1
|
Thirion A, Loots DT, Williams ME, Solomons R, Mason S. 1H-NMR metabolomics investigation of CSF from children with HIV reveals altered neuroenergetics due to persistent immune activation. Front Neurosci 2024; 18:1270041. [PMID: 38745940 PMCID: PMC11091326 DOI: 10.3389/fnins.2024.1270041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Background HIV can invade the central nervous system (CNS) early during infection, invading perivascular macrophages and microglia, which, in turn, release viral particles and immune mediators that dysregulate all brain cell types. Consequently, children living with HIV often present with neurodevelopmental delays. Methods In this study, we used proton nuclear magnetic resonance (1H-NMR) spectroscopy to analyze the neurometabolic profile of HIV infection using cerebrospinal fluid samples obtained from 17 HIV+ and 50 HIV- South African children. Results Nine metabolites, including glucose, lactate, glutamine, 1,2-propanediol, acetone, 3-hydroxybutyrate, acetoacetate, 2-hydroxybutyrate, and myo-inositol, showed significant differences when comparing children infected with HIV and those uninfected. These metabolites may be associated with activation of the innate immune response and disruption of neuroenergetics pathways. Conclusion These results elucidate the neurometabolic state of children infected with HIV, including upregulation of glycolysis, dysregulation of ketone body metabolism, and elevated reactive oxygen species production. Furthermore, we hypothesize that neuroinflammation alters astrocyte-neuron communication, lowering neuronal activity in children infected with HIV, which may contribute to the neurodevelopmental delay often observed in this population.
Collapse
Affiliation(s)
- Anicia Thirion
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Du Toit Loots
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Monray E. Williams
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Regan Solomons
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Shayne Mason
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
2
|
Naidoo KK, Highton AJ, Baiyegunhi OO, Bhengu SP, Dong KL, Bunders MJ, Altfeld M, Ndung’u T. Early Initiation of Antiretroviral Therapy Preserves the Metabolic Function of CD4+ T Cells in Subtype C Human Immunodeficiency Virus 1 Infection. J Infect Dis 2024; 229:753-762. [PMID: 37804102 PMCID: PMC10938216 DOI: 10.1093/infdis/jiad432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/19/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Immune dysfunction often persists in people living with human immunodeficiency virus (HIV) who are on antiretroviral therapy (ART), clinically manifesting as HIV-1-associated comorbid conditions. Early ART initiation may reduce incidence of HIV-1-associated immune dysfunction and comorbid conditions. Immunometabolism is a critical determinant of functional immunity. We investigated the effect of HIV-1 infection and timing of ART initiation on CD4+ T cell metabolism and function. METHODS Longitudinal blood samples from people living with HIV who initiated ART during hyperacute HIV-1 infection (HHI; before peak viremia) or chronic HIV-1 infection (CHI) were assessed for the metabolic and immune functions of CD4+ T cells. Metabolite uptake and mitochondrial mass were measured using fluorescent analogues and MitoTracker Green accumulation, respectively, and were correlated with CD4+ T cell effector functions. RESULTS Initiation of ART during HHI prevented dysregulation of glucose uptake by CD4+ T cells, but glucose uptake was reduced before and after ART initiation in CHI. Glucose uptake positively correlated with interleukin-2 and tumor necrosis factor-α production by CD4+ T cells. CHI was associated with elevated mitochondrial mass in effector memory CD4+ T cells that persisted after ART and correlated with PD-1 expression. CONCLUSIONS ART initiation in HHI largely prevented metabolic impairment of CD4+ T cells. ART initiation in CHI was associated with persistently dysregulated immunometabolism of CD4+ T cells, which was associated with impaired cellular functions and exhaustion.
Collapse
Affiliation(s)
- Kewreshini K Naidoo
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Andrew J Highton
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | | | - Sindiswa P Bhengu
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Krista L Dong
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Madeleine J Bunders
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Altfeld
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
- German Center for Infection Disease (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Thumbi Ndung’u
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Africa Health Research Institute, Durban, South Africa
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
- Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
3
|
Thirion A, Loots DT, Williams ME, Solomons R, Mason S. An exploratory investigation of the CSF metabolic profile of HIV in a South African paediatric cohort using GCxGC-TOF/MS. Metabolomics 2024; 20:33. [PMID: 38427142 PMCID: PMC10907482 DOI: 10.1007/s11306-024-02098-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024]
Abstract
INTRODUCTION Because cerebrospinal fluid (CSF) samples are difficult to obtain for paediatric HIV, few studies have attempted to profile neurometabolic dysregulation. AIM AND OBJECTIVE The aim of this exploratory study was to profile the neurometabolic state of CSF from a South African paediatric cohort using GCxGC-TOF/MS. The study included 54 paediatric cases (< 12 years), 42 HIV-negative controls and 12 HIV-positive individuals. RESULTS The results revealed distinct metabolic alterations in the HIV-infected cohort. In the PLS-DA model, 18 metabolites significantly discriminated between HIV-infected and control groups. In addition, fold-change analysis, Mann-Whitney U tests, and effect size measurements verified these findings. Notably, lactose, myo-inositol, and glycerol, although not significant by p-value alone, demonstrated practical significance based on the effect size. CONCLUSIONS This study provided valuable insights on the impact of HIV on metabolic pathways, including damage to the gut and blood-brain barrier, disruption of bioenergetics processes, gliosis, and a potential marker for antiretroviral therapy. Nevertheless, the study recognized certain constraints, notably a limited sample size and the absence of a validation cohort. Despite these limitations, the rarity of the study's focus on paediatric HIV research underscores the significance and unique contributions of its findings.
Collapse
Affiliation(s)
- Anicia Thirion
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, North West, South Africa
| | - Du Toit Loots
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, North West, South Africa
| | - Monray E Williams
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, North West, South Africa
| | - Regan Solomons
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505, South Africa
| | - Shayne Mason
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, North West, South Africa.
| |
Collapse
|
4
|
Akiso M, Ameka M, Naidoo K, Langat R, Kombo J, Sikuku D, Ndung’u T, Altfeld M, Anzala O, Mureithi M. Metabolic and mitochondrial dysregulation in CD4+ T cells from HIV-positive women on combination anti-retroviral therapy. PLoS One 2023; 18:e0286436. [PMID: 37816026 PMCID: PMC10564234 DOI: 10.1371/journal.pone.0286436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/20/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND For optimal functionality, immune cells require a robust and adaptable metabolic program that is fueled by dynamic mitochondrial activity. In this study, we investigate the metabolic alterations occurring in immune cells during HIV infection and antiretroviral therapy by analyzing the uptake of metabolic substrates and mitochondrial phenotypes. By delineating changes in immune cell metabolic programming during HIV, we may identify novel potential therapeutic targets to improve anti-viral immune responses. METHODS After consent and voluntary participation was confirmed, whole blood was drawn from HIV uninfected women and women with chronic HIV infection on long-term combination antiretroviral therapy (HIV/cART). Peripheral blood mononuclear cells-derived immune cells were directly incubated with different fluorescently tagged metabolites and markers of mitochondrial activity: FITC-2-NBDG (2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl) amino]-2-deoxy-D-glucose), FITC-BODIPY (4,4-Difluoro-5,7-Dimethyl-4-Bora-3a,4a-Diaza-s-Indacene-3-Hexadecanoic Acid), FITC-MitoTracker Green and APC-MitoTracker Deep Red. The uptake of glucose and fats and the mitochondrial mass and potential were measured using flow cytometry. All values are reported quantitatively as geometric means of fluorescence intensity. RESULTS During chronic HIV infection, cellular uptake of glucose increases in HIV+ dendritic cells in particular. CD4+ T cells had the lowest uptake of glucose and fats compared to all other cells regardless of HIV status, while CD8+ T cells took up more fatty acids. Interestingly, despite the lower utilization of glucose and fats in CD4+ T cells, mitochondrial mass increased in HIV+ CD4+ T cells compared to HIV negative CD4+ T-cells. HIV+ CD4+ T cells also had the highest mitochondrial potential. CONCLUSIONS Significant disparities in the utilization of substrates by leukocytes during chronic HIV/cART exist. Innate immune cells increased utilization of sugars and fats while adaptive immune cells displayed lower glucose and fat utilization despite having a higher mitochondrial activity. Our findings suggest that cART treated HIV-infected CD4+ T cells be dysfunctional or may prefer alternative fuel sources not included in these studies. This underscores the importance of understanding the metabolic effects of HIV treatment on immune function.
Collapse
Affiliation(s)
- Matrona Akiso
- Department of Medical Microbiology & Immunology, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| | - Magdalene Ameka
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| | - Kewreshini Naidoo
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Robert Langat
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
- Division of Surgical Outcomes and Precision Medicine Research, Department of Surgery, University of Minnesota Twin Cities, United States of America
| | - Janet Kombo
- Department of Medical Microbiology & Immunology, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| | - Delories Sikuku
- Department of Medical Microbiology & Immunology, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
| | - Thumbi Ndung’u
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Marcus Altfeld
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Virus Immunology Department, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Omu Anzala
- Department of Medical Microbiology & Immunology, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| | - Marianne Mureithi
- Department of Medical Microbiology & Immunology, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| |
Collapse
|
5
|
Cartwright SL, Schmied J, Karrow N, Mallard BA. Impact of heat stress on dairy cattle and selection strategies for thermotolerance: a review. Front Vet Sci 2023; 10:1198697. [PMID: 37408833 PMCID: PMC10319441 DOI: 10.3389/fvets.2023.1198697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/02/2023] [Indexed: 07/07/2023] Open
Abstract
Climate change is a problem that causes many environmental issues that impact the productivity of livestock species. One of the major issues associated with climate change is an increase of the frequency of hot days and heat waves, which increases the risk of heat stress for livestock species. Dairy cattle have been identified as being susceptible to heat stress due to their high metabolic heat load. Studies have shown heat stress impacts several biological processes that can result in large economic consequences. When heat stress occurs, dairy cattle employ several physiological and cellular mechanisms in order to dissipate heat and protect cells from damage. These mechanisms require an increase and diversion in energy toward protection and away from other biological processes. Therefore, in turn heat stress in dairy cattle can lead numerous issues including reductions in milk production and reproduction as well as increased risk for disease and mortality. This indicates a need to select dairy cattle that would be thermotolerant. Various selection strategies to confer thermotolerance have been discussed in the literature, including selecting for reduced milk production, crossbreeding with thermotolerant breeds, selecting based on physiological traits and most recently selecting for enhanced immune response. This review discusses the various issues associated with heat stress in dairy cattle and the pros and cons to the various selection strategies that have been proposed to select for thermotolerance in dairy cattle.
Collapse
Affiliation(s)
- Shannon L. Cartwright
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Julie Schmied
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Niel Karrow
- Centre of Genetics of Improvement of Livestock, Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Bonnie A. Mallard
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
- Centre of Genetics of Improvement of Livestock, Animal Biosciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
6
|
Ray A, Bonorden MJL, Pandit R, Nkhata KJ, Bishayee A. Infections and immunity: associations with obesity and related metabolic disorders. J Pathol Transl Med 2023; 57:28-42. [PMID: 36647284 PMCID: PMC9846011 DOI: 10.4132/jptm.2022.11.14] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/14/2022] [Indexed: 01/18/2023] Open
Abstract
About one-fourth of the global population is either overweight or obese, both of which increase the risk of insulin resistance, cardiovascular diseases, and infections. In obesity, both immune cells and adipocytes produce an excess of pro-inflammatory cytokines that may play a significant role in disease progression. In the recent coronavirus disease 2019 (COVID-19) pandemic, important pathological characteristics such as involvement of the renin-angiotensin-aldosterone system, endothelial injury, and pro-inflammatory cytokine release have been shown to be connected with obesity and associated sequelae such as insulin resistance/type 2 diabetes and hypertension. This pathological connection may explain the severity of COVID-19 in patients with metabolic disorders. Many studies have also reported an association between type 2 diabetes and persistent viral infections. Similarly, diabetes favors the growth of various microorganisms including protozoal pathogens as well as opportunistic bacteria and fungi. Furthermore, diabetes is a risk factor for a number of prion-like diseases. There is also an interesting relationship between helminths and type 2 diabetes; helminthiasis may reduce the pro-inflammatory state, but is also associated with type 2 diabetes or even neoplastic processes. Several studies have also documented altered circulating levels of neutrophils, lymphocytes, and monocytes in obesity, which likely modifies vaccine effectiveness. Timely monitoring of inflammatory markers (e.g., C-reactive protein) and energy homeostasis markers (e.g., leptin) could be helpful in preventing many obesity-related diseases.
Collapse
Affiliation(s)
- Amitabha Ray
- College of Medical Science, Alderson Broaddus University, Philippi, WV, USA,Corresponding Author: Amitabha Ray, MD, PhD, College of Medical Science, Alderson Broaddus University, 101 College Hill Drive, Philippi, WV 26416, USA Tel: +1-304-457-6587, Fax: +1-304-457-6308, E-mail:
| | | | - Rajashree Pandit
- Division of Medical & Behavioral Health, Pueblo Community College, Pueblo, CO, USA
| | | | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, USA
| |
Collapse
|
7
|
Abstract
Metabolic adaptation to viral infections critically determines the course and manifestations of disease. At the systemic level, a significant feature of viral infection and inflammation that ensues is the metabolic shift from anabolic towards catabolic metabolism. Systemic metabolic sequelae such as insulin resistance and dyslipidaemia represent long-term health consequences of many infections such as human immunodeficiency virus, hepatitis C virus and severe acute respiratory syndrome coronavirus 2. The long-held presumption that peripheral and tissue-specific 'immune responses' are the chief line of defence and thus regulate viral control is incomplete. This Review focuses on the emerging paradigm shift proposing that metabolic engagements and metabolic reconfiguration of immune and non-immune cells following virus recognition modulate the natural course of viral infections. Early metabolic footprints are likely to influence longer-term disease manifestations of infection. A greater appreciation and understanding of how local biochemical adjustments in the periphery and tissues influence immunity will ultimately lead to interventions that curtail disease progression and identify new and improved prognostic biomarkers.
Collapse
Affiliation(s)
- Clovis S Palmer
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA.
| |
Collapse
|
8
|
Dimitriou ID, Meiri D, Jitkova Y, Elford AR, Koritzinsky M, Schimmer AD, Ohashi PS, Sonenberg N, Rottapel R. Translational Control by 4E-BP1/2 Suppressor Proteins Regulates Mitochondrial Biosynthesis and Function during CD8 + T Cell Proliferation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2702-2712. [PMID: 35667842 DOI: 10.4049/jimmunol.2101090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/03/2022] [Indexed: 06/15/2023]
Abstract
CD8+ T cell proliferation and differentiation into effector and memory states are high-energy processes associated with changes in cellular metabolism. CD28-mediated costimulation of T cells activates the PI3K/AKT/mammalian target of rapamycin signaling pathway and induces eukaryotic translation initiation factor 4E-dependent translation through the derepression by 4E-BP1 and 4E-BP2. In this study, we demonstrate that 4E-BP1/2 proteins are required for optimum proliferation of mouse CD8+ T cells and the development of an antiviral effector function. We show that translation of genes encoding mitochondrial biogenesis is impaired in T cells derived from 4E-BP1/2-deficient mice. Our findings demonstrate an unanticipated role for 4E-BPs in regulating a metabolic program that is required for cell growth and biosynthesis during the early stages of CD8+ T cell expansion.
Collapse
Affiliation(s)
- Ioannis D Dimitriou
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - David Meiri
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yulia Jitkova
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Alisha R Elford
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Marianne Koritzinsky
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Pamela S Ohashi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Robert Rottapel
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada;
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada; and
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Gálvez I, Navarro MC, Martín-Cordero L, Otero E, Hinchado MD, Ortega E. The Influence of Obesity and Weight Loss on the Bioregulation of Innate/Inflammatory Responses: Macrophages and Immunometabolism. Nutrients 2022; 14:nu14030612. [PMID: 35276970 PMCID: PMC8840693 DOI: 10.3390/nu14030612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity is characterized by low-grade inflammation and more susceptibility to infection, particularly viral infections, as clearly demonstrated in COVID-19. In this context, immunometabolism and metabolic flexibility of macrophages play an important role. Since inflammation is an inherent part of the innate response, strategies for decreasing the inflammatory response must avoid immunocompromise the innate defenses against pathogen challenges. The concept “bioregulation of inflammatory/innate responses” was coined in the context of the effects of exercise on these responses, implying a reduction in excessive inflammatory response, together with the preservation or stimulation of the innate response, with good transitions between pro- and anti-inflammatory macrophages adapted to each individual’s inflammatory set-point in inflammatory diseases, particularly in obesity. The question now is whether these responses can be obtained in the context of weight loss by dietary interventions (low-fat diet or abandonment of the high-fat diet) in the absence of exercise, which can be especially relevant for obese individuals with difficulties exercising such as those suffering from persistent COVID-19. Results from recent studies are controversial and do not point to a clear anti-inflammatory effect of these dietary interventions, particularly in the adipose tissue. Further research focusing on the innate response is also necessary.
Collapse
Affiliation(s)
- Isabel Gálvez
- Immunophyisiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (I.G.); (M.C.N.); (L.M.-C.); (E.O.); (M.D.H.)
- Immunophysiology Research Group, Nursing Department, Faculty of Medicine and Health Sciences, University of Extremadura, 06071 Badajoz, Spain
| | - María Carmen Navarro
- Immunophyisiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (I.G.); (M.C.N.); (L.M.-C.); (E.O.); (M.D.H.)
- Immunophysiology Research Group, Physiology Department, Faculty of Sciences, University of Extremadura, 06071 Badajoz, Spain
| | - Leticia Martín-Cordero
- Immunophyisiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (I.G.); (M.C.N.); (L.M.-C.); (E.O.); (M.D.H.)
- Immunophysiology Research Group, Nursing Department, University Center of Plasencia, University of Extremadura, 10600 Plasencia, Spain
| | - Eduardo Otero
- Immunophyisiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (I.G.); (M.C.N.); (L.M.-C.); (E.O.); (M.D.H.)
- Immunophysiology Research Group, Physiology Department, Faculty of Sciences, University of Extremadura, 06071 Badajoz, Spain
| | - María Dolores Hinchado
- Immunophyisiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (I.G.); (M.C.N.); (L.M.-C.); (E.O.); (M.D.H.)
- Immunophysiology Research Group, Physiology Department, Faculty of Sciences, University of Extremadura, 06071 Badajoz, Spain
| | - Eduardo Ortega
- Immunophyisiology Research Group, Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 06071 Badajoz, Spain; (I.G.); (M.C.N.); (L.M.-C.); (E.O.); (M.D.H.)
- Immunophysiology Research Group, Physiology Department, Faculty of Sciences, University of Extremadura, 06071 Badajoz, Spain
- Correspondence: ; Tel.: +34-924-289-300
| |
Collapse
|
10
|
Non-invasive plasma glycomic and metabolic biomarkers of post-treatment control of HIV. Nat Commun 2021; 12:3922. [PMID: 34188039 PMCID: PMC8241829 DOI: 10.1038/s41467-021-24077-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 05/31/2021] [Indexed: 12/15/2022] Open
Abstract
Non-invasive biomarkers that predict HIV remission after antiretroviral therapy (ART) interruption are urgently needed. Such biomarkers can improve the safety of analytic treatment interruption (ATI) and provide mechanistic insights into the host pathways involved in post-ART HIV control. Here we report plasma glycomic and metabolic signatures of time-to-viral-rebound and probability-of-viral-remission using samples from two independent cohorts. These samples include a large number of post-treatment controllers, a rare population demonstrating sustained virologic suppression after ART-cessation. These signatures remain significant after adjusting for key demographic and clinical confounders. We also report mechanistic links between some of these biomarkers and HIV latency reactivation and/or myeloid inflammation in vitro. Finally, machine learning algorithms, based on selected sets of these biomarkers, predict time-to-viral-rebound with 74% capacity and probability-of-viral-remission with 97.5% capacity. In summary, we report non-invasive plasma biomarkers, with potential functional significance, that predict both the duration and probability of HIV remission after treatment interruption.
Collapse
|
11
|
Singh Y, Trautwein C, Fendel R, Krickeberg N, Berezhnoy G, Bissinger R, Ossowski S, Salker MS, Casadei N, Riess O. SARS-CoV-2 infection paralyzes cytotoxic and metabolic functions of the immune cells. Heliyon 2021; 7:e07147. [PMID: 34075347 PMCID: PMC8159709 DOI: 10.1016/j.heliyon.2021.e07147] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 03/10/2021] [Accepted: 05/24/2021] [Indexed: 12/29/2022] Open
Abstract
The SARS-CoV-2 virus is the causative agent of the global COVID-19 infectious disease outbreak, which can lead to acute respiratory distress syndrome (ARDS). However, it is still unclear how the virus interferes with immune cell and metabolic functions in the human body. In this study, we investigated the immune response in acute or convalescent COVID-19 patients. We characterized the peripheral blood mononuclear cells (PBMCs) using flow cytometry and found that CD8+ T cells were significantly subsided in moderate COVID-19 and convalescent patients. Furthermore, characterization of CD8+ T cells suggested that convalescent patients have significantly diminished expression of both perforin and granzyme A. Using 1H-NMR spectroscopy, we characterized the metabolic status of their autologous PBMCs. We found that fructose, lactate and taurine levels were elevated in infected (mild and moderate) patients compared with control and convalescent patients. Glucose, glutamate, formate and acetate levels were attenuated in COVID-19 (mild and moderate) patients. In summary, our report suggests that SARS-CoV-2 infection leads to disrupted CD8+ T cytotoxic functions and changes the overall metabolic functions of immune cells.
Collapse
Affiliation(s)
- Yogesh Singh
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstrasse 7, 72076, Tübingen, Germany
- NGS Competence Center Tübingen (NCCT), University of Tübingen, Calwerstrasse 7, 72076 Tübingen, Germany
- Research Institute of Women’s Health, University of Tübingen, Calwerstrasse 7/6, 72076, Tübingen, Germany
| | - Christoph Trautwein
- Werner Siemens Imaging Center, University of Tübingen, Röntgenweg 13, 72076, Tübingen, Germany
| | - Rolf Fendel
- Institute of Tropical Medicine, University Hospital of Tübingen, Wilhelmstrasse 27, 72076, Tübingen, Germany
| | - Naomi Krickeberg
- Institute of Tropical Medicine, University Hospital of Tübingen, Wilhelmstrasse 27, 72076, Tübingen, Germany
| | - Georgy Berezhnoy
- Werner Siemens Imaging Center, University of Tübingen, Röntgenweg 13, 72076, Tübingen, Germany
| | - Rosi Bissinger
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital of Tübingen, Germany
| | - Stephan Ossowski
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstrasse 7, 72076, Tübingen, Germany
- NGS Competence Center Tübingen (NCCT), University of Tübingen, Calwerstrasse 7, 72076 Tübingen, Germany
| | - Madhuri S. Salker
- Research Institute of Women’s Health, University of Tübingen, Calwerstrasse 7/6, 72076, Tübingen, Germany
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstrasse 7, 72076, Tübingen, Germany
- NGS Competence Center Tübingen (NCCT), University of Tübingen, Calwerstrasse 7, 72076 Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstrasse 7, 72076, Tübingen, Germany
- NGS Competence Center Tübingen (NCCT), University of Tübingen, Calwerstrasse 7, 72076 Tübingen, Germany
| | | |
Collapse
|
12
|
Dahou S, Smahi MCE, Nouari W, Dahmani Z, Benmansour S, Ysmail-Dahlouk L, Miliani M, Yebdri F, Fakir N, Laoufi MY, Chaib-Draa M, Tourabi A, Aribi M. L-Threoascorbic acid treatment promotes S. aureus-infected primary human endothelial cells survival and function, as well as intracellular bacterial killing, and immunomodulates the release of IL-1β and soluble ICAM-1. Int Immunopharmacol 2021; 95:107476. [PMID: 33676147 DOI: 10.1016/j.intimp.2021.107476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Vitamin C (ascorbic acid, AscH2) has been shown to enhance immunity. Here, we studied its immunomodulatory effect on human endothelial cells (ECs) during S. aureus infection. MATERIALS AND METHODS The ex vivo effects of AscH2 were performed on primary human umbilical vein endothelial cells (HUVECs) infected or not with S. aureus. RESULTS AscH2 treatment induced a marked downregulation of nitric oxide (NO) production and a moderate upregulation of arginase activity in S. aureus-infected HUVECs (respectively, p < 0.05 and p > 0.05). Although the upregulated release levels of soluble intercellular adhesion molecular 1 (sICAM-1/sCD54) and sE-selectin (sCD62E) molecules were not significantly different between treated and untreated S. aureus-infected HUVECs, AscH2 treatment induced reversing effect on sICAM-1 release when comparing to uninfected control HUVECs. Moreover, AscH2 treatment appears to have a significant effect on preventing HUVEC necrosis induced by S. aureus infection (p < 0.05). Furthermore, AscH2 treatment induced a significant upregulation of cell protective redox biomarker in S. aureus-infected, as shown by superoxide dismutase (SOD) activity (p < 0.05), but not by catalase activity (p > 0.05). Additionally, S. aureus infection markedly downregulated total bound calcium ions (bCa2+) levels as compared to control HUVECs, whereas, AscH2 treatment induced a slight upregulation of bCa2+ levels in infected HUVECs as compared to infected and untreated HUVECs (p > 0.05). On the other hand, AscH2 treatment downregulated increased total cellular cholesterol content (tccCHOL) levels in HUVECs induced by S. aureus infection (p < 0.05). In addition, AscH2 treatment markedly reversed S. aureus effect on upregulation of intracellular glucose (iGLU) levels within infected HUVECs (p < 0.05). Moreover, AscH2 treatment significantly downregulated S. aureus growth (p < 0.05), and significantly upregulated bacterial internalization and intracellular killing by HUVECs (p < 0.05), as well as their cell cycle activation (p < 0.01). Finally, AscH2 treatment has a slight effect on the production of interleukin 6 (IL-6), but induced a marked downregulation of that of IL-1β in S. aureus-infected HUVECs (respectively, p > 0.05, and p < 0.05). CONCLUSIONS Our outcomes demonstrated that, during S. aureus infection, AscH2 treatment promotes human ECs survival and function, as well as prevents inflammatory response exacerbation, while inducing bactericidal activity.
Collapse
Affiliation(s)
- Sara Dahou
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Mohammed Chems-Eddine Smahi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria; Neonatal Department of Specialized Maternal and Child Hospital of Tlemcen, 13000, Tlemcen, Algeria
| | - Wafa Nouari
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Zoheir Dahmani
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Souheila Benmansour
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria; Neonatal Department of Specialized Maternal and Child Hospital of Tlemcen, 13000, Tlemcen, Algeria
| | - Lamia Ysmail-Dahlouk
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Maroua Miliani
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Fadela Yebdri
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Nassima Fakir
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Mohammed Yassine Laoufi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria; Neonatal Department of Specialized Maternal and Child Hospital of Tlemcen, 13000, Tlemcen, Algeria
| | - Mouad Chaib-Draa
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Amina Tourabi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, 13000 Tlemcen, Algeria.
| |
Collapse
|
13
|
Qi C, Wang Y, Li P, Zhao J. Gamma Delta T Cells and Their Pathogenic Role in Psoriasis. Front Immunol 2021; 12:627139. [PMID: 33732249 PMCID: PMC7959710 DOI: 10.3389/fimmu.2021.627139] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 01/15/2021] [Indexed: 12/14/2022] Open
Abstract
γδT cells are an unconventional population of T lymphocytes that play an indispensable role in host defense, immune surveillance, and homeostasis of the immune system. They display unique developmental, distributional, and functional patterns and rapidly respond to various insults and contribute to diverse diseases. Although γδT cells make up only a small portion of the total T cell pool, emerging evidence suggest that aberrantly activated γδT cells may play a role in the pathogenesis of psoriasis. Dermal γδT cells are the major IL-17-producing cells in the skin that respond to IL-23 stimulation. Furthermore, γδT cells exhibit memory-cell-like characteristics that mediate repeated episodes of psoriatic inflammation. This review discusses the differentiation, development, distribution, and biological function of γδT cells and the mechanisms by which they contribute to psoriasis. Potential therapeutic approaches targeting these cells in psoriasis have also been detailed.
Collapse
Affiliation(s)
- Cong Qi
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Yazhuo Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Ruisch JE, Kloft M, Fazzi GE, Melenhorst J, Magee DR, Grabsch HI. Large negative lymph nodes - a surrogate for immune activation in rectal cancer patients? Pathol Res Pract 2020; 216:153106. [PMID: 32825969 DOI: 10.1016/j.prp.2020.153106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/01/2020] [Accepted: 07/07/2020] [Indexed: 11/24/2022]
Abstract
AIM The size of regional, tumor draining lymph nodes without metastasis (LNneg) found in rectal cancer resection specimens varies and seems to be related to patient survival. Yet, the histopathological features influencing LNneg size in rectal cancer have not been studied in detail. Our pilot study focused on investigating the relationship between lymph node (LN) size and LNneg microarchitecture in rectal cancer (RC) resection specimens. METHOD In this retrospective cohort study, resection specimens from 146 RC patients, treated with either surgery alone (n = 29) or neoadjuvant therapy followed by resection (n = 117), were included in the study. Histology of LNnegs was reviewed to establish number of lymphoid follicles and presence of intranodal fat. Longest long axis and area of each LN were measured digitally. RESULTS 1830 LNnegs were measured. The microarchitecture was analyzed in a subset of 680 LNnegs. 153 (22.5 %) LNnegs contained intranodal fat. After neoadjuvant treatment, presence of intranodal fat was related to smaller LNneg area (median (range) area of LNneg without intranodal fat: 4.51 mm2 (0.15-46.89 mm2), with intranodal fat: 3.46 mm2 (0.12-27.22 mm2), p = 0.048). A higher number of lymphoid follicles was related to a larger LNneg area in both patient groups (p < 0.001). CONCLUSION Our pilot data suggest that in rectal cancer the presence of large regional LNnegs may reflect increased immune activation due to tumor related antigens. Further studies are warranted to investigate whether histologically visible microarchitectural features of LNnegs such as lymphoid follicles translate to particular features in radiological images and hence could potentially help to identify LNneg with more certainty at the time of pre-treatment disease staging.
Collapse
Affiliation(s)
- J E Ruisch
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - M Kloft
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - G E Fazzi
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - J Melenhorst
- Department of Surgery, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - D R Magee
- School of Computing, University of Leeds, Leeds, United Kingdom; HeteroGenius Limited, Leeds, United Kingdom
| | - H I Grabsch
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands; Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
15
|
Fol M, Włodarczyk M, Druszczyńska M. Host Epigenetics in Intracellular Pathogen Infections. Int J Mol Sci 2020; 21:ijms21134573. [PMID: 32605029 PMCID: PMC7369821 DOI: 10.3390/ijms21134573] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 12/18/2022] Open
Abstract
Some intracellular pathogens are able to avoid the defense mechanisms contributing to host epigenetic modifications. These changes trigger alterations tothe chromatin structure and on the transcriptional level of genes involved in the pathogenesis of many bacterial diseases. In this way, pathogens manipulate the host cell for their own survival. The better understanding of epigenetic consequences in bacterial infection may open the door for designing new vaccine approaches and therapeutic implications. This article characterizes selected intracellular bacterial pathogens, including Mycobacterium spp., Listeria spp., Chlamydia spp., Mycoplasma spp., Rickettsia spp., Legionella spp. and Yersinia spp., which can modulate and reprogram of defense genes in host innate immune cells.
Collapse
Affiliation(s)
- Marek Fol
- Correspondence: ; Tel.: +48-42-635-44-72
| | | | | |
Collapse
|
16
|
Deguit CDT, Hough M, Hoh R, Krone M, Pilcher CD, Martin JN, Deeks SG, McCune JM, Hunt PW, Rutishauser RL. Some Aspects of CD8+ T-Cell Exhaustion Are Associated With Altered T-Cell Mitochondrial Features and ROS Content in HIV Infection. J Acquir Immune Defic Syndr 2019; 82:211-219. [PMID: 31513075 PMCID: PMC6746248 DOI: 10.1097/qai.0000000000002121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Reversing or preventing T-cell exhaustion is an important treatment goal in the context of HIV disease; however, the mechanisms that regulate HIV-specific CD8 T-cell exhaustion are incompletely understood. Since mitochondrial mass (MM), mitochondrial membrane potential (MMP), and cellular reactive oxygen species (ROS) content are altered in exhausted CD8 T cells in other settings, we hypothesized that similar lesions may arise in HIV infection. METHODS We sampled cryopreserved peripheral blood mononuclear cells from HIV-uninfected (n = 10) and HIV-infected participants with varying levels and mechanisms of viral control: viremic (VL > 2000 copies/mL; n = 8) or aviremic (VL < 40 copies/mL) due to antiretroviral therapy (n = 11) or natural control (n = 9). We characterized the MM, MMP, and ROS content of bulk CD8 T cells and MHC class I tetramer+ HIV-specific CD8 T cells by flow cytometry. RESULTS We observed higher MM, MMP, and ROS content across bulk effector-memory CD8 T-cell subsets in HIV-infected compared with HIV-uninfected participants. Among HIV-specific CD8 T cells, these features did not vary by the extent or mechanism of viral control but were significantly altered in cells displaying characteristics associated with exhaustion (eg, high PD-1 expression, low CD127 expression, and impaired proliferative capacity). CONCLUSIONS While we did not find that control of HIV replication in vivo correlates with the CD8 T-cell MM, MMP, or ROS content, we did find that some features of CD8 T-cell exhaustion are associated with alterations in mitochondrial state. Our findings support further studies to probe the relationship between mitochondrial dynamics and CD8 T-cell functionality in HIV infection.
Collapse
Affiliation(s)
- Christian Deo T. Deguit
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
- Current Address: Department of Biochemistry and Molecular Biology, University of the Philippines, Manila, Philippines
| | - Michelle Hough
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
- Current Address: Department of Medicine, University of Southern California, Los Angeles, CA, U.S.A
| | - Rebecca Hoh
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
| | - Melissa Krone
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, U.S.A
| | - Christopher D. Pilcher
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, U.S.A
| | - Steven G. Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
| | - Joseph M. McCune
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
- Current Address: Bill & Melinda Gates Foundation, Seattle, WA, U.S.A
| | - Peter W. Hunt
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
| | - Rachel L. Rutishauser
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
| |
Collapse
|
17
|
Lee MKS, Al-Sharea A, Shihata WA, Bertuzzo Veiga C, Cooney OD, Fleetwood AJ, Flynn MC, Claeson E, Palmer CS, Lancaster GI, Henstridge DC, Hamilton JA, Murphy AJ. Glycolysis Is Required for LPS-Induced Activation and Adhesion of Human CD14 +CD16 - Monocytes. Front Immunol 2019; 10:2054. [PMID: 31555280 PMCID: PMC6742687 DOI: 10.3389/fimmu.2019.02054] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/14/2019] [Indexed: 01/09/2023] Open
Abstract
Monocytes in humans consist of 3 subsets; CD14+CD16- (classical), CD14+CD16+ (intermediate) and CD14dimCD16+ (non-classical), which exhibit distinct and heterogeneous responses to activation. During acute inflammation CD14+CD16- monocytes are significantly elevated and migrate to the sites of injury via the adhesion cascade. The field of immunometabolism has begun to elucidate the importance of the engagement of specific metabolic pathways in immune cell function. Yet, little is known about monocyte metabolism and the role of metabolism in mediating monocyte activation and adherence to vessels. Accordingly, we aimed to determine whether manipulating the metabolism of CD14+CD16- monocytes alters their ability to become activated and adhere. We discovered that LPS stimulation increased the rate of glycolysis in human CD14+CD16- monocytes. Inhibition of glycolysis with 2-deoxy-D-glucose blunted LPS-induced activation and adhesion of monocytes. Mechanistically, we found that increased glycolysis was regulated by mTOR-induced glucose transporter (GLUT)-1. Furthermore, enhanced glycolysis increased accumulation of reactive oxygen species (ROS) and activation of p38 MAPK, which lead to activation and adhesion of monocytes. These findings reveal that glycolytic metabolism is critical for the activation of CD14+CD16- monocytes and contributes to our understanding of the interplay between metabolic substrate preference and immune cell function.
Collapse
Affiliation(s)
- Man K. S. Lee
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Diabetes, Monash University, Melbourne, VIC, Australia
| | - Annas Al-Sharea
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Diabetes, Monash University, Melbourne, VIC, Australia
| | - Waled A. Shihata
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Camilla Bertuzzo Veiga
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Olivia D. Cooney
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Andrew J. Fleetwood
- Department of Medicine, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Australian Institute of Musculoskeletal Science, University of Melbourne and Western Health, St. Albans, VIC, Australia
| | - Michelle C. Flynn
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Ellen Claeson
- Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Clovis S. Palmer
- Department of Infectious Disease, Burnet Institute, Melbourne, VIC, Australia
| | - Graeme I. Lancaster
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Darren C. Henstridge
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - John A. Hamilton
- Department of Medicine, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Australian Institute of Musculoskeletal Science, University of Melbourne and Western Health, St. Albans, VIC, Australia
| | - Andrew J. Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Alzahrani J, Hussain T, Simar D, Palchaudhuri R, Abdel-Mohsen M, Crowe SM, Mbogo GW, Palmer CS. Inflammatory and immunometabolic consequences of gut dysfunction in HIV: Parallels with IBD and implications for reservoir persistence and non-AIDS comorbidities. EBioMedicine 2019; 46:522-531. [PMID: 31327693 PMCID: PMC6710907 DOI: 10.1016/j.ebiom.2019.07.027] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/07/2019] [Accepted: 07/09/2019] [Indexed: 12/15/2022] Open
Abstract
The gastrointestinal mucosa is critical for maintaining the integrity and functions of the gut. Disruption of this barrier is a hallmark and a risk factor for many intestinal and chronic inflammatory diseases. Inflammatory bowel disease (IBD) and HIV infection are characterized by microbial translocation and systemic inflammation. Despite the clinical overlaps between HIV and IBD, significant differences exist such as the severity of gut damage and mechanisms of immune cell homeostasis. Studies have supported the role of metabolic activation of immune cells in promoting chronic inflammation in HIV and IBD. This inflammatory response persists in HIV+ persons even after long-term virologic suppression by antiretroviral therapy (ART). Here, we review gut dysfunction and microbiota changes during HIV infection and IBD, and discuss how this may induce metabolic reprogramming of monocytes, macrophages and T cells to impact disease outcomes. Drawing from parallels with IBD, we highlight how factors such as lipopolysaccharides, residual viral replication, and extracellular vesicles activate biochemical pathways that regulate immunometabolic processes essential for HIV persistence and non-AIDS metabolic comorbidities. This review highlights new mechanisms and support for the use of immunometabolic-based therapeutics towards HIV remission/cure, and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Jehad Alzahrani
- Life Sciences, Burnet Institute, Melbourne, Australia; School of Medical Science, RMIT University, Melbourne, Australia
| | - Tabinda Hussain
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - David Simar
- School of Medical Sciences, UNSW, Sydney, Australia
| | | | | | - Suzanne M Crowe
- Life Sciences, Burnet Institute, Melbourne, Australia; Department of Infectious Diseases, Monash University, Melbourne, Australia
| | | | - Clovis S Palmer
- Life Sciences, Burnet Institute, Melbourne, Australia; School of Medical Science, RMIT University, Melbourne, Australia; Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
19
|
O'Sullivan D. The metabolic spectrum of memory T cells. Immunol Cell Biol 2019; 97:636-646. [DOI: 10.1111/imcb.12274] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/13/2019] [Accepted: 05/22/2019] [Indexed: 12/27/2022]
Affiliation(s)
- David O'Sullivan
- Department of Immunometabolism Max Planck Institute of Immunobiology and Epigenetics Freiburg Germany
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to examine the evidence describing adipose tissue as a reservoir for HIV-1 and how this often expansive anatomic compartment contributes to HIV persistence. RECENT FINDINGS Memory CD4 T cells and macrophages, the major host cells for HIV, accumulate in adipose tissue during HIV/SIV infection of humans and rhesus macaques. Whereas HIV and SIV proviral DNA is detectable in CD4 T cells of multiple fat depots in virtually all infected humans and monkeys examined, viral RNA is less frequently detected, and infected macrophages may be less prevalent in adipose tissue. However, based on viral outgrowth assays, adipose-resident CD4 T cells are latently infected with virus that is replication-competent and infectious. Additionally, adipocytes interact with CD4 T cells and macrophages to promote immune cell activation and inflammation which may be supportive for HIV persistence. Antiviral effector cells, such as CD8 T cells and NK/NKT cells, are abundant in adipose tissue during HIV/SIV infection and typically exceed CD4 T cells, whereas B cells are largely absent from adipose tissue of humans and monkeys. Additionally, CD8 T cells in adipose tissue of HIV patients are activated and have a late differentiated phenotype, with unique TCR clonotypes of less diversity relative to blood CD8 T cells. With respect to the distribution of antiretroviral drugs in adipose tissue, data is limited, but there may be class-specific penetration of fat depots. The trafficking of infected immune cells within adipose tissues is a common event during HIV/SIV infection of humans and monkeys, but the virus may be mostly transcriptionally dormant. Viral replication may occur less in adipose tissue compared to other major reservoirs, such as lymphoid tissue, but replication competence and infectiousness of adipose latent virus are comparable to other tissues. Due to the ubiquitous nature of adipose tissue, inflammatory interactions among adipocytes and CD4 T cells and macrophages, and selective distribution of antiretroviral drugs, the sequestration of infected immune cells within fat depots likely represents a major challenge for cure efforts.
Collapse
Affiliation(s)
- Jacob Couturier
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St., MSB 2.112, Houston, TX, 77030, USA
| | - Dorothy E Lewis
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St., MSB 2.112, Houston, TX, 77030, USA.
| |
Collapse
|
21
|
Rao M, Dodoo E, Zumla A, Maeurer M. Immunometabolism and Pulmonary Infections: Implications for Protective Immune Responses and Host-Directed Therapies. Front Microbiol 2019; 10:962. [PMID: 31134013 PMCID: PMC6514247 DOI: 10.3389/fmicb.2019.00962] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022] Open
Abstract
The biology and clinical efficacy of immune cells from patients with infectious diseases or cancer are associated with metabolic programming. Host immune- and stromal-cell genetic and epigenetic signatures in response to the invading pathogen shape disease pathophysiology and disease outcomes. Directly linked to the immunometabolic axis is the role of the host microbiome, which is also discussed here in the context of productive immune responses to lung infections. We also present host-directed therapies (HDT) as a clinically viable strategy to refocus dysregulated immunometabolism in patients with infectious diseases, which requires validation in early phase clinical trials as adjuncts to conventional antimicrobial therapy. These efforts are expected to be continuously supported by newly generated basic and translational research data to gain a better understanding of disease pathology while devising new molecularly defined platforms and therapeutic options to improve the treatment of patients with pulmonary infections, particularly in relation to multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Ernest Dodoo
- Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt, Germany
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London, NIHR Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.,Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt, Germany
| |
Collapse
|
22
|
Persistent Replication of HIV, Hepatitis C Virus (HCV), and HBV Results in Distinct Gene Expression Profiles by Human NK Cells. J Virol 2019; 93:JVI.00575-18. [PMID: 30185599 DOI: 10.1128/jvi.00575-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 08/20/2018] [Indexed: 01/14/2023] Open
Abstract
Natural killer (NK) cells during chronic viral infection have been well studied in the past. We performed an unbiased next-generation RNA-sequencing approach to identify commonalities or differences of the effect of HIV, HCV, and HBV viremia on NK cell transcriptomes. Using cell sorting, we obtained CD3- CD56+ NK cells from blood of 6 HIV-, 8 HCV-, and 32 HBV-infected patients without treatment. After library preparation and sequencing, we used an in-house analytic pipeline to compare expression levels with matched healthy controls. In NK cells from HIV-, HCV-, and HBV-infected patients, transcriptome analysis identified 272, 53, and 56 differentially expressed genes, respectively (fold change, >1.5; q-value, 0.2). Interferon-stimulated genes were induced in NK cells from HIV/HCV patients, but not during HBV infection. HIV viremia downregulated ribosome assembly genes in NK cells. In HBV-infected patients, viral load and alanine aminotransferase (ALT) variation had little effect on genes related to NK effector function. In conclusion, we compare, for the first time, NK cell transcripts of viremic HIV, HCV, and HBV patients. We clearly demonstrate distinctive NK cell gene signatures in three different populations, suggestive for a different degree of functional alterations of the NK cell compartment compared to healthy individuals.IMPORTANCE Three viruses exist that can result in persistently high viral loads in immunocompetent humans: human immunodeficiency virus (HIV), hepatitis C virus, and hepatitis B virus. In the last decades, by using flow cytometry and in vitro assays on NK cells from patients with these types of infections, several impairments have been established, particularly during and possibly contributing to HIV viremia. However, the background of NK cell impairments in viremic patients is not well understood. In this study, we describe the NK cell transcriptomes of patients with high viral loads of different etiologies. We clearly demonstrate distinctive NK cell gene signatures with regard to interferon-stimulated gene induction and the expression of genes coding for activation markers or proteins involved in cytotoxic action, as well immunological genes. This study provides important details necessary to uncover the origin of functional and phenotypical differences between viremic patients and healthy subjects and provides many leads that can be confirmed using future in vitro manipulation experiments.
Collapse
|
23
|
Ong EZ, Gan ES, de Alwis R, Wijaya L, Ong XM, Zhang M, Wong AW, Cheung YB, Zellweger RM, Ooi EE, Low JG. Genomic signature of early T-cell response is associated with lower antibody titer threshold for sterilizing immunity. Antiviral Res 2019; 166:35-41. [PMID: 30940521 DOI: 10.1016/j.antiviral.2019.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/21/2019] [Accepted: 03/27/2019] [Indexed: 02/02/2023]
Abstract
Vaccination is an effective approach to reduce disease burden. High vaccination coverage blocks pathogen transmission to ensure herd immunity. However, the concept of herd immunity assumes that vaccinated individuals cannot be infected and mediate silent pathogen transmission. While the correlates of vaccine-mediated protection against disease have been examined, the correlates of sterilizing immunity that prevents infection have not been systematically defined. Here, we used full genome expression profiling to explore the molecular correlates of serological response and non-response to measles, mumps and rubella (MMR) vaccination as surrogates of infection and sterilizing immunity, respectively. We observed that the antibody titers needed to sterilize infection with the vaccine strains were higher than current WHO disease protection thresholds. In subjects with baseline antibodies below such sterilizing immunity thresholds, serological non-response to MMR vaccination was associated with gene expression profile indicative of early T-cell activation and signalling. Specifically, genes that regulate T-cell function and response were induced at day 1 post-vaccination in non-responders but not in responders. These findings suggest that rapid T-cell response prevented MMR vaccine infection to limit antigenic presentation and hence serological response. Collectively, our findings suggest an important role for T-cells in engendering sterilizing immunity.
Collapse
Affiliation(s)
- Eugenia Z Ong
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Esther S Gan
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Ruklanthi de Alwis
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Limin Wijaya
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Xin Mei Ong
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | | | - Abigail Wl Wong
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Yin Bun Cheung
- Center for Quantitative Medicine, Duke-NUS Medical School, Singapore; Department for International Health, University of Tampere, 33100, Finland
| | - Raphaël M Zellweger
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Eng Eong Ooi
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART), Singapore
| | - Jenny G Low
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Department of Infectious Diseases, Singapore General Hospital, Singapore.
| |
Collapse
|
24
|
Curto P, Santa C, Allen P, Manadas B, Simões I, Martinez JJ. A Pathogen and a Non-pathogen Spotted Fever Group Rickettsia Trigger Differential Proteome Signatures in Macrophages. Front Cell Infect Microbiol 2019; 9:43. [PMID: 30895174 PMCID: PMC6414445 DOI: 10.3389/fcimb.2019.00043] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/11/2019] [Indexed: 12/13/2022] Open
Abstract
We have previously reported that Rickettsia conorii and Rickettsia montanensis have distinct intracellular fates within THP-1 macrophages, suggesting that the ability to proliferate within macrophages may be a distinguishable factor between pathogenic and non-pathogenic Spotted fever group (SFG) members. To start unraveling the molecular mechanisms underlying the capacity (or not) of SFG Rickettsia to establish their replicative niche in macrophages, we have herein used quantitative proteomics by SWATH-MS to profile the alterations resulted by the challenge of THP-1 macrophages with R. conorii and R. montanensis. We show that the pathogenic, R. conorii, and the non-pathogenic, R. montanensis, member of SFG Rickettsia trigger differential proteomic signatures in macrophage-like cells upon infection. R. conorii specifically induced the accumulation of several enzymes of the tricarboxylic acid cycle, oxidative phosphorylation, fatty acid β-oxidation, and glutaminolysis, as well as of several inner and outer membrane mitochondrial transporters. These results suggest a profound metabolic rewriting of macrophages by R. conorii toward a metabolic signature of an M2-like, anti-inflammatory activation program. Moreover, several subunits forming the proteasome and immunoproteasome are found in lower abundance upon infection with both rickettsial species, which may help bacteria to escape immune surveillance. R. conorii-infection specifically induced the accumulation of several host proteins implicated in protein processing and quality control in ER, suggesting that this pathogenic Rickettsia may be able to increase the ER protein folding capacity. This work reveals novel aspects of macrophage-Rickettsia interactions, expanding our knowledge of how pathogenic rickettsiae explore host cells to their advantage.
Collapse
Affiliation(s)
- Pedro Curto
- PhD Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Cátia Santa
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Paige Allen
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Isaura Simões
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Juan J. Martinez
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
25
|
Affiliation(s)
- Clair M. Gardiner
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin 2 Ireland
| |
Collapse
|
26
|
Nieman DC, Lila MA, Gillitt ND. Immunometabolism: A Multi-Omics Approach to Interpreting the Influence of Exercise and Diet on the Immune System. Annu Rev Food Sci Technol 2019; 10:341-363. [PMID: 30633566 DOI: 10.1146/annurev-food-032818-121316] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Immunometabolism is an evolving field of scientific endeavor that merges immunology and metabolism and has provided valuable context when evaluating the influence of dietary interventions on exercise-induced immune dysfunction. Metabolomics, lipidomics, and proteomics provide a system-wide view of the metabolic response to exercise by simultaneously measuring and identifying a large number of small-molecule metabolites, lipids, and proteins. Many of these are involved with immune function and regulation and are sensitive to dietary influences, especially acute carbohydrate ingestion from either sugar beverages or fruits such as bananas. Emerging evidence using large multi-omics data sets supports the combined intake of fruit sugars and phytochemicals by athletes during heavy exertion as an effective strategy to improve metabolic recovery, augment viral defense, and counter postexercise inflammation and immune dysfunction at the cell level. Multi-omics methodologies have given investigators new outcome targets to assess the efficacy of various dietary interventions for physiologically stressed athletes.
Collapse
Affiliation(s)
- David C Nieman
- Human Performance Laboratory, Department of Health and Exercise Science, Appalachian State University, North Carolina Research Campus, Kannapolis, North Carolina 28081, USA;
| | - Mary Ann Lila
- Plants for Human Health Institute, Department of Food, Bioprocessing & Nutrition Sciences, North Carolina State University, North Carolina Research Campus, Kannapolis, North Carolina 28081, USA
| | - Nicholas D Gillitt
- Dole Nutrition Research Laboratory, Kannapolis, North Carolina 28081, USA
| |
Collapse
|
27
|
Crowe SM, Kintzios S, Kaltsas G, Palmer CS. A Bioelectronic System to Measure the Glycolytic Metabolism of Activated CD4+ T Cells. BIOSENSORS-BASEL 2019; 9:bios9010010. [PMID: 30634392 PMCID: PMC6468583 DOI: 10.3390/bios9010010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/30/2018] [Accepted: 01/01/2019] [Indexed: 01/02/2023]
Abstract
The evaluation of glucose metabolic activity in immune cells is becoming an increasingly standard task in immunological research. In this study, we described a sensitive, inexpensive, and non-radioactive assay for the direct and rapid measurement of the metabolic activity of CD4+ T cells in culture. A portable, custom-built Cell Culture Metabolite Biosensor device was designed to measure the levels of acidification (a proxy for glycolysis) in cell-free CD4+ T cell culture media. In this assay, ex vivo activated CD4+ T cells were incubated in culture medium and mini electrodes were placed inside the cell free culture filtrates in 96-well plates. Using this technique, the inhibitors of glycolysis were shown to suppress acidification of the cell culture media, a response similar to that observed using a gold standard lactate assay kit. Our findings show that this innovative biosensor technology has potential for applications in metabolic research, where acquisition of sufficient cellular material for ex vivo analyses presents a substantial challenge.
Collapse
Affiliation(s)
- Suzanne M Crowe
- Life Sciences Discipline, Burnet Institute, Melbourne, VIC 3001, Australia.
- Department of Infectious Diseases, Monash University, Melbourne, VIC 3004, Australia.
- Infectious Diseases Department, The Alfred hospital, Melbourne, VIC 3004, Australia.
| | - Spyridon Kintzios
- Laboratory of Cell Technology, School of Food Science, Biotechnology and Development, Agricultural University of Athens, 11855 Athens, Greece.
| | - Grigoris Kaltsas
- Department of Electrical and Electronics Engineering, microSENSES lab, University of West Attika, 12244 Athens, Greece.
| | - Clovis S Palmer
- Life Sciences Discipline, Burnet Institute, Melbourne, VIC 3001, Australia.
- Department of Infectious Diseases, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
28
|
Nicoli F, Paul S, Appay V. Harnessing the Induction of CD8 + T-Cell Responses Through Metabolic Regulation by Pathogen-Recognition-Receptor Triggering in Antigen Presenting Cells. Front Immunol 2018; 9:2372. [PMID: 30410483 PMCID: PMC6209652 DOI: 10.3389/fimmu.2018.02372] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/24/2018] [Indexed: 01/03/2023] Open
Abstract
Cytotoxic CD8+ T-cells are key players of the immune responses against viruses. During the priming of a CD8+ T-cell response, the activation of a naïve T-cell by a professional antigen presenting cell (APC) involves the induction of various intracellular and metabolic pathways. The modulation of these pathways at the level of APCs or T-cells offers great potential to enhance the induction of robust effector cells and the generation of long-lived memory cells. On the one hand, signaling through pathogen recognition receptors (PRRs) expressed by APCs can greatly influence T-cell priming, and the potential of several PRR ligands as adjuvants are being studied. On the other hand, the engagement of several metabolic processes, at play in APCs and T-cells upon stimulation, implies that modulating cellular metabolism can impact on priming efficacy. Here, we review recent efforts to understand the interplay between PRR mediated signaling and metabolic pathway modulation in this context, through three examples: interplay between TLR4 and fatty acid metabolism, between TLR9 and IDO, and between STING and autophagy. These initial works highlight the potential for harnessing the induction of antiviral CD8+ T-cell responses using synergistic modulation of metabolic and PRR pathways.
Collapse
Affiliation(s)
- Francesco Nicoli
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Stéphane Paul
- GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Victor Appay
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses, Paris, France.,International Research Center of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
29
|
Lack of Sprouty 1 and 2 enhances survival of effector CD8 + T cells and yields more protective memory cells. Proc Natl Acad Sci U S A 2018; 115:E8939-E8947. [PMID: 30126987 DOI: 10.1073/pnas.1808320115] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Identifying novel pathways that promote robust function and longevity of cytotoxic T cells has promising potential for immunotherapeutic strategies to combat cancer and chronic infections. We show that sprouty 1 and 2 (Spry1/2) molecules regulate the survival and function of memory CD8+ T cells. Spry1/2 double-knockout (DKO) ovalbumin (OVA)-specific CD8+ T cells (OT-I cells) mounted more vigorous autoimmune diabetes than WT OT-I cells when transferred to mice expressing OVA in their pancreatic β-islets. To determine the consequence of Spry1/2 deletion on effector and memory CD8+ T cell development and function, we used systemic infection with lymphocytic choriomeningitis virus (LCMV) Armstrong. Spry1/2 DKO LCMV gp33-specific P14 CD8+ T cells survive contraction better than WT cells and generate significantly more polyfunctional memory T cells. The larger number of Spry1/2 DKO memory T cells displayed enhanced infiltration into infected tissue, demonstrating that absence of Spry1/2 can result in increased recall capacity. Upon adoptive transfer into naive hosts, Spry1/2 DKO memory T cells controlled Listeria monocytogenes infection better than WT cells. The enhanced formation of more functional Spry1/2 DKO memory T cells was associated with significantly reduced mTORC1 activity and glucose uptake. Reduced p-AKT, p-FoxO1/3a, and T-bet expression was also consistent with enhanced survival and memory accrual. Collectively, loss of Spry1/2 enhances the survival of effector CD8+ T cells and results in the formation of more protective memory cells. Deleting Spry1/2 in antigen-specific CD8+ T cells may have therapeutic potential for enhancing the survival and functionality of effector and memory CD8+ T cells in vivo.
Collapse
|
30
|
Pan Y, Kupper TS. Metabolic Reprogramming and Longevity of Tissue-Resident Memory T Cells. Front Immunol 2018; 9:1347. [PMID: 29967608 PMCID: PMC6016524 DOI: 10.3389/fimmu.2018.01347] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 05/31/2018] [Indexed: 12/31/2022] Open
Abstract
Tissue-resident memory T cells (TRM) persist in peripheral tissues for long periods of time in the absence of antigenic stimulation. Upon re-encounter with cognate antigen, TRM trigger an immediate immune response at the local tissue microenvironment and provide the first line of host defense. TRM have been reported to play significant roles in host antimicrobial infection, cancer immunotherapy, and pathogenesis of a number of human autoimmune diseases, such as psoriasis, vitiligo, and atopic dermatitis. TRM display a distinct gene transcriptome with unique gene expression profiles related to cellular metabolism that is different from naive T cells (TN), central memory T cells (TCM), and effector memory T cells (TEM). Skin CD8+ TRM upregulate expression of genes associated with lipid uptake and metabolism and utilize mitochondria fatty acid β-oxidation to support their long-term survival (longevity) and function. In this review, we will summarize the recent progresses in the metabolic programming of TRM and will also discuss the potential to target the unique metabolic pathways of TRM to treat TRM-mediated diseases.
Collapse
Affiliation(s)
- Youdong Pan
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Thomas S Kupper
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
31
|
Palmer CS, Palchaudhuri R, Albargy H, Abdel-Mohsen M, Crowe SM. Exploiting immune cell metabolic machinery for functional HIV cure and the prevention of inflammaging. F1000Res 2018; 7:125. [PMID: 29445452 PMCID: PMC5791007 DOI: 10.12688/f1000research.11881.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2018] [Indexed: 12/31/2022] Open
Abstract
An emerging paradigm in immunology suggests that metabolic reprogramming and immune cell activation and functions are intricately linked. Viral infections, such as HIV infection, as well as cancer force immune cells to undergo major metabolic challenges. Cells must divert energy resources in order to mount an effective immune response. However, the fact that immune cells adopt specific metabolic programs to provide host defense against intracellular pathogens and how this metabolic shift impacts immune cell functions and the natural course of diseases have only recently been appreciated. A clearer insight into how these processes are inter-related will affect our understanding of several fundamental aspects of HIV persistence. Even in patients with long-term use of anti-retroviral therapies, HIV infection persists and continues to cause chronic immune activation and inflammation, ongoing and cumulative damage to multiple organs systems, and a reduction in life expectancy. HIV-associated fundamental changes to the metabolic machinery of the immune system can promote a state of “inflammaging”, a chronic, low-grade inflammation with specific immune changes that characterize aging, and can also contribute to the persistence of HIV in its reservoirs. In this commentary, we will bring into focus evolving concepts on how HIV modulates the metabolic machinery of immune cells in order to persist in reservoirs and how metabolic reprogramming facilitates a chronic state of inflammation that underlies the development of age-related comorbidities. We will discuss how immunometabolism is facilitating the changing paradigms in HIV cure research and outline the novel therapeutic opportunities for preventing inflammaging and premature development of age-related conditions in HIV
+ individuals.
Collapse
Affiliation(s)
- Clovis S Palmer
- Centre for Biomedical Research, Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, Australia.,Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
| | - Riya Palchaudhuri
- Centre for Biomedical Research, Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, Australia.,Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
| | - Hassan Albargy
- Centre for Biomedical Research, Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, Australia.,Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
| | | | - Suzanne M Crowe
- Centre for Biomedical Research, Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, Australia.,Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
| |
Collapse
|