1
|
Niu J, Meng G. Roles and Mechanisms of NLRP3 in Influenza Viral Infection. Viruses 2023; 15:1339. [PMID: 37376638 DOI: 10.3390/v15061339] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Pathogenic viral infection represents a major challenge to human health. Due to the vast mucosal surface of respiratory tract exposed to the environment, host defense against influenza viruses has perpetually been a considerable challenge. Inflammasomes serve as vital components of the host innate immune system and play a crucial role in responding to viral infections. To cope with influenza viral infection, the host employs inflammasomes and symbiotic microbiota to confer effective protection at the mucosal surface in the lungs. This review article aims to summarize the current findings on the function of NACHT, LRR and PYD domains-containing protein 3 (NLRP3) in host response to influenza viral infection involving various mechanisms including the gut-lung crosstalk.
Collapse
Affiliation(s)
- Junling Niu
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, University of Chinese Academy of Sciences, 320 Yueyang Road, Life Science Research Building B-205, Shanghai 200031, China
| | - Guangxun Meng
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, University of Chinese Academy of Sciences, 320 Yueyang Road, Life Science Research Building B-205, Shanghai 200031, China
| |
Collapse
|
2
|
Loh MK, Stickling C, Schrank S, Hanshaw M, Ritger AC, Dilosa N, Finlay J, Ferrara NC, Rosenkranz JA. Liposaccharide-induced sustained mild inflammation fragments social behavior and alters basolateral amygdala activity. Psychopharmacology (Berl) 2023; 240:647-671. [PMID: 36645464 DOI: 10.1007/s00213-023-06308-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 01/02/2023] [Indexed: 01/17/2023]
Abstract
RATIONALE Conditions with sustained low-grade inflammation have high comorbidity with depression and anxiety and are associated with social withdrawal. The basolateral amygdala (BLA) is critical for affective and social behaviors and is sensitive to inflammatory challenges. Large systemic doses of lipopolysaccharide (LPS) initiate peripheral inflammation, increase BLA neuronal activity, and disrupt social and affective measures in rodents. However, LPS doses commonly used in behavioral studies are high enough to evoke sickness syndrome, which can confound interpretation of amygdala-associated behaviors. OBJECTIVES AND METHODS The objectives of this study were to find a LPS dose that triggers mild peripheral inflammation but not observable sickness syndrome in adult male rats, to test the effects of sustained mild inflammation on BLA and social behaviors. To accomplish this, we administered single doses of LPS (0-100 μg/kg, intraperitoneally) and measured open field behavior, or repeated LPS (5 μg/kg, 3 consecutive days), and measured BLA neuronal firing, social interaction, and elevated plus maze behavior. RESULTS Repeated low-dose LPS decreased BLA neuron firing rate but increased the total number of active BLA neurons. Repeated low-dose LPS also caused early disengagement during social bouts and less anogenital investigation and an overall pattern of heightened social caution associated with reduced gain of social familiarity over the course of a social session. CONCLUSIONS These results provide evidence for parallel shifts in social interaction and amygdala activity caused by prolonged mild inflammation. This effect of inflammation may contribute to social symptoms associated with comorbid depression and chronic inflammatory conditions.
Collapse
Affiliation(s)
- Maxine K Loh
- Discipline of Cellular and Molecular Pharmacology, Department of Foundational Sciences and Humanities, Chicago Medical School, Rosalind Franklin University of Medicine and Science, IL, 60064, North Chicago, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Courtney Stickling
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Sean Schrank
- Discipline of Cellular and Molecular Pharmacology, Department of Foundational Sciences and Humanities, Chicago Medical School, Rosalind Franklin University of Medicine and Science, IL, 60064, North Chicago, USA.,Discipline of Neuroscience, Department of Foundational Sciences and Humanities, Chicago Medical School, Rosalind Franklin University of Medicine and Science, IL, North Chicago, USA
| | - Madison Hanshaw
- Discipline of Cellular and Molecular Pharmacology, Department of Foundational Sciences and Humanities, Chicago Medical School, Rosalind Franklin University of Medicine and Science, IL, 60064, North Chicago, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Alexandra C Ritger
- Discipline of Cellular and Molecular Pharmacology, Department of Foundational Sciences and Humanities, Chicago Medical School, Rosalind Franklin University of Medicine and Science, IL, 60064, North Chicago, USA.,Discipline of Neuroscience, Department of Foundational Sciences and Humanities, Chicago Medical School, Rosalind Franklin University of Medicine and Science, IL, North Chicago, USA
| | - Naijila Dilosa
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Joshua Finlay
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Nicole C Ferrara
- Discipline of Cellular and Molecular Pharmacology, Department of Foundational Sciences and Humanities, Chicago Medical School, Rosalind Franklin University of Medicine and Science, IL, 60064, North Chicago, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - J Amiel Rosenkranz
- Discipline of Cellular and Molecular Pharmacology, Department of Foundational Sciences and Humanities, Chicago Medical School, Rosalind Franklin University of Medicine and Science, IL, 60064, North Chicago, USA. .,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| |
Collapse
|
3
|
Skjellerudsveen BM, Omdal R, Hetta AK, Kvaløy JT, Aabakken L, Skoie IM, Grimstad T. Fatigue: a frequent and biologically based phenomenon in newly diagnosed celiac disease. Sci Rep 2022; 12:7281. [PMID: 35508622 PMCID: PMC9068783 DOI: 10.1038/s41598-022-11802-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 04/26/2022] [Indexed: 11/18/2022] Open
Abstract
Fatigue is increasingly recognized as a major complaint in patients with chronic inflammatory and autoimmune diseases. Although fatigue is assumed to represent a significant problem in celiac disease, existing knowledge is scarce, and opinions are conflicting. This study aimed to investigate the prevalence and severity of fatigue in patients with newly diagnosed celiac disease and compare it with healthy control subjects. Ninety patients with newly diagnosed celiac disease were compared with 90 age- and sex-matched healthy subjects. The primary endpoints were fatigue severity as measured by: the fatigue Visual Analog Scale (fVAS), the Fatigue Severity Scale (FSS), and the inverted Vitality subscale of the MOS36 (SF-36vs). Higher scores indicate more severe fatigue. Clinically relevant fatigue was determined using predefined cut-off values. Secondary endpoints were the associations between fatigue, and sex, age, depression, pain, and selected biochemical variables. The median (IQR) fVAS-scores were 43.0 (18.0–64.5) in patients, and 9.0 (2.0–16.0) in the control group (p < 0.001); and the FSS scores 3.8 (2.0–4.8) in patients, and 1.4 (1.0–1.9) in control subjects (p < 0.001). Inverted SF-36vs scores had a mean (SD) value of 58.8 (23.6) in patients, and 29.7 (14.3) in healthy subjects (p < 0.001). The presence of clinically relevant fatigue ranged from 41 to 50% in patients. Increased fatigue severity was associated with female sex, younger age, and elevated pain and depression scores, but not with levels of selected biochemical variables, including hemoglobin. Fatigue is a severe and frequent phenomenon in patients with untreated celiac disease.
Collapse
Affiliation(s)
| | - Roald Omdal
- Department of Internal Medicine, Stavanger University Hospital, Pb. 8100, 4068, Stavanger, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Anne Kristine Hetta
- Department of Internal Medicine, Stavanger University Hospital, Pb. 8100, 4068, Stavanger, Norway
| | - Jan Terje Kvaløy
- Department of Mathematics and Physics, University of Stavanger, Stavanger, Norway.,Department of Research, Stavanger University Hospital, Stavanger, Norway
| | - Lars Aabakken
- Department of Transplantation Medicine, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Inger Marie Skoie
- Department of Dermatology, Stavanger University Hospital, Stavanger, Norway
| | - Tore Grimstad
- Department of Internal Medicine, Stavanger University Hospital, Pb. 8100, 4068, Stavanger, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
4
|
Vithoulkas G. An integrated perspective on transmutation of acute inflammation into chronic and the role of the microbiome. J Med Life 2021; 14:740-747. [PMID: 35126742 PMCID: PMC8811668 DOI: 10.25122/jml-2021-0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/30/2021] [Indexed: 11/20/2022] Open
Abstract
The Continuum theory and the Levels of Health theory were separately proposed to explain the myriad responses to treatment and understand the process of health and disease in an individual. In light of accumulating evidence on the intricate relationship between the human immune system and microbiome, an attempt is made in this article to connect these two theories to explain the transmutation of the efficiently responding immune system (through the acute inflammatory response and high fever) to one involved in a low-grade chronic inflammatory process (resulting in chronic disease). There is already enough evidence to demonstrate the role of the microbiome in all chronic inflammatory diseases. In this article, we discuss the mechanism by which subjecting a healthy person to continuous drug treatment for acute inflammatory conditions (at a certain time) leads to transmutation to chronic disease. Although this hypothesis requires further experimental evidence, it calls for a reconsideration of the manner in which we treat acute infectious diseases in the population.
Collapse
Affiliation(s)
- George Vithoulkas
- University of the Aegean, Syros, Greece
- Postgraduate Doctors’ Training Institute, Health Care Ministry of the Chuvash Republic, Cheboksary, Russian Federation
| |
Collapse
|
5
|
Brain Perivascular Macrophages Do Not Mediate Interleukin-1-Induced Sickness Behavior in Rats. Pharmaceuticals (Basel) 2021; 14:ph14101030. [PMID: 34681254 PMCID: PMC8541198 DOI: 10.3390/ph14101030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/01/2021] [Accepted: 10/07/2021] [Indexed: 01/08/2023] Open
Abstract
Sickness behavior, characterized by on overall reduction in behavioral activity, is commonly observed after bacterial infection. Sickness behavior can also be induced by the peripheral administration of Gram-negative bacterial lipopolysaccharide (LPS) or interleukin-1beta (IL-1β), a pro-inflammatory cytokine released by LPS-activated macrophages. In addition to the microglia, the brain contains perivascular macrophages, which express the IL-1 type 1 receptor (IL-1R1). In the present study, we assessed the role of brain perivascular macrophages in mediating IL-1β-induced sickness behavior in rats. To do so, we used intracerebroventricular (icv) administration of an IL-1β-saporin conjugate, known to eliminate IL-R1-expressing brain cells, prior to systemic or central IL-1β injection. Icv IL-1β-saporin administration resulted in a reduction in brain perivascular macrophages, without altering subsequent icv or ip IL-1β-induced reductions in food intake, locomotor activity, and social interactions. In conclusion, the present work shows that icv IL-1β-saporin administration is an efficient way to target brain perivascular macrophages, and to determine whether these cells are involved in IL-1β-induced sickness behavior.
Collapse
|
6
|
Ohno M, Kakino A, Sekiya T, Nomura N, Shingai M, Sawamura T, Kida H. Critical role of oxidized LDL receptor-1 in intravascular thrombosis in a severe influenza mouse model. Sci Rep 2021; 11:15675. [PMID: 34344944 PMCID: PMC8333315 DOI: 10.1038/s41598-021-95046-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/12/2021] [Indexed: 11/10/2022] Open
Abstract
Although coagulation abnormalities, including microvascular thrombosis, are thought to contribute to tissue injury and single- or multiple-organ dysfunction in severe influenza, the detailed mechanisms have yet been clarified. This study evaluated influenza-associated abnormal blood coagulation utilizing a severe influenza mouse model. After infecting C57BL/6 male mice with intranasal applications of 500 plaque-forming units of influenza virus A/Puerto Rico/8/34 (H1N1; PR8), an elevated serum level of prothrombin fragment 1 + 2, an indicator for activated thrombin generation, was observed. Also, an increased gene expression of oxidized low-density lipoprotein (LDL) receptor-1 (Olr1), a key molecule in endothelial dysfunction in the progression of atherosclerosis, was detected in the aorta of infected mice. Body weight decrease, serum levels of cytokines and chemokines, viral load, and inflammation in the lungs of infected animals were similar between wild-type and Olr1 knockout (KO) mice. In contrast, the elevation of prothrombin fragment 1 + 2 levels in the sera and intravascular thrombosis in the lungs by PR8 virus infection were not induced in KO mice. Collectively, the results indicated that OLR1 is a critical host factor in intravascular thrombosis as a pathogeny of severe influenza. Thus, OLR1 is a promising novel therapeutic target for thrombosis during severe influenza.
Collapse
Affiliation(s)
- Marumi Ohno
- Laboratory for Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
| | - Akemi Kakino
- Department of Molecular Pathophysiology, School of Medicine, Shinshu University, Matsumoto, Japan
| | - Toshiki Sekiya
- Laboratory for Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
| | - Naoki Nomura
- Laboratory for Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
| | - Masashi Shingai
- Laboratory for Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
| | - Tatsuya Sawamura
- Department of Molecular Pathophysiology, School of Medicine, Shinshu University, Matsumoto, Japan
| | - Hiroshi Kida
- Laboratory for Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan.
| |
Collapse
|
7
|
Galmiche M, Achamrah N, Déchelotte P, Ribet D, Breton J. Role of microbiota-gut-brain axis dysfunctions induced by infections in the onset of anorexia nervosa. Nutr Rev 2021; 80:381-391. [PMID: 34010427 DOI: 10.1093/nutrit/nuab030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Anorexia nervosa (AN) is an eating disorder characterized by low food intake, severe body weight loss, intense fear of gaining weight, and dysmorphophobia. This chronic disease is associated with both psychiatric and somatic comorbidities. Over the years, clinical studies have accumulated evidence that viral or bacterial infections may promote the onset of eating disorders such as AN. This review aims to describe how infections and the subsequent immune responses affect food intake regulation in the short term and also how these processes may lead to long-term intestinal disorders, including gut barrier disruption and gut microbiota dysbiosis, even after the clearance of the pathogens. We discuss in particular how infection-mediated intestinal dysbiosis may promote the onset of several AN symptoms and comorbidities, including appetite dysregulation, functional gastrointestinal disorders, and mood disorders.
Collapse
Affiliation(s)
- Marie Galmiche
- M. Galmiche, N. Achamrah, P. Déchelotte, and J. Breton are with Nutrition Department, CHU Rouen, F-76000 Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, INSERM UMR 1073, Nutrition, Gut and Brain Laboratory, Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
| | - Najate Achamrah
- M. Galmiche, N. Achamrah, P. Déchelotte, and J. Breton are with Nutrition Department, CHU Rouen, F-76000 Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, INSERM UMR 1073, Nutrition, Gut and Brain Laboratory, Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
| | - Pierre Déchelotte
- M. Galmiche, N. Achamrah, P. Déchelotte, and J. Breton are with Nutrition Department, CHU Rouen, F-76000 Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, INSERM UMR 1073, Nutrition, Gut and Brain Laboratory, Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
| | - David Ribet
- M. Galmiche, N. Achamrah, P. Déchelotte, and J. Breton are with Nutrition Department, CHU Rouen, F-76000 Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, INSERM UMR 1073, Nutrition, Gut and Brain Laboratory, Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
| | - Jonathan Breton
- M. Galmiche, N. Achamrah, P. Déchelotte, and J. Breton are with Nutrition Department, CHU Rouen, F-76000 Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, INSERM UMR 1073, Nutrition, Gut and Brain Laboratory, Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
| |
Collapse
|
8
|
Lasselin J, Schedlowski M, Karshikoff B, Engler H, Lekander M, Konsman JP. Comparison of bacterial lipopolysaccharide-induced sickness behavior in rodents and humans: Relevance for symptoms of anxiety and depression. Neurosci Biobehav Rev 2020; 115:15-24. [PMID: 32433924 DOI: 10.1016/j.neubiorev.2020.05.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/24/2020] [Accepted: 05/02/2020] [Indexed: 12/26/2022]
Abstract
Increasing evidence from animal and human studies suggests that inflammation may be involved in mood disorders. Sickness behavior and emotional changes induced by experimental inflammatory stimuli have been extensively studied in humans and rodents to better understand the mechanisms underlying inflammation-driven mood alterations. However, research in animals and humans have remained compartmentalized and a comprehensive comparison of inflammation-induced sickness and depressive-like behavior between rodents and humans is lacking. Thus, here, we highlight similarities and differences in the effects of bacterial lipopolysaccharide administration on the physiological (fever and cytokines), behavioral and emotional components of the sickness response in rodents and humans, and discuss the translational challenges involved. We also emphasize the differences between observable sickness behavior and subjective sickness reports, and advocate for the need to obtain both subjective reports and objective measurements of sickness behavior in humans. We aim to provide complementary insights for translational clinical and experimental research on inflammation-induced behavioral and emotional changes, and their relevance for mood disorders such as depression.
Collapse
Affiliation(s)
- Julie Lasselin
- Stress Research Institute, Stockholm University, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Manfred Schedlowski
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, Essen, Germany
| | - Bianka Karshikoff
- Stress Research Institute, Stockholm University, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Harald Engler
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, Essen, Germany
| | - Mats Lekander
- Stress Research Institute, Stockholm University, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Pieter Konsman
- Institute for Cognitive and Integrative Neuroscience, CNRS UMR 5287, University of Bordeaux, France
| |
Collapse
|
9
|
Momota M, Lelliott P, Kubo A, Kusakabe T, Kobiyama K, Kuroda E, Imai Y, Akira S, Coban C, Ishii KJ. ZBP1 governs the inflammasome-independent IL-1α and neutrophil inflammation that play a dual role in anti-influenza virus immunity. Int Immunol 2020; 32:203-212. [PMID: 31630209 PMCID: PMC10689344 DOI: 10.1093/intimm/dxz070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/17/2019] [Indexed: 11/14/2022] Open
Abstract
Influenza A virus (IAV) triggers the infected lung to produce IL-1 and recruit neutrophils. Unlike IL-1β, however, little is known about IL-1α in terms of its mechanism of induction, action and physiological relevance to the host immunity against IAV infection. In particular, whether Z-DNA-binding protein 1 (ZBP1), a key molecule for IAV-induced cell death, is involved in the IL-1α induction, neutrophil infiltration and the physiological outcome has not been elucidated. Here, we show in a murine model that the IAV-induced IL-1α is mediated solely by ZBP1, in an NLRP3-inflammasome-independent manner, and is required for the optimal IL-1β production followed by the formation of neutrophil extracellular traps (NETs). During IAV infection, ZBP1 displays a dual role in anti-IAV immune responses mediated by neutrophils, resulting in either protective or pathological outcomes in vivo. Thus, ZBP1-mediated IL-1α production is the key initial step of IAV-infected NETs, regulating the duality of the consequent lung inflammation.
Collapse
Affiliation(s)
- Masatoshi Momota
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research Center (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Mockup Vaccine, Center for Vaccine and Adjuvant Research Center (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Vaccine Science, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Patrick Lelliott
- Malaria Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Atsuko Kubo
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research Center (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Takato Kusakabe
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research Center (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Mockup Vaccine, Center for Vaccine and Adjuvant Research Center (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Vaccine Science, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Kouji Kobiyama
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research Center (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Etsushi Kuroda
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research Center (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Vaccine Science, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Immunology, Hyogo College of Medicine, Hyogo, Japan
| | - Yumiko Imai
- Laboratory of Regulation of Intractable Infectious Diseases, Center for Vaccine and Adjuvant Research Center (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Shizuo Akira
- Host Defense, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Cevayir Coban
- Malaria Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Malaria Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research Center (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Mockup Vaccine, Center for Vaccine and Adjuvant Research Center (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Vaccine Science, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
10
|
Chaskiel L, Bristow AD, Bluthé RM, Dantzer R, Blomqvist A, Konsman JP. Interleukin-1 reduces food intake and body weight in rat by acting in the arcuate hypothalamus. Brain Behav Immun 2019; 81:560-573. [PMID: 31310797 DOI: 10.1016/j.bbi.2019.07.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 12/19/2022] Open
Abstract
A reduction in food intake is commonly observed after bacterial infection, a phenomenon that can be reproduced by peripheral administration of Gram-negative bacterial lipopolysaccharide (LPS) or interleukin-1beta (IL-1β), a pro-inflammatory cytokine released by LPS-activated macrophages. The arcuate nucleus of the hypothalamus (ARH) plays a major role in food intake regulation and expresses IL-1 type 1 receptor (IL-1R1) mRNA. In the present work, we tested the hypothesis that IL-1R1 expressing cells in the ARH mediate IL-1β and/or LPS-induced hypophagia in the rat. To do so, we developed an IL-1β-saporin conjugate, which eliminated IL-R1-expressing neurons in the hippocampus, and micro-injected it into the ARH prior to systemic IL-1β and LPS administration. ARH IL-1β-saporin injection resulted in loss of neuropeptide Y-containing cells and attenuated hypophagia and weight loss after intraperitoneal IL-1β, but not LPS, administration. In conclusion, the present study shows that ARH NPY-containing neurons express functional IL-1R1s that mediate peripheral IL-1β-, but not LPS-, induced hypophagia. Our present and previous findings indicate that the reduction of food intake after IL-1β and LPS are mediated by different neural pathways.
Collapse
Affiliation(s)
- Léa Chaskiel
- Psychoneuroimmunology, Nutrition and Genetics, UMR CNRS 5226-INRA 1286, University of Bordeaux, 33076 Bordeaux, France
| | - Adrian D Bristow
- National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, UK
| | - Rose-Marie Bluthé
- Psychoneuroimmunology, Nutrition and Genetics, UMR CNRS 5226-INRA 1286, University of Bordeaux, 33076 Bordeaux, France
| | - Robert Dantzer
- Department of Symptom Research, MD Anderson Cancer Center, The University of Texas, Houston, TX 770030, USA
| | - Anders Blomqvist
- Division of Neurobiology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, S-581 85 Linköping, Sweden
| | - Jan Pieter Konsman
- UMR CNRS 5287 Aquitaine Institute for Integrative and Cognitive Neuroscience, University of Bordeaux, 33076 Bordeaux, France.
| |
Collapse
|
11
|
Yan YQ, Fu YJ, Wu S, Qin HQ, Zhen X, Song BM, Weng YS, Wang PC, Chen XY, Jiang ZY. Anti-influenza activity of berberine improves prognosis by reducing viral replication in mice. Phytother Res 2018; 32:2560-2567. [PMID: 30306659 DOI: 10.1002/ptr.6196] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 08/13/2018] [Accepted: 08/16/2018] [Indexed: 12/14/2022]
Abstract
Berberine, a natural isoquinoline alkaloid isolated from the berberis species, has a wide array of biological properties such as anti-inflammatory, antibacterial, antifungal, and antihelminthic effects. We evaluated the antiviral effect of berberine against influenza A/FM1/1/47 (H1N1) in vivo and in vitro. The results showed that berberine strongly suppressed viral replication in A549 cells and in mouse lungs. Meanwhile, berberine relieved pulmonary inflammation and reduced necrosis, inflammatory cell infiltration, and pulmonary edema induced by viral infection in mice when compared with vehicle-treated mice. Berberine suppressed the viral infection-induced up-regulation of TLR7 signaling pathway, such as TLR7, MyD88, and NF-κB (p65), at both the mRNA and protein levels. Furthermore, berberine significantly inhibited the viral infection-induced increase in Th1/Th2 and Th17/Treg ratios as well as the production of inflammatory cytokines. Our data provide new insight into the potential of berberine as a therapeutic agent for viral infection via its antiviral activity.
Collapse
Affiliation(s)
- Yu-Qi Yan
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, GuangZhou, China.,Guangdong Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, GuangZhou, China.,Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, GuangZhou, China
| | - Ying-Jie Fu
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, GuangZhou, China.,Guangdong Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, GuangZhou, China.,Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, GuangZhou, China
| | - Sha Wu
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, GuangZhou, China.,Guangdong Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, GuangZhou, China.,Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, GuangZhou, China
| | - Hong-Qiong Qin
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, GuangZhou, China.,Guangdong Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, GuangZhou, China.,Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, GuangZhou, China
| | - Xiao Zhen
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, GuangZhou, China.,Guangdong Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, GuangZhou, China.,Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, GuangZhou, China
| | - Bi-Mei Song
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, GuangZhou, China.,Guangdong Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, GuangZhou, China.,Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, GuangZhou, China
| | - Yuan-Shu Weng
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, GuangZhou, China.,Guangdong Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, GuangZhou, China.,Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, GuangZhou, China
| | - Peng-Cheng Wang
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, GuangZhou, China.,Guangdong Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, GuangZhou, China.,Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, GuangZhou, China
| | - Xiao-Yin Chen
- School of Traditional Chinese Medicine, Jinan University, GuangZhou, China
| | - Zhen-You Jiang
- Department of Microbiology and Immunology, Basic Medicine College, Jinan University, GuangZhou, China.,Guangdong Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, GuangZhou, China.,Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Jinan University, GuangZhou, China
| |
Collapse
|
12
|
Zager A, Brandão WN, Margatho RO, Peron JP, Tufik S, Andersen ML, Kornum BR, Palermo-Neto J. The wake-promoting drug Modafinil prevents motor impairment in sickness behavior induced by LPS in mice: Role for dopaminergic D1 receptor. Prog Neuropsychopharmacol Biol Psychiatry 2018; 81:468-476. [PMID: 28499899 DOI: 10.1016/j.pnpbp.2017.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/04/2017] [Accepted: 05/08/2017] [Indexed: 01/08/2023]
Abstract
The wake-promoting drug Modafinil has been used for many years for treatment of Narcolepsy and Excessive Daytime Sleepiness, due to a dopamine-related psychostimulant action. Recent studies have indicated that Modafinil prevents neuroinflammation in animal models. Thus, the aim of the present study was to evaluate the effect of Modafinil pretreatment in the Lipopolysaccharide (LPS)-induced sickness and depressive-like behaviors. Adult male C57BL/6J mice were pretreated with Vehicle or Modafinil (90mg/Kg) and, 30min later, received a single saline or LPS (2mg/Kg) administration, and were submitted to the open field and elevated plus maze test 2h later. After 24h, mice were subjected to tail suspension test, followed by either flow cytometry with whole brain for CD11b+CD45+ cells or qPCR in brain areas for cytokine gene expression. Modafinil treatment prevented the LPS-induced motor impairment, anxiety-like and depressive-like behaviors, as well as the increase in brain CD11b+CD45high cells induced by LPS. Our results indicate that Modafinil pretreatment also decreased the IL-1β gene upregulation caused by LPS in brain areas, which is possibly correlated with the preventive behavioral effects. The pharmacological blockage of the dopaminergic D1R by the drug SCH-23390 counteracted the effect of Modafinil on locomotion and anxiety-like behavior, but not on depressive-like behavior and brain immune cells. The dopaminergic D1 receptor signaling is essential to the Modafinil effects on LPS-induced alterations in locomotion and anxiety, but not on depression and brain macrophages. This evidence suggests that Modafinil treatment might be useful to prevent inflammation-related behavioral alterations, possibly due to a neuroimmune mechanism.
Collapse
Affiliation(s)
- Adriano Zager
- Neuroimmunomodulation Research Group, Department of Pathology, School of Veterinary Medicine, University of São Paulo (USP), São Paulo, Brazil.
| | - Wesley Nogueira Brandão
- Neuroimmune Interactions Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Rafael Oliveira Margatho
- Neuroimmunomodulation Research Group, Department of Pathology, School of Veterinary Medicine, University of São Paulo (USP), São Paulo, Brazil
| | - Jean Pierre Peron
- Neuroimmune Interactions Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Sergio Tufik
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil
| | - Monica Levy Andersen
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil
| | - Birgitte Rahbek Kornum
- Molecular Sleep Laboratory, Department of Clinical Biochemistry, Glostrup Research Institute-Rigshospitalet, Glostrup, Denmark
| | - João Palermo-Neto
- Neuroimmunomodulation Research Group, Department of Pathology, School of Veterinary Medicine, University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
13
|
Impact of chemotherapy on cancer-related fatigue and cytokines in 1312 patients: a systematic review of quantitative studies. Curr Opin Support Palliat Care 2018; 10:165-79. [PMID: 27043288 DOI: 10.1097/spc.0000000000000205] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE OF REVIEW Cancer-related fatigue (CRF) is the most common psychosomatic distress experienced by cancer patients before, during and after chemotherapy. Its impact on functional status and Health Related Quality of Life is a great concern among patients, healthcare professionals and researchers. The primary objective of this systematic review is to determine whether the different chemotherapies affect the association of CRF with individual pro- and anti-inflammatory cytokines. The PRISMA statement guideline has been followed to systematically search and screen article from PubMed and Embase. RECENT FINDINGS This review has examined 14 studies which included a total of 1312 patients. These studies assayed 20 different kinds of cytokines. The cytokines interleukin-6, interleukin-1RA, TGF-β and sTNF-R2 were associated with CRF in patients receiving anthracycline-based chemotherapy. However, only interleukin-13 was identified in the taxane-based chemotherapy. Similarly, different sets of cytokines were linked with CRF in patients with chemotherapy regimens containing platinum, cyclophosphamides, topotecan or bleomycin. SUMMARY This review has identified that cytokines are differentially linked with CRF according to the various types of chemotherapy regimens.
Collapse
|
14
|
Holzer P, Farzi A, Hassan AM, Zenz G, Jačan A, Reichmann F. Visceral Inflammation and Immune Activation Stress the Brain. Front Immunol 2017; 8:1613. [PMID: 29213271 PMCID: PMC5702648 DOI: 10.3389/fimmu.2017.01613] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 11/07/2017] [Indexed: 12/20/2022] Open
Abstract
Stress refers to a dynamic process in which the homeostasis of an organism is challenged, the outcome depending on the type, severity, and duration of stressors involved, the stress responses triggered, and the stress resilience of the organism. Importantly, the relationship between stress and the immune system is bidirectional, as not only stressors have an impact on immune function, but alterations in immune function themselves can elicit stress responses. Such bidirectional interactions have been prominently identified to occur in the gastrointestinal tract in which there is a close cross-talk between the gut microbiota and the local immune system, governed by the permeability of the intestinal mucosa. External stressors disturb the homeostasis between microbiota and gut, these disturbances being signaled to the brain via multiple communication pathways constituting the gut-brain axis, ultimately eliciting stress responses and perturbations of brain function. In view of these relationships, the present article sets out to highlight some of the interactions between peripheral immune activation, especially in the visceral system, and brain function, behavior, and stress coping. These issues are exemplified by the way through which the intestinal microbiota as well as microbe-associated molecular patterns including lipopolysaccharide communicate with the immune system and brain, and the mechanisms whereby overt inflammation in the GI tract impacts on emotional-affective behavior, pain sensitivity, and stress coping. The interactions between the peripheral immune system and the brain take place along the gut-brain axis, the major communication pathways of which comprise microbial metabolites, gut hormones, immune mediators, and sensory neurons. Through these signaling systems, several transmitter and neuropeptide systems within the brain are altered under conditions of peripheral immune stress, enabling adaptive processes related to stress coping and resilience to take place. These aspects of the impact of immune stress on molecular and behavioral processes in the brain have a bearing on several disturbances of mental health and highlight novel opportunities of therapeutic intervention.
Collapse
Affiliation(s)
- Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria.,BioTechMed-Graz, Graz, Austria
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Ahmed M Hassan
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Geraldine Zenz
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Angela Jačan
- CBmed GmbH-Center for Biomarker Research in Medicine, Graz, Austria
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| |
Collapse
|
15
|
Hennessy E, Gormley S, Lopez-Rodriguez AB, Murray C, Murray C, Cunningham C. Systemic TNF-α produces acute cognitive dysfunction and exaggerated sickness behavior when superimposed upon progressive neurodegeneration. Brain Behav Immun 2017; 59:233-244. [PMID: 27633985 PMCID: PMC5176008 DOI: 10.1016/j.bbi.2016.09.011] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 09/01/2016] [Accepted: 09/12/2016] [Indexed: 12/13/2022] Open
Abstract
Inflammation influences chronic neurodegeneration but its precise roles are not yet clear. Systemic inflammation caused by infection, trauma or co-morbidity can alter the brain's inflammatory status, produce acute cognitive impairments, such as delirium, and drive new pathology and accelerated decline. Consistent with this, elevated systemic TNF-α is associated with more rapid cognitive decline over 6months in Alzheimer's disease patients. In the current study we challenged normal animals and those with existing progressive neurodegeneration (ME7 prion disease) with TNF-α (i.p.) to test the hypothesis that this cytokine has differential effects on cognitive function, sickness behavior and features of underlying pathology contingent on the animals' baseline condition. TNF-α (50μg/kg) had no impact on performance of normal animals (normal brain homogenate; NBH) on working memory (T-maze) but produced acute impairments in ME7 animals similarly challenged. Plasma TNF-α and CCL2 levels were equivalent in NBH and ME7 TNF-challenged animals but hippocampal and hypothalamic transcription of IL-1β, TNF-α and CCL2 and translation of IL-1β were higher in ME7+TNF-α than NBH+TNF-α animals. TNF-α produced an exaggerated sickness behavior response (hypothermia, weight loss, inactivity) in ME7 animals compared to that in NBH animals. However a single challenge with this dose was not sufficient to produce de novo neuronal death, synaptic loss or tau hyperphosphorylation that was distinguishable from that arising from ME7 alone. The data indicate that acutely elevated TNF-α has robust acute effects on brain function, selectively in the degenerating brain, but more sustained levels may be required to significantly impact on underlying neurodegeneration.
Collapse
Affiliation(s)
- Edel Hennessy
- School of Biochemistry & Immunology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Shane Gormley
- School of Biochemistry & Immunology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Ana Belen Lopez-Rodriguez
- School of Biochemistry & Immunology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Caoimhe Murray
- School of Biochemistry & Immunology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Carol Murray
- School of Biochemistry & Immunology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Colm Cunningham
- School of Biochemistry & Immunology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
16
|
Araki R, Nishida S, Hiraki Y, Li F, Matsumoto K, Yabe T. Kamikihito Ameliorates Lipopolysaccharide-Induced Sickness Behavior via Attenuating Neural Activation, but Not Inflammation, in the Hypothalamic Paraventricular Nucleus and Central Nucleus of the Amygdala in Mice. Biol Pharm Bull 2016; 39:289-94. [PMID: 26830488 DOI: 10.1248/bpb.b15-00707] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sickness behavior is a series of behavioral and psychological changes that develop in those stricken with cancers and inflammatory diseases. The etiological mechanism of sickness behavior is not known in detail, and consequently there are no established standard therapies. Kamikihito (KKT), a Kampo (traditional Japanese herbal) medicine composed of 14 herbs, has been used clinically to treat psychiatric dysfunction. Previously, we found that KKT ameliorated sickness behavior in mice inoculated with murine colon 26 adenocarcinoma cells. In this study, we examined the effects of KKT on bacterial endotoxin lipopolysaccharide (LPS)-induced sickness behavior in mice. The administration of LPS caused the emotional aspects of sickness behavior, such as loss of object exploration, social interaction deficit, and depressive-like behavior. LPS also induced mRNA expression for cyclooxygenase (COX)-2, interleukin (IL)-1β and IL-6, and increased the number of c-Fos immunopositive cells in the hypothalamus and amygdala. KKT ameliorated the behavioral changes and reversed the increases in c-Fos immunopositive cells in the two brain regions, but did not influence the mRNA expression. These results suggest that KKT ameliorates sickness behavior via the suppression of neural activation without anti-inflammatory effects, and that KKT has the potential to treat sickness behavior.
Collapse
Affiliation(s)
- Ryota Araki
- Laboratory of Functional Biomolecules and Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | | | | | | | | | | |
Collapse
|
17
|
Söderholm S, Fu Y, Gaelings L, Belanov S, Yetukuri L, Berlinkov M, Cheltsov AV, Anders S, Aittokallio T, Nyman TA, Matikainen S, Kainov DE. Multi-Omics Studies towards Novel Modulators of Influenza A Virus-Host Interaction. Viruses 2016; 8:v8100269. [PMID: 27690086 PMCID: PMC5086605 DOI: 10.3390/v8100269] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 09/13/2016] [Accepted: 09/22/2016] [Indexed: 12/20/2022] Open
Abstract
Human influenza A viruses (IAVs) cause global pandemics and epidemics. These viruses evolve rapidly, making current treatment options ineffective. To identify novel modulators of IAV–host interactions, we re-analyzed our recent transcriptomics, metabolomics, proteomics, phosphoproteomics, and genomics/virtual ligand screening data. We identified 713 potential modulators targeting 199 cellular and two viral proteins. Anti-influenza activity for 48 of them has been reported previously, whereas the antiviral efficacy of the 665 remains unknown. Studying anti-influenza efficacy and immuno/neuro-modulating properties of these compounds and their combinations as well as potential viral and host resistance to them may lead to the discovery of novel modulators of IAV–host interactions, which might be more effective than the currently available anti-influenza therapeutics.
Collapse
Affiliation(s)
- Sandra Söderholm
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland.
- Finnish Institute of Occupational Health, Helsinki 00250, Finland.
| | - Yu Fu
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| | - Lana Gaelings
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| | - Sergey Belanov
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| | - Laxman Yetukuri
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| | - Mikhail Berlinkov
- Institute of Mathematics and Computer Science, Ural Federal University, Yekaterinburg 620083, Russia.
| | - Anton V Cheltsov
- Q-Mol L.L.C. in Silico Pharmaceuticals, San Diego, CA 92037, USA.
| | - Simon Anders
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
- Department of Mathematics and Statistics, University of Turku, Turku 20014, Finland.
| | | | - Sampsa Matikainen
- Finnish Institute of Occupational Health, Helsinki 00250, Finland.
- Department of Rheumatology, Helsinki University Hospital, University of Helsinki, Helsinki 00015, Finland.
| | - Denis E Kainov
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| |
Collapse
|
18
|
Sickness: From the focus on cytokines, prostaglandins, and complement factors to the perspectives of neurons. Neurosci Biobehav Rev 2015; 57:30-45. [PMID: 26363665 DOI: 10.1016/j.neubiorev.2015.07.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/06/2015] [Accepted: 07/31/2015] [Indexed: 12/29/2022]
Abstract
Systemic inflammation leads to a variety of physiological (e.g. fever) and behavioral (e.g. anorexia, immobility, social withdrawal, depressed mood, disturbed sleep) responses that are collectively known as sickness. While these phenomena have been studied for the past few decades, the neurobiological mechanisms by which sickness occurs remain unclear. In this review, we first revisit how the body senses and responds to infections and injuries by eliciting systemic inflammation. Next, we focus on how peripheral inflammatory molecules such as cytokines, prostaglandins, and activated complement factors communicate with the brain to trigger neuroinflammation and sickness. Since depression also involves inflammation, we further elaborate on the interrelationship between sickness and depression. Finally, we discuss how immune activation can modulate neurons in the brain, and suggest future perspectives to help unravel how changes in neuronal functions relate to sickness responses.
Collapse
|
19
|
Ramos I, Fernandez-Sesma A. Modulating the Innate Immune Response to Influenza A Virus: Potential Therapeutic Use of Anti-Inflammatory Drugs. Front Immunol 2015; 6:361. [PMID: 26257731 PMCID: PMC4507467 DOI: 10.3389/fimmu.2015.00361] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/04/2015] [Indexed: 12/27/2022] Open
Abstract
Infection by influenza A viruses (IAV) is frequently characterized by robust inflammation that is usually more pronounced in the case of avian influenza. It is becoming clearer that the morbidity and pathogenesis caused by IAV are consequences of this inflammatory response, with several components of the innate immune system acting as the main players. It has been postulated that using a therapeutic approach to limit the innate immune response in combination with antiviral drugs has the potential to diminish symptoms and tissue damage caused by IAV infection. Indeed, some anti-inflammatory agents have been shown to be effective in animal models in reducing IAV pathology as a proof of principle. The main challenge in developing such therapies is to selectively modulate signaling pathways that contribute to lung injury while maintaining the ability of the host cells to mount an antiviral response to control virus replication. However, the dissection of those pathways is very complex given the numerous components regulated by the same factors (i.e., NF kappa B transcription factors) and the large number of players involved in this regulation, some of which may be undescribed or unknown. This article provides a comprehensive review of the current knowledge regarding the innate immune responses associated with tissue damage by IAV infection, the understanding of which is essential for the development of effective immunomodulatory drugs. Furthermore, we summarize the recent advances on the development and evaluation of such drugs as well as the lessons learned from those studies.
Collapse
Affiliation(s)
- Irene Ramos
- Department of Microbiology, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| | - Ana Fernandez-Sesma
- Department of Microbiology, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| |
Collapse
|
20
|
Stankiewicz AM, Goscik J, Swiergiel AH, Majewska A, Wieczorek M, Juszczak GR, Lisowski P. Social stress increases expression of hemoglobin genes in mouse prefrontal cortex. BMC Neurosci 2014; 15:130. [PMID: 25472829 PMCID: PMC4269175 DOI: 10.1186/s12868-014-0130-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 11/18/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In order to better understand the effects of social stress on the prefrontal cortex, we investigated gene expression in mice subjected to acute and repeated social encounters of different duration using microarrays. RESULTS The most important finding was identification of hemoglobin genes (Hbb-b1, Hbb-b2, Hba-a1, Hba-a2, Beta-S) as potential markers of chronic social stress in mice. Expression of these genes was progressively increased in animals subjected to 8 and 13 days of repeated stress and was correlated with altered expression of Mgp (Mglap), Fbln1, 1500015O10Rik (Ecrg4), SLC16A10, and Mndal. Chronic stress increased also expression of Timp1 and Ppbp that are involved in reaction to vascular injury. Acute stress did not affect expression of hemoglobin genes but it altered expression of Fam107a (Drr1) and Agxt2l1 (Etnppl) that have been implicated in psychiatric diseases. CONCLUSIONS The observed up-regulation of genes associated with vascular system and brain injury suggests that stressful social encounters may affect brain function through the stress-induced dysfunction of the vascular system.
Collapse
Affiliation(s)
- Adrian M Stankiewicz
- Department of Animal Behavior, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552, Magdalenka, Poland.
| | - Joanna Goscik
- Faculty of Computer Science, Bialystok University of Technology, Wiejska 45A, 15-351, Bialystok, Poland.
| | - Artur H Swiergiel
- Department of Human and Animal Physiology, Institute of Biology, University of Gdansk, 80-308, Gdansk, Poland.
| | - Alicja Majewska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland.
| | - Marek Wieczorek
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Pomorska, 141/143, Poland.
| | - Grzegorz R Juszczak
- Department of Animal Behavior, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552, Magdalenka, Poland.
| | - Paweł Lisowski
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552, Magdalenka, Poland. .,iPS Cell-Based Disease Modeling Group, Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, 13092, Berlin, Germany.
| |
Collapse
|
21
|
Protective effect of mangiferin against lipopolysaccharide-induced depressive and anxiety-like behaviour in mice. Eur J Pharmacol 2014; 740:337-45. [DOI: 10.1016/j.ejphar.2014.07.031] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 05/12/2014] [Accepted: 07/10/2014] [Indexed: 12/13/2022]
|
22
|
Araki R, Hiraki Y, Yabe T. Genipin attenuates lipopolysaccharide-induced persistent changes of emotional behaviors and neural activation in the hypothalamic paraventricular nucleus and the central amygdala nucleus. Eur J Pharmacol 2014; 741:1-7. [PMID: 25084220 DOI: 10.1016/j.ejphar.2014.07.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 12/11/2022]
Abstract
Sickness behavior is a series of behavioral and psychological changes that develop in inflammatory disease, including infections and cancers. Administration of the bacterial endotoxin lipopolysaccharide (LPS) induces sickness behavior in rodents. Genipin, an aglycon derived from an iridoid glycoside geniposide extracted from the fruit of Gardenia jasminoides, has anti-inflammatory and antidepressant activities. However, the effects of genipin on inflammation-induced changes in emotional behaviors are unknown. In this study, we examined the effects of genipin on LPS-induced inflammation in BV-2 cells and sickness behavior in mice. Pretreatment with genipin inhibited LPS-induced increases in NO production and reduced the mRNA levels of inflammation-related genes (iNOS, COX-2, IL-1β and IL-6) in BV-2 cells. Oral administration of genipin ameliorated LPS-induced depressive-like behavior in the forced swim test and social behavior deficits 24h after LPS administration in mice. LPS-induced expression of mRNAs for inflammation-related genes and the number of c-fos immunopositive cells decreased in the paraventricular nucleus (PVN) of the hypothalamus and the central nucleus of the amygdala (CeA), suggesting that genipin attenuates LPS-induced changes of emotional behaviors through inhibition of neural activation and inflammatory responses in the PVN and CeA. These novel pharmacological effects of genipin may be useful for treatment of patients with sickness behavior.
Collapse
Affiliation(s)
- Ryota Araki
- Department of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan.
| | - Yosuke Hiraki
- Department of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | - Takeshi Yabe
- Department of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan.
| |
Collapse
|
23
|
Sanders CJ, Johnson B, Frevert CW, Thomas PG. Intranasal influenza infection of mice and methods to evaluate progression and outcome. Methods Mol Biol 2014; 1031:177-88. [PMID: 23824900 DOI: 10.1007/978-1-62703-481-4_20] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In vivo influenza infection models are critical for understanding viral dynamics and host responses during infection. Mouse models are extremely useful for infection studies requiring a high number of test animals. The vast array of gene knockout mice available is particularly helpful in investigating a particular gene's contributions to infection. Thus, more in vivo scientific experimentation of influenza has been done on mice than any other animal model. Here, we describe the technique of intranasal inoculation of mice and methods for assessing the severity of disease and humane endpoints, and discuss data acquired from infection of female C57BL/6J mice.
Collapse
Affiliation(s)
- Catherine J Sanders
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | | | | |
Collapse
|
24
|
Immunoglobulin-Mediated Neuro-Cognitive Impairment: New Data and a Comprehensive Review. Clin Rev Allergy Immunol 2013; 45:248-55. [DOI: 10.1007/s12016-013-8357-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Arnett SV, Clark IA. Inflammatory fatigue and sickness behaviour - lessons for the diagnosis and management of chronic fatigue syndrome. J Affect Disord 2012; 141:130-42. [PMID: 22578888 DOI: 10.1016/j.jad.2012.04.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 04/09/2012] [Accepted: 04/09/2012] [Indexed: 12/18/2022]
Abstract
Persistent and severe fatigue is a common part of the presentation of a diverse range of disease processes. There is a growing body of evidence indicating a common inflammatory pathophysiology underlying many conditions where fatigue is a primary patient concern, including chronic fatigue syndrome. This review explores current models of how inflammatory mediators act on the central nervous system to produce fatigue and sickness behaviour, and the commonality of these processes in conditions as diverse as surgical trauma, infection, various cancers, inflammatory bowel disease, connective tissue diseases and autoimmune diseases. We also discuss evidence indicating chronic fatigue syndrome may have important pathophysiological similarities with cytokine mediated sickness behaviour, and what lessons can be applied from sickness behaviour to chronic fatigue syndrome with regards to the diagnosis and management.
Collapse
Affiliation(s)
- S V Arnett
- Research School of Biology, Australian National University, Australia.
| | | |
Collapse
|
26
|
Abstract
INTRODUCTION Depression is associated with inflammation, Th1 and Th17 responses, oxidative and nitrosative stress (O&NS), autoimmune responses against neoantigenic determinants, and neuroprogression (i.e., neurodegeneration, impaired plasticity and reduced neurogenesis). These pathways involve increased monocytic activation and interleukin-1 (IL-1) levels. AREAS COVERED This review will highlight the putative role of IL-1 in depression and the potential use of IL-1 signaling blockade as a treatment of depression. Electronic databases, i.e., Scopus, PUBMED and Google Scholar were employed using keywords: depression, depressive-like, interleukin-1, and interleukin-1 receptor antagonist (IL-1RA). EXPERT OPINION Ample studies show that depression is accompanied by increased levels of IL-1 and IL-1RA, which attenuates the pro-inflammatory activities of IL-1. In some, but not all studies, antidepressant treatment decreased IL-1β levels. In translational models, IL-1β administration elicits depressive-like behaviors, neuroinflammation and neuroprogression, whereas treatment with IL-1RA yields antidepressant-like effects and attenuates neuroprogression. Anakinra, an IL-1RA, targets not only IL-1 signaling, but also Th1, Th17, O&NS and neuroprogressive pathways and therefore may be advanced to clinical Phase-II trials in depression due to medical conditions associated with an elevated IL-1/IL-1RA ratio.
Collapse
|
27
|
Richey L, Doremus-Fitzwater TL, Buck HM, Deak T. Acute illness-induced behavioral alterations are similar to those observed during withdrawal from acute alcohol exposure. Pharmacol Biochem Behav 2012; 103:284-94. [PMID: 22921768 DOI: 10.1016/j.pbb.2012.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 06/27/2012] [Accepted: 08/07/2012] [Indexed: 01/09/2023]
Abstract
Exposure to an immunogen results in a constellation of behavioral changes collectively referred to as "sickness behaviors," with alterations in cytokine expression previously shown to contribute to this sickness response. Since behaviors observed during ethanol withdrawal are strikingly similar to sickness behaviors, we hypothesized that behavioral manifestations of ethanol withdrawal might be an expression of sickness behaviors induced by ethanol-related changes in peripheral and/or central cytokine expression. Accordingly, behaviors exhibited during a modified social investigation test were first characterized in male rats following an acute injection of lipopolysaccharide (LPS; 100μg/kg). Subsequently, behavioral changes after either a high (4-g/kg; Experiment 2) or low dose (0.5g/kg; Experiment 3) of ethanol were also examined in the same social investigation test, as well as in the forced-swim test (FST; Experiment 4). Results from these experiments demonstrated similar reductions in both exploration and social investigatory behavior during acute illness and ethanol withdrawal, while a seemingly paradoxical decrease in immobility was observed in the FST during acute ethanol withdrawal. In follow-up studies, neither indomethacin (Experiment 5) nor interleukin-1 receptor antagonist (Experiment 6) pre-exposure reversed the ethanol withdrawal-induced behavioral changes observed in this social investigation test. Taken together, these studies demonstrate that the behavioral sequelae of acute illness and ethanol withdrawal are similar in nature, while antagonist studies suggest that these behavioral alterations are not reversed by blockade of IL-1 receptors or inhibition of prostaglandin synthesis. Though a direct mechanistic link between cytokines and the expression of acute ethanol withdrawal-related behaviors has yet to be found, future studies examining the involvement of brain cytokines as potential mediators of ethanol effects are greatly needed.
Collapse
Affiliation(s)
- Laura Richey
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, USA
| | | | | | | |
Collapse
|
28
|
Haba R, Shintani N, Onaka Y, Wang H, Takenaga R, Hayata A, Baba A, Hashimoto H. Lipopolysaccharide affects exploratory behaviors toward novel objects by impairing cognition and/or motivation in mice: Possible role of activation of the central amygdala. Behav Brain Res 2011; 228:423-31. [PMID: 22209851 DOI: 10.1016/j.bbr.2011.12.027] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 12/10/2011] [Accepted: 12/16/2011] [Indexed: 12/14/2022]
Abstract
Lipopolysaccharide (LPS) produces a series of systemic and psychiatric changes called sickness behavior. In the present study, we characterized the LPS-induced decrease in novel object exploratory behaviors in BALB/c mice. As already reported, LPS (0.3-5 μg/mouse) induced dose- and time-dependent decreases in locomotor activity, food intake, social interaction, and exploration for novel objects, and an increase in immobility in the forced-swim test. Although the decrease in locomotor activity was ameliorated by 10h postinjection, novel object exploratory behaviors remained decreased at 24h and were observed even with the lowest dose of LPS. In an object exploration test, LPS shortened object exploration time but did not affect moving time or the frequency of object exploration. Although pre-exposure to the same object markedly decreased the duration of exploration and LPS did not change this reduction, LPS significantly impaired the exploration of a novel object that replaced the familiar one. LPS did not affect anxiety-like behaviors in open-field and elevated plus-maze tests. An LPS-induced increase in the number of c-Fos-immunoreactive cells was observed in several brain regions within 6h of LPS administration, but the number of cells quickly returned to control levels, except in the central amygdala where the increase continued for 24h. These results suggest that LPS most prominently affects object exploratory behaviors by impairing cognition and/or motivation including continuous attention and curiosity toward objects, and that this may be associated with activation of brain nuclei such as the central amygdala.
Collapse
Affiliation(s)
- Ryota Haba
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Moon ML, McNeil LK, Freund GG. Macrophages make me sick: how macrophage activation states influence sickness behavior. Psychoneuroendocrinology 2011; 36:1431-40. [PMID: 21855222 PMCID: PMC3199305 DOI: 10.1016/j.psyneuen.2011.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 06/16/2011] [Accepted: 07/03/2011] [Indexed: 12/18/2022]
Abstract
The macrophage (MΦ) is an essential cellular first responder in the innate immune system, sensing, alerting, removing and destroying intrinsic and extrinsic pathogens. While congenital aplasia of granulocytes, T or B lymphocytes leads to serious disease, lack of MΦs is incompatible with life. The MΦ, however, is not a monomorphic entity. These constructers, repairers and defenders of the body are diverse in form and function. What controls MΦ phenotype is beginning to be understood and involves a complex interplay of origination, location and microenvironment. Common to all MΦ developmental pathways are pro-inflammatory and anti-inflammatory cytokines. MΦs respond to these bioactives in distinct ways developing recently recognized activation phenotypes that canonically support bacterial clearance (classical activation), parasite defense/tissue repair (alternative activation) and anti-inflammation (deactivation). Critically, the same cytokines which orchestrate immune defense and homeostasis dramatically impact sense of well being and cognition by eliciting sickness symptoms. Such behaviors are the manifestation of pro/anti-inflammatory cytokine action in the brain and are a direct consequence of MΦ function. This review describes the "new" archetypal MΦ activation states, delineates microglia phenotypic plasticity and explores the importance of these macrophage activation states to sickness behavior.
Collapse
Affiliation(s)
- Morgan L. Moon
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Leslie K. McNeil
- Department of Pathology, University of Illinois, Urbana, IL 61801, USA
| | - Gregory G. Freund
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
- Department of Pathology, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
30
|
Jost S, Quillay H, Reardon J, Peterson E, Simmons RP, Parry BA, Bryant NNP, Binder WD, Altfeld M. Changes in cytokine levels and NK cell activation associated with influenza. PLoS One 2011; 6:e25060. [PMID: 21966414 PMCID: PMC3179484 DOI: 10.1371/journal.pone.0025060] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 08/23/2011] [Indexed: 01/12/2023] Open
Abstract
Several studies have highlighted the important role played by murine natural killer (NK) cells in the control of influenza infection. However, human NK cell responses in acute influenza infection, including infection with the 2009 pandemic H1N1 influenza virus, are poorly documented. Here, we examined changes in NK cell phenotype and function and plasma cytokine levels associated with influenza infection and vaccination. We show that absolute numbers of peripheral blood NK cells, and particularly those of CD56bright NK cells, decreased upon acute influenza infection while this NK cell subset expanded following intramuscular influenza vaccination. NK cells exposed to influenza antigens were activated, with higher proportions of NK cells expressing CD69 in study subjects infected with seasonal influenza strains. Vaccination led to increased levels of CD25+ NK cells, and notably CD56bright CD25+ NK cells, whereas decreased amounts of this subset were present in the peripheral blood of influenza infected individuals, and predominantly in study subjects infected with the 2009 pandemic H1N1 influenza virus. Finally, acute influenza infection was associated with low plasma concentrations of inflammatory cytokines, including IFN-γ, MIP-1β, IL-2 and IL-15, and high levels of the anti-inflammatory cytokines IL-10 and IL-1ra. Altogether, these data suggest a role for the CD56bright NK cell subset in the response to influenza, potentially involving their recruitment to infected tissues and a local production and/or uptake of inflammatory cytokines.
Collapse
Affiliation(s)
- Stephanie Jost
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Heloise Quillay
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jeff Reardon
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Eric Peterson
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rachel P. Simmons
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Blair A. Parry
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Nancy N. P. Bryant
- Massachusetts General Hospital Medical Walk-In Clinic, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - William D. Binder
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Marcus Altfeld
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
31
|
Abstract
BACKGROUND Fatigue is common, disabling yet underappreciated, in patients with chronic diseases, including inflammatory bowel disease (IBD). AIMS To examine the literature and determine the prevalence and patterns of fatigue in IBD patients, to identify opportunities and directions for future research in this area. METHODS A systematic review using PubMed and Ovid Medline databases was conducted using search terms 'fatigue', 'Crohn', 'colitis' and 'inflammatory bowel disease'. A review of fatigue in other similar chronic diseases was also performed. RESULTS Ten studies were found to include data on fatigue in IBD patients; all were conducted between 1999 and 2009. However, only one study (in children) measured fatigue in IBD patients as a primary outcome. In patients in remission, the prevalence of fatigue in IBD patients ranges from 41 to 48%. Data are sparse and conflicting on whether fatigue severity is proportional to disease severity/activity. CONCLUSIONS Despite the clinical reality of fatigue, there are few published studies examining fatigue in IBD as a primary outcome. More data are needed on the prevalence, correlation between disease activity and fatigue severity, and putative pathogenic pathways involved in fatigue pathogenesis, before ultimately elucidating targeted therapies for fatigue in IBD patients.
Collapse
Affiliation(s)
- D R van Langenberg
- Monash University Department of Gastroenterology & Hepatology, Medicine, Box Hill Hospital, Box Hill, Melbourne, Vic., Australia.
| | | |
Collapse
|
32
|
Loftis JM, Huckans M, Morasco BJ. Neuroimmune mechanisms of cytokine-induced depression: current theories and novel treatment strategies. Neurobiol Dis 2009; 37:519-33. [PMID: 19944762 DOI: 10.1016/j.nbd.2009.11.015] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 11/05/2009] [Accepted: 11/18/2009] [Indexed: 01/16/2023] Open
Abstract
The relationships between immune and neural function are an increasingly important area of study for neuropsychiatric disorders, in particular depression. This is exemplified by the growing number of publications on cytokines and depression during the last 10 years, as compared to earlier decades. This review summarizes the current theories and novel treatment strategies for depression, with a focus on cytokine-induced depression. Neuroimmune mechanisms are now viewed as central to the development of depressive symptoms and emerging evidence is beginning to identify the neural circuits involved in cytokine-induced depression. The current diagnostic categories for depression, as defined by the Diagnostic and Statistical Manual of Mental Disorders, however, are not etiologically or biologically derived, and it has been proposed that "depression", likely reflects multiple pathogeneses leading to varying symptom constellations. As we move toward a better biological understanding of depression-related symptom constellations or syndromes, the term "depression" may prove inadequately broad, and an integration of interdisciplinary literatures will increase in importance. Future research should aim to characterize these depression-related symptom constellations or syndromes better with the goal of optimizing treatment strategies.
Collapse
Affiliation(s)
- Jennifer M Loftis
- Research and Development Service, Behavioral Health and Clinical Neurosciences Division, Portland VA Medical Center, 3710 SW US Veterans Hospital Rd., Portland, OR 97239, USA.
| | | | | |
Collapse
|
33
|
Zager A, Andersen ML, Lima MMS, Reksidler AB, Machado RB, Tufik S. Modulation of sickness behavior by sleep: the role of neurochemical and neuroinflammatory pathways in mice. Eur Neuropsychopharmacol 2009; 19:589-602. [PMID: 19394204 DOI: 10.1016/j.euroneuro.2009.03.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 02/19/2009] [Accepted: 03/24/2009] [Indexed: 01/02/2023]
Abstract
Activation of the immune system elicits several behavioral changes that are collectively called sickness behavior and consists in a strategy to overcome infection. Sleep deprivation can increase susceptibility to pathogens and to behavioral alterations. Thus, the present study aimed to determine how paradoxical sleep deprivation (PSD) affects the behavioral and neurochemical responses to lipopolysaccharide (LPS, potent activator of the immune response). Adult inbred mice were paradoxical sleep deprived (72 h), whereas the control group was kept in their home cages. Both groups received either an injection of saline or LPS (5, 10 or 20 microg/animal ip) before behavioral tasks and tissue collection. During the recovery sleep period, LPS provoked a strong inhibition of sleep rebound due to a suppression of paradoxical sleep. PSD increased the susceptibility of mice to LPS-induced immobility in the open field, which was capable of affecting the anxiety-like behavior also. These altered behavioral responses to LPS were accompanied by reduction in dopamine turnover within the striatum and increased expression of cyclooxygenase-2 in the cortex. The study provides some insights into how the sleep-wake cycle affects the expression of sickness behavior induced by LPS.
Collapse
Affiliation(s)
- Adriano Zager
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
INTRODUCTION An increasing amount of data suggests that dysregulation of the immune system, including the cytokine network, is associated with the etiology and pathophysiology of mood disorders. Genes encoding cytokines are highly polymorphic and single nucleotide polymorphisms, associated with increased or reduced cytokine production, have been described. The aim of this study was to define the genetic immunologic scenario associated with major depressive disorder (MDD) and bipolar disorder. METHODS Eighty-four Italian outpatients affected by bipolar disorder type I, bipolar disorder type II, or MDD, and 363 healthy controls were enrolled into the study. We analyzed allele and genotype distribution of -308 (G/A) tumor necrosis factor-a (TNF-a), +874 (T/A) interferon-g (IFN-g), -174 (G/C) interleukin (IL)-6, and -1082 (G/A) IL-10 promoter polymorphisms by Polymerase Chain Reaction Sequence Specific Primers technique. RESULTS We observed different genotype and allele distributions of TNF-a, IFN-g, and IL-10 polymorphisms in the three groups of patients analyzed. In particular, bipolar II patients were characterized by an absence of adenine (A) high producer allele of TNF-a (P<.001) and a lower percentage of TT high producer genotype of IFN-g (P<.001); bipolar I individuals showed reduced percentage of AA low producer genotype of IL-10 (P<.001). Both bipolar I and bipolar II patients not carrying guanine (G) high producer IL-6 allele showed a lower mean age at onset (P=.048). CONCLUSION These data support the existence of a genetic profile related to pro-inflammatory cytokines in patients affected by mood disorders. The differences observed across the three clinical phenotypes suggest the presence of different pathogenetic mechanisms involved in the susceptibility of phenotypically different mood disorders.
Collapse
|
35
|
Pecchi E, Dallaporta M, Jean A, Thirion S, Troadec JD. Prostaglandins and sickness behavior: old story, new insights. Physiol Behav 2009; 97:279-92. [PMID: 19275907 DOI: 10.1016/j.physbeh.2009.02.040] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Revised: 02/23/2009] [Accepted: 02/26/2009] [Indexed: 12/31/2022]
Abstract
Previous evidence has shown that prostaglandins play a key role in the development of sickness behavior observed during inflammatory states. In particular, prostaglandin E2 (PGE2) is produced in the brain by a variety of inflammatory signals such as endotoxins or cytokines. Its injection has been also shown to induce symptoms of sickness behavior. The role of cyclooxygenase enzymes (COX), the rate-limiting enzymes converting arachidonic acid into prostaglandins, in sickness behavior has been extensively studied, and it has been demonstrated that strategies aiming at inhibiting these enzymes limit anorexia, body weight loss and fever in animals with inflammatory diseases. However, inhibiting COX activity may lead to negative gastric or cardiovascular effects, since COX enzymes play a role in the synthesis of others prostanoids with various and sometimes contrasting properties. Recently, prostaglandin E synthases (PGES), which specifically catalyze the final step of PGE2 biosynthesis, were characterized. Among these enzymes, the microsomal prostaglandin E synthase-1 (mPGES-1) was of a particular interest since it was shown to be up-regulated by inflammatory signals in a variety of cell types. Moreover, mPGES-1 was shown to be crucial for correct immune-to-brain communication and induction of fever and anorexia by pro-inflammatory agents. This review takes stock of previous knowledge and recent advances in understanding the role of prostaglandins and of their specific synthesizing enzymes in the molecular mechanisms underlying sickness behavior. The review concludes with a short summary of key questions that remain to be addressed and points out therapeutic developments in this research field.
Collapse
Affiliation(s)
- Emilie Pecchi
- Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille, UMR 6231 CNRS, USC INRA 2027, Université Paul Cézanne et Université de la Méditerranée, Marseille, France
| | | | | | | | | |
Collapse
|
36
|
Kallapur SG, Nitsos I, Moss TJM, Polglase GR, Pillow JJ, Cheah FC, Kramer BW, Newnham JP, Ikegami M, Jobe AH. IL-1 mediates pulmonary and systemic inflammatory responses to chorioamnionitis induced by lipopolysaccharide. Am J Respir Crit Care Med 2009; 179:955-61. [PMID: 19234101 DOI: 10.1164/rccm.200811-1728oc] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
RATIONALE Chorioamnionitis frequently associates with preterm delivery and increased amniotic fluid IL-1, and causes fetal lung and systemic inflammation. However, chorioamnionitis is also associated with a paradoxical reduction in the incidence of surfactant deficiency-related respiratory distress syndrome in preterm infants. OBJECTIVES To identify the role of IL-1 signaling in the mediation of pulmonary and systemic inflammation and lung maturation in a fetal sheep model of lipopolysaccharide (LPS) induced chorioamnionitis. METHODS After confirming the efficacy of recombinant human IL-1 receptor antagonist (rhIL-1ra), fetal sheep were exposed to intraamniotic (IA) injections of Escherichia coli LPS with or without prior IA injections of rhIL-1ra. Preterm lambs were delivered at 82% of term gestation. MEASUREMENTS AND MAIN RESULTS rhIL-1ra decreased IA LPS-induced lung inflammation assessed by decreased lung neutrophil and monocyte influx, inducible nitric oxide synthase expression, lung IL-6 and IL-1beta mRNA expression, and airway myeloperoxidase concentrations. rhIL-1ra inhibited IA LPS-induced fetal systemic inflammation assessed by decreased plasma IL-8, protein carbonyls, blood neutrophilia, and the expression of serum amyloid A3 mRNA in the liver. rhIL-1ra also partially blocked the lung maturational effects of IA LPS. Therefore blockade of IL-1 signaling in the amniotic compartment inhibited fetal lung and systemic inflammation and lung maturation in response to LPS-induced chorioamnionitis. CONCLUSIONS IL-1 plays a central role in the pathogenesis of chorioamnionitis-induced fetal inflammatory responses.
Collapse
Affiliation(s)
- Suhas G Kallapur
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Dawe K. Ob or not ob? Is leptin involved in hypoxia-induced social withdrawal? Brain Behav Immun 2009; 23:167-8. [PMID: 19041392 DOI: 10.1016/j.bbi.2008.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Accepted: 11/03/2008] [Indexed: 11/26/2022] Open
Affiliation(s)
- Karen Dawe
- Department of Social Medicine, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol BS8 2PS, UK.
| |
Collapse
|
38
|
Activation of the maternal immune system alters cerebellar development in the offspring. Brain Behav Immun 2009; 23:116-23. [PMID: 18755264 PMCID: PMC2614890 DOI: 10.1016/j.bbi.2008.07.012] [Citation(s) in RCA: 200] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Revised: 07/19/2008] [Accepted: 07/29/2008] [Indexed: 11/23/2022] Open
Abstract
A common pathological finding in autism is a localized deficit in Purkinje cells (PCs). Cerebellar abnormalities have also been reported in schizophrenia. Using a mouse model that exploits a known risk factor for these disorders, maternal infection, we asked if the offspring of pregnant mice given a mid-gestation respiratory infection have cerebellar pathology resembling that seen in these disorders. We also tested the effects of maternal immune activation in the absence of virus by injection of the synthetic dsRNA, poly(I:C). We infected pregnant mice with influenza on embryonic day 9.5 (E9.5), or injected poly(I:C) i.p. on E12.5, and assessed the linear density of PCs in the cerebellum of adult or postnatal day 11 (P11) offspring. To study granule cell migration, we also injected BrdU on P11. Adult offspring of influenza- or poly(I:C)-exposed mice display a localized deficit in PCs in lobule VII of the cerebellum, as do P11 offspring. Coincident with this are heterotopic PCs, as well as delayed migration of granule cells in lobules VI and VII. The cerebellar pathology observed in the offspring of influenza- or poly(I:C)-exposed mice is strikingly similar to that observed in autism. The poly(I:C) findings indicate that deficits are likely caused by the activation of the maternal immune system. Finally, our data suggest that cerebellar abnormalities occur during embryonic development, and may be an early deficit in autism and schizophrenia.
Collapse
|
39
|
Otitis Media and Sinusitis. ALLERGY FRONTIERS: CLINICAL MANIFESTATIONS 2009. [PMCID: PMC7121333 DOI: 10.1007/978-4-431-88317-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Otitis media (OM) and sinusitis are common and costly maladies that are often preceded by the development of a viral upper respiratory infection (vURI). Although antibiotics have been shown to be somewhat effective in the treatment of these disorders, increasing concern over the emergence of pathogen resistence to these agents underscores the need for the development of other treatment options, including agents to treat and/or prevent vURIs. Earlier research implicated roles for cytopathology, cellular infiltration, and inflammatory mediators such as bradykinin, in the pathogenesis of vURIs and its complications, including OM and sinusitis, but these factors are now recognized as late events with specific and limited contributions to disease expression. Current therapies are relatively ineffective and aimed at reducing symptoms rather than moderating underlying mechanisms. Nasal elevations of proinflammatory cytokines and leukotrienes track symptom expression during vURIs, and it is hypothesized that these chemicals orchestrate a common response to infection with many different viruses causing vURIs. Moreover, recent evidence demonstrates that specific cytokine gene polymorphisms may modulate the severity of illness and incidence of complications during episodes of vURI. Additionally, other evidence supports a role for neurogenic inflammation in the development of complications. Future studies should dissect the role of proinflammatory cytokines, leukotrienes, and neuropeptides in the expression of symptoms, signs, pathophysiologies, and complications of vURIs.
Collapse
|
40
|
Clark IA, Budd AC, Alleva LM. Sickness behaviour pushed too far--the basis of the syndrome seen in severe protozoal, bacterial and viral diseases and post-trauma. Malar J 2008; 7:208. [PMID: 18854046 PMCID: PMC2576339 DOI: 10.1186/1475-2875-7-208] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Accepted: 10/14/2008] [Indexed: 12/03/2022] Open
Abstract
Certain distinctive components of the severe systemic inflammatory syndrome are now well-recognized to be common to malaria, sepsis, viral infections, and post-trauma illness. While their connection with cytokines has been appreciated for some time, the constellation of changes that comprise the syndrome has simply been accepted as an empirical observation, with no theory to explain why they should coexist. New data on the effects of the main pro-inflammatory cytokines on the genetic control of sickness behaviour can be extended to provide a rationale for why this syndrome contains many of its accustomed components, such as reversible encephalopathy, gene silencing, dyserythropoiesis, seizures, coagulopathy, hypoalbuminaemia and hypertriglyceridaemia. It is thus proposed that the pattern of pathology that comprises much of the systemic inflammatory syndrome occurs when one of the usually advantageous roles of pro-inflammatory cytokines – generating sickness behaviour by moderately repressing genes (Dbp, Tef, Hlf, Per1, Per2 and Per3, and the nuclear receptor Rev-erbα) that control circadian rhythm – becomes excessive. Although reversible encephalopathy and gene silencing are severe events with potentially fatal consequences, they can be viewed as having survival advantages through lowering energy demand. In contrast, dyserythropoiesis, seizures, coagulopathy, hypoalbuminaemia and hypertriglyceridaemia may best be viewed as unfortunate consequences of extreme repression of these same genetic controls when the pro-inflammatory cytokines that cause sickness behaviour are produced excessively. As well as casting a new light on the previously unrationalized coexistence of these aspects of systemic inflammatory diseases, this concept is consistent with the case for a primary role for inflammatory cytokines in their pathogenesis across this range of diseases.
Collapse
Affiliation(s)
- Ian A Clark
- School of Biochemistry and Molecular Biology, Australian National University, Canberra, Australia.
| | | | | |
Collapse
|
41
|
Saito M, Watanabe S. Differential modulation of lipopolysaccharide- and zymosan-induced hypophagia by dexamethasone treatment. Pharmacol Biochem Behav 2008; 90:428-33. [DOI: 10.1016/j.pbb.2008.03.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Revised: 03/25/2008] [Accepted: 03/29/2008] [Indexed: 10/22/2022]
|
42
|
Teeling JL, Felton LM, Deacon RMJ, Cunningham C, Rawlins JNP, Perry VH. Sub-pyrogenic systemic inflammation impacts on brain and behavior, independent of cytokines. Brain Behav Immun 2007; 21:836-50. [PMID: 17367989 DOI: 10.1016/j.bbi.2007.01.012] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Revised: 01/11/2007] [Accepted: 01/23/2007] [Indexed: 12/26/2022] Open
Abstract
Systemic inflammation impacts on the brain and gives rise to behavioral changes, often referred to as 'sickness behavior'. These symptoms are thought to be mainly mediated by pro-inflammatory cytokines. We have investigated the communication pathways between the immune system and brain following sub-pyrogenic inflammation. Low grade systemic inflammation was induced in mice using lipopolysaccharide (LPS); 1-100 microg/kg to mimic aspects of bacterial infection. Changes in fever, open-field activity, burrowing and consumption of glucose solution were assessed and immune activation was studied in the periphery and brain by measuring cytokine production, and immunohistochemistry to study changes in immune cell phenotype. Sub-pyrogenic inflammation resulted in changes in a species-typical, untrained behavior (burrowing) that depends on the integrity of the hippocampus. Increased expression of cytokines was observed in the periphery and selected regions of the brain which coincided with changes in behavior. However, peripheral neutralization of LPS-induced pro-inflammatory cytokines IL-1beta, IL-6 and TNF-alpha did not abrogate the LPS-induced behavioral changes nor affect CNS cytokine synthesis. In contrast, pretreatment of mice with indomethacin completely prevented LPS-induced behavior changes, without affecting cytokine levels. Taken together, these experiments suggest a key role for prostaglandins, rather than cytokines, in communicating to the brain.
Collapse
Affiliation(s)
- J L Teeling
- CNS Inflammation Group, School of Biological Sciences, University of Southampton, Bassett Crescent East, UK.
| | | | | | | | | | | |
Collapse
|
43
|
van den Biggelaar AHJ, Gussekloo J, de Craen AJM, Frölich M, Stek ML, van der Mast RC, Westendorp RGJ. Inflammation and interleukin-1 signaling network contribute to depressive symptoms but not cognitive decline in old age. Exp Gerontol 2007; 42:693-701. [PMID: 17350781 DOI: 10.1016/j.exger.2007.01.011] [Citation(s) in RCA: 158] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2006] [Accepted: 01/30/2007] [Indexed: 11/23/2022]
Abstract
The association between inflammation and neuropsychiatric symptoms in old age is generally accepted but poorly understood. The purpose of this study was to examine whether inflammation precedes depressive symptoms and cognitive decline in old age, and to identify specific inflammatory pathways herein. We measured serum C-reactive protein (CRP) and lipopolysaccharide-induced production of Interleukin (IL)-1beta, IL-6, Tumor Necrosis Factor (TNF)-alpha, IL-1 receptor antagonist (ra), and IL-10 levels in 85-year-old participants free from neuropsychiatric symptoms at baseline (n=267). Participants were prospectively followed for depressive symptoms (Geriatric Depression Scale) and cognitive functioning (Mini Mental State Examination) from 85 to 90 years. Higher baseline CRP levels preceded accelerated increase in depressive symptoms (p<0.001). A higher production capacity of the pro-inflammatory cytokine IL-1beta preceded a greater increase of depressive symptoms (p=0.06), whereas that of its natural antagonist IL-1ra preceded a smaller increase of depressive symptoms (p=0.003). There was no relation of CRP, IL-1beta, and IL-1ra with cognitive decline. Our findings show that in old age inflammatory processes contribute to the development of depressive symptoms but not cognitive decline. A high innate IL-1ra to IL-1beta production capacity reflects a better ability to neutralize inflammation and may therefore protect against depressive symptoms.
Collapse
|
44
|
van Os E, van den Broek WW, Mulder PGH, ter Borg PCJ, Bruijn JA, van Buuren HR. Depression in patients with primary biliary cirrhosis and primary sclerosing cholangitis. J Hepatol 2007; 46:1099-103. [PMID: 17399846 DOI: 10.1016/j.jhep.2007.01.036] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 12/31/2006] [Accepted: 01/01/2007] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIMS Former studies reported a high prevalence of depression in patients with primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC). These studies hypothesized that the presence of depression could explain the fatigue experienced by these patients. METHODS Our aim was to study the prevalence of depression in a Dutch population with PBC and PSC. In addition, to investigating the effects of using an additional diagnostic structured psychiatric interview, after screening with the Beck Depression Inventory (BDI), a self-report severity scale instrument used in former studies. Patients with PBC and PSC (n=92) completed the BDI. Patients with scores of 10 or higher (n=39) were interviewed using a structured psychiatric interview. Patients with scores lower than 10 were at random (30/53, 57%) also interviewed using a structured psychiatric interview. RESULTS Of the 92 patients that were included 42% had depressive symptoms according to the BDI. However, of these patients only 3.7% had a depressive syndrome according to the DSM-IV criteria as assessed with the structured psychiatric interview. CONCLUSIONS The prevalence of a depressive disorder in patients with PBC and PSC is not higher than in the general population. Fatigue in patients with PBC and PSC cannot be explained by depression.
Collapse
Affiliation(s)
- Erik van Os
- Department of Psychiatry, Erasmus Medical Centre, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
45
|
Schubert C, Hong S, Natarajan L, Mills PJ, Dimsdale JE. The association between fatigue and inflammatory marker levels in cancer patients: a quantitative review. Brain Behav Immun 2007; 21:413-27. [PMID: 17178209 DOI: 10.1016/j.bbi.2006.11.004] [Citation(s) in RCA: 246] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Revised: 11/03/2006] [Accepted: 11/03/2006] [Indexed: 11/16/2022] Open
Abstract
Increased cytokine and neopterin levels may be responsible for cancer-related fatigue, the most common complaint among cancer patients. We quantitatively reviewed empirical findings on this topic, focusing on studies not using immunotherapy. PubMed, PsychINFO and BIOSIS were searched for articles published until July 2006. Studies remained unweighted or were weighted according to study quality and sample size. The correlation coefficient r was used for statistical analyses. Heterogeneity among the studies was examined using the I(2) index. Eighteen studies (1037 participants) of moderately high methodological quality were located and statistically analyzed. Most studies measured more than one inflammatory marker, resulting in a total of 58 correlation estimates. In 31 of these, we had to input a null correlation because results had been simply reported as nonsignificant and no further statistical information was available. General analyses based on weighting according to sample size showed a significantly positive correlation between fatigue and circulating levels of inflammatory markers (r=0.11, p<0.0001). Analyses of individual inflammatory markers revealed significantly positive correlations between fatigue and IL-6 (r=0.12, p=0.004), fatigue and IL-1 ra (r=0.24, p=0.0005), and fatigue and neopterin (r=0.22, p=0.0001). Fatigue did not correlate significantly with IL-1 beta (r=0.05, p=0.42) or TNF-alpha (r=0.04, p=0.34). Given its preliminary nature due to the limited available data, this quantitative review showed a positive association between cancer-related fatigue and circulating levels of IL-6, IL-1 ra and neopterin. Future studies examining the relationship between cancer related fatigue and inflammation would benefit from multiple rather than single blood sampling and from repeated daily ratings of the multidimensional nature of fatigue.
Collapse
Affiliation(s)
- Christian Schubert
- Clinical Department of Medical Psychology and Psychotherapy, Innsbruck Medical University, Innsbruck, Austria.
| | | | | | | | | |
Collapse
|
46
|
Silverman MN, Macdougall MG, Hu F, Pace TWW, Raison CL, Miller AH. Endogenous glucocorticoids protect against TNF-alpha-induced increases in anxiety-like behavior in virally infected mice. Mol Psychiatry 2007; 12:408-17. [PMID: 17389906 PMCID: PMC3392959 DOI: 10.1038/sj.mp.4001921] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Endogenous glucocorticoids restrain proinflammatory cytokine responses to immune challenges such as viral infection. In addition, proinflammatory cytokines induce behavioral alterations including changes in locomotor/exploratory activity. Accordingly, we examined proinflammatory cytokines and open-field behavior in virally infected mice rendered glucocorticoid deficient by adrenalectomy (ADX). Mice were infected with murine cytomegalovirus (MCMV), and open-field behavior (36 h post-infection) and plasma concentrations of tumor necrosis factor (TNF)-alpha and interleukin (IL)-6 (42 h post-infection) were assessed. Compared to sham-ADX-MCMV-infected animals, ADX-MCMV-infected mice exhibited significant reductions in total distance moved, number of center entries, and time spent in center. These behavioral alterations were accompanied by significantly higher plasma concentrations of TNF-alpha and IL-6, both of which were correlated with degree of behavioral change. To examine the role of TNF-alpha in these behavioral alterations, open-field behavior was compared in wild-type (WT) and TNF-R1-knockout (KO), ADX-MCMV-infected mice. TNF-R1-KO mice exhibited significantly attenuated decreases in number of rearings, number of center entries and time spent in center, but not distance moved, which correlated with plasma IL-6. Given the potential role of brain cytokines in these findings, mRNA expression of TNF-alpha, IL-1 and IL-6 was assessed in various brain regions. Although MCMV induced increases in proinflammatory cytokine mRNA throughout the brain (especially in ADX animals), no remarkable differences were found between WT and TNF-R1-KO mice. These results demonstrate that endogenous glucocorticoids restrain proinflammatory cytokine responses to viral infection and their impact on locomotor/exploratory activity. Moreover, TNF-alpha appears to mediate cytokine-induced changes in open-field behaviors, especially those believed to reflect anxiety.
Collapse
Affiliation(s)
- MN Silverman
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - MG Macdougall
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - F Hu
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - TWW Pace
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - CL Raison
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - AH Miller
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
47
|
Santoro TJ, Tomita M, Larson SJ. The potential impact of sickness-motivated behavior on the expression of neuropsychiatric disturbances in systemic lupus erythematosus. Med Hypotheses 2007; 69:502-7. [PMID: 17399911 DOI: 10.1016/j.mehy.2007.01.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Accepted: 01/09/2007] [Indexed: 11/18/2022]
Abstract
Activation of the peripheral immune system is often accompanied by changes in cognition, ingestive behavior, sleep pattern, and sexual drive; collectively referred to as sickness behavior. Mounting evidence suggests that sickness behavior may be a purposeful attempt on the part of an organism to conserve energy and thereby facilitate recuperation. Illnesses characterized by chronic, uncontrolled immune reactivity such as systemic lupus erythematosus are also frequently associated with impaired emotionality and cognition; which, unlike sickness behavior, are conventionally thought to emanate from fixed structural lesions of the brain. Clinical observations, however, indicate that the neuropsychiatric disturbances in lupus may wax and wane in intensity and suggest the hypothesis that sickness-motivated behavior may significantly influence the neuropsychiatric manifestations of systemic lupus erythematosus and, perhaps, those of other autoimmune diseases associated with neuroinflammation. The hypothesis that patients with systemic lupus erythematosus undergo a reorganization of their motivational priorities, which influences cognitive performance and emotional output, may be examined using validated behavior paradigms in autoimmune MRL-MpJ-Tnfrsf6(lpr) (MRL-lpr/lpr) mice that spontaneously develop a lupus-like illness accompanied by disturbances in cognition and emotionality. Confirming that sickness-motivated behavior contributes to the aberrations in cognition and emotionality exhibited by an experimental model of systemic lupus erythematosus might have important therapeutic and prognostic implications by invoking the possibility that similar motivational effects may be influencing cognitive and/or emotional output in patients with neuropsychiatric lupus.
Collapse
Affiliation(s)
- Thomas J Santoro
- Department of Graduate Medical Education, University of Illinois College of Medicine at Peoria, One Illini Drive, Box 1649, Peoria, IL 61605, United States.
| | | | | |
Collapse
|
48
|
Jayasekera JP, Vinuesa CG, Karupiah G, King NJC. Enhanced antiviral antibody secretion and attenuated immunopathology during influenza virus infection in nitric oxide synthase-2-deficient mice. J Gen Virol 2006; 87:3361-3371. [PMID: 17030871 DOI: 10.1099/vir.0.82131-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
NOS2 gene-deficient (NOS2−/−) mice are less susceptible than wild-type (NOS2+/+) mice to infection with Influenza A virus. Virus titres in the lungs of influenza-infected NOS2−/− mice are significantly lower than those in NOS2+/+ mice, with enhanced viral clearance in NOS2−/− mice dependent on gamma interferon (IFN-γ). The current study was undertaken to ascertain the role of specific components of the immune response in promoting virus clearance in influenza-infected NOS2−/− mice. Levels of T cell- and natural killer cell-mediated cytotoxicity in the lungs of virus-infected mice were not significantly different between NOS2+/+ and NOS2−/− mice. However, virus-infected NOS2−/− mice produced higher levels of virus-specific IgG2a antibody. Furthermore, more viable B cells and plasmablasts, along with greater levels of IFN-γ, were found in NOS2−/− splenocyte cultures stimulated with B-cell mitogens. In addition to the early reduction in virus titres, clinical symptoms and proinflammatory cytokine production were attenuated in NOS2−/− mice. Thus, NOS2−/− B cells are capable of responding rapidly to influenza virus infection by proliferating and preferentially producing antibody of the IgG2a subtype. The relationship between viral load and the development of immunopathology is discussed.
Collapse
Affiliation(s)
- Jerome P Jayasekera
- Department of Pathology, Bosch Institute, School of Biomedical Sciences, Blackburn Building D06, University of Sydney, NSW 2006, Australia
| | - Carola G Vinuesa
- Division of Immunology and Genetics, John Curtin School of Medical Research (JCSMR), Australian National University, Canberra, ACT 2601, Australia
| | - Gunasegaran Karupiah
- Division of Immunology and Genetics, John Curtin School of Medical Research (JCSMR), Australian National University, Canberra, ACT 2601, Australia
| | - Nicholas J C King
- Department of Pathology, Bosch Institute, School of Biomedical Sciences, Blackburn Building D06, University of Sydney, NSW 2006, Australia
| |
Collapse
|
49
|
Elander L, Engström L, Hallbeck M, Blomqvist A. IL-1beta and LPS induce anorexia by distinct mechanisms differentially dependent on microsomal prostaglandin E synthase-1. Am J Physiol Regul Integr Comp Physiol 2006; 292:R258-67. [PMID: 16946079 DOI: 10.1152/ajpregu.00511.2006] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent work demonstrated that the febrile response to peripheral immune stimulation with proinflammatory cytokine IL-1beta or bacterial wall lipopolysaccharide (LPS) is mediated by induced synthesis of prostaglandin E(2) by the terminal enzyme microsomal prostaglandin E synthase-1 (mPGES-1). The present study examined whether a similar mechanism might also mediate the anorexia induced by these inflammatory agents. Transgenic mice with a deletion of the Ptges gene, which encodes mPGES-1, and wild-type controls were injected intraperitoneally with IL-1beta, LPS, or saline. Mice were free fed, and food intake was continuously monitored with an automated system for 12 h. Body weight was recorded every 24 h for 4 days. The IL-1beta induced anorexia in wild-type but not knock-out mice, and so it was almost completely dependent on mPGES-1. In contrast, LPS induced anorexia of the same magnitude in both phenotypes, and hence it was independent of mPGES-1. However, when the mice were prestarved for 22 h, LPS induced anorexia and concomitant body weight loss in the knock-out animals that was attenuated compared with the wild-type controls. These data suggest that IL-1beta and LPS induce anorexia by distinct immune-to-brain signaling pathways and that the anorexia induced by LPS is mediated by a mechanism different from the fever induced by LPS. However, nutritional state and/or motivational factors also seem to influence the pathways for immune signaling to the brain. Furthermore, both IL-1beta and LPS caused reduced meal size but not meal frequency, suggesting that both agents exerted an anhedonic effect during these experimental conditions.
Collapse
Affiliation(s)
- Louise Elander
- Division of Cell Biology, Department of Biomedicine and Surgery, Faculty of Health Sciences, Linköping University, S-58185 Linköping, Sweden
| | | | | | | |
Collapse
|
50
|
Abstract
Administration of cytokines to animals can elicit many effects on the brain, particularly neuroendocrine and behavioral effects. Cytokine administration also alters neurotransmission, which may underlie these effects. The most well studied effect is the activation of the hypothalamo-pituitary-adrenocortical (HPA) axis, especially that by interleukin-1 (IL-1). Peripheral and central administration of IL-1 also induces norepinephrine (NE) release in the brain, most markedly in the hypothalamus. Small changes in brain dopamine (DA) are occasionally observed, but these effects are not regionally selective. IL-1 also increases brain concentrations of tryptophan, and the metabolism of serotonin (5-HT) throughout the brain in a regionally nonselective manner. Increases of tryptophan and 5-HT, but not NE, are also elicited by IL-6, which also activates the HPA axis, although it is much less potent in these respects than IL-1. IL-2 has modest effects on DA, NE and 5-HT. Like IL-6, tumor necrosis factor-α (TNFα) activates the HPA axis, but affects NE and tryptophan only at high doses. The interferons (IFN's) induce fever and HPA axis activation in man, but such effects are weak or absent in rodents. The reported effects of IFN's on brain catecholamines and serotonin have been very varied. However, interferon-γ, and to a lesser extent, interferon-α, have profound effects on the catabolism of tryptophan, effectively reducing its concentration in plasma, and may thus limit brain 5-HT synthesis.Administration of endotoxin (LPS) elicits responses similar to those of IL-1. Bacterial and viral infections induce HPA activation, and also increase brain NE and 5-HT metabolism and brain tryptophan. Typically, there is also behavioral depression. These effects are strikingly similar to those of IL-1, suggesting that IL-1 secretion, which accompanies many infections, may mediate these responses. Studies with IL-1 antagonists, support this possibility, although in most cases the antagonism is incomplete, suggesting the existence of multiple mechanisms. Because LPS is known to stimulate the secretion of IL-1, IL-6 and TNFα, it seems likely that these cytokines mediate at least some of the responses, but studies with antagonists indicate that there are multiple mechanisms. The neurochemical responses to cytokines are likely to underlie the endocrine and behavioral responses. The NE response to IL-1 appears to be instrumental in the HPA activation, but other mechanisms exist. Neither the noradrenergic nor the serotonergic systems appear to be involved in the major behavioral responses. The significance of the serotonin response is unknown.
Collapse
Affiliation(s)
- Adrian J Dunn
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center, 1501 Kings Highway, P.O. Box 33932, Shreveport, LA 71130-3932, USA
| |
Collapse
|