1
|
Zakrocka I, Urbańska EM, Załuska W, Kronbichler A. Kynurenine Pathway after Kidney Transplantation: Friend or Foe? Int J Mol Sci 2024; 25:9940. [PMID: 39337426 PMCID: PMC11432217 DOI: 10.3390/ijms25189940] [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: 08/21/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Kidney transplantation significantly improves the survival of patients with end-stage kidney disease (ESKD) compared to other forms of kidney replacement therapy. However, kidney transplant recipients' outcomes are not fully satisfactory due to increased risk of cardiovascular diseases, infections, and malignancies. Immune-related complications remain the biggest challenge in the management of kidney graft recipients. Despite the broad spectrum of immunosuppressive agents available and more detailed methods used to monitor their effectiveness, chronic allograft nephropathy remains the most common cause of kidney graft rejection. The kynurenine (KYN) pathway is the main route of tryptophan (Trp) degradation, resulting in the production of a plethora of substances with ambiguous properties. Conversion of Trp to KYN by the enzyme indoleamine 2,3-dioxygenase (IDO) is the rate-limiting step determining the formation of the next agents from the KYN pathway. IDO activity, as well as the production of subsequent metabolites of the pathway, is highly dependent on the balance between pro- and anti-inflammatory conditions. Moreover, KYN pathway products themselves possess immunomodulating properties, e.g., modify the activity of IDO and control other immune-related processes. KYN metabolites were widely studied in neurological disorders but recently gained the attention of researchers in the context of immune-mediated diseases. Evidence that this route of Trp degradation may represent a peripheral tolerogenic pathway with significant implications for transplantation further fueled this interest. Our review aimed to present recent knowledge about the role of the KYN pathway in the pathogenesis, diagnosis, monitoring, and treatment of kidney transplant recipients' complications.
Collapse
Affiliation(s)
- Izabela Zakrocka
- Department of Nephrology, Medical University of Lublin, 20-093 Lublin, Poland; (I.Z.); (W.Z.)
| | - Ewa M. Urbańska
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Wojciech Załuska
- Department of Nephrology, Medical University of Lublin, 20-093 Lublin, Poland; (I.Z.); (W.Z.)
| | - Andreas Kronbichler
- Department of Internal Medicine IV, Nephrology and Hypertension, Medical University Innsbruck, 6020 Innsbruck, Austria
- Department of Health, Medicine and Caring Sciences, Linköping University, 581 83 Linköping, Sweden
| |
Collapse
|
2
|
Al-Qahtani Z, Al-Kuraishy HM, Ali NH, Elewa YHA, Batiha GES. Kynurenine pathway in type 2 diabetes: Role of metformin. Drug Dev Res 2024; 85:e22243. [PMID: 39129450 DOI: 10.1002/ddr.22243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 03/08/2024] [Accepted: 07/29/2024] [Indexed: 08/13/2024]
Abstract
The Kynurenine pathway (KP) which is involved in the synthesis of nicotinamide adenine dinucleotide (NAD) from tryptophan (Trp) is intricate in the development of insulin resistance (IR) and type 2 diabetes (T2D). Inflammatory reactions in response to cardiometabolic disorders can induce the development of IR through the augmentation of KP. However, kynurenine (KYN), a precursor of kynurenic acid (KA) is increased following physical exercise and involved in the reduction of IR. Consequently, KP metabolites KA and KYN have anti-diabetogenic effects while other metabolites have diabetogenic effects. KP modulators, either inhibitors or activators, affect glucose homeostasis and insulin sensitivity in T2D in a bidirectional way, either protective or detrimental, that is not related to the KP effect. However, metformin through inhibition of inflammatory signaling pathways can reduce the activation of KP in T2D. These findings indicated a strong controversy regarding the role of KP in T2D. Therefore, the objectives of this mini review were to clarify how KP induces the development of IR and T2D. In addition, this review aimed to find the mechanistic role of antidiabetic drug metformin on the KP, and how KP modulators affect the pathogenesis of T2D.
Collapse
Affiliation(s)
- Zainah Al-Qahtani
- Internal Medicine Department, Neurology Section, College of Medicine, King Khaled university, Abha, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Naif H Ali
- Department of internal medicine, Medical College, Najran University, Najran, Saudi Arabia
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhur University, Damanhur, Egypt
| |
Collapse
|
3
|
Zhang K, Mishra A, Jagannath C. New insight into arginine and tryptophan metabolism in macrophage activation during tuberculosis. Front Immunol 2024; 15:1363938. [PMID: 38605962 PMCID: PMC11008464 DOI: 10.3389/fimmu.2024.1363938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Arginine and tryptophan are pivotal in orchestrating cytokine-driven macrophage polarization and immune activation. Specifically, interferon-gamma (IFN-γ) stimulates inducible nitric oxide synthase (iNOS) expression), leading to the conversion of arginine into citrulline and nitric oxide (NO), while Interleukin-4 (IL4) promotes arginase activation, shifting arginine metabolism toward ornithine. Concomitantly, IFN-γ triggers indoleamine 2,3-dioxygenase 1 (IDO1) and Interleukin-4 induced 1 (IL4i1), resulting in the conversion of tryptophan into kynurenine and indole-3-pyruvic acid. These metabolic pathways are tightly regulated by NAD+-dependent sirtuin proteins, with Sirt2 and Sirt5 playing integral roles. In this review, we present novel insights that augment our understanding of the metabolic pathways of arginine and tryptophan following Mycobacterium tuberculosis infection, particularly their relevance in macrophage responses. Additionally, we discuss arginine methylation and demethylation and the role of Sirt2 and Sirt5 in regulating tryptophan metabolism and arginine metabolism, potentially driving macrophage polarization.
Collapse
Affiliation(s)
- Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| |
Collapse
|
4
|
Pires AS, Gupta S, Barton SA, Vander Wall R, Tan V, Heng B, Phillips JK, Guillemin GJ. Temporal Profile of Kynurenine Pathway Metabolites in a Rodent Model of Autosomal Recessive Polycystic Kidney Disease. Int J Tryptophan Res 2022; 15:11786469221126063. [PMID: 36329761 PMCID: PMC9623391 DOI: 10.1177/11786469221126063] [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: 07/08/2022] [Accepted: 08/27/2022] [Indexed: 11/28/2022] Open
Abstract
Autosomal recessive polycystic kidney disease (ARPKD) is an early onset genetic disorder characterized by numerous renal cysts resulting in end stage renal disease. Our study aimed to determine if metabolic reprogramming and tryptophan (Trp) metabolism via the kynurenine pathway (KP) is a critical dysregulated pathway in PKD. Using the Lewis polycystic kidney (LPK) rat model of PKD and Lewis controls, we profiled temporal trends for KP metabolites in plasma, urine, and kidney tissues from 6- and 12-week-old mixed sex animals using liquid and gas chromatography, minimum n = 5 per cohort. A greater kynurenine (KYN) concentration was observed in LPK kidney and plasma of 12-week rats compared to age matched Lewis controls (P ⩽ .05). LPK kidneys also showed an age effect (P ⩽ .05) with KYN being greater in 12-week versus 6-week LPK. The metabolites xanthurenic acid (XA), 3-hydroxykynurenine (3-HK), and 3-hydroxyanthranilic acid (3-HAA) were significantly greater in the plasma of 12-week LPK rats compared to age matched Lewis controls (P ⩽ .05). Plasma XA and 3-HK also showed an age effect (P ⩽ .05) being greater in 12-week versus 6-week LPK. We further describe a decrease in Trp levels in LPK plasma and kidney (strain effect P ⩽ .05). There were no differences in KP metabolites in urine between cohorts. Using the ratio of product and substrates in the KP, a significant age-strain effect (P ⩽ .05) was observed in the activity of the KYN/Trp ratio (tryptophan-2,3-dioxygenase [TDO] or indoleamine-2,3-dioxygenase [IDO] activity), kynurenine 3-monooxygenase (KMO), KAT A (kynurenine aminotransferase A), KAT B, total KAT, total KYNU (kynureninase), KYNU A, KYNU B, and total KYNU within LPK kidneys, supporting an activated KP. Confirmation of the activation of these enzymes will require verification through orthogonal techniques. In conclusion, we have demonstrated an up-regulation of the KP in alignment with progression of renal impairment in the LPK rat model, suggesting that KP activation may be a critical contributor to the pathobiology of PKD.
Collapse
Affiliation(s)
- Ananda Staats Pires
- Neuroinflammation Group, Macquarie
Medical School, Centre for Motor Neuron Disease Research, Faculty of Medicine,
Health and Human Sciences, Macquarie University, Sydney, NSW, Australia,Laboratório de Bioenergética e Estresse
Oxidativo, Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade
Federal de Santa Catarina, Florianópolis, Brasil
| | - Shabarni Gupta
- Autonomic and Sensory Neuroscience
Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences,
Macquarie University, Sydney, NSW, Australia
| | - Sean A Barton
- Autonomic and Sensory Neuroscience
Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences,
Macquarie University, Sydney, NSW, Australia
| | - Roshana Vander Wall
- Autonomic and Sensory Neuroscience
Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences,
Macquarie University, Sydney, NSW, Australia
| | - Vanessa Tan
- Neuroinflammation Group, Macquarie
Medical School, Centre for Motor Neuron Disease Research, Faculty of Medicine,
Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Benjamin Heng
- Neuroinflammation Group, Macquarie
Medical School, Centre for Motor Neuron Disease Research, Faculty of Medicine,
Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Jacqueline K Phillips
- Autonomic and Sensory Neuroscience
Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences,
Macquarie University, Sydney, NSW, Australia,Jacqueline K Phillips, Autonomic and
Sensory Neuroscience Group, Macquarie Medical School,Department of Biomedical
Science, Faculty of Medicine, Health and Human Sciences, Macquarie University,
Level 1, 75 Talavera Road, Sydney, NSW 2109, Australia.
| | - Gilles J Guillemin
- Neuroinflammation Group, Macquarie
Medical School, Centre for Motor Neuron Disease Research, Faculty of Medicine,
Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
5
|
Klaessens S, Stroobant V, De Plaen E, Van den Eynde BJ. Systemic tryptophan homeostasis. Front Mol Biosci 2022; 9:897929. [PMID: 36188218 PMCID: PMC9515494 DOI: 10.3389/fmolb.2022.897929] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/01/2022] [Indexed: 11/27/2022] Open
Abstract
Tryptophan is an essential amino acid, which is not only a building block for protein synthesis, but also a precursor for the biosynthesis of co-enzymes and neuromodulators, such as NAD/NADP(H), kynurenic acid, melatonin and serotonin. It also plays a role in immune homeostasis, as local tryptophan catabolism impairs T-lymphocyte mediated immunity. Therefore, tryptophan plasmatic concentration needs to be stable, in spite of large variations in dietary supply. Here, we review the main checkpoints accounting for tryptophan homeostasis, including absorption, transport, metabolism and elimination, and we discuss the physiopathology of disorders associated with their dysfunction. Tryptophan is catabolized along the kynurenine pathway through the action of two enzymes that mediate the first and rate-limiting step of the pathway: indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO). While IDO1 expression is restricted to peripheral sites of immune modulation, TDO is massively expressed in the liver and accounts for 90% of tryptophan catabolism. Recent data indicated that the stability of the TDO protein is regulated by tryptophan and that this regulation allows a tight control of tryptophanemia. TDO is stabilized when tryptophan is abundant in the plasma, resulting in rapid degradation of dietary tryptophan. In contrast, when tryptophan is scarce, TDO is degraded by the proteasome to avoid excessive tryptophan catabolism. This is triggered by the unmasking of a degron in a non-catalytic tryptophan-binding site, resulting in TDO ubiquitination by E3 ligase SKP1-CUL1-F-box. Deficiency in TDO or in the hepatic aromatic transporter SLC16A10 leads to severe hypertryptophanemia, which can disturb immune and neurological homeostasis.
Collapse
Affiliation(s)
- Simon Klaessens
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
- *Correspondence: Simon Klaessens, ; Benoit J. Van den Eynde,
| | - Vincent Stroobant
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
| | - Etienne De Plaen
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
| | - Benoit J. Van den Eynde
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology, Wavre, Belgium
- Nuffield Department of Clinical Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
- *Correspondence: Simon Klaessens, ; Benoit J. Van den Eynde,
| |
Collapse
|
6
|
Abd El-Fattah EE. IDO/kynurenine pathway in cancer: possible therapeutic approaches. Lab Invest 2022; 20:347. [PMID: 35918736 PMCID: PMC9344609 DOI: 10.1186/s12967-022-03554-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/25/2022] [Indexed: 11/10/2022]
Abstract
Cancer is one of the leading causes of death in both men and women worldwide. One of the main changes associated with cancer progression, metastasis, recurrence, and chemoresistance is the change in the tumor immune microenvironment, especially immunosuppression. Cancer immunosuppression appears in multiple forms, such as inhibition of immuno-stimulant cells with downregulation of immuno-stimulant mediators or through stimulation of immuno-suppressive cells with upregulation of immunosuppressive mediators. One of the most immunosuppressive mediators that approved potency in lung cancer progression is indoleamine 2,3-dioxygenase (IDO) and its metabolite kynurenine (Kyn). The current review tries to elucidate the role of IDO/Kyn on cancer proliferation, apoptosis, angiogenesis, oxidative stress, and cancer stemness. Besides, our review investigates the new therapeutic modalities that target IDO/Kyn pathway and thus as drug candidates for targeting lung cancer and drugs that potentiate IDO/Kyn pathway and thus can be cancer-promoting agents.
Collapse
Affiliation(s)
- Eslam E Abd El-Fattah
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt.
| |
Collapse
|
7
|
Skariah S, Sultan AA, Mordue DG. IFN-induced cell-autonomous immune mechanisms in the control of intracellular protozoa. Parasitol Res 2022; 121:1559-1571. [DOI: 10.1007/s00436-022-07514-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 04/04/2022] [Indexed: 10/18/2022]
|
8
|
Ghosh A, Joseph B, Anil S. Nitric Oxide in the Management of Respiratory Consequences in COVID-19: A Scoping Review of a Different Treatment Approach. Cureus 2022; 14:e23852. [PMID: 35530860 PMCID: PMC9072273 DOI: 10.7759/cureus.23852] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2022] [Indexed: 12/13/2022] Open
Abstract
The severe acute respiratory distress syndrome coronavirus 2 (SARS-CoV-2) virus causing COVID-19 significantly affects the respiratory functions of infected individuals by massively disrupting the pulmonary oxygenation and activating the synthesis of proinflammatory cytokines, inducing severe oxidative stress, enhanced vascular permeability, and endothelial dysfunction which have rendered researchers and clinicians to depend on prophylactic treatment due to the unavailability of proper disease management approaches. Previous studies have indicated that nitric oxide (NO) application appears to be significant concerning the antiviral activities, antioxidant, and anti-inflammatory properties in relieving disease-related symptoms. To identify, explore, and map the literature on the role of nitric oxide in the management of respiratory consequences in COVID-19 through this scoping review, Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed during the search to answer the focal question: "What are the potential uses of nitric oxide in the management of respiratory failure in COVID-19?" Administering exogenous NO in the form of inhaled gas or stimulating the system to produce NO appears to be a suitable option to manage COVID-19-induced pneumonia and respiratory illness. This treatment modality seems to attenuate respiratory distress among patients suffering from severe infections or patients with comorbidities. Exogenous NO at different doses effectively reduces systemic hyperinflammation and oxidative stress, improves arterial oxygenation, and restores pulmonary alveolar cellular integrity to prevent the lungs and other organs from further damage. This therapy could pave the way for better management of COVID-19 before the onset of disease-related complications.
Collapse
Affiliation(s)
- Arunibha Ghosh
- Neurosciences, S.N.Pradhan Centre for Neurosciences, University of Calcutta, Kolkata, IND
| | | | - Sukumaran Anil
- Dentistry, Oral Health Institute, Hamad Medical Corporation, Doha, QAT.,Dentistry, College of Dental Medicine, Qatar University, Doha, QAT
| |
Collapse
|
9
|
Biswas P, Dai Y, Stuehr DJ. Indoleamine dioxygenase and tryptophan dioxygenase activities are regulated through GAPDH- and Hsp90-dependent control of their heme levels. Free Radic Biol Med 2022; 180:179-190. [PMID: 35051612 PMCID: PMC11389873 DOI: 10.1016/j.freeradbiomed.2022.01.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/08/2021] [Accepted: 01/11/2022] [Indexed: 01/15/2023]
Abstract
Indoleamine-2, 3-dioxygenase (IDO1) and Tryptophan-2, 3-dioxygense (TDO) are heme-containing dioxygenases that catalyze the conversion of tryptophan to N-formyl-kynurenine and thus enable generation of l-kynurenine and related metabolites that govern the immune response and broadly impact human biology. Given that TDO and IDO1 activities are directly proportional to their heme contents, it is important to understand their heme delivery and insertion processes. Early studies established that TDO and IDO1 heme levels are sub-saturating in vivo and subject to change but did not identify the cellular mechanisms that provide their heme or enable dynamic changes in their heme contents. We investigated the potential involvement of GAPDH and chaperone Hsp90, based on our previous studies linking these proteins to intracellular heme allocation. We studied heme delivery and insertion into IDO1 and TDO expressed in both normal and heme-deficient HEK293T cells and into IDO1 naturally expressed in HeLa cells in response to IFN-γ, and also investigated the interactions of TDO and IDO1 with GAPDH and Hsp90 in cells and among their purified forms. We found that GAPDH delivered both mitochondrially-generated and exogenous heme to apo-IDO1 and apo-TDO in cells, potentially through a direct interaction with either enzyme. In contrast, we found Hsp90 interacted with apo-IDO1 but not with apo-TDO, and was only needed to drive heme insertion into apo-IDO1. By uncovering the cellular processes that allocate heme to IDO1 and TDO, our study provides new insight on how their activities and l-kynurenine production may be controlled in health and disease.
Collapse
Affiliation(s)
- Pranjal Biswas
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - Yue Dai
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
10
|
Sathyasaikumar KV, Pérez de la Cruz V, Pineda B, Vázquez Cervantes GI, Ramírez Ortega D, Donley DW, Severson PL, West BL, Giorgini F, Fox JH, Schwarcz R. Cellular Localization of Kynurenine 3-Monooxygenase in the Brain: Challenging the Dogma. Antioxidants (Basel) 2022; 11:315. [PMID: 35204197 PMCID: PMC8868204 DOI: 10.3390/antiox11020315] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023] Open
Abstract
Kynurenine 3-monooxygenase (KMO), a key player in the kynurenine pathway (KP) of tryptophan degradation, regulates the synthesis of the neuroactive metabolites 3-hydroxykynurenine (3-HK) and kynurenic acid (KYNA). KMO activity has been implicated in several major brain diseases including Huntington's disease (HD) and schizophrenia. In the brain, KMO is widely believed to be predominantly localized in microglial cells, but verification in vivo has not been provided so far. Here, we examined KP metabolism in the brain after depleting microglial cells pharmacologically with the colony stimulating factor 1 receptor inhibitor PLX5622. Young adult mice were fed PLX5622 for 21 days and were euthanized either on the next day or after receiving normal chow for an additional 21 days. Expression of microglial marker genes was dramatically reduced on day 22 but had fully recovered by day 43. In both groups, PLX5622 treatment failed to affect Kmo expression, KMO activity or tissue levels of 3-HK and KYNA in the brain. In a parallel experiment, PLX5622 treatment also did not reduce KMO activity, 3-HK and KYNA in the brain of R6/2 mice (a model of HD with activated microglia). Finally, using freshly isolated mouse cells ex vivo, we found KMO only in microglia and neurons but not in astrocytes. Taken together, these data unexpectedly revealed that neurons contain a large proportion of functional KMO in the adult mouse brain under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Korrapati V. Sathyasaikumar
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA;
| | - Verónica Pérez de la Cruz
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (V.P.d.l.C.); (G.I.V.C.); (D.R.O.)
| | - Benjamín Pineda
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico;
| | - Gustavo Ignacio Vázquez Cervantes
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (V.P.d.l.C.); (G.I.V.C.); (D.R.O.)
| | - Daniela Ramírez Ortega
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (V.P.d.l.C.); (G.I.V.C.); (D.R.O.)
| | - David W. Donley
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY 82071, USA; (D.W.D.); (J.H.F.)
| | - Paul L. Severson
- Plexxikon Inc., South San Francisco, CA 94080, USA; (P.L.S.); (B.L.W.)
| | - Brian L. West
- Plexxikon Inc., South San Francisco, CA 94080, USA; (P.L.S.); (B.L.W.)
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7JA, UK;
| | - Jonathan H. Fox
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY 82071, USA; (D.W.D.); (J.H.F.)
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA;
| |
Collapse
|
11
|
Dual-Sensitive Gold-Nanocubes Platform with Synergistic Immunotherapy for Inducing Immune Cycle Using NIR-Mediated PTT/NO/IDO. Pharmaceuticals (Basel) 2022; 15:ph15020138. [PMID: 35215251 PMCID: PMC8880079 DOI: 10.3390/ph15020138] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/22/2022] [Accepted: 01/23/2022] [Indexed: 11/17/2022] Open
Abstract
Currently, the combination therapies based on immunotherapy have been rapidly developed, but the response rate has not always increased as expected. Nano-platform has become a potential strategy which can trigger multi-functions to increase immunotherapeutic efficacy via activating T-cells and photothermal effect. Herein, to avoid the self-degradation and provide pH-sensitive property, S-nitrosoglutathione (GSNO) was loaded in gold nanocubes (AuNCs) with polyacrylic acid (PAA) coating. Subsequently, the layer-by-layer (LbL) assembly of iron oxide nanoparticles (Fe3O4) and betanin can provide the conjugation of 1-methyl-D-tryptophan (1-M-DT) on the nanoparticle to form an NO gas-photothermal-immune nano-platform (GAPFBD) for achieving combinatory therapy of NO gas, photothermal therapy (PTT), and indoleamine 2,3-dioxygenase (IDO) immunotherapy. After irradiation by 808-nm laser, the GSNO was released under a lower pH environment due to the structural transformation of PAA and then transformed into NO production of 64.5 ± 1.6% under PTT. The combination of PTT and NO gas therapy can effectively eliminate cancer cells, resulting in a large amount of tumor-associated antigens (TAAs) compared to the individual treatment in vitro. Additionally, the released 1-M-DT inhibited IDO and combined with TAAs to enhance maturation of dendritic cells (DCs), indicating the excellent synergistic effect of PTT and NO with IDO inhibitors. These results revealed that this dual-sensitive nanoparticle presented a combination strategy of PTT/NO/IDO for the synergistic effect to promote DC maturation.
Collapse
|
12
|
Virok DP, Tömösi F, Keller-Pintér A, Szabó K, Bogdanov A, Poliska S, Rázga Z, Bruszel B, Cseh Z, Kókai D, Paróczai D, Endrész V, Janáky T, Burián K. Indoleamine 2,3-Dioxygenase Cannot Inhibit Chlamydia trachomatis Growth in HL-60 Human Neutrophil Granulocytes. Front Immunol 2021; 12:717311. [PMID: 34819931 PMCID: PMC8606673 DOI: 10.3389/fimmu.2021.717311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 10/18/2021] [Indexed: 12/05/2022] Open
Abstract
Aims Neutrophil granulocytes are the major cells involved in Chlamydia trachomatis (C. trachomatis)-mediated inflammation and histopathology. A key protein in human intracellular antichlamydial defense is the tryptophan-degrading enzyme indoleamine 2,3-dioxygenase (IDO) which limits the growth of the tryptophan auxotroph Chlamydia. Despite its importance, the role of IDO in the intracellular defense against Chlamydia in neutrophils is not well characterized. Methods Global gene expression screen was used to evaluate the effect of C. trachomatis serovar D infection on the transcriptome of human neutrophil granulocytes. Tryptophan metabolite concentrations in the Chlamydia-infected and/or interferon-gamma (IFNG)-treated neutrophils were measured by ultra-high-performance liquid chromatography–tandem mass spectrometry (UHPLC–MS/MS). Results Our results indicate that the C. trachomatis infection had a major impact on neutrophil gene expression, inducing 1,295 genes and repressing 1,510 genes. A bioinformatics analysis revealed that important factors involved in the induction of neutrophil gene expression were the interferon-related transcription factors such as IRF1-5, IRF7-9, STAT2, ICSB, and ISGF3. One of the upregulated genes was ido1, a known infection- and interferon-induced host gene. The tryptophan-degrading activity of IDO1 was not induced significantly by Chlamydia infection alone, but the addition of IFNG greatly increased its activity. Despite the significant IDO activity in IFNG-treated cells, C. trachomatis growth was not affected by IFNG. This result was in contrast to what we observed in HeLa human cervical epithelial cells, where the IFNG-mediated inhibition of C. trachomatis growth was significant and the IFNG-induced IDO activity correlated with growth inhibition. Conclusions IDO activity was not able to inhibit chlamydial growth in human neutrophils. Whether the IDO activity was not high enough for inhibition or other chlamydial growth-promoting host mechanisms were induced in the infected and interferon-treated neutrophils needs to be further investigated.
Collapse
Affiliation(s)
- Dezső P Virok
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Ferenc Tömösi
- Department of Medical Chemistry, Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | | | - Kitti Szabó
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - Anita Bogdanov
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Szilárd Poliska
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Zsolt Rázga
- Department of Pathology, University of Szeged, Szeged, Hungary
| | - Bella Bruszel
- Department of Medical Chemistry, Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Zsuzsanna Cseh
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Dávid Kókai
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Dóra Paróczai
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Valéria Endrész
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Tamás Janáky
- Department of Medical Chemistry, Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Katalin Burián
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
13
|
Tryptophan: From Diet to Cardiovascular Diseases. Int J Mol Sci 2021; 22:ijms22189904. [PMID: 34576067 PMCID: PMC8472285 DOI: 10.3390/ijms22189904] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/02/2021] [Accepted: 09/11/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease (CVD) is one of the major causes of mortality worldwide. Inflammation is the underlying common mechanism involved in CVD. It has been recently related to amino acid metabolism, which acts as a critical regulator of innate and adaptive immune responses. Among different metabolites that have emerged as important regulators of immune and inflammatory responses, tryptophan (Trp) metabolites have been shown to play a pivotal role in CVD. Here, we provide an overview of the fundamental aspects of Trp metabolism and the interplay between the dysregulation of the main actors involved in Trp metabolism such as indoleamine 2, 3-dioxygenase 1 (IDO) and CVD, including atherosclerosis and myocardial infarction. IDO has a prominent and complex role. Its activity, impacting on several biological pathways, complicates our understanding of its function, particularly in CVD, where it is still under debate. The discrepancy of the observed IDO effects could be potentially explained by its specific cell and tissue contribution, encouraging further investigations regarding the role of this enzyme. Thus, improving our understanding of the function of Trp as well as its derived metabolites will help to move one step closer towards tailored therapies aiming to treat CVD.
Collapse
|
14
|
Hautbergue T, Antigny F, Boët A, Haddad F, Masson B, Lambert M, Delaporte A, Menager JB, Savale L, Pavec JL, Fadel E, Humbert M, Junot C, Fenaille F, Colsch B, Mercier O. Right Ventricle Remodeling Metabolic Signature in Experimental Pulmonary Hypertension Models of Chronic Hypoxia and Monocrotaline Exposure. Cells 2021; 10:1559. [PMID: 34205639 PMCID: PMC8235667 DOI: 10.3390/cells10061559] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Over time and despite optimal medical management of patients with pulmonary hypertension (PH), the right ventricle (RV) function deteriorates from an adaptive to maladaptive phenotype, leading to RV failure (RVF). Although RV function is well recognized as a prognostic factor of PH, no predictive factor of RVF episodes has been elucidated so far. We hypothesized that determining RV metabolic alterations could help to understand the mechanism link to the deterioration of RV function as well as help to identify new biomarkers of RV failure. METHODS In the current study, we aimed to characterize the metabolic reprogramming associated with the RV remodeling phenotype during experimental PH induced by chronic-hypoxia-(CH) exposure or monocrotaline-(MCT) exposure in rats. Three weeks after PH initiation, we hemodynamically characterized PH (echocardiography and RV catheterization), and then we used an untargeted metabolomics approach based on liquid chromatography coupled to high-resolution mass spectrometry to analyze RV and LV tissues in addition to plasma samples from MCT-PH and CH-PH rat models. RESULTS CH exposure induced adaptive RV phenotype as opposed to MCT exposure which induced maladaptive RV phenotype. We found that predominant alterations of arginine, pyrimidine, purine, and tryptophan metabolic pathways were detected on the heart (LV+RV) and plasma samples regardless of the PH model. Acetylspermidine, putrescine, guanidinoacetate RV biopsy levels, and cytosine, deoxycytidine, deoxyuridine, and plasmatic thymidine levels were correlated to RV function in the CH-PH model. It was less likely correlated in the MCT model. These pathways are well described to regulate cell proliferation, cell hypertrophy, and cardioprotection. These findings open novel research perspectives to find biomarkers for early detection of RV failure in PH.
Collapse
Affiliation(s)
- Thaïs Hautbergue
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - Fabrice Antigny
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Angèle Boët
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Réanimation des Cardiopathies Congénitales, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - François Haddad
- Cardiovascular Medicine, Stanford Hospital, Stanford University, Stanford, CA 94305, USA;
| | - Bastien Masson
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Mélanie Lambert
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Amélie Delaporte
- Service d’Anesthésie, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France;
| | - Jean-Baptiste Menager
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - Laurent Savale
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Jérôme Le Pavec
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - Elie Fadel
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - Marc Humbert
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Christophe Junot
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - François Fenaille
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - Benoit Colsch
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - Olaf Mercier
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| |
Collapse
|
15
|
Queiroz RF, Suarna C, Corcilius L, Sergeant GE, Shengule S, Payne RJ, Ayer A, Stocker R. Preparation, validation and use of a vasoactive tryptophan-derived hydroperoxide and relevant control compounds. Nat Protoc 2021; 16:3382-3418. [PMID: 34117477 DOI: 10.1038/s41596-021-00541-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 03/22/2021] [Indexed: 11/09/2022]
Abstract
The L-tryptophan-derived tricyclic hydroperoxide cis-WOOH was recently identified as a novel and biologically important factor for regulating vascular tone and blood pressure under inflammatory conditions and potentially other cellular redox signaling events. cis-WOOH is highly labile and currently not available commercially. In this protocol, we provide procedures for the synthesis, purification, quantification and characterization of cis-WOOH, its epimer trans-WOOH and their respective alcohols (cis-WOH and trans-WOH). Photo-oxidation of L-tryptophan (L-Trp) results in a mixture containing cis-WOOH and trans-WOOH, which are separated and purified by semi-preparative HPLC. cis-WOH and trans-WOH are then produced by sodium borohydride reduction and purified by semi-preparative HPLC. Characterization of cis-WOOH and trans-WOOH and the reduced alcohol variants is achieved using HPLC, fluorescence, NMR and liquid chromatography-tandem mass spectrometry. The protocol provides instructions for storage and quantification, as well as ways to test the stability of these hydroperoxides in commonly used buffers and media. Finally, we describe examples of how to monitor the formation of cis-WOOH in biological samples. The protocol ensures reasonable yield (11%) and purity (>99%) of cis-WOOH and control compounds in 5-6 d and outlines conditions under which cis-WOOH is stable for several months.
Collapse
Affiliation(s)
- Raphael F Queiroz
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,Department of Natural Sciences, Southwest Bahia State University, Vitoria da Conquista, Brazil
| | - Cacang Suarna
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Leo Corcilius
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia
| | - Genevieve E Sergeant
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia
| | - Sudhir Shengule
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,Starpharma Pty Ltd, Abbotsford, Melbourne, Victoria, Australia
| | - Richard J Payne
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Brisbane, Queensland, Australia
| | - Anita Ayer
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia. .,The Heart Research Institute, Sydney, New South Wales, Australia. .,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| | - Roland Stocker
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia. .,The Heart Research Institute, Sydney, New South Wales, Australia. .,School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
16
|
The kynurenine pathway in major depression: What we know and where to next. Neurosci Biobehav Rev 2021; 127:917-927. [PMID: 34029552 DOI: 10.1016/j.neubiorev.2021.05.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/27/2022]
Abstract
Major depression is a serious psychiatric disorder, occurring in up to 20 % of the population. Despite its devastating burden, the neurobiological changes associated with depression are not fully understood. A growing body of evidence suggests the kynurenine pathway is implicated in the pathophysiology of depression. In this review, we bring together the literature examining elements of the kynurenine pathway in depression and explore the implications for the pathophysiology and treatment of depression, while highlighting the gaps in the current knowledge. Current research indicates an increased potential for neurotoxic activity of the kynurenine pathway in peripheral blood samples but an increased activation of the putative neuroprotective arm in some brain regions in depression. The disconnect between these findings requires further investigation, with a greater research effort on elucidating the central effects of the kynurenine pathway in driving depression symptomology. Research investigating the benefits of targeting the kynurenine pathway centred on human brain findings and the heterogenous subtypes of depression will help guide the identification of effective drug targets in depression.
Collapse
|
17
|
Pieretti JC, Rubilar O, Weller RB, Tortella GR, Seabra AB. Nitric oxide (NO) and nanoparticles - Potential small tools for the war against COVID-19 and other human coronavirus infections. Virus Res 2021; 291:198202. [PMID: 33086123 PMCID: PMC7568847 DOI: 10.1016/j.virusres.2020.198202] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022]
Abstract
The endogenous free radical nitric oxide (NO) plays a pivotal role in the immunological system. NO has already been reported as a potential candidate for use in the treatment of human coronavirus infections, including COVID-19. In fact, inhaled NO has been used in clinical settings for its antiviral respiratory action, and in the regulation of blood pressure to avoid clot formation. In this mini-review, we discuss recent progress concerning the antivirus activity of NO in clinical, pre-clinical and research settings, and its beneficial effects in the treatment of clinical complications in patients infected with coronaviruses and other respiratory viral diseases, including COVID-19. We also highlight promising therapeutic effects of NO donors allied to nanomaterials to combat COVID-19 and other human coronavirus infections. Nanomaterials can be designed to deliver sustained, localized NO release directly at the desired application site, enhancing the beneficial effects of NO and minimizing the side effects. Challenges and perspectives are presented to open new fields of research.
Collapse
Affiliation(s)
- Joana C Pieretti
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil
| | - Olga Rubilar
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile; Centro de Excelencia en Investigación Biotecnologica Aplicada al Medio Ambiente (CIBAMA-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Richard B Weller
- Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Gonzalo R Tortella
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile; Centro de Excelencia en Investigación Biotecnologica Aplicada al Medio Ambiente (CIBAMA-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Amedea B Seabra
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil.
| |
Collapse
|
18
|
Stancioiu F, Papadakis GZ, Kteniadakis S, Izotov BN, Coleman MD, Spandidos DA, Tsatsakis A. A dissection of SARS‑CoV2 with clinical implications (Review). Int J Mol Med 2020; 46:489-508. [PMID: 32626922 PMCID: PMC7307812 DOI: 10.3892/ijmm.2020.4636] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
We are being confronted with the most consequential pandemic since the Spanish flu of 1918‑1920 to the extent that never before have 4 billion people quarantined simultaneously; to address this global challenge we bring to the forefront the options for medical treatment and summarize SARS‑CoV2 structure and functions, immune responses and known treatments. Based on literature and our own experience we propose new interventions, including the use of amiodarone, simvastatin, pioglitazone and curcumin. In mild infections (sore throat, cough) we advocate prompt local treatment for the naso‑pharynx (inhalations; aerosols; nebulizers); for moderate to severe infections we propose a tried‑and‑true treatment: the combination of arginine and ascorbate, administered orally or intravenously. The material is organized in three sections: i) Clinical aspects of COVID‑19; acute respiratory distress syndrome (ARDS); known treatments; ii) Structure and functions of SARS‑CoV2 and proposed antiviral drugs; iii) The combination of arginine‑ascorbate.
Collapse
Affiliation(s)
| | | | | | - Boris Nikovaevich Izotov
- Department of Analytical and Forensic Medical Toxicology, Sechenov University, 119991 Moscow, Russia
| | - Michael D. Coleman
- School of Life and Health Sciences, Aston University, B4 7ET Birmingham, UK
| | | | - Aristidis Tsatsakis
- Department of Analytical and Forensic Medical Toxicology, Sechenov University, 119991 Moscow, Russia
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
19
|
Di Meo S, Venditti P. Evolution of the Knowledge of Free Radicals and Other Oxidants. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9829176. [PMID: 32411336 PMCID: PMC7201853 DOI: 10.1155/2020/9829176] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/31/2020] [Indexed: 12/11/2022]
Abstract
Free radicals are chemical species (atoms, molecules, or ions) containing one or more unpaired electrons in their external orbitals and generally display a remarkable reactivity. The evidence of their existence was obtained only at the beginning of the 20th century. Chemists gradually ascertained the involvement of free radicals in organic reactions and, in the middle of the 20th century, their production in biological systems. For several decades, free radicals were thought to cause exclusively damaging effects . This idea was mainly supported by the finding that oxygen free radicals readily react with all biological macromolecules inducing their oxidative modification and loss of function. Moreover, evidence was obtained that when, in the living organism, free radicals are not neutralized by systems of biochemical defences, many pathological conditions develop. However, after some time, it became clear that the living systems not only had adapted to the coexistence with free radicals but also developed methods to turn these toxic substances to their advantage by using them in critical physiological processes. Therefore, free radicals play a dual role in living systems: they are toxic by-products of aerobic metabolism, causing oxidative damage and tissue dysfunction, and serve as molecular signals activating beneficial stress responses. This discovery also changed the way we consider antioxidants. Their use is usually regarded as helpful to counteract the damaging effects of free radicals but sometimes is harmful as it can block adaptive responses induced by low levels of radicals.
Collapse
Affiliation(s)
- Sergio Di Meo
- Università degli Studi di Napoli Federico II Dipartimento di Biologia, Complesso, Universitario Monte Sant'Angelo, Via Cinthia, I-80126 Napoli, Italy
| | - Paola Venditti
- Università degli Studi di Napoli Federico II Dipartimento di Biologia, Complesso, Universitario Monte Sant'Angelo, Via Cinthia, I-80126 Napoli, Italy
| |
Collapse
|
20
|
Kang JS. Dietary restriction of amino acids for Cancer therapy. Nutr Metab (Lond) 2020; 17:20. [PMID: 32190097 PMCID: PMC7071719 DOI: 10.1186/s12986-020-00439-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/06/2020] [Indexed: 12/14/2022] Open
Abstract
Biosyntheses of proteins, nucleotides and fatty acids, are essential for the malignant proliferation and survival of cancer cells. Cumulating research findings show that amino acid restrictions are potential strategies for cancer interventions. Meanwhile, dietary strategies are popular among cancer patients. However, there is still lacking solid rationale to clarify what is the best strategy, why and how it is. Here, integrated analyses and comprehensive summaries for the abundances, signalling and functions of amino acids in proteomes, metabolism, immunity and food compositions, suggest that, intermittent dietary lysine restriction with normal maize as an intermittent staple food for days or weeks, might have the value and potential for cancer prevention or therapy. Moreover, dietary supplements were also discussed for cancer cachexia including dietary immunomodulatory.
Collapse
Affiliation(s)
- Jian-Sheng Kang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| |
Collapse
|
21
|
Boros FA, Vécsei L. Immunomodulatory Effects of Genetic Alterations Affecting the Kynurenine Pathway. Front Immunol 2019; 10:2570. [PMID: 31781097 PMCID: PMC6851023 DOI: 10.3389/fimmu.2019.02570] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/16/2019] [Indexed: 12/15/2022] Open
Abstract
Several enzymes and metabolites of the kynurenine pathway (KP) have immunomodulatory effects. Modulation of the activities and levels of these molecules might be of particular importance under disease conditions when the amelioration of overreacting immune responses is desired. Results obtained by the use of animal and tissue culture models indicate that by eliminating or decreasing activities of key enzymes of the KP, a beneficial shift in disease outcome can be attained. This review summarizes experimental data of models in which IDO, TDO, or KMO activity modulation was achieved by interventions affecting enzyme production at a genomic level. Elimination of IDO activity was found to improve the outcome of sepsis, certain viral infections, chronic inflammation linked to diabetes, obesity, aorta aneurysm formation, and in anti-tumoral processes. Similarly, lack of TDO activity was advantageous in the case of anti-tumoral immunity, while KMO inhibition was found to be beneficial against microorganisms and in the combat against tumors, as well. On the other hand, the complex interplay among KP metabolites and immune function in some cases requires an increase in a particular enzyme activity for the desired immune response modulation, as was shown by the exacerbation of liver fibrosis due to the elimination of IDO activity and the detrimental effects of TDO inhibition in a mouse model of autoimmune gastritis. The relevance of these studies concerning possible human applications are discussed and highlighted. Finally, a brief overview is presented on naturally occurring genetic variants affecting immune functions via modulation of KP enzyme activity.
Collapse
Affiliation(s)
- Fanni A. Boros
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
- MTA-SZTE Neuroscience Research Group of the Hungarian Academy of Sciences, University of Szeged, Szeged, Hungary
- Department of Neurology, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| |
Collapse
|
22
|
Mondanelli G, Iacono A, Allegrucci M, Puccetti P, Grohmann U. Immunoregulatory Interplay Between Arginine and Tryptophan Metabolism in Health and Disease. Front Immunol 2019; 10:1565. [PMID: 31354721 PMCID: PMC6629926 DOI: 10.3389/fimmu.2019.01565] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/24/2019] [Indexed: 12/30/2022] Open
Affiliation(s)
- Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Alberta Iacono
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Massimo Allegrucci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
23
|
Bando H, Lee Y, Sakaguchi N, Pradipta A, Sakamoto R, Tanaka S, Ma JS, Sasai M, Yamamoto M. Toxoplasma Effector GRA15-Dependent Suppression of IFN-γ-Induced Antiparasitic Response in Human Neurons. Front Cell Infect Microbiol 2019; 9:140. [PMID: 31119110 PMCID: PMC6504700 DOI: 10.3389/fcimb.2019.00140] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/16/2019] [Indexed: 01/06/2023] Open
Abstract
Toxoplasma gondii is an important human and animal pathogen that causes life-threatening toxoplasmosis. The host immune system produces interferon-γ (IFN-γ) to inhibit T. gondii proliferation. IFN-γ-inducible indole-2,3-dioxygenase 1 (IDO1), which mediates tryptophan degradation, has a major role in anti-T. gondii immune responses in various human cells. In response to the host's immune system, T. gondii secretes many virulence molecules into the host cells to suppress IFN-γ-dependent antiparasitic immune responses. The GRA15-induced proparasitic mechanism for suppressing IDO1-dependent immune responses has previously been tested only in human hepatocyte and monocyte co-cultures. Thus, whether human cells other than hepatocytes contain this virulence mechanism remains unclear. Here, we show that the GRA15-dependent virulence mechanism for suppressing the IDO1-dependent anti-T. gondii response operates in human neuronal cell lines and primary human neurons. Analysis of various human cell lines revealed that IL-1β-induced iNOS-dependent reduction of IDO1 mRNA expression occurred in brain cell lines (A172; glioblastoma, IMR-32; neuroblastoma, and T98G; glioblastoma) and liver cell lines (Huh7 and HepG2), but not in other cell lines. Moreover, co-culturing type II T. gondii-infected THP-1 human monocytes with the brain cell lines inhibited the IDO1-mediated anti-T. gondii response in a GRA15-dependent manner. These data suggest that a GRA15-dependent virulence mechanism antagonizes the IDO1-dependent host immune response in human brain cells.
Collapse
Affiliation(s)
- Hironori Bando
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Suita, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Youngae Lee
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Suita, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Naoya Sakaguchi
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Suita, Japan
| | - Ariel Pradipta
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Suita, Japan
| | - Ryoma Sakamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Suita, Japan
| | - Shun Tanaka
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Suita, Japan
| | - Ji Su Ma
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Suita, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Suita, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Suita, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
24
|
Mondanelli G, Iacono A, Carvalho A, Orabona C, Volpi C, Pallotta MT, Matino D, Esposito S, Grohmann U. Amino acid metabolism as drug target in autoimmune diseases. Autoimmun Rev 2019; 18:334-348. [DOI: 10.1016/j.autrev.2019.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 10/30/2018] [Indexed: 12/14/2022]
|
25
|
Lemos H, Huang L, Prendergast GC, Mellor AL. Immune control by amino acid catabolism during tumorigenesis and therapy. Nat Rev Cancer 2019; 19:162-175. [PMID: 30696923 DOI: 10.1038/s41568-019-0106-z] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Immune checkpoints arise from physiological changes during tumorigenesis that reprogramme inflammatory, immunological and metabolic processes in malignant lesions and local lymphoid tissues, which constitute the immunological tumour microenvironment (TME). Improving clinical responses to immune checkpoint blockade will require deeper understanding of factors that impact local immune balance in the TME. Elevated catabolism of the amino acids tryptophan (Trp) and arginine (Arg) is a common TME hallmark at clinical presentation of cancer. Cells catabolizing Trp and Arg suppress effector T cells and stabilize regulatory T cells to suppress immunity in chronic inflammatory diseases of clinical importance, including cancers. Processes that induce Trp and Arg catabolism in the TME remain incompletely defined. Indoleamine 2,3 dioxygenase (IDO) and arginase 1 (ARG1), which catabolize Trp and Arg, respectively, respond to inflammatory cues including interferons and transforming growth factor-β (TGFβ) cytokines. Dying cells generate inflammatory signals including DNA, which is sensed to stimulate the production of type I interferons via the stimulator of interferon genes (STING) adaptor. Thus, dying cells help establish local conditions that suppress antitumour immunity to promote tumorigenesis. Here, we review evidence that Trp and Arg catabolism contributes to inflammatory processes that promote tumorigenesis, impede immune responses to therapy and might promote neurological comorbidities associated with cancer.
Collapse
Affiliation(s)
- Henrique Lemos
- Institute of Cellular Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle-upon-Tyne, UK
| | - Lei Huang
- Institute of Cellular Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle-upon-Tyne, UK
| | | | - Andrew L Mellor
- Institute of Cellular Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle-upon-Tyne, UK.
| |
Collapse
|
26
|
Lim YJ, Foo TC, Yeung AWS, Tu X, Ma Y, Hawkins CL, Witting PK, Jameson GNL, Terentis AC, Thomas SR. Human Indoleamine 2,3-Dioxygenase 1 Is an Efficient Mammalian Nitrite Reductase. Biochemistry 2019; 58:974-986. [PMID: 30585477 DOI: 10.1021/acs.biochem.8b01231] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The heme enzyme indoleamine 2,3-dioxygenase-1 (IDO1) catalyzes the first reaction of l-tryptophan oxidation along the kynurenine pathway. IDO1 is a central immunoregulatory enzyme with important implications for inflammation, infectious disease, autoimmune disorders, and cancer. Here we demonstrate that IDO1 is a mammalian nitrite reductase capable of chemically reducing nitrite to nitric oxide (NO) under hypoxia. Ultraviolet-visible absorption and resonance Raman spectroscopy showed that incubation of dithionite-reduced, ferrous-IDO1 protein (FeII-IDO1) with nitrite under anaerobic conditions resulted in the time-dependent formation of an FeII-nitrosyl IDO1 species, which was inhibited by substrate l-tryptophan, dependent on the concentration of nitrite or IDO1, and independent of the concentration of the reductant, dithionite. The bimolecular rate constant for IDO1 nitrite reductase activity was determined as 5.4 M-1 s-1 (pH 7.4, 23 °C), which was comparable to that measured for myoglobin (3.6 M-1 s-1; pH 7.4, 23 °C), an efficient and biologically important mammalian heme-based nitrite reductase. IDO1 nitrite reductase activity was pH-dependent but differed with myoglobin in that it showed a reduced proton dependency at pH >7. Electron paramagnetic resonance studies measuring NO production showed that the conventional IDO1 dioxygenase reducing cofactors, ascorbate and methylene blue, enhanced IDO1's nitrite reductase activity and the time- and IDO1 concentration-dependent release of NO in a manner inhibited by l-tryptophan or the IDO inhibitor 1-methyl-l-tryptophan. These data identify IDO1 as an efficient mammalian nitrite reductase that is capable of generating NO under anaerobic conditions. IDO1's nitrite reductase activity may have important implications for the enzyme's biological actions when expressed within hypoxic tissues.
Collapse
Affiliation(s)
| | - Timothy C Foo
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | | | | | | - Clare L Hawkins
- Department of Biomedical Sciences , University of Copenhagen , Copenhagen N DK-2200 , Denmark
| | - Paul K Witting
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health , University of Sydney , Sydney , NSW 2006 , Australia
| | - Guy N L Jameson
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Parkville , VIC 3010 , Australia
| | - Andrew C Terentis
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | |
Collapse
|
27
|
Jesus LB, Santos AB, Jesus EEV, Santos RGD, Grangeiro MS, Bispo-da-Silva A, Arruda MR, Argolo DS, Pinheiro AM, El-Bachá RS, Costa SL, Costa MFD. IDO, COX and iNOS have an important role in the proliferation of Neospora caninum in neuron/glia co-cultures. Vet Parasitol 2019; 266:96-102. [PMID: 30736955 DOI: 10.1016/j.vetpar.2019.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 01/03/2019] [Accepted: 01/05/2019] [Indexed: 12/23/2022]
Abstract
Central nervous system (CNS) is the main site for encystment of Neospora caninum in different animal species. In this tissue, glial cells (astrocytes and microglia) modulate responses to aggression in order to preserve homeostasis and neuronal function. Previous data showed that when primary cultures of glial cells are infected with N. caninum, they develop gliosis and the immune response is characterized by the release of TNF and IL-10, followed by the control of parasite proliferation. In order to elucidate this control, three enzymatic systems involved in parasite-versus-host interactions were observed on a model of neuron/glia co/cultures obtained from rat brains. Indoleamine 2,3-dioxygenase (IDO), induced nitric oxide synthase (iNOS) responsible for the catabolism of tryptophan and arginine, respectively, and cycloxigenase (COX) were studied comparing their modulation by respective inhibitors with the number of tachyzoites or the immune response measured by the release of IL-10 and TNF. Cells were treated with the inhibitors of iNOS (1.5 mM L-NAME), IDO (1 mM 1-methyl tryptophan), COX-1 (1 μM indomethacin) and COX-2 (1 μM nimesulide) before infection with tachyzoites of N. caninum (1:1 cell: parasite). After 72 h of infection, immunocytochemistry showed astrogliosis and a significant increase in the number and length of neurites, compared with uninfected co-cultures, while an increase of IL-10 and TNF was verified. N. caninum did not change iNOS activity, but the inhibition of the basal levels of this enzyme stimulated parasite proliferation. Additionally, a significant increase of about 40% was verified in the IDO activity, whose inhibition caused 1.2-fold increase in parasitic growth. For COX-2 activity, infection of cultures stimulated a significant increase in release of PGE2 and its inhibition by nimesulide allowed the parasitic growth. These data indicate that iNOS, IDO and COX-2 control the proliferation of N. caninum in this in vitro model. On the other hand, the release of IL-10 by glia besides modulating the inflammation also allow the continuity of parasitism.
Collapse
Affiliation(s)
- L B Jesus
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - A B Santos
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - E E V Jesus
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - R G D Santos
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - M S Grangeiro
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - A Bispo-da-Silva
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - M R Arruda
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - D S Argolo
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil
| | - A M Pinheiro
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; Centro de Ciências Agrárias Ambientais e Biológica, Universidade do Recôncavo da Bahia - URBA, R. Ruy Barbosa 710 Centro, CEP 44380-000, Cruz das Almas, Bahia, Brazil
| | - R S El-Bachá
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; INCT de Neurociência Translacional (INNT)- CNPq, Brazil
| | - S L Costa
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; INCT de Neurociência Translacional (INNT)- CNPq, Brazil.
| | - M F D Costa
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, CEP 41100-100, Salvador, Bahia, Brazil; INCT de Neurociência Translacional (INNT)- CNPq, Brazil.
| |
Collapse
|
28
|
Inducible Nitric Oxide Synthase Is a Key Host Factor for Toxoplasma GRA15-Dependent Disruption of the Gamma Interferon-Induced Antiparasitic Human Response. mBio 2018; 9:mBio.01738-18. [PMID: 30301855 PMCID: PMC6178625 DOI: 10.1128/mbio.01738-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Although Toxoplasma virulence mechanisms targeting gamma interferon (IFN-γ)-induced cell-autonomous antiparasitic immunity have been extensively characterized in mice, the virulence mechanisms in humans remain uncertain, partly because cell-autonomous immune responses against Toxoplasma differ markedly between mice and humans. Despite the identification of inducible nitric oxide synthase (iNOS) as an anti-Toxoplasma host factor in mice, here we show that iNOS in humans is a pro-Toxoplasma host factor that promotes the growth of the parasite. The GRA15 Toxoplasma effector-dependent disarmament of IFN-γ-induced parasite growth inhibition was evident when parasite-infected monocytes were cocultured with hepatocytes. Interleukin-1β (IL-1β), produced from monocytes in a manner dependent on GRA15 and the host's NLRP3 inflammasome, combined with IFN-γ to strongly stimulate iNOS expression in hepatocytes; this dramatically reduced the levels of indole 2,3-dioxygenase 1 (IDO1), a critically important IFN-γ-inducible anti-Toxoplasma protein in humans, thus allowing parasite growth. Taking the data together, Toxoplasma utilizes human iNOS to antagonize IFN-γ-induced IDO1-mediated cell-autonomous immunity via its GRA15 virulence factor.IMPORTANCE Toxoplasma, an important intracellular parasite of humans and animals, causes life-threatening toxoplasmosis in immunocompromised individuals. Gamma interferon (IFN-γ) is produced in the host to inhibit the proliferation of this parasite and eventually cause its death. Unlike mouse disease models, which involve well-characterized virulence strategies that are used by Toxoplasma to suppress IFN-γ-dependent immunity, the strategies used by Toxoplasma in humans remain unclear. Here, we show that GRA15, a Toxoplasma effector protein, suppresses the IFN-γ-induced indole-2,3-dioxygenase 1-dependent antiparasite immune response in human cells. Because NLRP3-dependent production of IL-1β and nitric oxide (NO) in Toxoplasma-infected human cells is involved in the GRA15-dependent virulence mechanism, blocking NO or IL-1β production in the host could represent a novel therapeutic approach for treating human toxoplasmosis.
Collapse
|
29
|
Eleftheriadis T. What May Constrain the Success of Indoleamine 2,3-Dioxygenase 1 Inhibitors in Cancer Immunotherapy? Front Immunol 2018; 9:1879. [PMID: 30150994 PMCID: PMC6099423 DOI: 10.3389/fimmu.2018.01879] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 07/30/2018] [Indexed: 01/06/2023] Open
|
30
|
Langrzyk A, Nowak WN, Stępniewski J, Jaźwa A, Florczyk-Soluch U, Józkowicz A, Dulak J. Critical View on Mesenchymal Stromal Cells in Regenerative Medicine. Antioxid Redox Signal 2018; 29:169-190. [PMID: 28874054 DOI: 10.1089/ars.2017.7159] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE The belief in the potency of stem cells has resulted in the medical applications of numerous cell types for organ repair, often with the low adherence to methodological stringency. Such uncritical enthusiasm is mainly presented in the approaches employing so-called mesenchymal stem cells (MSC), for the treatment of numerous, unrelated conditions. However, it should be stressed that such broad clinical applications of MSC are mostly based on the belief that MSC can efficiently differentiate into multiple cell types, not only osteoblasts, chondrocytes and adipose cells. Recent Advances: Studies employing lineage tracing established more promising markers to characterize MSC identity and localization in vivo and confirmed the differences between MSC isolated from various organs. Furthermore, preclinical and clinical experiments proved that transdifferentiation of MSC is unlikely to contribute to repair of numerous tissues, including the heart. Therefore, the salvage hypotheses, like MSC fusion with cells in target organs or the paracrine mechanisms, were proposed to justify the widespread application of MSC and to explain transient, if any, effects. CRITICAL ISSUES The lack of standardization concerning the cells markers, their origin and particularly the absence of stringent functional characterization of MSC, leads to propagation of the worrying hype despite the lack of convincing therapeutic efficiency of MSC. FUTURE DIRECTIONS The adherence to rigorous methodological rules is necessary to prevent the application of procedures which can be dangerous for patients and scientific research on the medical application of stem cells. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
| | - Witold N Nowak
- 2 Cardiovascular Division, King's College London , London, United Kingdom .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Jacek Stępniewski
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Agnieszka Jaźwa
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Urszula Florczyk-Soluch
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Alicja Józkowicz
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Józef Dulak
- 1 Kardio-Med Silesia , Zabrze, Poland .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| |
Collapse
|
31
|
Grayfer L, Kerimoglu B, Yaparla A, Hodgkinson JW, Xie J, Belosevic M. Mechanisms of Fish Macrophage Antimicrobial Immunity. Front Immunol 2018; 9:1105. [PMID: 29892285 PMCID: PMC5985312 DOI: 10.3389/fimmu.2018.01105] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/02/2018] [Indexed: 12/13/2022] Open
Abstract
Overcrowding conditions and temperatures shifts regularly manifest in large-scale infections of farmed fish, resulting in economic losses for the global aquaculture industries. Increased understanding of the functional mechanisms of fish antimicrobial host defenses is an important step forward in prevention of pathogen-induced morbidity and mortality in aquaculture setting. Like other vertebrates, macrophage-lineage cells are integral to fish immune responses and for this reason, much of the recent fish immunology research has focused on fish macrophage biology. These studies have revealed notable similarities as well as striking differences in the molecular strategies by which fish and higher vertebrates control their respective macrophage polarization and functionality. In this review, we address the current understanding of the biological mechanisms of teleost macrophage functional heterogeneity and immunity, focusing on the key cytokine regulators that control fish macrophage development and their antimicrobial armamentarium.
Collapse
Affiliation(s)
- Leon Grayfer
- Department of Biological Sciences, George Washington University, Washington, DC, United States
| | - Baris Kerimoglu
- Department of Biological Sciences, George Washington University, Washington, DC, United States
| | - Amulya Yaparla
- Department of Biological Sciences, George Washington University, Washington, DC, United States
| | | | - Jiasong Xie
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Miodrag Belosevic
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
32
|
Volpini X, Ambrosio LF, Fozzatti L, Insfran C, Stempin CC, Cervi L, Motran CC. Trypanosoma cruzi Exploits Wnt Signaling Pathway to Promote Its Intracellular Replication in Macrophages. Front Immunol 2018; 9:859. [PMID: 29743880 PMCID: PMC5930390 DOI: 10.3389/fimmu.2018.00859] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 04/06/2018] [Indexed: 01/04/2023] Open
Abstract
During the acute phase of Trypanosoma cruzi infection, macrophages can act as host cells for the parasites as well as effector cells in the early anti-parasitic immune response. Thus, the targeting of specific signaling pathways could modulate macrophages response to restrict parasite replication and instruct an appropriate adaptive response. Recently, it has become evident that Wnt signaling has immunomodulatory functions during inflammation and infection. Here, we tested the hypothesis that during T. cruzi infection, the activation of Wnt signaling pathway in macrophages plays a role in modulating the inflammatory/tolerogenic response and therefore regulating the control of parasite replication. In this report, we show that early after T. cruzi infection of bone marrow-derived macrophages (BMM), β-catenin was activated and Wnt3a, Wnt5a, and some Frizzled receptors as well as Wnt/β-catenin pathway’s target genes were upregulated, with Wnt proteins signaling sustaining the activation of Wnt/β-catenin pathway and then activating the Wnt/Ca+2 pathway. Wnt signaling pathway activation was critical to sustain the parasite’s replication in BMM; since the treatments with specific inhibitors of β-catenin transcriptional activation or Wnt proteins secretion limited the parasite replication. Mechanistically, inhibition of Wnt signaling pathway armed BMM to fight against T. cruzi by inducing the production of pro-inflammatory cytokines and indoleamine 2,3-dioxygenase activity and by downregulating arginase activity. Likewise, in vivo pharmacological inhibition of the Wnts’ interaction with its receptors controlled the parasite replication and improved the survival of lethally infected mice. It is well established that T. cruzi infection activates a plethora of signaling pathways that ultimately regulate immune mediators to determine the modulation of a defined set of effector functions in macrophages. In this study, we have revealed a new signaling pathway that is activated by the interaction between protozoan parasites and host innate immunity, establishing a new conceptual framework for the development of new therapies.
Collapse
Affiliation(s)
- Ximena Volpini
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, Argentina
| | - Laura F Ambrosio
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, Argentina
| | - Laura Fozzatti
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, Argentina
| | - Constanza Insfran
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, Argentina
| | - Cinthia C Stempin
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, Argentina
| | - Laura Cervi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, Argentina
| | - Claudia Cristina Motran
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, Argentina
| |
Collapse
|
33
|
Endothelial indoleamine 2,3-dioxygenase-1 regulates the placental vascular tone and is deficient in intrauterine growth restriction and pre-eclampsia. Sci Rep 2018; 8:5488. [PMID: 29615752 PMCID: PMC5883010 DOI: 10.1038/s41598-018-23896-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 03/21/2018] [Indexed: 11/16/2022] Open
Abstract
Indoleamine 2,3-dioxygenase-1 (IDO1) mediates the degradation of L-tryptophan (L-Trp) and is constitutively expressed in the chorionic vascular endothelium of the human placenta with highest levels in the microvasculature. Given that endothelial expression of IDO1 has been shown to regulate vascular tone and blood pressure in mice under the condition of systemic inflammation, we asked whether IDO1 is also involved in the regulation of placental blood flow and if yes, whether this function is potentially impaired in intrauterine growth restriction (IUGR) and pre-eclampsia (PE). In the large arteries of the chorionic plate L-Trp induced relaxation only after upregulation of IDO1 using interferon gamma and tumor necrosis factor alpha. However, ex vivo placental perfusion of pre-constricted cotyledonic vasculature with L-Trp decreases the vessel back pressure without prior IDO1 induction. Further to this finding, IDO1 protein expression and activity is reduced in IUGR and PE when compared to gestational age–matched control tissue. These data suggest that L-Trp catabolism plays a role in the regulation of placental vascular tone, a finding which is potentially linked to placental and fetal growth. In this context our data suggest that IDO1 deficiency is related to the pathogenesis of IUGR and PE.
Collapse
|
34
|
Hornyák L, Dobos N, Koncz G, Karányi Z, Páll D, Szabó Z, Halmos G, Székvölgyi L. The Role of Indoleamine-2,3-Dioxygenase in Cancer Development, Diagnostics, and Therapy. Front Immunol 2018; 9:151. [PMID: 29445380 PMCID: PMC5797779 DOI: 10.3389/fimmu.2018.00151] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 01/17/2018] [Indexed: 01/03/2023] Open
Abstract
Tumors are composed of abnormally transformed cell types and tissues that differ from normal tissues in their genetic and epigenetic makeup, metabolism, and immunology. Molecular compounds that modulate the immune response against neoplasms offer promising new strategies to combat cancer. Inhibitors targeting the indoleamine-2,3-dioxygenase 1 enzyme (IDO1) represent one of the most potent therapeutic opportunities to inhibit tumor growth. Herein, we assess the biochemical role of IDO1 in tumor metabolism and immune surveillance, and review current diagnostic and therapeutic approaches that are intended to increase the effectiveness of immunotherapies against highly aggressive and difficult-to-treat IDO-expressing cancers.
Collapse
Affiliation(s)
- Lilla Hornyák
- MTA-DE Momentum Genome Architecture and Recombination Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nikoletta Dobos
- Department of Biopharmacy, Faculty of Pharmacology, University of Debrecen, Debrecen, Hungary
| | - Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Karányi
- MTA-DE Momentum Genome Architecture and Recombination Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dénes Páll
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szabó
- Department of Emergency Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Halmos
- Department of Biopharmacy, Faculty of Pharmacology, University of Debrecen, Debrecen, Hungary
| | - Lóránt Székvölgyi
- MTA-DE Momentum Genome Architecture and Recombination Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
35
|
Matysik-Woźniak A, Paduch R, Turski WA, Maciejewski R, Jünemann AG, Rejdak R. Effects of tryptophan, kynurenine and kynurenic acid exerted on human reconstructed corneal epithelium in vitro. Pharmacol Rep 2017; 69:722-729. [DOI: 10.1016/j.pharep.2017.02.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 02/21/2017] [Accepted: 02/24/2017] [Indexed: 11/28/2022]
|
36
|
de Araújo EF, Feriotti C, Galdino NADL, Preite NW, Calich VLG, Loures FV. The IDO-AhR Axis Controls Th17/Treg Immunity in a Pulmonary Model of Fungal Infection. Front Immunol 2017; 8:880. [PMID: 28791025 PMCID: PMC5523665 DOI: 10.3389/fimmu.2017.00880] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/10/2017] [Indexed: 12/20/2022] Open
Abstract
In infectious diseases, the enzyme indoleamine 2,3 dioxygenase-1 (IDO1) that catalyzes the tryptophan (Trp) degradation along the kynurenines (Kyn) pathway has two main functions, the control of pathogen growth by reducing available Trp and immune regulation mediated by the Kyn-mediated expansion of regulatory T (Treg) cells via aryl hydrocarbon receptor (AhR). In pulmonary paracoccidioidomycosis (PCM) caused by the dimorphic fungus Paracoccidioides brasiliensis, IDO1 was shown to control the disease severity of both resistant and susceptible mice to the infection; however, only in resistant mice, IDO1 is induced by TGF-β signaling that confers a stable tolerogenic phenotype to dendritic cells (DCs). In addition, in pulmonary PCM, the tolerogenic function of plasmacytoid dendritic cells was linked to the IDO1 activity. To further evaluate the function of IDO1 in pulmonary PCM, IDO1-deficient (IDO1-/-) C57BL/6 mice were intratracheally infected with P. brasiliensis yeasts and the infection analyzed at three postinfection periods regarding several parameters of disease severity and immune response. The fungal loads and tissue pathology of IDO1-/- mice were higher than their wild-type controls resulting in increased mortality rates. The evaluation of innate lymphoid cells showed an upregulated differentiation of the innate lymphoid cell 3 phenotype accompanied by a decreased expansion of ILC1 and NK cells in the lungs of infected IDO1-/- mice. DCs from these mice expressed elevated levels of costimulatory molecules and cytokine IL-6 associated with reduced production of IL-12, TNF-α, IL-1β, TGF-β, and IL-10. This response was concomitant with a marked reduction in AhR production. The absence of IDO1 expression caused an increased influx of activated Th17 cells to the lungs with a simultaneous reduction in Th1 and Treg cells. Accordingly, the suppressive cytokines IL-10, TGF-β, IL-27, and IL-35 appeared in reduced levels in the lungs of IDO1-/- mice. In conclusion, the immunological balance mediated by the axis IDO/AhR is fundamental to determine the balance between Th17/Treg cells and control the severity of pulmonary PCM.
Collapse
Affiliation(s)
- Eliseu Frank de Araújo
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Claudia Feriotti
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Nycolas Willian Preite
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Vera Lúcia Garcia Calich
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Flávio Vieira Loures
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
37
|
Koshiguchi M, Komazaki H, Hirai S, Egashira Y. Ferulic acid suppresses expression of tryptophan metabolic key enzyme indoleamine 2, 3-dioxygenase via NFκB and p38 MAPK in lipopolysaccharide-stimulated microglial cells. Biosci Biotechnol Biochem 2017; 81:966-971. [DOI: 10.1080/09168451.2016.1274636] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Abstract
Ferulic acid (FA) is a phenol compound found in plants that has anti-inflammatory properties. Indoleamine 2, 3-dioxygenase (IDO) is a tryptophan catabolic enzyme induced in immune cells, including glial cells, during inflammation. Enhanced IDO expression leads to reduced tryptophan levels and increased levels of toxic metabolites, including quinolinic acid. Therefore, inhibition of IDO expression may be effective in suppressing progression of neurodegenerative diseases. In this study, we examined the effect of FA in microglial cells on IDO expression levels and related inflammatory signal molecules. FA suppressed LPS-induced IDO mRNA expression and also suppressed nuclear translocation of NF-κB and phosphorylation of p38 MAPK. However, FA did not affect the production of LPS-induced inflammatory mediators and phosphorylation of JNK. Our results indicate that FA suppresses LPS-induced IDO mRNA expression, which may be mediated by inhibition of the NF-κB and p38 MAPK pathways in microglial cells.
Collapse
Affiliation(s)
- Manami Koshiguchi
- Laboratory of Food and Nutrition, Graduate School of Horticulture, Chiba University, Chiba, Japan
| | - Hitoshi Komazaki
- Laboratory of Food and Nutrition, Graduate School of Horticulture, Chiba University, Chiba, Japan
| | - Shizuka Hirai
- Laboratory of Food and Nutrition, Graduate School of Horticulture, Chiba University, Chiba, Japan
| | - Yukari Egashira
- Laboratory of Food and Nutrition, Graduate School of Horticulture, Chiba University, Chiba, Japan
| |
Collapse
|
38
|
Strasser B, Becker K, Fuchs D, Gostner JM. Kynurenine pathway metabolism and immune activation: Peripheral measurements in psychiatric and co-morbid conditions. Neuropharmacology 2017; 112:286-296. [DOI: 10.1016/j.neuropharm.2016.02.030] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 02/17/2016] [Accepted: 02/23/2016] [Indexed: 12/14/2022]
|
39
|
Huntingtons Disease Mice Infected with Toxoplasma gondii Demonstrate Early Kynurenine Pathway Activation, Altered CD8+ T-Cell Responses, and Premature Mortality. PLoS One 2016; 11:e0162404. [PMID: 27611938 PMCID: PMC5017698 DOI: 10.1371/journal.pone.0162404] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/10/2016] [Indexed: 12/03/2022] Open
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a polyglutamine-repeat expansion in the huntingtin protein. Activation of the kynurenine pathway of tryptophan degradation is implicated in the pathogenesis of HD. Indoleamine-2,3-dioxygenase (IDO) catalyzes the oxidation of tryptophan to kynurenine, the first step in this pathway. The prevalent, neuroinvasive protozoal pathogen Toxoplasma gondii (T. gondii) results in clinically silent life-long infection in immune-competent individuals. T. gondii infection results in activation of IDO which provides some protection against the parasite by depleting tryptophan which the parasite cannot synthesize. The kynurenine pathway may therefore represent a point of synergism between HD and T. gondii infection. We show here that IDO activity is elevated at least four-fold in frontal cortex and striata of non-infected N171-82Q HD mice at 14-weeks corresponding to early–advanced HD. T. gondii infection at 5 weeks resulted in elevation of cortical IDO activity in HD mice. HD-infected mice died significantly earlier than wild-type infected and HD control mice. Prior to death, infected HD mice demonstrated decreased CD8+ T-lymphocyte proliferation in brain and spleen compared to wild-type infected mice. We demonstrate for the first time that HD mice have an altered response to an infectious agent that is characterized by premature mortality, altered immune responses and early activation of IDO. Findings are relevant to understanding how T. gondii infection may interact with pathways mediating neurodegeneration in HD.
Collapse
|
40
|
Kettle AJ, Winterbourn CC. Myeloperoxidase: a key regulator of neutrophil oxidant production. Redox Rep 2016; 3:3-15. [PMID: 27414766 DOI: 10.1080/13510002.1997.11747085] [Citation(s) in RCA: 476] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
41
|
Lewis GD, Ngo D, Hemnes AR, Farrell L, Domos C, Pappagianopoulos PP, Dhakal BP, Souza A, Shi X, Pugh ME, Beloiartsev A, Sinha S, Clish CB, Gerszten RE. Metabolic Profiling of Right Ventricular-Pulmonary Vascular Function Reveals Circulating Biomarkers of Pulmonary Hypertension. J Am Coll Cardiol 2016; 67:174-189. [PMID: 26791065 DOI: 10.1016/j.jacc.2015.10.072] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/06/2015] [Accepted: 10/13/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Pulmonary hypertension and associated right ventricular (RV) dysfunction are important determinants of morbidity and mortality, which are optimally characterized by invasive hemodynamic measurements. OBJECTIVES This study sought to determine whether metabolite profiling could identify plasma signatures of right ventricular-pulmonary vascular (RV-PV) dysfunction. METHODS We measured plasma concentrations of 105 metabolites using targeted mass spectrometry in 71 individuals (discovery cohort) who underwent comprehensive physiological assessment with right-sided heart catheterization and radionuclide ventriculography at rest and during exercise. Our findings were validated in a second cohort undergoing invasive hemodynamic evaluations (n = 71), as well as in an independent cohort with or without known pulmonary arterial (PA) hypertension (n = 30). RESULTS In the discovery cohort, 21 metabolites were associated with 2 or more hemodynamic indicators of RV-PV function (i.e., resting right atrial pressure, mean PA pressure, pulmonary vascular resistance [PVR], and PVR and PA pressure-flow response [ΔPQ] during exercise). We identified novel associations of RV-PV dysfunction with circulating indoleamine 2,3-dioxygenase (IDO)-dependent tryptophan metabolites (TMs), tricarboxylic acid intermediates, and purine metabolites and confirmed previously described associations with arginine-nitric oxide metabolic pathway constituents. IDO-TM levels were inversely related to RV ejection fraction and were particularly well correlated with exercise PVR and ΔPQ. Multisite sampling demonstrated transpulmonary release of IDO-TMs. IDO-TMs also identified RV-PV dysfunction in a validation cohort with known risk factors for pulmonary hypertension and in patients with established PA hypertension. CONCLUSIONS Metabolic profiling identified reproducible signatures of RV-PV dysfunction, highlighting both new biomarkers and pathways for further functional characterization.
Collapse
Affiliation(s)
- Gregory D Lewis
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
| | - Debby Ngo
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anna R Hemnes
- Vanderbilt University Pulmonary Unit, Nashville, Tennessee
| | - Laurie Farrell
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Carly Domos
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Paul P Pappagianopoulos
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bishnu P Dhakal
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amanda Souza
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Xu Shi
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Arkadi Beloiartsev
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sumita Sinha
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Robert E Gerszten
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Broad Institute of MIT and Harvard, Cambridge, Massachusetts; Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
42
|
Remus JL, Dantzer R. Inflammation Models of Depression in Rodents: Relevance to Psychotropic Drug Discovery. Int J Neuropsychopharmacol 2016; 19:pyw028. [PMID: 27026361 PMCID: PMC5043641 DOI: 10.1093/ijnp/pyw028] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/23/2016] [Indexed: 01/06/2023] Open
Abstract
Inflammation and depression are closely inter-related; inflammation induces symptoms of depression and, conversely, depressed mood and stress favor an inflammatory phenotype. The mechanisms that mediate the ability of inflammation to induce symptoms of depression are intensively studied at the preclinical level. This review discusses how it has been possible to build animal models of inflammation-induced depression based on clinical data and to explore critical mechanisms downstream of inflammation. Namely, we focus on the ability of inflammation to increase the activity of the tryptophan-degrading enzyme, indoleamine 2,3 dioxygenase, which leads to the production of kynurenine and downstream neuroactive metabolites. By acting on glutamatergic neurotransmission, these neuroactive metabolites play a key role in the development of depression-like behaviors. An important outcome of the preclinical research on inflammation-induced depression is the identification of potential novel targets for antidepressant treatments, which include targeting the kynurenine system and production of downstream metabolites, altering transport of kynurenine into the brain, and modulating glutamatergic transmission.
Collapse
Affiliation(s)
- Jennifer L Remus
- Laboratory of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Robert Dantzer
- Laboratory of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
43
|
Dantzer R. Role of the Kynurenine Metabolism Pathway in Inflammation-Induced Depression: Preclinical Approaches. Curr Top Behav Neurosci 2016; 31:117-138. [PMID: 27225497 DOI: 10.1007/7854_2016_6] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Physically ill patients with chronic inflammation often present with symptoms of depression. Our understanding of the pathophysiology of inflammation-associated depression has benefited from preclinical studies on the mechanisms of sickness and clinical studies on the symptoms of sickness and depression that develop in patients treated with immunotherapy. Sickness behavior develops when the immune system is activated by pathogen- or damage-associated molecular patterns. It is a normal biological response to infection and cell injury. It helps the organism to mobilize its immune and metabolic defenses to fight the danger. Depression emerges on the background of sickness when the inflammatory response is too intense and long lasting or the resolution process is deficient. The transition from sickness to depression is mediated by activation of the kynurenine metabolism pathway that leads to the formation of neurotoxic kynurenine metabolites including quinolinic acid, an agonist of N-methyl-D-aspartate receptors. The neuroimmune processes and molecular factors that have been identified in the studies of inflammation-associated depression represent potential new targets for the development of innovative therapies for the treatment of major depressive disorders.
Collapse
Affiliation(s)
- Robert Dantzer
- Department of Symptom Research, The University of Texas, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
44
|
Dogruman-Al F, Engin AB, Bukan N, Evirgen-Bostanci S, Çeber K. Late-stage systemic immune effectors in Plasmodium berghei ANKA infection: biopterin and oxidative stress. Pteridines 2015. [DOI: 10.1515/pterid-2014-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
To investigate the involvement of systemic oxidative stress in the pathogenesis of murine cerebral malaria, mice were infected with the Plasmodium berghei (P. berghei) ANKA 6653 strain. Serum tryptophan (Trp), kynurenine and urinary biopterin, liver, brain, spleen and serum superoxide dismutase (SOD), glutathione peroxidase (GPx), malondialdehyde (MDA) and nitrite and nitrate (NOx) levels were measured on day 7 post-inoculation. Our data showed a significant decrease in SOD and an increase in GPx activity and MDA level in all the examined biological materials (p<0.05), except spleen. Conversely, GPx activities in spleen were depleted, while SOD and MDA levels remained unchanged. Increased MDA levels might indicate increased peroxynitrite production, lipid peroxidation and oxidative stress. Also, elevated urinary biopterin, which was accompanied by increased NOx (p<0.05), may support the inhibition of Trp degradation (p>0.05). The excessive NO synthesis in P. berghei infection may be related to the up-regulation of inducible NO synthase, which was in accordance with the increased biopterin excretion. Thus, the large quantities of released toxic redox active radicals attack cell membranes and induce lipid peroxidation. Although P. berghei infection did not demonstrate systemic Trp degradation and related indoleamine-2,3-dioxygenase activity, it may cause multi-organ failure and death, owing to host-derived severe oxidative stress.
Collapse
Affiliation(s)
- Funda Dogruman-Al
- Faculty of Medicine, Department of Medical Microbiology, Gazi University, 06500, Besevler, Ankara, Turkey
| | - Ayşe Başak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, 06330, Hipodrom, Ankara, Turkey
| | - Neslihan Bukan
- Faculty of Medicine, Department of Medical Biochemistry, Gazi University, 06500, Besevler, Ankara, Turkey
| | | | - Kemal Çeber
- Microbiology Laboratory, Mersin State Hospital, Mersin, Turkey
| |
Collapse
|
45
|
Abstract
IDO1 (indoleamine 2,3-dioxygenase 1) is a member of a unique class of mammalian haem dioxygenases that catalyse the oxidative catabolism of the least-abundant essential amino acid, L-Trp (L-tryptophan), along the kynurenine pathway. Significant increases in knowledge have been recently gained with respect to understanding the fundamental biochemistry of IDO1 including its catalytic reaction mechanism, the scope of enzyme reactions it catalyses, the biochemical mechanisms controlling IDO1 expression and enzyme activity, and the discovery of enzyme inhibitors. Major advances in understanding the roles of IDO1 in physiology and disease have also been realised. IDO1 is recognised as a prominent immune regulatory enzyme capable of modulating immune cell activation status and phenotype via several molecular mechanisms including enzyme-dependent deprivation of L-Trp and its conversion into the aryl hydrocarbon receptor ligand kynurenine and other bioactive kynurenine pathway metabolites, or non-enzymatic cell signalling actions involving tyrosine phosphorylation of IDO1. Through these different modes of biochemical signalling, IDO1 regulates certain physiological functions (e.g. pregnancy) and modulates the pathogenesis and severity of diverse conditions including chronic inflammation, infectious disease, allergic and autoimmune disorders, transplantation, neuropathology and cancer. In the present review, we detail the current understanding of IDO1’s catalytic actions and the biochemical mechanisms regulating IDO1 expression and activity. We also discuss the biological functions of IDO1 with a focus on the enzyme's immune-modulatory function, its medical implications in diverse pathological settings and its utility as a therapeutic target.
Collapse
|
46
|
Mancuso R, Hernis A, Agostini S, Rovaris M, Caputo D, Fuchs D, Clerici M. Indoleamine 2,3 Dioxygenase (IDO) Expression and Activity in Relapsing-Remitting Multiple Sclerosis. PLoS One 2015; 10:e0130715. [PMID: 26110930 PMCID: PMC4482492 DOI: 10.1371/journal.pone.0130715] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/23/2015] [Indexed: 01/09/2023] Open
Abstract
Background Interferon gamma (IFN-γ) production induces the transcription of indoleamine 2,3 dioxygenase (IDO) resulting in the reduction of T-cell activation and proliferation through the depletion of tryptophan and the elicitation of Treg lymphocytes. IDO was shown to be involved in the pathogenesis of autoimmune diseases; we investigated whether changes in IDO gene expression and activity could be indicative of onset of relapse in multiple sclerosis (MS) patients. Methods IDO and interferon-γ (IFN-γ) gene expression, serum IDO activity (Kynurenine/Tryptophan ratio) and serum neopterin concentration – a protein released by macrophages upon IFN-γ stimulation – were measured in 51 individuals: 36 relapsing remitting (RR)-MS patients (21 in acute phase -AMS, 15 in stable phase -SMS) and 15 healthy controls (HC). PBMCs samples in AMS patients were collected before (BT-AMS) and during glucocorticoids-based therapy (DT-AMS). Results IDO expression was increased and IFN-γ was decreased (p<0.001) in BT-AMS compared to SMS patients. Glucocorticoids-induced disease remission resulted in a significant reduction of IDO and IFN-γ gene expression, IDO catalytic activity (p<0.001). Serum neopterin concentration followed the same trend as IDO expression and activity. Conclusions Measurement of IDO gene expression and activity in blood could be a useful marker to monitor the clinical course of RR-MS. Therapeutic interventions modulating IDO activity may be beneficial in MS.
Collapse
Affiliation(s)
- Roberta Mancuso
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
- * E-mail:
| | - Ambra Hernis
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
| | - Simone Agostini
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
| | - Marco Rovaris
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
| | - Domenico Caputo
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innrain 80–82, A-6020, Innsbruck, Austria
| | - Mario Clerici
- Don C. Gnocchi Foundation ONLUS, Piazza Morandi 3, 20100, Milan, Italy
- Department of Physiopathology and Transplantation, University of Milano, Via Fratelli Cervi 93, 20090, Segrate, Milano, Italy
| |
Collapse
|
47
|
Zoller H, Jenal A, Staettermayer AF, Schroecksnadel S, Ferenci P, Fuchs D. Tryptophan Breakdown in Patients with HCV Infection is Influenced by IL28B Polymorphism. Pharmaceuticals (Basel) 2015; 8:337-50. [PMID: 26096654 PMCID: PMC4491665 DOI: 10.3390/ph8020337] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 12/17/2022] Open
Abstract
Until recently, the standard treatment of chronic hepatitis C virus (HCV) infection was a combination therapy with PEG-IFN-α plus ribavirin. Previous studies have proven that several markers predict the outcome of such therapy, e.g., pretreatment plasma levels of interferon inducible protein IP-10, HCV RNA and IL28B-related single nucleotide polymorphisms (SNP). Altered activity of tryptophan metabolizing enzyme indoleamine 2,3-dioxygenase (IDO) has been also shown in patients suffering from HCV infection. In this study, we investigated whether IL28B SNP in patients infected with HCV is related to the tryptophan breakdown rate. Before therapy, serum tryptophan and kynurenine concentrations were determined in 25 patients with established HCV infection and the kynurenine to tryptophan ratio (KYN/TRP) was calculated as an estimate of the tryptophan breakdown rate. In parallel, neopterin and nitrite concentrations were determined. A significant difference of serum KYN/TRP existed between the three IL28B polymorphism groups: C/C genotype had the highest and T/T genotype had the lowest KYN/TRP (p < 0.05). Likewise, C/C genotype was associated with higher KYN/TRP than non-C/C genotype (p = 0.01). There was a smaller difference between the three groups regarding the absolute kynurenine concentrations, the C/C genotype being associated with higher kynurenine concentrations. None of the other comparisons revealed any statistical significance. In conclusion, patients with C/C genotype presented with the highest tryptophan breakdown rate already before antiretroviral therapy with IFN-α/ribavirin. The differences in tryptophan metabolism might relate to HCV clearance and also to side effects of IFN-α therapy.
Collapse
Affiliation(s)
- Heinz Zoller
- Department of Internal Medicine, Biocenter, Innsbruck Medical University, Innsbruck 6020, Austria
| | - Annina Jenal
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck 6020, Austria
| | | | - Sebastian Schroecksnadel
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck 6020, Austria
| | - Peter Ferenci
- Department of Medicine III, Medical University of Vienna, Vienna 1090, Austria
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck 6020, Austria.
| |
Collapse
|
48
|
Bone Marrow Mesenchymal Stem Cells Alleviate Extracellular Kynurenine Levels, as Detected by High-Performance Liquid Chromatography. Inflammation 2015; 38:1450-7. [PMID: 25854176 DOI: 10.1007/s10753-015-0119-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Endothelial cell dysfunction plays an important role in the occurrence and development of sepsis, which is a consequence of the interaction between coagulation and inflammation. Kynurenine (KYN) is an endothelium-derived relaxing factor that makes a large contribution to sepsis pathophysiology. In this study, we investigated the influence of bone marrow mesenchymal stem cells (BMSCs) on KYN production by cultured endothelial cells. KYN and tryptophan (TRP) concentrations in cell supernatants were simultaneously measured with a high-performance liquid chromatography (HPLC) system equipped with a fluorescence detector (FLD) and an ultraviolet detector (UVD). Our results revealed that lipopolysaccharide-stimulated endothelial cells produced more KYN, which was accompanied by a parallel decrease in TRP. When co-cultured with BMSCs, KYN and TRP production were significantly decreased compared to lipopolysaccharide (LPS)-induction alone. Our results suggest that BMSCs can attenuate endothelial cell damage by decreasing KYN as detected with HPLC. This method is the first to be capable of capturing functional changes in the cells and is simple, fast, and suitable for cellular level research purposes.
Collapse
|
49
|
Tryptophan catabolism is unaffected in chronic granulomatous disease. Nature 2014; 514:E16-7. [DOI: 10.1038/nature13844] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/04/2014] [Indexed: 12/18/2022]
|
50
|
Schmidt SV, Schultze JL. New Insights into IDO Biology in Bacterial and Viral Infections. Front Immunol 2014; 5:384. [PMID: 25157255 PMCID: PMC4128074 DOI: 10.3389/fimmu.2014.00384] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/28/2014] [Indexed: 12/24/2022] Open
Abstract
Initially, indoleamine-2,3-dioxygenase (IDO) has been introduced as a bactericidal effector mechanism and has been linked to T-cell immunosuppression and tolerance. In recent years, evidence has been accumulated that IDO also plays an important role during viral infections including HIV, influenza, and hepatitis B and C. Moreover, novel aspects about the role of IDO in bacterial infections and sepsis have been revealed. Here, we review these recent findings highlighting the central role of IDO and tryptophan metabolism in many major human infections. Moreover, we also shed light on issues concerning human-specific and mouse-specific host–pathogen interactions that need to be considered when studying the biology of IDO in the context of infections.
Collapse
Affiliation(s)
- Susanne V Schmidt
- Genomics and Immunoregulation, LIMES-Institute, University of Bonn , Bonn , Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, LIMES-Institute, University of Bonn , Bonn , Germany
| |
Collapse
|