Cardiff, UK
| | | | | | - Gareth W Jones
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, UK
| | - Simon A Jones
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrecht Universität, Kiel, Germany
| | - Anwen Williams
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Ernest Choy
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| |
Collapse
|
22
|
Martínez-Morcillo FJ, Cantón-Sandoval J, Martínez-Menchón T, Corbalán-Vélez R, Mesa-Del-Castillo P, Pérez-Oliva AB, García-Moreno D, Mulero V. Non-canonical roles of NAMPT and PARP in inflammation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 115:103881. [PMID: 33038343 DOI: 10.1016/j.dci.2020.103881] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 06/11/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is the most important hydrogen carrier in cell redox reactions. It is involved in mitochondrial function and metabolism, circadian rhythm, the immune response and inflammation, DNA repair, cell division, protein-protein signaling, chromatin remodeling and epigenetics. Recently, NAD+ has been recognized as the molecule of life, since, by increasing NAD+ levels in old or sick animals, it is possible to improve their health and lengthen their lifespan. In this review, we summarize the contribution of NAD+ metabolism to inflammation, with special emphasis in the major NAD+ biosynthetic enzyme, nicotinamide phosphoribosyl transferase (NAMPT), and the NAD+-consuming enzyme, poly(ADP-ribose) polymerase (PARP). The extracurricular roles of these enzymes, i.e. the proinflammatory role of NAMPT after its release, and the ability of PARP to promote a novel form of cell death, known as parthanatos, upon hyperactivation are revised and discussed in the context of several chronic inflammatory diseases.
Collapse
Affiliation(s)
- Francisco J Martínez-Morcillo
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain
| | - Joaquín Cantón-Sandoval
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain
| | - Teresa Martínez-Menchón
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain; Servicio de Dermatología, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Raúl Corbalán-Vélez
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain; Servicio de Dermatología, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Pablo Mesa-Del-Castillo
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain; Servicio de Reumatología, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Ana B Pérez-Oliva
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain.
| | - Diana García-Moreno
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain.
| | - Victoriano Mulero
- Departmento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain.
| |
Collapse
|
23
|
Navas LE, Carnero A. NAD + metabolism, stemness, the immune response, and cancer. Signal Transduct Target Ther 2021; 6:2. [PMID: 33384409 PMCID: PMC7775471 DOI: 10.1038/s41392-020-00354-w] [Citation(s) in RCA: 273] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/11/2020] [Accepted: 09/27/2020] [Indexed: 02/07/2023] Open
Abstract
NAD+ was discovered during yeast fermentation, and since its discovery, its important roles in redox metabolism, aging, and longevity, the immune system and DNA repair have been highlighted. A deregulation of the NAD+ levels has been associated with metabolic diseases and aging-related diseases, including neurodegeneration, defective immune responses, and cancer. NAD+ acts as a cofactor through its interplay with NADH, playing an essential role in many enzymatic reactions of energy metabolism, such as glycolysis, oxidative phosphorylation, fatty acid oxidation, and the TCA cycle. NAD+ also plays a role in deacetylation by sirtuins and ADP ribosylation during DNA damage/repair by PARP proteins. Finally, different NAD hydrolase proteins also consume NAD+ while converting it into ADP-ribose or its cyclic counterpart. Some of these proteins, such as CD38, seem to be extensively involved in the immune response. Since NAD cannot be taken directly from food, NAD metabolism is essential, and NAMPT is the key enzyme recovering NAD from nicotinamide and generating most of the NAD cellular pools. Because of the complex network of pathways in which NAD+ is essential, the important role of NAD+ and its key generating enzyme, NAMPT, in cancer is understandable. In the present work, we review the role of NAD+ and NAMPT in the ways that they may influence cancer metabolism, the immune system, stemness, aging, and cancer. Finally, we review some ongoing research on therapeutic approaches.
Collapse
Affiliation(s)
- Lola E Navas
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cancer, Sevilla, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain. .,CIBER de Cancer, Sevilla, Spain.
| |
Collapse
|
24
|
Zhu J, Wang J, Huang J, Du W, He Y, Pan H, Luo J. MicroRNA-140-5p regulates the proliferation, apoptosis and inflammation of RA FLSs by repressing STAT3. Exp Ther Med 2020; 21:171. [PMID: 33456538 PMCID: PMC7792473 DOI: 10.3892/etm.2020.9602] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
Ectopic expression of microRNA (miRNA) in rheumatoid arthritis (RA) fibroblast-like synoviocyte (RA FLS) is associated with the development of rheumatoid arthritis. The present study aimed to evaluate the effects of miRNA-140-5p (miR-140) on the properties of RA FLSs. It was found that miR-140 expression was decreased in 33 RA patients and extracted RA FLS samples, when compared to the corresponding healthy controls. Abnormally increased miR-140 expression in RA FLSs attenuated cell proliferation and increased cell apoptosis. Additionally, reduced pro-inflammatory cytokine production was observed in RA FLSs transfected with a miR-140 precursor. Furthermore, the 3'-UTR of the signal transducer and activator of transcription (STAT) 3 gene was identified as a target of miR-140. Notably, restoration of STAT3 expression rescued the regulatory effect of miR-140 on the proliferation, apoptosis and inflammatory cytokine production of RA FLSs. Therefore, the current findings indicated that miR-140 is a crucial modulator of both proliferation and apoptosis, shedding light on the etiology behind RA FLS viability, which is modulated by an interplay between miR-140 and STAT3 in the context of RA.
Collapse
Affiliation(s)
- Jiehua Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianglin Wang
- School of Laboratory Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jialin Huang
- School of Laboratory Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Wensheng Du
- Department of Laboratory Medicine, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yingzhong He
- Department of Laboratory Medicine, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hongfei Pan
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Junmin Luo
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
25
|
Polyakova YV, Zavodovsky BV, Sivordova LE, Akhverdyan YR, Zborovskaya IA. Visfatin and Rheumatoid Arthritis: Pathogenetic Implications and Clinical Utility. Curr Rheumatol Rev 2020; 16:224-239. [DOI: 10.2174/1573397115666190409112621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
Objective:
Analysis and generalization of data related to visfatin involvement in the
pathogenesis of inflammation at various stages of rheumatoid arthritis.
Data Synthesis:
Visfatin is an adipocytokine which has also been identified in non-adipose tissues.
It influences directly on the maturation of B cells, which are involved in autoantibody production
and T cell activation. Visfatin can promote inflammation via regulation of pro-inflammatory cytokines
including TNF, IL-1β and IL-6. The concentration of circulating visfatin in rheumatoid arthritis
patients is higher compared to healthy individuals. Several studies suggest that visfatin level is
associated with rheumatoid arthritis activity, and its elevation may precede clinical signs of the relapse.
In murine collagen-induced arthritis, visfatin levels were also found to be elevated both in
inflamed synovial cells and in joint vasculature. Visfatin blockers have been shown to confer fast
and long-term attenuation of pathological processes; however, most of their effects are transient.
Other factors responsible for hyperactivation of the immune system can participate in this process
at a later stage. Treatment of rheumatoid arthritis with a combination of these blockers and inhibitors
of other mediators of inflammation can potentially improve treatment outcomes compared to
current therapeutic strategies. Recent advances in the treatment of experimental arthritis in mice as
well as the application of emerging treatment strategies obtained from oncology for rheumatoid arthritis
management could be a source of novel adipokine-mediated anti-rheumatic drugs.
Conclusion:
The ongoing surge of interest in anticytokine therapy makes further study of visfatin
highly relevant as it may serve as a base for innovational RA treatment.
Collapse
Affiliation(s)
- Yulia V. Polyakova
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Boris V. Zavodovsky
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Larisa E. Sivordova
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Yuri R. Akhverdyan
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Irina A. Zborovskaya
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| |
Collapse
|
26
|
Han DF, Li Y, Xu HY, Li RH, Zhao D. An Update on the Emerging Role of Visfatin in the Pathogenesis of Osteoarthritis and Pharmacological Intervention. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:8303570. [PMID: 32831881 PMCID: PMC7429770 DOI: 10.1155/2020/8303570] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/10/2020] [Accepted: 06/18/2020] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is one of the most common degenerative joint diseases that affects millions of people worldwide, mainly the aging population. Despite numerous published reports, little is known about the pathology of this disease, and no feasible treatment plan exists to stop OA progression. Recently, extensive basic and clinical studies have shown that adipokines play a key role in OA development. Moreover, some drugs associated with adipokines have shown chondroprotective and anti-inflammatory effects on OA. Visfatin has been shown to play a detrimental role in the progression of OA. It increases the production of matrix metalloproteinases and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS), induces the production of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α, affects the differentiation of mesenchymal stem cells to adipocytes, and induces osteophyte formation by inhibiting osteoclastogenesis. Although some side effects of chemical visfatin inhibitors have been reported, they were shown to be successful in the treatment of diabetes, cancer, and other diseases that can utilize Chinese herbs, further suggesting that similar therapeutic strategies could be used in OA prevention and treatment. Here, we describe the pathophysiological mechanism of visfatin in OA and discuss some potential pharmacological interventions using Chinese herbs.
Collapse
Affiliation(s)
- Dong-Feng Han
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yang Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Hui-Ying Xu
- Department of Ultrasound, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Rong-Hang Li
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ding Zhao
- Department of Orthopedic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
27
|
Park JW, Kim OH, Lee SC, Kim KH, Hong HE, Seo H, Choi HJ, Kim SJ. Serum level of visfatin can reflect the severity of inflammation in patients with acute cholecystitis. Ann Surg Treat Res 2020; 99:26-36. [PMID: 32676479 PMCID: PMC7332317 DOI: 10.4174/astr.2020.99.1.26] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/02/2020] [Accepted: 04/07/2020] [Indexed: 12/29/2022] Open
Abstract
Purpose Visfatin is a key cytokine released from the pe ripheral blood mononuclear cells (PBMCs) as well as adipose tissue, and it is involved in immune response as well as inflammation. In this study, we investigated whether the serum visfatin level could be a prognostic factor for predicting the severity of inflammation in patients with acute cholecystitis. Methods We examined the blood samples and gallbladder specimens from patients who underwent laparoscopic cholecystectomy for either acute (n = 18) or chronic cholecystitis (n = 18). We determined the visfatin levels of these samples using various procedures such as real-time polymerase chain reaction, enzyme-linked immunosorbent assay, western blotting, and immunohistochemistry. Results The patients with acute cholecystitis exhibited higher mRNA expression of visfatin in PBMCs, higher serum levels of visfatin, and increased protein expression of visfatin in the gallbladder specimens than in patients with chronic cholecystitis. In the in vitro model of acute cholecystitis, the mRNA expression of visfatin showed the fastest increase among the other pro-inflammatory mediators studied, including interleukin (IL)-10, tumor necrosis factor-α, IL-6, intracellular adhesion molecule-1, and ascular cell adhesion molecule-1. Inhibition of visfatin using siRNA abrogated the inhibitory effects of lipopolysaccharide (LPS) on the expression of ABCG1 in GBECs, suggesting that visfatin is significantly involved in the LPS-driven suppression of ABCG1. Conclusion Taken together, we concluded that visfatin is a pro-inflammatory mediators that is upregulated during acute cholecystitis and is expected to be increased within a short time after inflammation. Therefore, measuring the serum level of visfatin would be helpful in predicting the inflammatory severity in the patients with acute cholecystitis.
Collapse
Affiliation(s)
- Jae Woo Park
- Department of Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Ok-Hee Kim
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sang Chul Lee
- Department of Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kee-Hwan Kim
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ha-Eun Hong
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Haeyeon Seo
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ho Joong Choi
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Say-June Kim
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
28
|
Audrito V, Messana VG, Deaglio S. NAMPT and NAPRT: Two Metabolic Enzymes With Key Roles in Inflammation. Front Oncol 2020; 10:358. [PMID: 32266141 PMCID: PMC7096376 DOI: 10.3389/fonc.2020.00358] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) and nicotinate phosphoribosyltransferase (NAPRT) are two intracellular enzymes that catalyze the first step in the biosynthesis of NAD from nicotinamide and nicotinic acid, respectively. By fine tuning intracellular NAD levels, they are involved in the regulation/reprogramming of cellular metabolism and in the control of the activity of NAD-dependent enzymes, including sirtuins, PARPs, and NADases. However, during evolution they both acquired novel functions as extracellular endogenous mediators of inflammation. It is well-known that cellular stress and/or damage induce release in the extracellular milieu of endogenous molecules, called alarmins or damage-associated molecular patterns (DAMPs), which modulate immune functions through binding pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs), and activate inflammatory responses. Increasing evidence suggests that extracellular (e)NAMPT and eNAPRT are novel soluble factors with cytokine/adipokine/DAMP-like actions. Elevated eNAMPT were reported in several metabolic and inflammatory disorders, including obesity, diabetes, and cancer, while eNAPRT is emerging as a biomarker of sepsis and septic shock. This review will discuss available data concerning the dual role of this unique family of enzymes.
Collapse
Affiliation(s)
- Valentina Audrito
- Laboratory of Tumor Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Vincenzo Gianluca Messana
- Laboratory of Tumor Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Deaglio
- Laboratory of Tumor Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
29
|
Visfatin and chemerin levels correspond with inflammation and might reflect the bridge between metabolism, inflammation and fibrosis in patients with systemic sclerosis. Postepy Dermatol Alergol 2019; 36:551-565. [PMID: 31839772 PMCID: PMC6906965 DOI: 10.5114/ada.2018.79104] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 08/24/2018] [Indexed: 02/06/2023] Open
Abstract
Introduction Adipokines are regulatory molecules which act as mediators of the inflammatory, fibrotic and metabolic processes by interacting with the immune system. Aim We hypothesized that chemerin and visfatin by pro-inflammatory properties play a significant role in inflammation in systemic sclerosis. To address this hypothesis, we determined serum chemerin and visfatin levels in SSc patients, compared with the control group and defined the correlations with clinical and laboratory parameters in SSc patients. Material and methods The study included 48 Caucasian female patients with SSc and 38 healthy subjects of the control group. Serum concentrations of selected adipokines were measured using commercially available ELISA Kits. Results Patients with SSc had higher chemerin levels (209.38 ±55.35 ng/ml) than the control group (182.71 ±33.94 ng/ml) and the difference was statistically significant (Z = 2.14, p = 0.032). The highest chemerin levels were found in dcSSc patients (242.46 ±95.82 ng/ml). We indicated a positive correlation of chemerin and visfatin with levels of inflammatory markers: CRP (r = 0.35, p = 0.013 for chemerin; r = 0.41, p = 0.003 for visfatin) and ESR (r = 0.31, p = 0.03 for chemerin; r = 0.30, p = 0.03 for visfatin). What is more, chemerin manifested a statistically significant positive correlation with the concentration of complement component C3 (r = 0.47, p = 0.001) and C4 (r = 0.29, p = 0.049), whereas visfatin correlated with C4 levels (r = 0.32, p = 0.029). Conclusions The results of our study indicate that chemerin and visfatin as pro-inflammatory cytokines might represent new markers corresponding with inflammation in systemic sclerosis and might reflect the bridge between metabolism, inflammation and potentially, chemerin may also link inflammation with skin and lung fibrosis.
Collapse
|
30
|
Hormonal regulation of visfatin gene in avian Leghorn male hepatoma (LMH) cells. Comp Biochem Physiol A Mol Integr Physiol 2019; 240:110592. [PMID: 31669171 DOI: 10.1016/j.cbpa.2019.110592] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/07/2019] [Accepted: 10/22/2019] [Indexed: 01/08/2023]
Abstract
Visfain has been extensively studied in mammals and has been shown to play an important role in obesity and insulin resistance. However, there is a paucity of information on visfatin regulation in non-mammalian species. After characterization of chicken visfatin gene, we undertook this study to determine its hormonal regulation in avian (non-mammalian) liver cells. Addition of 5 ng/mL TNFα, 100 ng/mL leptin, 1, 3, 10 or 100 ng/mL T3 for 24 h upregulated visfatin gene expression by 1.2, 1.8, 1.95, 1.75, 1.80, and 2.45 folds (P < .05), respectively, compared to untreated LMH cells. Administration of 10 ng/mL of orexin A significantly down regulated visfatin gene expression by 1.35 folds compared to control cells. In contrast, treatment with IL-6 or orexin B for 24 h did not influence visfatin mRNA abundance. These pro-inflammatory cytokines and obesity-related hormones modulate the expression of CRP, INSIG2, and nuclear orphan receptors. Hepatic CRP gene expression was significantly upregulated by IL-6, TNFα, orexin B, and T3 and down regulated by leptin and orexin A. LXR mRNA abundances were increased by orexin A, decreased by orexin B, and T3, and did not affected by IL6, TNFα, or leptin. The expression of FXR gene was induced by IL-6, leptin, and T3, but it was not influenced by TNFα, orexin A or B. CXR gene expression was up regulated by TNFα, leptin, orexin B, and T3, down regulated by 5 ng/mL orexin A, and did not affected by IL-6. INSIG2 mRNA levels were increased by TNFα (5 ng/mL), leptin (100 ng/mL), and T3 (1, 3, 10, and 100 ng/mL), decreased by orexin A, and remained unchanged with IL-6 or orexin B treatment. Together, this is the first report showing hormonal regulation of visfatin in avian hepatocyte cells and suggesting a potential role of CRP, INSIG2, and nuclear orphan receptor LXR, FXR, and CXR in mediating these hormonal effects.
Collapse
|
31
|
Audrito V, Managò A, Gaudino F, Sorci L, Messana VG, Raffaelli N, Deaglio S. NAD-Biosynthetic and Consuming Enzymes as Central Players of Metabolic Regulation of Innate and Adaptive Immune Responses in Cancer. Front Immunol 2019; 10:1720. [PMID: 31402913 PMCID: PMC6671870 DOI: 10.3389/fimmu.2019.01720] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/09/2019] [Indexed: 12/15/2022] Open
Abstract
Cancer cells, particularly in solid tumors, are surrounded by non-neoplastic elements, including endothelial and stromal cells, as well as cells of immune origin, which can support tumor growth by providing the right conditions. On the other hand, local hypoxia, and lack of nutrients induce tumor cells to reprogram their metabolism in order to survive, proliferate, and disseminate: the same conditions are also responsible for building a tumor-suppressive microenvironment. In addition to tumor cells, it is now well-recognized that metabolic rewiring occurs in all cellular components of the tumor microenvironment, affecting epigenetic regulation of gene expression and influencing differentiation/proliferation decisions of these cells. Nicotinamide adenine dinucleotide (NAD) is an essential co-factor for energy transduction in metabolic processes. It is also a key component of signaling pathways, through the regulation of NAD-consuming enzymes, including sirtuins and PARPs, which can affect DNA plasticity and accessibility. In addition, both NAD-biosynthetic and NAD-consuming enzymes can be present in the extracellular environment, adding a new layer of complexity to the system. In this review we will discuss the role of the “NADome” in the metabolic cross-talk between cancer and infiltrating immune cells, contributing to cancer growth and immune evasion, with an eye to therapeutic implications.
Collapse
Affiliation(s)
- Valentina Audrito
- Department of Medical Sciences, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| | - Antonella Managò
- Department of Medical Sciences, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| | - Federica Gaudino
- Department of Medical Sciences, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| | - Leonardo Sorci
- Division of Bioinformatics and Biochemistry, Department of Materials, Environmental Sciences and Urban Planning, Polytechnic University of Marche, Ancona, Italy
| | - Vincenzo Gianluca Messana
- Department of Medical Sciences, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, Italy.,Italian Institute for Genomic Medicine, Turin, Italy
| |
Collapse
|
32
|
Naguib A, Elsawy N, Aboul-enein F, Hossam N. The relation between serum visfatin levels and cardiovascular involvement in rheumatoid arthritis. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2011.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Abir Naguib
- Department of Physical, Medicine Rheumatology and Rehabilitation Faculty of Medicine Alexandria UniversityEgypt
| | - Noha Elsawy
- Department of Physical, Medicine Rheumatology and Rehabilitation Faculty of Medicine Alexandria UniversityEgypt
| | - Fatma Aboul-enein
- Department of Cardiology and Angiology Faculty of Medicine Alexandria University Egypt
| | - Nermin Hossam
- Department of Clinical Pathology Faculty of Medicine Alexandria University Egypt
| |
Collapse
|
33
|
Franco-Trepat E, Alonso-Pérez A, Guillán-Fresco M, Jorge-Mora A, Gualillo O, Gómez-Reino JJ, Gómez Bahamonde R. Visfatin as a therapeutic target for rheumatoid arthritis. Expert Opin Ther Targets 2019; 23:607-618. [DOI: 10.1080/14728222.2019.1617274] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Eloi Franco-Trepat
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Ana Alonso-Pérez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - María Guillán-Fresco
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Alberto Jorge-Mora
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Oreste Gualillo
- Research laboratory 9 (NEIRID LAB), Institute of Medical Research, SERGAS, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Juan J. Gómez-Reino
- Rheumatology Group, Institute IDIS, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Rodolfo Gómez Bahamonde
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| |
Collapse
|
34
|
Type-II endometrial cancer: role of adipokines. Arch Gynecol Obstet 2019; 300:239-249. [PMID: 31062150 DOI: 10.1007/s00404-019-05181-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 04/24/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Type-II endometrial cancer is an estrogen independent and one of the most lethal types of cancer having poor prognosis. Adipokines play a crucial role in the triggering Type-II EMC. In addition, adipokines modulators, therefore, may have beneficial effects in the treatment of Type-II endometrial cancer, which was clinically evidenced. AREAS COVERED This review presents the role of various adipokines involved and also the suitable modulators to treat Type-II endometrial cancer. CONCLUSION In the present review, we try to discuss the role of individual adipokines in the pathogenesis of Type-II endometrial cancer, and also the possible beneficial effects of adipokines modulator in the treatment of Type-II endometrial cancer.
Collapse
|
35
|
Essential Role of Visfatin in Lipopolysaccharide and Colon Ascendens Stent Peritonitis-Induced Acute Lung Injury. Int J Mol Sci 2019; 20:ijms20071678. [PMID: 30987270 PMCID: PMC6480124 DOI: 10.3390/ijms20071678] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 03/22/2019] [Accepted: 03/27/2019] [Indexed: 12/21/2022] Open
Abstract
Acute lung injury (ALI) is a life-threatening syndrome characterized by acute and severe hypoxemic respiratory failure. Visfatin, which is known as an obesity-related cytokine with pro-inflammatory activities, plays a role in regulation of inflammatory cytokines. The mechanisms of ALI remain unclear in critically ill patients. Survival in ALI patients appear to be influenced by the stress generated by mechanical ventilation and by ALI-associated factors that initiate the inflammatory response. The objective for this study was to understand the mechanisms of how visfatin regulates inflammatory cytokines and promotes ALI. The expression of visfatin was evaluated in ALI patients and mouse sepsis models. Moreover, the underlying mechanisms were investigated using human bronchial epithelial cell lines, BEAS-2B and NL-20. An increase of serum visfatin was discovered in ALI patients compared to normal controls. Results from hematoxylin and eosin (H&E) and immunohistochemistry staining also showed that visfatin protein was upregulated in mouse sepsis models. Moreover, lipopolysaccharide (LPS) induced visfatin expression, activated the STAT3/NFκB pathway, and increased the expression of pro-inflammatory cytokines, including IL1-β, IL-6, and TNF-α in human bronchial epithelial cell lines NL-20 and BEAS-2B. Co-treatment of visfatin inhibitor FK866 reversed the activation of the STAT3/NFκB pathway and the increase of pro-inflammatory cytokines induced by LPS. Our study provides new evidence for the involvement of visfatin and down-stream events in acute lung injury. Further studies are required to confirm whether the anti-visfatin approaches can improve ALI patient survival by alleviating the pro-inflammatory process.
Collapse
|
36
|
Park J, Choi S, Hwang S, Kim S, Choi J, Jung K, Kwon JY, Kong Y, Cho M, Park S. CR6-interacting factor 1 controls autoimmune arthritis by regulation of signal transducer and activator of transcription 3 pathway and T helper type 17 cells. Immunology 2019; 156:413-421. [PMID: 30585643 PMCID: PMC6418438 DOI: 10.1111/imm.13042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 10/31/2018] [Accepted: 12/11/2018] [Indexed: 12/17/2022] Open
Abstract
CR6-interacting factor 1 (CRIF1) is a nuclear protein that interacts with other nuclear factors and androgen receptors, and is implicated in the regulation of cell cycle progression and cell growth. In this study, we examined whether CRIF1 exerts an immunoregulatory effect by modulating the differentiation and function of pathogenic T cells. To this end, the role of CRIF1 in rheumatoid arthritis, a systemic autoimmune disease characterized by hyperplasia of synovial tissue and progressive destruction of articular cartilage structure by pathogenic immune cells [such as T helper type 17 (Th17) cells], was investigated. p3XFLAG-CMV-10-CRIF1 was administered to mice with collagen-induced arthritis 8 days after collagen type II immunization and the disease severity and histologic evaluation, and osteoclastogenesis were assessed. CRIF1 over-expression in mice with collagen-induced arthritis attenuated the clinical and histological signs of inflammatory arthritis. Furthermore, over-expression of CRIF1 in mice with arthritis significantly reduced the number of signal transducer and activator of transcription 3-mediated Th17 cells in the spleen as well as osteoclast differentiation from bone marrow cells. To investigate the impact of loss of CRIF1 in T cells, we generated a conditional CRIF1 gene ablation model using CD4-cre transgenic mice and examined the frequency of Th17 cells and regulatory T cells. Deficiency of CRIF1 in CD4+ cells promoted the production of interleukin-17 and reduced the frequency of regulatory T cells. These results suggest a role for CRIF1 in modulating the activities of Th17 cells and osteoclasts in rheumatoid arthritis.
Collapse
Affiliation(s)
- Jin‐Sil Park
- The Rheumatism Research CenterCatholic Research Institute of Medical ScienceThe Catholic University of KoreaSeoulSouth Korea
| | - Si‐Young Choi
- The Rheumatism Research CenterCatholic Research Institute of Medical ScienceThe Catholic University of KoreaSeoulSouth Korea
| | - Sun‐Hee Hwang
- The Rheumatism Research CenterCatholic Research Institute of Medical ScienceThe Catholic University of KoreaSeoulSouth Korea
| | - Sung‐Min Kim
- The Rheumatism Research CenterCatholic Research Institute of Medical ScienceThe Catholic University of KoreaSeoulSouth Korea
| | - JeongWon Choi
- The Rheumatism Research CenterCatholic Research Institute of Medical ScienceThe Catholic University of KoreaSeoulSouth Korea
| | - Kyung‐Ah Jung
- The Rheumatism Research CenterCatholic Research Institute of Medical ScienceThe Catholic University of KoreaSeoulSouth Korea
| | - Ji Ye Kwon
- The Rheumatism Research CenterCatholic Research Institute of Medical ScienceThe Catholic University of KoreaSeoulSouth Korea
| | - Young‐Yun Kong
- School of Biological SciencesSeoul National UniversitySeoulSouth Korea
| | - Mi‐La Cho
- The Rheumatism Research CenterCatholic Research Institute of Medical ScienceThe Catholic University of KoreaSeoulSouth Korea
- Department of Medical Life ScienceCollege of MedicineThe Catholic University of KoreaSeoulKorea
- Department of Biomedicine & Health Sciences, College of MedicineThe Catholic University of KoreaSeoulKorea
| | - Sung‐Hwan Park
- The Rheumatism Research CenterCatholic Research Institute of Medical ScienceThe Catholic University of KoreaSeoulSouth Korea
- Divison of RheumatologyDepartment of Internal MedicineThe Catholic University of KoreaSeoulSouth Korea
| |
Collapse
|
37
|
Effects of Diet-Induced Obesity on Tracheal Responsiveness to Methacholine, Tracheal Visfatin Level, and Lung Histological Changes in Ovalbumin-Sensitized Female Wistar Rats. Inflammation 2018; 41:846-858. [PMID: 29380115 DOI: 10.1007/s10753-018-0738-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Many studies have shown a close relationship between obesity and asthma severity. In the present study, the effects of diet-induced obesity were examined on airway responsiveness to methacholine in addition to visfatin level in female Wistar rats' tracheae after sensitization with ovalbumin. The rats were divided into four groups: control with normal diet (ND), ovalbumin (OVA)-sensitized with normal diet (S + ND), high-fat diet (HFD), and OVA-sensitized with a high-fat diet (S + HFD). The animals were fed for 8 weeks with standard pelts or high-fat diet and then sensitized and challenged with OVA or saline for another 4 weeks. At the end of the study, the tracheae were isolated and assessed for airway responsiveness and visfatin protein levels. Diet-induced obesity groups developed increased weight and obesity indices (p < 0.001). After sensitization with OVA and diet-induced obesity, there were marked leftward shifts in methacholine concentration-response curves in S + HFD group compared to other groups. Also, maximum response was the highest (p < 0.05 to p < 0.001), EC50 was the lowest (p < 0.05 to p < 0.001), and visfatin protein level was the highest (p < 0.05 to p < 0.01) in S + HFD. According to results, diet-induced obesity caused airway hyperresponsiveness to methacholine and enhanced visfatin protein levels in the tracheae of ovalbumin-sensitized female rats. Our results suggested that, in obese ovalbumin-sensitized conditions in female rats, the local production of adipocytokines, such as visfatin, may be increased, resulting in the deterioration of inflammation in lungs. This finding shows a possible mechanism for the altered phenotype in obesity-ovalbumin sensitization conditions in female rats.
Collapse
|
38
|
Jones SA, Jenkins BJ. Recent insights into targeting the IL-6 cytokine family in inflammatory diseases and cancer. Nat Rev Immunol 2018; 18:773-789. [DOI: 10.1038/s41577-018-0066-7] [Citation(s) in RCA: 721] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Chougule D, Nadkar M, Venkataraman K, Rajadhyaksha A, Hase N, Jamale T, Kini S, Khadilkar P, Anand V, Madkaikar M, Pradhan V. Adipokine interactions promote the pathogenesis of systemic lupus erythematosus. Cytokine 2018; 111:20-27. [PMID: 30098476 DOI: 10.1016/j.cyto.2018.08.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Adipokines are chemical mediators released from adipose tissue involved in regulation of appetite, insulin sensitivity, immune system and inflammatory responses. Adipokines contributes to low grade inflammatory response in autoimmune disease like Systemic Lupus Erythematosus (SLE) but the pathophysiology is yet not clear. The aim of this study is to understand role of adipokine interactions in SLE disease pathogenesis. METHODS Sixty newly diagnosed treatment naïve SLE patients fulfilling the ACR criteria and forty age-sex matched healthy subjects were enrolled in thiscase-control study. Disease activity in SLE patients was evaluated using SELENA-SLEDAI. Array of adipokines, C1q circulating immune complexes (C1q-CIC), anti-C1q, anti-ribososmal P0 (anti-RibP0) and anti-mitochondrial antibodies (AMA) levels were detected by ELISA. Antinuclear antibodies (ANA) and anti-dsDNA autoantibodieswere detected by Indirect Immunofluorescence (IIF), while antigen specificities were detected by Immunoassay blot. Serum levels of C3 and C4 complement factors were assessed by nephlometer. RESULTS Statistically significant elevation in progranulin, adipsin and resistin levels was seen among SLE patients when compared to healthy controls (p < 0.0001). Leptin and omentin levels were significantly reduced in SLE patients (p < 0.0001). There was no statistically significant difference in serum adiponectin, chemerin and visfatin levels when these two groups were compared (p > 0.05). Adiponectin, adipsin and resistin levels were elevated in SLE patients with renal manifestations (p < 0.05). Reduced leptin levels were significantly associated with presence of renal manifestations (p < 0.05). Adiponectin levels positively correlated with disease activity (r = 0.294, p = 0.027) whereas negatively correlated with C3 levels (r = -0.439, p = 0.0007). A positive correlation was observed between hypocomplementemia and leptin levels (p < 0.05). Leptin levels were negatively correlated with disease activity, anti-dsDNA, C1q-CIC and anti-C1q levels (p < 0.05). A significant positive correlation was observed between progranulin levels and anti-ribosomal P0 antibodies (r = 0.499, p < 0.0001). CONCLUSION Adipokines levels and associated clinical manifestations suggest involvement of adipokines in disease pathogenesis of SLE. SLE disease activity and complement components may suggest regulatory effect of adipokines (adiponectin and leptin) on disease pathogenesis. Further studies on adipokines in SLE patients with renal manifestations may propose them as prognostic markers in renal damage. TRIAL REGISTRATION NA.
Collapse
Affiliation(s)
- Durga Chougule
- Department of Clinical & Experimental Immunology, National Institute of Immunohaematology (Indian Council of Medical Research), 13th Floor, KEM Hospital Campus, Parel, Mumbai, India
| | - Milind Nadkar
- Department of Medicine, Seth GS Medical College and KEM Hospital, Parel, Mumbai, India
| | | | - Anjali Rajadhyaksha
- Department of Medicine, Seth GS Medical College and KEM Hospital, Parel, Mumbai, India.
| | - Niwrutti Hase
- Department of Nephrology, Seth GS Medical College and KEM Hospital, Parel, Mumbai, India
| | - Tukaram Jamale
- Department of Nephrology, Seth GS Medical College and KEM Hospital, Parel, Mumbai, India
| | - Seema Kini
- Department of Medicine, TNMC & BYLN, Mumbai, India
| | - Prasad Khadilkar
- Department of Clinical & Experimental Immunology, National Institute of Immunohaematology (Indian Council of Medical Research), 13th Floor, KEM Hospital Campus, Parel, Mumbai, India
| | - Vidya Anand
- Chennai Meenakshi Multi-speciality Hospital, Chennai, India
| | - Manisha Madkaikar
- Department of Clinical & Experimental Immunology, National Institute of Immunohaematology (Indian Council of Medical Research), 13th Floor, KEM Hospital Campus, Parel, Mumbai, India
| | - Vandana Pradhan
- Department of Clinical & Experimental Immunology, National Institute of Immunohaematology (Indian Council of Medical Research), 13th Floor, KEM Hospital Campus, Parel, Mumbai, India.
| |
Collapse
|
40
|
Audrito V, Managò A, La Vecchia S, Zamporlini F, Vitale N, Baroni G, Cignetto S, Serra S, Bologna C, Stingi A, Arruga F, Vaisitti T, Massi D, Mandalà M, Raffaelli N, Deaglio S. Nicotinamide Phosphoribosyltransferase (NAMPT) as a Therapeutic Target in BRAF-Mutated Metastatic Melanoma. J Natl Cancer Inst 2018; 110:290-303. [DOI: 10.1093/jnci/djx198] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Valentina Audrito
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Antonella Managò
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Sofia La Vecchia
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Federica Zamporlini
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Nicoletta Vitale
- Department of Molecular Biotechnologies and Health Science, University of Turin, Italy
| | - Gianna Baroni
- Division of Pathological Anatomy, Department of Surgery and Translational Medicine, University of Florence, Italy
| | - Simona Cignetto
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Sara Serra
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Cinzia Bologna
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Aureliano Stingi
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Francesca Arruga
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Tiziana Vaisitti
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Daniela Massi
- Division of Pathological Anatomy, Department of Surgery and Translational Medicine, University of Florence, Italy
| | - Mario Mandalà
- Unit of Medical Oncology, Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Nadia Raffaelli
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Italy
- Italian Institute for Genomic Medicine, Turin, Italy
| |
Collapse
|
41
|
Lokau J, Agthe M, Flynn CM, Garbers C. Proteolytic control of Interleukin-11 and Interleukin-6 biology. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017. [DOI: 10.1016/j.bbamcr.2017.06.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
42
|
Vora M, Alattia LA, Ansari J, Ong M, Cotelingam J, Coppola D, Shackelford R. Nicotinamide Phosphoribosyl Transferase a Reliable Marker of Progression in Cervical Dysplasia. Anticancer Res 2017; 37:4821-4825. [PMID: 28870901 DOI: 10.21873/anticanres.11889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/29/2017] [Accepted: 07/12/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND/AIM Nicotinamide phosphoribosyl transferase (Nampt) catalyses the rate-limiting step of the mammalian nicotinamide adenine dinucleotide (NAD) salvage pathway. Nampt is highly expressed in several epithelial and mesenchymal neoplasms, where is promotes cell-cycle progression ans chemotherapy resistance. To our knowledge, alterations in Nampt expression have not been examined in cervical intraepithelial neoplasia (CIN) or squamous cell carcinoma (SCC). MATERIALS AND METHODS We performed immunohistochemical analysis for Nampt using tissue microarrays on 14 samples of benign cervical squamous epithelium and 15 CIN I, 15 CIN II, and 13 samples of CIN III. The SCCs included 5 low-grade, 67 intermediate-grade, and 81 high-grade tumors. RESULTS Nampt levels increased with increased CIN grades were compared to benign cervical squamous epithelium. Similarly, Nampt levels increased with increasing SCC grade. CONCLUSION Nampt expression is a reliable marker of progression in cervical dysplasia and SCC.
Collapse
Affiliation(s)
- Moiz Vora
- Department of Pathology and Translational Pathobiology, LSU Health, Shreveport, LA, U.S.A
| | - Lubna A Alattia
- Department of Pathology and Translational Pathobiology, LSU Health, Shreveport, LA, U.S.A
| | - Junaid Ansari
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, U.S.A
| | - Menchu Ong
- Department of Pathology and Translational Pathobiology, LSU Health, Shreveport, LA, U.S.A
| | - James Cotelingam
- Department of Pathology and Translational Pathobiology, LSU Health, Shreveport, LA, U.S.A
| | - Domenico Coppola
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - Rodney Shackelford
- Department of Pathology and Translational Pathobiology, LSU Health, Shreveport, LA, U.S.A.
| |
Collapse
|
43
|
Shackelford RE, Abdulsattar J, Wei EX, Cotelingam J, Coppola D, Herrera GA. Increased Nicotinamide Phosphoribosyltransferase and Cystathionine-β-Synthase in Renal Oncocytomas, Renal Urothelial Carcinoma, and Renal Clear Cell Carcinoma. Anticancer Res 2017; 37:3423-3427. [PMID: 28668830 DOI: 10.21873/anticanres.11709] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 05/22/2017] [Accepted: 05/24/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Renal oncocytomas (ROs), and clear cell (RCC) and urothelial carcinomas (UC), are common renal neoplasms. Nicotinamide phosphoribosyltransferase (Nampt) catalyzes the rate-limiting step of NAD+ synthesis and its expression is increased in several tumors. Nampt concomitantly regulates hydrogen sulfide (H2S)-synthesizing enzyme levels, including cystathionine-β-synthase (CBS). MATERIALS AND METHODS We used tissue microarrays to examine Nampt and the H2S-synthesizing enzyme CBS protein levels in benign kidney, RCC, UC and ROs. RESULTS Compared to benign kidney, all three neoplasms showed increased Nampt and CBS protein levels, with the levels increasing in RCC at higher Fuhrman grades. CONCLUSION H2S is known to ameliorate chronic renal failure but, as yet, no role for H2S in renal neoplasia has been demonstrated. Here, we showed, for the first time, that Nampt, CBS and, likely, H2S likely play a role in malignant and benign neoplastic renal disease.
Collapse
Affiliation(s)
- Rodney E Shackelford
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, LA, U.S.A.
| | - Jehan Abdulsattar
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, LA, U.S.A
| | - Eric X Wei
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, LA, U.S.A
| | - James Cotelingam
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, LA, U.S.A
| | - Domenico Coppola
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - Guillermo A Herrera
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, LA, U.S.A
| |
Collapse
|
44
|
Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM. JAK-STAT Signaling as a Target for Inflammatory and Autoimmune Diseases: Current and Future Prospects. Drugs 2017; 77:521-546. [PMID: 28255960 PMCID: PMC7102286 DOI: 10.1007/s40265-017-0701-9] [Citation(s) in RCA: 799] [Impact Index Per Article: 99.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Janus kinase/signal transduction and activator of transcription (JAK-STAT) signaling pathway is implicated in the pathogenesis of inflammatory and autoimmune diseases including rheumatoid arthritis, psoriasis, and inflammatory bowel disease. Many cytokines involved in the pathogenesis of autoimmune and inflammatory diseases use JAKs and STATs to transduce intracellular signals. Mutations in JAK and STAT genes cause a number of immunodeficiency syndromes, and polymorphisms in these genes are associated with autoimmune diseases. The success of small-molecule JAK inhibitors (Jakinibs) in the treatment of rheumatologic disease demonstrates that intracellular signaling pathways can be targeted therapeutically to treat autoimmunity. Tofacitinib, the first rheumatologic Jakinib, is US Food and Drug Administration (FDA) approved for rheumatoid arthritis and is currently under investigation for other autoimmune diseases. Many other Jakinibs are in preclinical development or in various phases of clinical trials. This review describes the JAK-STAT pathway, outlines its role in autoimmunity, and explains the rationale/pre-clinical evidence for targeting JAK-STAT signaling. The safety and clinical efficacy of the Jakinibs are reviewed, starting with the FDA-approved Jakinib tofacitinib, and continuing on to next-generation Jakinibs. Recent and ongoing studies are emphasized, with a focus on emerging indications for JAK inhibition and novel mechanisms of JAK-STAT signaling blockade.
Collapse
Affiliation(s)
- Shubhasree Banerjee
- Rheumatology Fellowship and Training Branch, National Institute of Arthritis Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | - Ann Biehl
- Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Massimo Gadina
- Translational Immunology Section, National Institute of Arthritis Musculoskeletal and Skin diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarfaraz Hasni
- Lupus Clinical Research Program, National Institute of Arthritis Musculoskeletal and Skin diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniella M Schwartz
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis Musculoskeletal and Skin diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
45
|
Türer ÇC, Balli U, Güven B, Çetinkaya BÖ, Keleş GÇ. Visfatin levels in gingival crevicular fluid and serum before and after non-surgical treatment for periodontal diseases. J Oral Sci 2016; 58:491-499. [PMID: 28025432 DOI: 10.2334/josnusd.16-0116] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
The purpose of this study was to evaluate visfatin levels at different stages of periodontal disease and in healthy tissues. In addition, the effect of non-surgical periodontal therapy on visfatin levels in gingival crevicular fluid and serum was investigated. Forty-five patients were divided into three groups based on clinical and radiographical findings. Group 1 comprised periodontally healthy individuals (n = 15); group 2 comprised patients with gingivitis (n = 15); and group 3 was composed of patients with generalized chronic periodontitis (n = 15). Gingival crevicular fluid and serum samples were collected before treatment and at 1, 3, and 6 months after treatment. Visfatin levels were measured by enzyme-linked immunosorbent assays. Gingival crevicular fluid and serum visfatin levels were higher in patients with chronic periodontitis than those with gingivitis or healthy controls (P < 0.016). In addition, visfatin levels were higher in the gingivitis group than in healthy controls (P < 0.016). Non-surgical periodontal treatment resulted in a significant reduction in gingival crevicular fluid and serum visfatin levels. Furthermore, visfatin levels increased with inflammation and decreased following periodontal treatment. Our findings suggest that visfatin is an inflammatory biomarker of periodontal disease.(J Oral Sci 58, 491-499, 2016).
Collapse
Affiliation(s)
- Çiğdem Coşkun Türer
- Department of Periodontology, Faculty of Dentistry, Bülent Ecevit University
| | | | | | | | | |
Collapse
|
46
|
Junker S, Frommer KW, Krumbholz G, Tsiklauri L, Gerstberger R, Rehart S, Steinmeyer J, Rickert M, Wenisch S, Schett G, Müller-Ladner U, Neumann E. Expression of adipokines in osteoarthritis osteophytes and their effect on osteoblasts. Matrix Biol 2016; 62:75-91. [PMID: 27884778 DOI: 10.1016/j.matbio.2016.11.005] [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] [Received: 08/25/2016] [Revised: 11/09/2016] [Accepted: 11/09/2016] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Osteophyte formation in osteoarthritis (OA) is mediated by increased osteoblast activity, which is -in turn- regulated by the Wnt signaling pathway. Obesity is regarded a risk factor in OA, yet little is known about the interaction between adipose tissue-derived factors, the adipokines, and bone formation, although adipokines are associated with the pathogenesis of OA. Therefore, the effect of adipokines on bone and cartilage forming cells and osteophyte development was analyzed. METHODS Human OA osteophytes were histologically characterized and adipokine expression was evaluated by immunohistochemistry. Osteoblasts and chondrocytes were isolated from OA tissue and stimulated with adiponectin, resistin, or visfatin. Cytokine and osteoblast/chondrocyte markers were quantified and activation of Wnt and p38 MAPK signaling was analyzed. RESULTS Adiponectin, resistin, and visfatin were expressed in OA osteophytes by various articular cell types. Stimulation of OA osteoblasts with adiponectin and of OA chondrocytes with visfatin led to an increased release of proinflammatory mediators but not to osteoblast differentiation or activation. Additionally, visfatin increased matrix degrading factors in chondrocytes. Wnt signaling was not altered by adipokines, but adiponectin induced p38 MAPK signaling in osteoblasts. CONCLUSION Adipokines are present in OA osteophytes, and adiponectin and visfatin increase the release of proinflammatory mediators by osteoblasts and chondrocytes. The effects of adiponectin were mediated by p38 MAPK but not Wnt signaling in osteoblasts. Therefore, the results support the idea that adipokines do not directly influence osteophyte development but the proinflammatory conditions in OA.
Collapse
Affiliation(s)
- Susann Junker
- Dept Internal Medicine and Rheumatology, Justus-Liebig-University Giessen, Kerckhoff-Klinik Bad Nauheim, Germany
| | - Klaus W Frommer
- Dept Internal Medicine and Rheumatology, Justus-Liebig-University Giessen, Kerckhoff-Klinik Bad Nauheim, Germany
| | - Grit Krumbholz
- Dept Internal Medicine and Rheumatology, Justus-Liebig-University Giessen, Kerckhoff-Klinik Bad Nauheim, Germany
| | - Lali Tsiklauri
- Dept Internal Medicine and Rheumatology, Justus-Liebig-University Giessen, Kerckhoff-Klinik Bad Nauheim, Germany
| | - Rüdiger Gerstberger
- Dept Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Germany
| | - Stefan Rehart
- Dept Orthopaedics and Trauma Surgery, Agaplesion-Markus-Hospital, Frankfurt, Germany
| | - Jürgen Steinmeyer
- Dept Orthopaedics and Orthopaedic Surgery, University Hospital Giessen and Marburg, Giessen, Germany
| | - Markus Rickert
- Dept Orthopaedics and Orthopaedic Surgery, University Hospital Giessen and Marburg, Giessen, Germany
| | - Sabine Wenisch
- Clinic for Small Animals, Institute for Veterinary Anatomy, Histology und Embryology, Justus-Liebig-University Giessen, Germany
| | - Georg Schett
- Medical Clinic 3, Rheumatology and Immunology, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ulf Müller-Ladner
- Dept Internal Medicine and Rheumatology, Justus-Liebig-University Giessen, Kerckhoff-Klinik Bad Nauheim, Germany
| | - Elena Neumann
- Dept Internal Medicine and Rheumatology, Justus-Liebig-University Giessen, Kerckhoff-Klinik Bad Nauheim, Germany.
| |
Collapse
|
47
|
Kendal-Wright CE. Stretching, Mechanotransduction, and Proinflammatory Cytokines in the Fetal Membranes. Reprod Sci 2016; 14:35-41. [DOI: 10.1177/1933719107310763] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Claire E. Kendal-Wright
- Developmental and Reproduction Biology, John A. Burns
School of Medicine, and the Pacific Biomedical Research Center, Honolulu,
Hawaii,
| |
Collapse
|
48
|
Mishra V, Shettar L, Bajaj M, Math AS, Thakur SL. Interlinking Periodontitis and Type 2 Diabetes Mellitus by Assessment of Crevicular Visfatin Levels in Health and in Disease Before and After Initial Periodontal Therapy. J Clin Diagn Res 2016; 10:ZC67-71. [PMID: 27656567 PMCID: PMC5028447 DOI: 10.7860/jcdr/2016/18656.8283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 04/05/2016] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Visfatin is a new adipocytokine associated with both chronic periodontitis and type 2 diabetes mellitus independently. AIM We aimed to estimate and compare the changes in the levels of visfatin in the Gingival Crevicular Fluid (GCF) of healthy subjects and in subjects with periodontitis with or without controlled Type 2 Diabetes Mellitus (T2DM) after administration of non-surgical periodontal therapy. MATERIALS AND METHODS Forty two subjects were equally divided into Group 1 (healthy), Group 2 (systemically healthy with chronic periodontitis), Group 3 (subjects with chronic periodontitis having controlled T2DM). Defined clinical parameters were recorded at baseline and at one month follow-up period. Visfatin was assessed using enzyme linked immunosorbent assay. One way ANOVA and Tukey's multiple post hoc procedures were used. Pearson's correlation coefficient was used for correlation. RESULTS Significant increase in the visfatin levels was seen with the highest values observed in diabetes with periodontal disease. Visfatin responded to non-surgical periodontal therapy as observed by significant decrease in levels after one month but even at this period diabetics showed the highest levels. CONCLUSION Visfatin levels are highest in individuals with both periodontal disease and diabetes even after periodontal therapy. Individuals with T2DM may be at higher risk of developing periodontal disease.
Collapse
Affiliation(s)
- Vandita Mishra
- Senior Lecturer, Department of Periodontics, Sri Sai College of Dental Surgery, Vikarabad, Telangana, India
| | - Leena Shettar
- Professor and Head, Department of Periodontics, S.D.M. College of Dental Sciences and Hospital, Dharwad, Karnataka, India
| | - Mahesh Bajaj
- Student, Department of Periodontics, SDM College of Dental Sciences and Hospital, Dharwad, Karnataka, India
| | - Abhishek Savir Math
- Reader, Department of Prosthodontics, Sri Sai College of Dental Surgery, Vikarabad, Telangana, India
| | - Srinath L. Thakur
- Principal and Professor, Department of Periodontics, S.D.M. College of Dental Sciences and Hospital, Dharwad, Karnataka, India
| |
Collapse
|
49
|
Harwardt T, Lukas S, Zenger M, Reitberger T, Danzer D, Übner T, Munday DC, Nevels M, Paulus C. Human Cytomegalovirus Immediate-Early 1 Protein Rewires Upstream STAT3 to Downstream STAT1 Signaling Switching an IL6-Type to an IFNγ-Like Response. PLoS Pathog 2016; 12:e1005748. [PMID: 27387064 PMCID: PMC4936752 DOI: 10.1371/journal.ppat.1005748] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/16/2016] [Indexed: 12/24/2022] Open
Abstract
The human cytomegalovirus (hCMV) major immediate-early 1 protein (IE1) is best known for activating transcription to facilitate viral replication. Here we present transcriptome data indicating that IE1 is as significant a repressor as it is an activator of host gene expression. Human cells induced to express IE1 exhibit global repression of IL6- and oncostatin M-responsive STAT3 target genes. This repression is followed by STAT1 phosphorylation and activation of STAT1 target genes normally induced by IFNγ. The observed repression and subsequent activation are both mediated through the same region (amino acids 410 to 445) in the C-terminal domain of IE1, and this region serves as a binding site for STAT3. Depletion of STAT3 phenocopies the STAT1-dependent IFNγ-like response to IE1. In contrast, depletion of the IL6 receptor (IL6ST) or the STAT kinase JAK1 prevents this response. Accordingly, treatment with IL6 leads to prolonged STAT1 instead of STAT3 activation in wild-type IE1 expressing cells, but not in cells expressing a mutant protein (IE1dl410-420) deficient for STAT3 binding. A very similar STAT1-directed response to IL6 is also present in cells infected with a wild-type or revertant hCMV, but not an IE1dl410-420 mutant virus, and this response results in restricted viral replication. We conclude that IE1 is sufficient and necessary to rewire upstream IL6-type to downstream IFNγ-like signaling, two pathways linked to opposing actions, resulting in repressed STAT3- and activated STAT1-responsive genes. These findings relate transcriptional repressor and activator functions of IE1 and suggest unexpected outcomes relevant to viral pathogenesis in response to cytokines or growth factors that signal through the IL6ST-JAK1-STAT3 axis in hCMV-infected cells. Our results also reveal that IE1, a protein considered to be a key activator of the hCMV productive cycle, has an unanticipated role in tempering viral replication. Our previous work has shown that the human cytomegalovirus (hCMV) major immediate-early 1 protein (IE1) modulates host cell signaling pathways involving proteins of the signal transducer and activator of transcription (STAT) family. IE1 has also long been known to facilitate viral replication by activating transcription. In this report we demonstrate that IE1 is as significant a repressor as it is an activator of host gene expression. Many genes repressed by IE1 are normally induced via STAT3 signaling triggered by interleukin 6 (IL6) or related cytokines, whereas many genes activated by IE1 are normally induced via STAT1 signaling triggered by interferon gamma (IFNγ). Our results suggest that the repression of STAT3- and the activation of STAT1-responsive genes by IE1 are coupled. By targeting STAT3, IE1 rewires upstream STAT3 to downstream STAT1 signaling. Consequently, genes normally induced by IL6 are repressed while genes normally induced by IFNγ become responsive to IL6 in the presence of IE1. We also demonstrate that, by switching an IL6 to an IFNγ-like response, IE1 tempers viral replication. These results suggest an unanticipated dual role for IE1 in either promoting or limiting hCMV propagation and demonstrate how a key viral regulatory protein merges two central cellular signaling pathways to divert cytokine responses relevant to hCMV pathogenesis.
Collapse
Affiliation(s)
- Thomas Harwardt
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Simone Lukas
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Marion Zenger
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Tobias Reitberger
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Daniela Danzer
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Theresa Übner
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Diane C. Munday
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Michael Nevels
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
- * E-mail: (MN); (CP)
| | - Christina Paulus
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
- * E-mail: (MN); (CP)
| |
Collapse
|
50
|
Rosti V, Campanelli R, Massa M, Viarengo G, Villani L, Poletto V, Bonetti E, Catarsi P, Magrini U, Grolla AA, Travelli C, Genazzani AA, Barosi G. Increased plasma nicotinamide phosphoribosyltransferase is associated with a hyperproliferative phenotype and restrains disease progression in MPN-associated myelofibrosis. Am J Hematol 2016; 91:709-13. [PMID: 27074203 DOI: 10.1002/ajh.24388] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 04/09/2016] [Indexed: 12/27/2022]
Abstract
Myeloproliferative neoplasm (MPN)-associated myelofibrosis is a clonal, neoplastic disorder of the hematopoietic stem cells, in which inflammation and immune dysregulation play an important role. Extracellular nicotinamide phosphoribosyltransferase (eNAMPT), also known as visfatin, is a cytokine implicated in a number of inflammatory and neoplastic diseases. Here plasma levels of eNAMPT in patients with MPN-associated myelofibrosis and their effects on disease phenotype and outcomes were examined. The concordance of eNAMPT levels with the marker of general inflammation high-sensitivity C-reactive protein (hs-CRP) was also studied. A total of 333 MPN-associated myelofibrosis patients (187 males and 146 females) and 31 age- and gender-matched normal-weight healthy subjects were enrolled in the study main body. Levels of eNAMPT and hs-CRP were simultaneously assayed in 209 MPN-associated myelofibrosis patients. Twenty-four polycythemia vera or essential thrombocythemia patients were used as controls. eNAMPT was over expressed in MPN-associated myelofibrosis, and eNAMPT expression was correlated with higher white blood cell count, higher hemoglobin, and higher platelet count, suggesting that eNAMPT is an indispensable permissive agent for myeloproliferation of MPN-associated myelofibrosis. The lack of correlation between eNAMPT and hs-CRP revealed that eNAMPT in MPN-associated myelofibrosis does not behave as a canonical inflammatory cytokine. In addition, higher levels of eNAMPT predicted longer time to blast transformation, and protected against progression toward thrombocytopenia and large splenomegaly. In conclusion, in MPN-associated myelofibrosis high levels of eNAMPT mark the myeloproliferative potential and, at variance with a high number of cancers, are protective against disease progression. Am. J. Hematol. 91:709-713, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vittorio Rosti
- Center for the Study of Myelofibrosis. Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation; Pavia 27100 Italy
| | - Rita Campanelli
- Center for the Study of Myelofibrosis. Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation; Pavia 27100 Italy
| | - Margherita Massa
- Biotechnology Research Area; IRCCS Policlinico S. Matteo Foundation; Pavia 27100 Italy
| | - Gianluca Viarengo
- Immunohematology and Transfusion Service; IRCCS Policlinico S. Matteo Foundation; Pavia Italy
| | - Laura Villani
- Center for the Study of Myelofibrosis. Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation; Pavia 27100 Italy
| | - Valentina Poletto
- Center for the Study of Myelofibrosis. Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation; Pavia 27100 Italy
| | - Elisa Bonetti
- Center for the Study of Myelofibrosis. Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation; Pavia 27100 Italy
| | - Paolo Catarsi
- Center for the Study of Myelofibrosis. Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation; Pavia 27100 Italy
| | - Umberto Magrini
- Center for the Study of Myelofibrosis. Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation; Pavia 27100 Italy
| | - Ambra A. Grolla
- Department of Pharmaceutical Sciences; Università Del Piemonte Orientale; Novara Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences; Università Del Piemonte Orientale; Novara Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences; Università Del Piemonte Orientale; Novara Italy
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis. Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation; Pavia 27100 Italy
| |
Collapse
|