151
|
Watanabe S, Alexander M, Misharin AV, Budinger GRS. The role of macrophages in the resolution of inflammation. J Clin Invest 2019; 129:2619-2628. [PMID: 31107246 DOI: 10.1172/jci124615] [Citation(s) in RCA: 543] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Macrophages are tissue-resident or infiltrated immune cells critical for innate immunity, normal tissue development, homeostasis, and repair of damaged tissue. Macrophage function is a sum of their ontogeny, the local environment in which they reside, and the type of injuries or pathogen to which they are exposed. In this Review, we discuss the role of macrophages in the restoration of tissue function after injury, highlighting important questions about how they respond to and modify the local microenvironment to restore homeostasis.
Collapse
Affiliation(s)
- Satoshi Watanabe
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA.,Department of Respiratory Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Michael Alexander
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Alexander V Misharin
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - G R Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
152
|
Calender A, Lim CX, Weichhart T, Buisson A, Besnard V, Rollat-Farnier PA, Bardel C, Roy P, Cottin V, Devouassoux G, Finat A, Pinson S, Lebecque S, Nunes H, Israel-Biet D, Bentaher A, Valeyre D, Pacheco Y. Exome sequencing and pathogenicity-network analysis of five French families implicate mTOR signalling and autophagy in familial sarcoidosis. Eur Respir J 2019; 54:13993003.00430-2019. [PMID: 31023854 DOI: 10.1183/13993003.00430-2019] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 04/14/2019] [Indexed: 12/18/2022]
|
153
|
Wilson JL, Weichhart T. TORching a semaphore for alternative macrophage activation. Nat Immunol 2019; 19:512-514. [PMID: 29777214 DOI: 10.1038/s41590-018-0117-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jayne Louise Wilson
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Thomas Weichhart
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
154
|
|
155
|
Ieronymaki E, Theodorakis EM, Lyroni K, Vergadi E, Lagoudaki E, Al-Qahtani A, Aznaourova M, Neofotistou-Themeli E, Eliopoulos AG, Vaporidi K, Tsatsanis C. Insulin Resistance in Macrophages Alters Their Metabolism and Promotes an M2-Like Phenotype. THE JOURNAL OF IMMUNOLOGY 2019; 202:1786-1797. [PMID: 30718296 DOI: 10.4049/jimmunol.1800065] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 01/10/2019] [Indexed: 12/14/2022]
Abstract
Obesity and insulin resistance influences metabolic processes, but whether it affects macrophage metabolism is not known. In this study, we demonstrate that chronic exposure of macrophages to insulin either in culture or in vivo in diet-induced, glucose-intolerant mice rendered them resistant to insulin signals marked by failure to induce Akt2 phosphorylation. Similarly, macrophages lacking Akt2 or IGF1 receptor were also resistant to insulin signals. Insulin-resistant macrophages had increased basal mTORC1 activity, possessed an M2-like phenotype, and reduced LPS responses. Moreover, they exhibited increased glycolysis and increased expression of key glycolytic enzymes. Inhibition of mTORC1 reversed the M2-like phenotype and suppressed glycolysis in insulin-resistant macrophages. In the context of polymicrobial sepsis, mice harboring insulin-resistant macrophages exhibited reduced sepsis-induced lung injury. Thus, macrophages obtain resistance to insulin characterized by increased glycolysis and a unique M2-like phenotype, termed M-insulin resistant, which accounts for obesity-related changes in macrophage responses and a state of trained immunity.
Collapse
Affiliation(s)
- Eleftheria Ieronymaki
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, 71110 Crete, Greece
| | - Emmanouel M Theodorakis
- Laboratory of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Konstantina Lyroni
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, 71110 Crete, Greece
| | - Eleni Vergadi
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, 71110 Crete, Greece
| | - Eleni Lagoudaki
- Laboratory of Pathology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Ahmed Al-Qahtani
- Department of Infection and Immunity, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; and
| | - Marina Aznaourova
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Elpida Neofotistou-Themeli
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Aristides G Eliopoulos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, 71110 Crete, Greece.,Department of Basic Sciences, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Katerina Vaporidi
- Laboratory of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Christos Tsatsanis
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece; .,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, 71110 Crete, Greece
| |
Collapse
|
156
|
Cullivan S, De La Harpe Golden P, Doyle D, Doddakula KK, Burke L, Murphy DM. Coexistent sarcoidosis and lymphangioleiomyomatosis in a patient with cystic lung disease. Respirol Case Rep 2019; 7:e00389. [PMID: 30519468 PMCID: PMC6261928 DOI: 10.1002/rcr2.389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/25/2018] [Accepted: 10/31/2018] [Indexed: 11/12/2022] Open
Abstract
A 45-year-old lady presented acutely with pleuritic chest pain, haemoptysis, and dyspnoea. Her background was significant for a 1.4 cm renal angiomyolipoma, and she was an ex-smoker without any relevant family history. A computed tomography (CT) pulmonary angiogram was negative for a pulmonary embolism but demonstrated diffuse cystic change throughout both lungs. A bronchoscopy confirmed a normal endobronchial tree, and pulmonary function tests demonstrated moderate airways obstruction, with reversibility and a normal diffusion capacity for carbon monoxide (DLCO). A video-assisted thoracoscopic surgery (VATS) lung biopsy showed non-caseating granulomas, and serum angiotensin converting enzyme (ACE) was elevated consistent with a diagnosis of pulmonary sarcoidosis. Further sectioning indicated focal areas that stained positive for Human Melanoma Black 45 (HMB-45), confirming lymphangioleiomyomatosis (LAM). A diagnosis of cystic lung disease secondary to coexistent sarcoidosis and LAM was made.
Collapse
Affiliation(s)
- Sarah Cullivan
- Department of Respiratory MedicineCork University HospitalIreland
| | | | - Deirdre Doyle
- Department of RadiologyCork University HospitalIreland
| | | | - Louise Burke
- Department of PathologyCork University HospitalIreland
| | | |
Collapse
|
157
|
Nathan N, Sileo C, Calender A, Pacheco Y, Rosental PA, Cavalin C, Macchi O, Valeyre D, Clement A. Paediatric sarcoidosis. Paediatr Respir Rev 2019; 29:53-59. [PMID: 30917882 DOI: 10.1016/j.prrv.2018.05.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/09/2018] [Indexed: 12/15/2022]
Abstract
Paediatric sarcoidosis is an extremely rare disease characterized by a granulomatous inflammation. The estimated incidence is 0.6-1.02/100,000 children, but in the absence of international registers, the disease is probably under-reported. Its pathophysiologic basis is not clearly understood but the current hypothesis is a combination of a genetic predisposition and an environmental exposure that could be either organic or mineral. Contrary to adult forms of the disease, general symptoms are often at the forefront at diagnosis. In its most frequent form, paediatric sarcoidosis is a multi-organ disorder affecting preferentially the lungs, the lymphatic system and the liver, but all organs can be affected. This review aims to provide an overview of current knowledge on sarcoidosis in children, providing a summary of the data available from cohort studies on the presentation, the management and the evolution of the disease in this specific population.
Collapse
Affiliation(s)
- Nadia Nathan
- Service de Pneumologie Pédiatrique et Centre de référence des maladies respiratoires rares, AP-HP, Hôpital Trousseau, Unité INSERM UMR-S933, Sorbonne Université, Paris, France.
| | - Chiara Sileo
- Service de Radiologie Pédiatrique, AP-HP, Hôpital Trousseau, Paris, France.
| | - Alain Calender
- Service de Génétique, CHU Lyon, Equipe EA7426 (PI3) Inflammation et immunité de l'épithélium respiratoire, Lyon, France.
| | - Yves Pacheco
- Service de Génétique, CHU Lyon, Equipe EA7426 (PI3) Inflammation et immunité de l'épithélium respiratoire, Lyon, France.
| | - Paul-André Rosental
- SLICOSIS Groupe, European Research Council (ERC)/SILICOSIS project, Centre for European Studies and Comparative Politics, Sciences Po, Paris, France.
| | - Catherine Cavalin
- SLICOSIS Groupe, European Research Council (ERC)/SILICOSIS project, Centre for European Studies and Comparative Politics, Sciences Po, Paris, France.
| | - Odile Macchi
- SLICOSIS Groupe, European Research Council (ERC)/SILICOSIS project, Centre for European Studies and Comparative Politics, Sciences Po, Paris, France.
| | - Dominique Valeyre
- AP-HP Service de pneumologie, Hôpital Avicenne, EA2363, Université Paris 13, Bobigny, France.
| | - Annick Clement
- Service de Pneumologie Pédiatrique et Centre de référence des maladies respiratoires rares, AP-HP, Hôpital Trousseau, Unité INSERM UMR-S933, Sorbonne Université, Paris, France.
| | | | | |
Collapse
|
158
|
Sohrabi Y, Lagache SMM, Schnack L, Godfrey R, Kahles F, Bruemmer D, Waltenberger J, Findeisen HM. mTOR-Dependent Oxidative Stress Regulates oxLDL-Induced Trained Innate Immunity in Human Monocytes. Front Immunol 2019; 9:3155. [PMID: 30723479 PMCID: PMC6350618 DOI: 10.3389/fimmu.2018.03155] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/20/2018] [Indexed: 01/04/2023] Open
Abstract
Introduction: Cells of the innate immune system particularly monocytes and macrophages have been recognized as pivotal players both during the initial insult as well as the chronic phase of atherosclerosis. It has recently been shown that oxidized low-density lipoprotein (oxLDL) induces a long-term pro-inflammatory response in monocytes due to epigenetic and metabolic reprogramming, an emerging new concept called trained innate immunity. Changes in the cellular redox state are crucial events in the regulation of many physiologic functions in macrophages including transcription, differentiation and inflammatory response. Here we have analyzed the role of reactive oxygen species (ROS) in regulating this proinflammatory monocyte priming in response to oxLDL-treatment. Methods and Results: Human monocytes were isolated and incubated with oxLDL for 24 h. After 5 days of resting, oxLDL treated cells produced significantly more inflammatory cytokines upon restimulation with the TLR2-agonist Pam3cys. Furthermore, oxLDL incubation induced persistent mTOR activation, ROS formation, HIF1α accumulation and HIF1α target gene expression, while pharmacologic mTOR inhibition or siRNA mediated inhibition of the mTORC1 subunit Raptor prevented ROS formation and proinflammatory priming. mTOR dependent ROS formation was associated with increased expression of NAPDH oxidases and necessary for the emergence of the primed phenotype as antioxidant treatment blocked oxLDL priming. Inhibition of cytosolic ROS formation could also block mTOR activation and HIF1α accumulation suggesting a positive feedback loop between mTOR and cytosolic ROS. Although mitochondrial ROS scavenging did not block HIF1α-accumulation at an early time point (24 h), it was persistently reduced on day 6. Therefore, mitochondrial ROS formation appears to occur initially downstream of the mTOR-cytoROS-HIF1α feedback loop but seems to be a crucial factor that controls the long-term activation of the mTOR-HIF1α-axis. Conclusion: In summary, our data demonstrate that mTOR dependent ROS production controls the oxLDL-induced trained innate immunity phenotype in human monocyte derived macrophages. Pharmacologic modulation of these pathways might provide a potential approach to modulate inflammation, associated with aberrant monocyte activation, during atherosclerosis development.
Collapse
Affiliation(s)
- Yahya Sohrabi
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany
| | - Sina M M Lagache
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany
| | - Lucia Schnack
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany
| | - Rinesh Godfrey
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany.,Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, Netherlands
| | - Florian Kahles
- Department of Internal Medicine I-Cardiology, University Hospital Aachen, Aachen, Germany
| | - Dennis Bruemmer
- Department of Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute Division of Cardiology, University of Pittsburgh Medical Center (UMPC) and University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Johannes Waltenberger
- Medical Faculty, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, Münster, Germany
| | - Hannes M Findeisen
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany
| |
Collapse
|
159
|
Snyder JP, Amiel E. Regulation of Dendritic Cell Immune Function and Metabolism by Cellular Nutrient Sensor Mammalian Target of Rapamycin (mTOR). Front Immunol 2019; 9:3145. [PMID: 30692999 PMCID: PMC6339945 DOI: 10.3389/fimmu.2018.03145] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cell (DC) activation is characterized by an acute increase in glucose metabolic flux that is required to fuel the high anabolic rates associated with DC activation. Inhibition of glycolysis significantly attenuates most aspects of DC immune effector function including antigen presentation, inflammatory cytokine production, and T cell stimulatory capacity. The cellular nutrient sensor mammalian/mechanistic Target of Rapamycin (mTOR) is an important upstream regulator of glycolytic metabolism and plays a central role in coordinating DC metabolic changes and immune responses. Because mTOR signaling can be activated by a variety of immunological stimuli, including signaling through the Toll-like Receptor (TLR) family of receptors, mTOR is involved in orchestrating many aspects of the DC metabolic response to microbial stimuli. It has become increasingly clear that mTOR's role in promoting or attenuating inflammatory processes in DCs is highly context-dependent and varies according to specific cellular subsets and the immunological conditions being studied. This review will address key aspects of the complex role of mTOR in regulating DC metabolism and effector function.
Collapse
Affiliation(s)
- Julia P Snyder
- Predoctoral student of the Cellular, Molecular, and Biomedical (CMB) Sciences Graduate Program at the University of Vermont, Burlington, VT, United States
| | - Eyal Amiel
- Department of Biomedical and Health Sciences, College of Nursing and Health Sciences, University of Vermont, Burlington, VT, United States
| |
Collapse
|
160
|
Jung J, Zeng H, Horng T. Metabolism as a guiding force for immunity. Nat Cell Biol 2019; 21:85-93. [PMID: 30602764 DOI: 10.1038/s41556-018-0217-x] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022]
Abstract
Recent studies indicate that cellular metabolism plays a key role in supporting immune cell maintenance and development. Here, we review how metabolism guides immune cell activation and differentiation to distinct cellular states, and how differential regulation of metabolism allows for context-dependent support during activation and lineage commitment. We discuss emerging principles of metabolic support of immune cell function in physiology and disease, as well as their general relevance to the field of cell biology.
Collapse
Affiliation(s)
- Jonathan Jung
- Department of Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,School of Medicine, University of Glasgow, Glasgow, UK
| | - Hu Zeng
- Division of Rheumatology, Mayo Clinic, Rochester, MN, USA. .,Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| | - Tiffany Horng
- Department of Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA. .,ShanghaiTech University, Shanghai, China.
| |
Collapse
|
161
|
Damsky W, Thakral D, Emeagwali N, Galan A, King B. Tofacitinib Treatment and Molecular Analysis of Cutaneous Sarcoidosis. N Engl J Med 2018; 379:2540-2546. [PMID: 30586518 PMCID: PMC6351852 DOI: 10.1056/nejmoa1805958] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There is evidence that Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling plays a role in the pathogenesis of sarcoidosis. We treated a patient with cutaneous sarcoidosis with the JAK inhibitor tofacitinib; the patient had not previously had a response to medications and had not received systemic glucocorticoids. This treatment resulted in clinical and histologic remission of her skin disease. Sequencing of RNA and immunohistochemical examination of skin-lesion samples obtained from the patient before and during therapy and immunohistochemical testing of lesion samples obtained from other patients with cutaneous sarcoidosis support a role for JAK-STAT signaling in cutaneous sarcoidosis. (Funded by the Ranjini and Ajay Poddar Resource Fund for Dermatologic Diseases Research and others.).
Collapse
Affiliation(s)
- William Damsky
- From the Departments of Dermatology (W.D., D.T., A.G., B.K.), Immunobiology (W.D.), and Pathology (A.G.) and the Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine (N.E.), Yale School of Medicine, New Haven, CT
| | - Durga Thakral
- From the Departments of Dermatology (W.D., D.T., A.G., B.K.), Immunobiology (W.D.), and Pathology (A.G.) and the Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine (N.E.), Yale School of Medicine, New Haven, CT
| | - Nkiruka Emeagwali
- From the Departments of Dermatology (W.D., D.T., A.G., B.K.), Immunobiology (W.D.), and Pathology (A.G.) and the Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine (N.E.), Yale School of Medicine, New Haven, CT
| | - Anjela Galan
- From the Departments of Dermatology (W.D., D.T., A.G., B.K.), Immunobiology (W.D.), and Pathology (A.G.) and the Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine (N.E.), Yale School of Medicine, New Haven, CT
| | - Brett King
- From the Departments of Dermatology (W.D., D.T., A.G., B.K.), Immunobiology (W.D.), and Pathology (A.G.) and the Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine (N.E.), Yale School of Medicine, New Haven, CT
| |
Collapse
|
162
|
Paschoal VA, Belchior T, Amano MT, Burgos-Silva M, Peixoto AS, Magdalon J, Vieira TS, Andrade ML, Moreno MF, Chimin P, Câmara NO, Festuccia WT. Constitutive Activation of the Nutrient Sensor mTORC1 in Myeloid Cells Induced by Tsc1 Deletion Protects Mice from Diet-Induced Obesity. Mol Nutr Food Res 2018; 62:e1800283. [DOI: 10.1002/mnfr.201800283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 06/27/2018] [Indexed: 11/11/2022]
Affiliation(s)
- Vivian A. Paschoal
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo 05508000 Brazil
| | - Thiago Belchior
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo 05508000 Brazil
| | - Mariane T. Amano
- Department of Immunology, Institute of Biomedical Sciences; University of São Paulo; São Paulo 05508000 Brazil
| | - Marina Burgos-Silva
- Department of Immunology, Institute of Biomedical Sciences; University of São Paulo; São Paulo 05508000 Brazil
| | - Albert S. Peixoto
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo 05508000 Brazil
| | - Juliana Magdalon
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo 05508000 Brazil
- Israelita Albert Einstein Hospital; São Paulo 05652-900 Brazil
| | - Thayna S. Vieira
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo 05508000 Brazil
| | - Maynara L. Andrade
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo 05508000 Brazil
| | - Mayara F. Moreno
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo 05508000 Brazil
| | - Patricia Chimin
- Department of Physical Education; Physical Education and Sports Center; Londrina State University; Londrina 86051-990 Parana Brazil
| | - Niels O. Câmara
- Department of Immunology, Institute of Biomedical Sciences; University of São Paulo; São Paulo 05508000 Brazil
| | - William T. Festuccia
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo 05508000 Brazil
| |
Collapse
|
163
|
Cornejo CM, Haun P, English J, Rosenbach M. Immune checkpoint inhibitors and the development of granulomatous reactions. J Am Acad Dermatol 2018; 81:1165-1175. [PMID: 30092327 DOI: 10.1016/j.jaad.2018.07.051] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/21/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022]
Abstract
Immune checkpoint inhibitors (ICPIs) have emerged as a frontline treatment for a growing list of malignancies. Disruption of the negative regulatory immune checkpoints by ICPIs has been associated with many immune-related adverse events. Granulomatous reactions, such as sarcoidosis-like reactions, granulomatous panniculitis, granuloma annulare, and granulomatous dermatitis, are uncommon but increasingly recognized immune-related adverse events seen in patients treated with ICPIs. The frequency and significance of these eruptions, including whether they portend responsiveness to treatment, remain unclear. Additionally, understanding the role of immune checkpoint blockade in these reactions may provide mechanistic insight into the relevant signaling pathways involved in sarcoidosis and other granulomatous disorders.
Collapse
Affiliation(s)
- Christine M Cornejo
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Paul Haun
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joseph English
- Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Misha Rosenbach
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
164
|
Regulation of Metabolic Disease-Associated Inflammation by Nutrient Sensors. Mediators Inflamm 2018; 2018:8261432. [PMID: 30116154 PMCID: PMC6079375 DOI: 10.1155/2018/8261432] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/21/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022] Open
Abstract
Visceral obesity is frequently associated with the development of type 2 diabetes (T2D), a highly prevalent chronic disease that features insulin resistance and pancreatic β-cell dysfunction as important hallmarks. Recent evidence indicates that the chronic, low-grade inflammation commonly associated with visceral obesity plays a major role connecting the excessive visceral fat deposition with the development of insulin resistance and pancreatic β-cell dysfunction. Herein, we review the mechanisms by which nutrients modulate obesity-associated inflammation.
Collapse
|
165
|
Oh MH, Collins SL, Sun IH, Tam AJ, Patel CH, Arwood ML, Chan-Li Y, Powell JD, Horton MR. mTORC2 Signaling Selectively Regulates the Generation and Function of Tissue-Resident Peritoneal Macrophages. Cell Rep 2018; 20:2439-2454. [PMID: 28877476 DOI: 10.1016/j.celrep.2017.08.046] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 06/27/2017] [Accepted: 08/11/2017] [Indexed: 12/31/2022] Open
Abstract
Tissue-resident macrophages play critical roles in sentinel and homeostatic functions as well as in promoting inflammation and immunity. It has become clear that the generation of these cells is highly dependent upon tissue-specific cues derived from the microenvironment that, in turn, regulate unique differentiation programs. Recently, a role for GATA6 has emerged in the differentiation programming of resident peritoneal macrophages. We identify a critical role for mTOR in integrating cues from the tissue microenvironment in regulating differentiation and metabolic reprogramming. Specifically, inhibition of mTORC2 leads to enhanced GATA6 expression in a FOXO1 dependent fashion. Functionally, inhibition of mTORC2 promotes peritoneal resident macrophage generation in the resolution phase during zymosan-induced peritonitis. Also, mTORC2-deficient peritoneal resident macrophages displayed increased functionality and metabolic reprogramming. Notably, mTORC2 activation distinguishes tissue-resident macrophage proliferation and differentiation from that of M2 macrophages. Overall, our data implicate a selective role for mTORC2 in the differentiation of tissue-resident macrophages.
Collapse
Affiliation(s)
- Min-Hee Oh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Samuel L Collins
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Im-Hong Sun
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ada J Tam
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Chirag H Patel
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Matthew L Arwood
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yee Chan-Li
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jonathan D Powell
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Maureen R Horton
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
166
|
Sellares J, Strambu I, Crouser ED, Freudenberg MA, Gulati M, Hart S, Herzog E, Kolb M, Weichhart T, Drake WP, Spitzer G, Singh N, Culver DA. New advances in the development of sarcoidosis models: a synopsis of a symposium sponsored by the Foundation for Sarcoidosis Research. SARCOIDOSIS VASCULITIS AND DIFFUSE LUNG DISEASES 2018; 35:2-4. [PMID: 32476873 DOI: 10.36141/svdld.v35i1.7032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 02/08/2018] [Indexed: 01/14/2023]
Abstract
Sarcoidosis is a complex disease with variable phenotypes that will require a multisystem approach to understand pathophysiology. One of the most challenging problems in sarcoidosis research is the absence of valid and widely accepted experimental models that accurately simulate human disease. The Foundation of Sarcoidosis Research (FSR) has funded five projects for the development of novel experimental models for sarcoidosis, presented and discussed in a workshop organized during the European Respiratory Society Congress held in Milan from September 9th to 13th. The experimental, in vivo or in sillico models presented may be quite helpful for investigating specific pathogenic and therapeutic questions, addressing especially severe forms of sarcoidosis. (Sarcoidosis Vasc Diffuse Lung Dis 2018; 35: 2-4).
Collapse
Affiliation(s)
- Jacobo Sellares
- Programa Multidisciplinar de Enfermedades Pulmonares Intersticiales DIfusas. Servei de Pneumologia. Hospital Clínic. Barcelona, Spain.,CiBER Enfermedades Respiratorias, Spain.,Institut Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Irina Strambu
- Pulmonology Department, University of Medicine and Pharmacy "Carol Davila", Bucharest, Romania
| | - Elliot D Crouser
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, the Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Marina A Freudenberg
- Department of Pneumology, Centre for Medicine, Medical Centre - University of Freiburg, Freiburg, Germany
| | - Mridu Gulati
- Yale University School of Medicine, New Haven, Connecticut, USA
| | - Simon Hart
- Respiratory Research Group, Hull York Medical School, Castle Hill Hospital, Cottingham, United Kingdom
| | - Erika Herzog
- Yale University School of Medicine, New Haven, Connecticut, USA
| | - Martin Kolb
- Division of Respirology, McMaster University, Hamilton, Ontario, Canada
| | - Thomas Weichhart
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Wonder P Drake
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, USA
| | | | - Noopur Singh
- Foundation for Sarcoidosis Research, Chicago, IL, USA
| | - Daniel A Culver
- Department of Pulmonary Medicine and Department of Pathobiology, Respiratory Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
167
|
Díaz Á, Sagasti C, Casaravilla C. Granulomatous responses in larval taeniid infections. Parasite Immunol 2018. [DOI: 10.1111/pim.12523] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Á. Díaz
- Área/Cátedra de Inmunología; Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias); Universidad de la República; Montevideo Uruguay
| | - C. Sagasti
- Área/Cátedra de Inmunología; Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias); Universidad de la República; Montevideo Uruguay
| | - C. Casaravilla
- Área/Cátedra de Inmunología; Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias); Universidad de la República; Montevideo Uruguay
| |
Collapse
|
168
|
Abstract
The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR)-dependent pathway is one of the most integral pathways linked to cell metabolism, proliferation, differentiation, and survival. This pathway is dysregulated in a variety of diseases, including neoplasia, immune-mediated diseases, and fibroproliferative diseases such as pulmonary fibrosis. The mTOR kinase is frequently referred to as the master regulator of this pathway. Alterations in mTOR signaling are closely associated with dysregulation of autophagy, inflammation, and cell growth and survival, leading to the development of lung fibrosis. Inhibitors of mTOR have been widely studied in cancer therapy, as they may sensitize cancer cells to radiation therapy. Studies also suggest that mTOR inhibitors are promising modulators of fibroproliferative diseases such as idiopathic pulmonary fibrosis (IPF) and radiation-induced pulmonary fibrosis (RIPF). Therefore, mTOR represents an attractive and unique therapeutic target in pulmonary fibrosis. In this review, we discuss the pathological role of mTOR kinase in pulmonary fibrosis and examine how mTOR inhibitors may mitigate fibrotic progression.
Collapse
|
169
|
Calender A, Rollat Farnier PA, Buisson A, Pinson S, Bentaher A, Lebecque S, Corvol H, Abou Taam R, Houdouin V, Bardel C, Roy P, Devouassoux G, Cottin V, Seve P, Bernaudin JF, Lim CX, Weichhart T, Valeyre D, Pacheco Y, Clement A, Nathan N. Whole exome sequencing in three families segregating a pediatric case of sarcoidosis. BMC Med Genomics 2018; 11:23. [PMID: 29510755 PMCID: PMC5839022 DOI: 10.1186/s12920-018-0338-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 02/19/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sarcoidosis (OMIM 181000) is a multi-systemic granulomatous disorder of unknown origin. Despite multiple genome-wide association (GWAS) studies, no major pathogenic pathways have been identified to date. To find out relevant sarcoidosis predisposing genes, we searched for de novo and recessive mutations in 3 young probands with sarcoidosis and their healthy parents using a whole-exome sequencing (WES) methodology. METHODS From the SARCFAM project based on a national network collecting familial cases of sarcoidosis, we selected three families (trios) in which a child, despite healthy parents, develop the disease before age 15 yr. Each trio was genotyped by WES (Illumina HiSEQ 2500) and we selected the gene variants segregating as 1) new mutations only occurring in affected children and 2) as recessive traits transmitted from each parents. The identified coding variants were compared between the three families. Allelic frequencies and in silico functional results were analyzed using ExAC, SIFT and Polyphenv2 databases. The clinical and genetic studies were registered by the ClinicalTrials.gov - Protocol Registration and Results System (PRS) ( https://clinicaltrials.gov ) receipt under the reference NCT02829853 and has been approved by the ethical committee (CPP LYON SUD EST - 2 - REF IRB 00009118 - September 21, 2016). RESULTS We identified 37 genes sharing coding variants occurring either as recessive mutations in at least 2 trios or de novo mutations in one of the three affected children. The genes were classified according to their potential roles in immunity related pathways: 9 to autophagy and intracellular trafficking, 6 to G-proteins regulation, 4 to T-cell activation, 4 to cell cycle and immune synapse, 2 to innate immunity. Ten of the 37 genes were studied in a bibliographic way to evaluate the functional link with sarcoidosis. CONCLUSIONS Whole exome analysis of case-parent trios is useful for the identification of genes predisposing to complex genetic diseases as sarcoidosis. Our data identified 37 genes that could be putatively linked to a pediatric form of sarcoidosis in three trios. Our in-depth focus on 10 of these 37 genes may suggest that the formation of the characteristic lesion in sarcoidosis, granuloma, results from combined deficits in autophagy and intracellular trafficking (ex: Sec16A, AP5B1 and RREB1), G-proteins regulation (ex: OBSCN, CTTND2 and DNAH11), T-cell activation (ex: IDO2, IGSF3), mitosis and/or immune synapse (ex: SPICE1 and KNL1). The significance of these findings needs to be confirmed by functional tests on selected gene variants.
Collapse
Affiliation(s)
- Alain Calender
- Genetics Department, Hospices Civils de LYON (HCL), University Hospital, East Pathology Center, LYON, B-A3, 59 Bld Pinel, 69677 BRON Cedex, France
- Inflammation & Immunity of the Respiratory Epithelium - EA7426 (PI3) – South Medical University Hospital – Lyon 1 Claude Bernard University, 165 Chemin du Grand Revoyet, 69310 Pierre-Bénite, France
| | | | - Adrien Buisson
- Genetics Department, Hospices Civils de LYON (HCL), University Hospital, East Pathology Center, LYON, B-A3, 59 Bld Pinel, 69677 BRON Cedex, France
| | - Stéphane Pinson
- Genetics Department, Hospices Civils de LYON (HCL), University Hospital, East Pathology Center, LYON, B-A3, 59 Bld Pinel, 69677 BRON Cedex, France
| | - Abderrazzaq Bentaher
- Inflammation & Immunity of the Respiratory Epithelium - EA7426 (PI3) – South Medical University Hospital – Lyon 1 Claude Bernard University, 165 Chemin du Grand Revoyet, 69310 Pierre-Bénite, France
| | - Serge Lebecque
- Cancer Research Center, INSERM U-1052, CNRS 5286, 69008 Lyon, France
| | - Harriet Corvol
- Pediatric pulmonology and Reference Center for rare lung diseases RespiRare, Hôpital Trousseau, AP-HP, INSERM UMR-S938, Sorbonne University, Paris, France
| | - Rola Abou Taam
- Pediatric pulmonology and Reference Center for rare lung diseases RespiRare, Hôpital Necker, Paris, France
| | - Véronique Houdouin
- Pediatric pulmonology and Reference Center for rare lung diseases RespiRare, Hôpital Robert Debré, INSERM U-1142, University Paris Diderot VII, Paris, France
| | - Claire Bardel
- Department of biostatistics, University Hospital, Hospices Civils de LYON (HCL), Lyon, France
| | - Pascal Roy
- Department of biostatistics, University Hospital, Hospices Civils de LYON (HCL), Lyon, France
| | - Gilles Devouassoux
- Department of Pulmonology, University Hospital, Hôpital Croix Rousse, Lyon, France
| | - Vincent Cottin
- Department of Pulmonology, University Hospital, Hôpital Louis Pradel, Lyon, France
| | - Pascal Seve
- Department of Internal medicine, University Hospital, Hôpital Croix Rousse, Lyon, France
| | | | - Clarice X. Lim
- Medical University of Vienna, Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Währinger Straße 10, 1090 Vienna, Austria
| | - Thomas Weichhart
- Medical University of Vienna, Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Währinger Straße 10, 1090 Vienna, Austria
| | - Dominique Valeyre
- EA2363, University Paris 13, COMUE Sorbonne-Paris-Cité, 74 rue Marcel Cachin, 93009 Bobigny, France
- Assistance Publique Hôpitaux de Paris, Department of Pulmonology, Avicenne University Hospital, 93009 Bobigny, France
| | - Yves Pacheco
- Inflammation & Immunity of the Respiratory Epithelium - EA7426 (PI3) – South Medical University Hospital – Lyon 1 Claude Bernard University, 165 Chemin du Grand Revoyet, 69310 Pierre-Bénite, France
| | - Annick Clement
- AP-HP Pediatric pulmonology and Reference Center for rare lung diseases RespiRare, Hôpital Trousseau, INSERM UMR-S933, Sorbonne University, Paris, France
| | - Nadia Nathan
- AP-HP Pediatric pulmonology and Reference Center for rare lung diseases RespiRare, Hôpital Trousseau, INSERM UMR-S933, Sorbonne University, Paris, France
| | - in the frame of GSF (Groupe Sarcoïdose France)
- Genetics Department, Hospices Civils de LYON (HCL), University Hospital, East Pathology Center, LYON, B-A3, 59 Bld Pinel, 69677 BRON Cedex, France
- Department of biostatistics, University Hospital, Hospices Civils de LYON (HCL), Lyon, France
- Inflammation & Immunity of the Respiratory Epithelium - EA7426 (PI3) – South Medical University Hospital – Lyon 1 Claude Bernard University, 165 Chemin du Grand Revoyet, 69310 Pierre-Bénite, France
- Cancer Research Center, INSERM U-1052, CNRS 5286, 69008 Lyon, France
- Pediatric pulmonology and Reference Center for rare lung diseases RespiRare, Hôpital Trousseau, AP-HP, INSERM UMR-S938, Sorbonne University, Paris, France
- Pediatric pulmonology and Reference Center for rare lung diseases RespiRare, Hôpital Necker, Paris, France
- Pediatric pulmonology and Reference Center for rare lung diseases RespiRare, Hôpital Robert Debré, INSERM U-1142, University Paris Diderot VII, Paris, France
- Department of Pulmonology, University Hospital, Hôpital Croix Rousse, Lyon, France
- Department of Pulmonology, University Hospital, Hôpital Louis Pradel, Lyon, France
- Department of Internal medicine, University Hospital, Hôpital Croix Rousse, Lyon, France
- Histology and Tumor Biology, ER2 UPMC, Hôpital Tenon, Paris, France
- Medical University of Vienna, Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Währinger Straße 10, 1090 Vienna, Austria
- EA2363, University Paris 13, COMUE Sorbonne-Paris-Cité, 74 rue Marcel Cachin, 93009 Bobigny, France
- Assistance Publique Hôpitaux de Paris, Department of Pulmonology, Avicenne University Hospital, 93009 Bobigny, France
- AP-HP Pediatric pulmonology and Reference Center for rare lung diseases RespiRare, Hôpital Trousseau, INSERM UMR-S933, Sorbonne University, Paris, France
| |
Collapse
|
170
|
Pagán AJ, Ramakrishnan L. TORmented macrophages spontaneously form granulomas. Nat Immunol 2018; 18:252-253. [PMID: 28198828 DOI: 10.1038/ni.3689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Antonio J Pagán
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Lalita Ramakrishnan
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
171
|
Abstract
Granulomas are organized aggregates of macrophages, often with characteristic morphological changes, and other immune cells. These evolutionarily ancient structures form in response to persistent particulate stimuli-infectious or noninfectious-that individual macrophages cannot eradicate. Granulomas evolved as protective responses to destroy or sequester particles but are frequently pathological in the context of foreign bodies, infections, and inflammatory diseases. We summarize recent findings that suggest that the granulomatous response unfolds in a stepwise program characterized by a series of macrophage activations and transformations that in turn recruit additional cells and produce structural changes. We explore why different granulomas vary and the reasons that granulomas are protective and pathogenic. Understanding the mechanisms and role of granuloma formation may uncover new therapies for the multitude of granulomatous diseases that constitute serious medical problems while enhancing the protective function of granulomas in infections.
Collapse
Affiliation(s)
- Antonio J Pagán
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; , .,MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Lalita Ramakrishnan
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; , .,MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| |
Collapse
|
172
|
Th17-lineage cells in pulmonary sarcoidosis and Löfgren's syndrome: Friend or foe? J Autoimmun 2018; 87:82-96. [DOI: 10.1016/j.jaut.2017.12.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 12/12/2017] [Indexed: 01/17/2023]
|
173
|
Rhinoscleroma pathogenesis: The type K3 capsule of Klebsiella rhinoscleromatis is a virulence factor not involved in Mikulicz cells formation. PLoS Negl Trop Dis 2018; 12:e0006201. [PMID: 29381692 PMCID: PMC5806929 DOI: 10.1371/journal.pntd.0006201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 02/09/2018] [Accepted: 12/31/2017] [Indexed: 12/16/2022] Open
Abstract
Rhinoscleroma is a human specific chronic granulomatous infection of the nose and upper airways caused by the Gram-negative bacterium Klebsiella pneumoniae subsp. rhinoscleromatis. Although considered a rare disease, it is endemic in low-income countries where hygienic conditions are poor. A hallmark of this pathology is the appearance of atypical foamy monocytes called Mikulicz cells. However, the pathogenesis of rhinoscleroma remains poorly investigated. Capsule polysaccharide (CPS) is a prominent virulence factor in bacteria. All K. rhinoscleromatis strains are of K3 serotype, suggesting that CPS can be an important driver of rhinoscleroma disease. In this study, we describe the creation of the first mutant of K. rhinoscleromatis, inactivated in its capsule export machinery. Using a murine model recapitulating the formation of Mikulicz cells in lungs, we observed that a K. rhinoscleromatis CPS mutant (KR cps-) is strongly attenuated and that mice infected with a high dose of KR cps- are still able to induce Mikulicz cells formation, unlike a K. pneumoniae capsule mutant, and to partially recapitulate the characteristic strong production of IL-10. Altogether, the results of this study show that CPS is a virulence factor of K. rhinoscleromatis not involved in the specific appearance of Mikulicz cells.
Collapse
|
174
|
Yoon BR, Oh YJ, Kang SW, Lee EB, Lee WW. Role of SLC7A5 in Metabolic Reprogramming of Human Monocyte/Macrophage Immune Responses. Front Immunol 2018; 9:53. [PMID: 29422900 PMCID: PMC5788887 DOI: 10.3389/fimmu.2018.00053] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/09/2018] [Indexed: 12/20/2022] Open
Abstract
Amino acids (AAs) are necessary nutrients which act not only as building blocks in protein synthesis but also in crucial anabolic cellular signaling pathways. It has been demonstrated that SLC7A5 is a critical transporter that mediates uptake of several essential amino acids in highly proliferative tumors and activated T cells. However, the dynamics and relevance of SLC7A5 activity in monocytes/macrophages is still poorly understood. We provide evidence that SLC7A5-mediated leucine influx contributes to pro-inflammatory cytokine production via mTOR complex 1 (mTORC1)-induced glycolytic reprograming in activated human monocytes/macrophages. Moreover, expression of SLC7A5 is significantly elevated in monocytes derived from patients with rheumatoid arthritis (RA), a chronic inflammatory disease, and was also markedly induced by LPS stimulation of both monocytes and macrophages from healthy individuals. Further, pharmacological blockade or silencing of SLC7A5 led to a significant reduction of IL-1β downstream of leucine-mediated mTORC1 activation. Inhibition of SLC7A5-mediated leucine influx was linked to downregulation of glycolytic metabolism as evidenced by the decreased extracellular acidification rate, suggesting a regulatory role for this molecule in glycolytic reprograming. Furthermore, the expression of SLC7A5 on circulating monocytes from RA patients positively correlated with clinical parameters, suggesting that SLC7A5-mediated AA influx is related to inflammatory conditions.
Collapse
Affiliation(s)
- Bo Ruem Yoon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoon-Jeong Oh
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Seong Wook Kang
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Eun Bong Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Won-Woo Lee
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, South Korea.,Seoul National University Hospital Biomedical Research Institute, Seoul, South Korea
| |
Collapse
|
175
|
Mandale MS, Bhavthankar JD, Humbe JG, Kaorey NN. Rhinoscleroma: Report of an Erratic Palatal Swelling. Contemp Clin Dent 2018; 9:S365-S368. [PMID: 30294174 PMCID: PMC6169265 DOI: 10.4103/ccd.ccd_282_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Clinicians come across cases of palatal swellings that present with ambiguous features. They may vary in their etiology from numerous infectious and noninfectious causes to a wide array of neoplasms. Accurate diagnosis of such lesions is vital for their prompt and precise management. Rhinoscleroma (RS), as its name suggests, is a persistent, specific, granulomatous disease that results in sclerosis of the affected organ - most frequently the nose. Although its occurrence in the adjacent sites has been reported, the clinical findings did not offer much deviation from the expected. Reported here is a case of RS involving the palate which not only detoured from its usual site and course of spread but also gave off a confusing façade in terms of its clinical presentation.
Collapse
Affiliation(s)
- Mandakini Subhash Mandale
- Department of Oral Pathology and Microbiology, Government Dental College and Hospital, Aurangabad, Maharashtra, India
| | - Jyoti Dilip Bhavthankar
- Department of Oral Pathology and Microbiology, Government Dental College and Hospital, Aurangabad, Maharashtra, India
| | - Jayanti Govind Humbe
- Department of Oral Pathology and Microbiology, Government Dental College and Hospital, Aurangabad, Maharashtra, India
| | - Nivedita Nishant Kaorey
- Department of Oral Pathology and Microbiology, Government Dental College and Hospital, Aurangabad, Maharashtra, India
| |
Collapse
|
176
|
The Pathogenesis of Pulmonary Sarcoidosis and Implications for Treatment. Chest 2017; 153:1432-1442. [PMID: 29224832 DOI: 10.1016/j.chest.2017.11.030] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/20/2017] [Accepted: 11/26/2017] [Indexed: 12/31/2022] Open
Abstract
Thoracic sarcoidosis is the most common form of sarcoidosis, encompassing a heterogeneous group of patients with a wide range of clinical features and associated outcomes. The distinction between isolated thoracic lymphadenopathy and pulmonary involvement matters. Morbidity is often higher, and long-term outcomes are worse for the latter. Although inflammatory infiltrates in pulmonary sarcoidosis may resolve, persistent disease activity is common and can result in lung fibrosis. Given the distinct clinical features and natural history of pulmonary sarcoidosis, its pathogenesis may differ in important ways from other sarcoidosis manifestations. This review highlights recent advances in the pathogenesis of pulmonary sarcoidosis, including the nature of the sarcoidosis antigen, the role of serum amyloid A and other host factors that contribute to alterations in innate immunity, factors that shape adaptive T-cell profiles in the lung, and how these mechanisms influence the maintenance of granulomatous inflammation in sarcoidosis. We discuss questions raised by recent findings, including the role of innate immunity in the pathogenesis, the meaning of immune cell exhaustion, and mechanisms that may contribute to lung fibrosis in sarcoidosis. We conclude with a reflection on when and how immunosuppressive therapies may be helpful for pulmonary sarcoidosis, a consideration of nonpharmacologic management strategies, and a survey of potential novel therapeutic targets for this vexing disease.
Collapse
|
177
|
Horn V, Triantafyllopoulou A. DNA damage signaling and polyploid macrophages in chronic inflammation. Curr Opin Immunol 2017; 50:55-63. [PMID: 29202328 DOI: 10.1016/j.coi.2017.11.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/30/2017] [Accepted: 11/17/2017] [Indexed: 01/01/2023]
Abstract
Whole genome duplications, an important step in cancer development, also occur in the macrophage lineage in disease: large multinucleated macrophages found within compact, ordered aggregates of immune cells, called granulomas, are a well-known histologic entity. Very recent work suggests that granuloma macrophages remarkably acquire epithelial cell features and the genotoxic stress response instructs granuloma macrophage genome duplications, suggesting that granuloma macrophages and pre-malignant epithelial cells may share common mechanisms of adaptation to chronic genotoxic stress. Exploring these mechanisms is key for a better understanding of the pathogenesis of chronic inflammatory diseases. Here we review the mechanisms of macrophage polyploidization, the role of DNA damage signaling in this process and the function of polyploid macrophages, with a focus on chronic inflammation.
Collapse
Affiliation(s)
- Veronika Horn
- Department of Rheumatology and Clinical Immunology, Charité University Medical Center, D-10117 Berlin, Germany; Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Antigoni Triantafyllopoulou
- Department of Rheumatology and Clinical Immunology, Charité University Medical Center, D-10117 Berlin, Germany; Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, D-79106 Freiburg, Germany; German Rheumatism Research Center, A Leibniz Institute, D-10117 Berlin, Germany; Institute of Microbiology, Charité University Medical Center, D-12203 Berlin, Germany.
| |
Collapse
|
178
|
mTOR signaling in immune cells and its implications for cancer immunotherapy. Cancer Lett 2017; 408:182-189. [DOI: 10.1016/j.canlet.2017.08.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/22/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
|
179
|
Kaiser Y, Lakshmikanth T, Chen Y, Mikes J, Eklund A, Brodin P, Achour A, Grunewald J. Mass Cytometry Identifies Distinct Lung CD4 + T Cell Patterns in Löfgren's Syndrome and Non-Löfgren's Syndrome Sarcoidosis. Front Immunol 2017; 8:1130. [PMID: 28955342 PMCID: PMC5601005 DOI: 10.3389/fimmu.2017.01130] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/28/2017] [Indexed: 11/27/2022] Open
Abstract
Sarcoidosis is a granulomatous disorder of unknown etiology, characterized by accumulation of activated CD4+ T cells in the lungs. Disease phenotypes Löfgren’s syndrome (LS) and “non-LS” differ in terms of clinical manifestations, genetic background, HLA association, and prognosis, but the underlying inflammatory mechanisms largely remain unknown. Bronchoalveolar lavage fluid cells from four HLA-DRB1*03+ LS and four HLA-DRB1*03− non-LS patients were analyzed by mass cytometry, using a panel of 33 unique markers. Differentially regulated CD4+ T cell populations were identified using the Citrus algorithm, and t-stochastic neighborhood embedding was applied for dimensionality reduction and single-cell data visualization. We identified 19 individual CD4+ T cell clusters differing significantly in abundance between LS and non-LS patients. Seven clusters more frequent in LS patients were characterized by significantly higher expression of regulatory receptors CTLA-4, PD-1, and ICOS, along with low expression of adhesion marker CD44. In contrast, 12 clusters primarily found in non-LS displayed elevated expression of activation and effector markers HLA-DR, CD127, CD39, as well as CD44. Hierarchical clustering further indicated functional heterogeneity and diverse origins of T cell receptor Vα2.3/Vβ22-restricted cells in LS. Finally, a near-complete overlap of CD8 and Ki-67 expression suggested larger influence of CD8+ T cell activity on sarcoid inflammation than previously appreciated. In this study, we provide detailed characterization of pulmonary T cells and immunological parameters that define separate disease pathways in LS and non-LS. With direct association to clinical parameters, such as granuloma persistence, resolution, or chronic inflammation, these results provide a valuable foundation for further exploration and potential clinical application.
Collapse
Affiliation(s)
- Ylva Kaiser
- Respiratory Medicine Unit, Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Tadepally Lakshmikanth
- Science for Life Laboratory, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Yang Chen
- Science for Life Laboratory, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Jaromir Mikes
- Science for Life Laboratory, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Eklund
- Respiratory Medicine Unit, Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Petter Brodin
- Science for Life Laboratory, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden.,Department of Neonatology, Karolinska University Hospital, Stockholm, Sweden
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Grunewald
- Respiratory Medicine Unit, Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
180
|
Jones RG, Pearce EJ. MenTORing Immunity: mTOR Signaling in the Development and Function of Tissue-Resident Immune Cells. Immunity 2017; 46:730-742. [PMID: 28514674 DOI: 10.1016/j.immuni.2017.04.028] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 04/24/2017] [Accepted: 04/28/2017] [Indexed: 12/31/2022]
Abstract
Tissue-resident immune cells must balance survival in peripheral tissues with the capacity to respond rapidly upon infection or tissue damage, and in turn couple these responses with intrinsic metabolic control and conditions in the tissue microenvironment. The serine/threonine kinase mammalian/mechanistic target of rapamycin (mTOR) is a central integrator of extracellular and intracellular growth signals and cellular metabolism and plays important roles in both innate and adaptive immune responses. This review discusses the function of mTOR signaling in the differentiation and function of tissue-resident immune cells, with focus on the role of mTOR as a metabolic sensor and its impact on metabolic regulation in innate and adaptive immune cells. We also discuss the impact of metabolic constraints in tissues on immune homeostasis and disease, and how manipulating mTOR activity with drugs such as rapamycin can modulate immunity in these contexts.
Collapse
Affiliation(s)
- Russell G Jones
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada.
| | - Edward J Pearce
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
181
|
Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC, Abraham RT. The PI3K Pathway in Human Disease. Cell 2017; 170:605-635. [PMID: 28802037 PMCID: PMC5726441 DOI: 10.1016/j.cell.2017.07.029] [Citation(s) in RCA: 1740] [Impact Index Per Article: 217.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/17/2017] [Accepted: 07/20/2017] [Indexed: 02/08/2023]
Abstract
Phosphoinositide 3-kinase (PI3K) activity is stimulated by diverse oncogenes and growth factor receptors, and elevated PI3K signaling is considered a hallmark of cancer. Many PI3K pathway-targeted therapies have been tested in oncology trials, resulting in regulatory approval of one isoform-selective inhibitor (idelalisib) for treatment of certain blood cancers and a variety of other agents at different stages of development. In parallel to PI3K research by cancer biologists, investigations in other fields have uncovered exciting and often unpredicted roles for PI3K catalytic and regulatory subunits in normal cell function and in disease. Many of these functions impinge upon oncology by influencing the efficacy and toxicity of PI3K-targeted therapies. Here we provide a perspective on the roles of class I PI3Ks in the regulation of cellular metabolism and in immune system functions, two topics closely intertwined with cancer biology. We also discuss recent progress developing PI3K-targeted therapies for treatment of cancer and other diseases.
Collapse
Affiliation(s)
- David A Fruman
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697-3900, USA.
| | - Honyin Chiu
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697-3900, USA
| | - Benjamin D Hopkins
- Meyer Cancer Center, Weill Cornell Medical College, 413 E. 69(th) Street, New York, NY 10021, USA
| | - Shubha Bagrodia
- Oncology R&D Group, Pfizer Worldwide Research and Development, 10646/CB4 Science Center Drive, San Diego, CA 92121, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medical College, 413 E. 69(th) Street, New York, NY 10021, USA
| | - Robert T Abraham
- Oncology R&D Group, Pfizer Worldwide Research and Development, 10646/CB4 Science Center Drive, San Diego, CA 92121, USA
| |
Collapse
|
182
|
Di Marco F, Palumbo G, Terraneo S, Imeri G, Lesma E, Sverzellati N, Peron A, Gualandri L, Canevini MP, Centanni S. Lymphangioleiomyomatosis, multifocal micronodular pneumocyte hyperplasia, and sarcoidosis: more pathological findings in the same chest CT, or a single pathological pathway? BMC Pulm Med 2017; 17:107. [PMID: 28754097 PMCID: PMC5534042 DOI: 10.1186/s12890-017-0447-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 07/18/2017] [Indexed: 02/07/2023] Open
Abstract
Background Autoimmune hepatitis/primary biliary cirrhosis overlap syndrome, lymphangioleiomyomatosis/tuberous sclerosis complex (LAM-TSC), and sarcoidosis are three rare diseases. Here we present, to the best of our knowledge, the first description of a patient with the coexistence of these three diseases. Case presentation A 47-year-old woman affected by LAM-TSC and primary biliary cirrosis/autoimmune hepatitis overlap syndrome. During her follow up a high resolution chest CT scan (HRTC) confirmed the presence of both multiple cysts and micronodular opacities consistent with multifocal micronodular pneumocytes hyperlasia (MMPH), and revealed multiple hilar-mediastinal symmetrical lymphadenopathies suggestive of sarcoidosis. Simultaneously, subcutaneous nodules appeared on her forearm bilaterally. Cutaneous biopsy showed granulomatous dermatitis with sarcoid-like granulomas. A diagnosis of stage I pulmonary sarcoidosis was made. No treatment for sarcoidosis was initiated since the patient had neither systemic involvement, nor respiratory impairment. Conclusions The presence of more than one rare disease should challenge the concept of a potential common underlying mechanism, since the a priori probability of the concomitant presence of different conditions with different pathogenic mechanisms - especially if rare diseases - is low. We speculate that the dysregulation of the pathway involving mTOR and MAPK and their interaction might play a role in the pathogenesis of other diseases, including sarcoidosis.
Collapse
Affiliation(s)
- Fabiano Di Marco
- Respiratory Unit, Ospedale San Paolo, Department of Health Sciences, Università degli Studi di Milano, Via A. di Rudinì, 8, 20142, Milan, Italy.
| | - Giuseppina Palumbo
- Respiratory Unit, Ospedale San Paolo, Department of Health Sciences, Università degli Studi di Milano, Via A. di Rudinì, 8, 20142, Milan, Italy
| | - Silvia Terraneo
- Respiratory Unit, Ospedale San Paolo, Department of Health Sciences, Università degli Studi di Milano, Via A. di Rudinì, 8, 20142, Milan, Italy
| | - Gianluca Imeri
- Respiratory Unit, Ospedale San Paolo, Department of Health Sciences, Università degli Studi di Milano, Via A. di Rudinì, 8, 20142, Milan, Italy
| | - Elena Lesma
- Laboratories of Pharmacology, Università degli Studi di Milano, Milan, Italy
| | - Nicola Sverzellati
- Radiology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Angela Peron
- Epilepsy Center, Ospedale San Paolo, Department of Health Sciences, Università degli Studi di Milano ASST Santi Paolo e Carlo, Milan, Italy
| | - Lorenzo Gualandri
- Dermatologic Clinic, Ospedale San Paolo, ASST Santi Paolo e Carlo, Milan, Italy
| | - Maria Paola Canevini
- Epilepsy Center, Ospedale San Paolo, Department of Health Sciences, Università degli Studi di Milano ASST Santi Paolo e Carlo, Milan, Italy
| | - Stefano Centanni
- Respiratory Unit, Ospedale San Paolo, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
183
|
Can NT, Grenert JP, Vohra P. Concomitant Epstein-Barr Virus-associated smooth muscle tumor and granulomatous inflammation of the liver. Pathol Res Pract 2017; 213:1306-1309. [PMID: 28756985 DOI: 10.1016/j.prp.2017.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/12/2017] [Accepted: 07/02/2017] [Indexed: 11/17/2022]
Abstract
Epstein-Barr Virus-associated smooth muscle tumor (EBV-SMT) is a rare mesenchymal tumor typically seen in immunocompromised patients. Here, we report a case of EBV-SMT and associated granulomatous inflammation in the liver of a 32-year-old man with history of human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS). To our knowledge, an association of these two lesions has not been previously reported. We review the literature and discuss pathogenesis, differential diagnosis and immunohistochemical (IHC) stains helpful for the diagnosis of this rare entity. Finally, we consider possible explanations for the concomitant presence of these lesions.
Collapse
Affiliation(s)
- Nhu Thuy Can
- Department of Pathology, University of California, San Francisco, CA, USA
| | - James P Grenert
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Poonam Vohra
- Department of Pathology, University of California, San Francisco, CA, USA.
| |
Collapse
|
184
|
Linke M, Fritsch SD, Sukhbaatar N, Hengstschläger M, Weichhart T. mTORC1 and mTORC2 as regulators of cell metabolism in immunity. FEBS Lett 2017; 591:3089-3103. [PMID: 28600802 DOI: 10.1002/1873-3468.12711] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/24/2017] [Accepted: 06/02/2017] [Indexed: 12/20/2022]
Abstract
The mechanistic target of rapamycin (mTOR) pathway is an evolutionarily conserved signaling pathway that senses intra- and extracellular nutrients, growth factors, and pathogen-associated molecular patterns to regulate the function of innate and adaptive immune cell populations. In this review, we focus on the role of the mTOR complex 1 (mTORC1) and mTORC2 in the regulation of the cellular energy metabolism of these immune cells to regulate and support immune responses. In this regard, mTORC1 and mTORC2 generally promote an anabolic response by stimulating protein synthesis, glycolysis, mitochondrial functions, and lipid synthesis to influence proliferation and survival, effector and memory responses, innate training and tolerance as well as hematopoietic stem cell maintenance and differentiation. Deactivation of mTOR restores cell homeostasis after immune activation and optimizes antigen presentation and memory T-cell generation. These findings show that the mTOR pathway integrates spatiotemporal information of the environmental and cellular energy status by regulating cellular metabolic responses to guide immune cell activation. Elucidation of the metabolic control mechanisms of immune responses will help to generate a systemic understanding of the immune system.
Collapse
Affiliation(s)
- Monika Linke
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Austria
| | - Stephanie Deborah Fritsch
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Austria
| | - Nyamdelger Sukhbaatar
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Austria
| | - Markus Hengstschläger
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Austria
| | - Thomas Weichhart
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Austria
| |
Collapse
|
185
|
|