1
|
Farkas AM, Youssef D, Tran MA, Balan S, Newman JH, Audenet F, Anastos H, Velazquez LG, Peros A, Ananthanarayanan A, Daza J, Gonzalez-Gugel E, Sadanala K, Theorell J, Galsky MD, Horowitz A, Sfakianos JP, Bhardwaj N. Natural Killer Cell Dysfunction In Human Bladder Cancer Is Caused By Tissue-Specific Suppression of SLAMF6 Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591366. [PMID: 38746459 PMCID: PMC11092609 DOI: 10.1101/2024.04.30.591366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
NK cells are innate lymphocytes critical for surveillance of viruses and tumors, however the mechanisms underlying NK cell dysfunction in cancer are incompletely understood. We assessed the effector function of NK cells from bladder cancer patients and found severe dysfunction in NK cells derived from tumors versus peripheral blood. While both peripheral and tumor-infiltrating NK cells exhibited conserved patterns of inhibitory receptor over-expression, this did not explain the observed defects in NK surveillance in bladder tumors. Rather, TME-specific TGF-β and metabolic perturbations such as hypoxia directly suppressed NK cell function. Specifically, an oxygen-dependent reduction in signaling through SLAMF6 was mechanistically responsible for poor NK cell function, as tumor-infiltrating NK cells cultured ex vivo under normoxic conditions exhibited complete restoration of function, while deletion of SLAMF6 abrogated NK cell cytolytic function even under normoxic conditions. Collectively, this work highlights the role of tissue-specific factors in dictating NK cell function, and implicates SLAMF6 signaling as a rational target for immuno-modulation to improve NK cell function in bladder cancer.
Collapse
|
2
|
Tran MA, Youssef D, Shroff S, Chowhan D, Beaumont KG, Sebra R, Mehrazin R, Wiklund P, Lin JJ, Horowitz A, Farkas AM, Galsky MD, Sfakianos JP, Bhardwaj N. Urine scRNAseq reveals new insights into the bladder tumor immune microenvironment. J Exp Med 2024; 221:e20240045. [PMID: 38847806 PMCID: PMC11157455 DOI: 10.1084/jem.20240045] [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: 01/07/2024] [Revised: 04/04/2024] [Accepted: 05/21/2024] [Indexed: 06/10/2024] Open
Abstract
Due to bladder tumors' contact with urine, urine-derived cells (UDCs) may serve as a surrogate for monitoring the tumor microenvironment (TME) in bladder cancer (BC). However, the composition of UDCs and the extent to which they mirror the tumor remain poorly characterized. We generated the first single-cell RNA-sequencing of BC patient UDCs with matched tumor and peripheral blood mononuclear cells (PBMC). BC urine was more cellular than healthy donor (HD) urine, containing multiple immune populations including myeloid cells, CD4+ and CD8+ T cells, natural killer (NK) cells, B cells, and dendritic cells (DCs) in addition to tumor and stromal cells. Immune UDCs were transcriptionally more similar to tumor than blood. UDCs encompassed cytotoxic and activated CD4+ T cells, exhausted and tissue-resident memory CD8+ T cells, macrophages, germinal-center-like B cells, tissue-resident and adaptive NK cells, and regulatory DCs found in tumor but lacking or absent in blood. Our findings suggest BC UDCs may be surrogates for the TME and serve as therapeutic biomarkers.
Collapse
Affiliation(s)
- Michelle A. Tran
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dina Youssef
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sanjana Shroff
- Department of Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Disha Chowhan
- Department of Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristin G. Beaumont
- Department of Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Department of Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Reza Mehrazin
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Wiklund
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jenny J. Lin
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amir Horowitz
- Department of Immunology and Immunotherapy, The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam M. Farkas
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D. Galsky
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John P. Sfakianos
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Extramural Member, Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| |
Collapse
|
3
|
Zhang Y, Meng Y, Wang S, Zu Y, Zhao X. Exploring pectin-casein micelles as novel carriers for oral drug delivery of artesunate in the treatment of systemic lupus erythematosus. Int J Biol Macromol 2024; 271:132523. [PMID: 38788864 DOI: 10.1016/j.ijbiomac.2024.132523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/06/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024]
Abstract
The oral route of administration is considered the optimal choice for treating chronic diseases due to its convenience and non-invasiveness, which can help prevent physical and mental harm to patients undergoing long-term treatment. However, challenges such as safety, gastrointestinal stability, and bioavailability of oral drugs often limit their effectiveness. Natural biomacromolecule micelles, known for their safety, stability, biocompatibility, and diverse functions, have emerged as promising carriers for oral treatment of chronic diseases like systemic lupus erythematosus (SLE) with fat-soluble drugs. This study introduces an innovative approach by developing an oral delivery system using chemically synthesized natural biomacromolecules to load artesunate for treating SLE. By synthesizing amphiphilic polymer micelles from pectin and casein through a carbodiimide reaction, a more stable structure is achieved. The hydrophobic core of these micelles encapsulates artesunate, resulting in the formation of an oral delivery system (PC-AS) with several advantages, including high drug loading and encapsulation efficiency, small particle size, negative potential, strong stability in the gastrointestinal tract, low toxicity and side effects, strong adhesion in the small intestine, and high bioavailability. These advantages facilitate efficient absorption of artesunate in the gastrointestinal tract, leading to improved bioavailability and effective alleviation of SLE-like symptoms in MRL/lpr mice. By utilizing chemically synthesized natural macromolecular micelles for delivering artesunate in the treatment of SLE, this study overcomes the oral barriers associated with the original drug and presents a novel solution for the long-term oral treatment of chronic diseases.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China; Engineering Research Center of Microbial Resources Development and Green Recycling, University of Shaanxi Province, College of Life Sciences, Yan'an University, Yan'an 716000, Shaanxi, PR China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China.
| | - Yongbin Meng
- Engineering Research Center of Microbial Resources Development and Green Recycling, University of Shaanxi Province, College of Life Sciences, Yan'an University, Yan'an 716000, Shaanxi, PR China.
| | - Siying Wang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China.
| | - Yuangang Zu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Xiuhua Zhao
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China.
| |
Collapse
|
4
|
Boothby M, Cho SH. Hypoxia and the Hypoxia-Inducible Factors in Lymphocyte Differentiation and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:115-141. [PMID: 39017842 DOI: 10.1007/978-3-031-62731-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Molecular oxygen doubles as a biomolecular building block and an element required for energy generation and metabolism in aerobic organisms. A variety of systems in mammalian cells sense the concentration of oxygen to which they are exposed and are tuned to the range present in our blood and tissues. The ability to respond to insufficient O2 in tissues is central to regulation of erythroid lineage cells, but challenges also are posed for immune cells by a need to adjust to very different oxygen concentrations. Hypoxia-inducible factors (HIFs) provide a major means of making such adjustments. For adaptive immunity, lymphoid lineages are initially defined in bone marrow niches; T lineage cells arise in the thymus, and B cells complete maturation in the spleen. Lymphocytes move from these first stops into microenvironments (bloodstream, lymphatics, and tissues) with distinct oxygenation in each. Herein, evidence pertaining to functions of the HIF transcription factors (TFs) in lymphocyte differentiation and function is reviewed. For the CD4+ and CD8+ subsets of T cells, the case is very strong that hypoxia and HIFs regulate important differentiation events and functions after the naïve lymphocytes emerge from the thymus. In the B lineage, the data indicate that HIF1 contributes to a balanced regulation of B-cell fates after antigen (Ag) activation during immunity. A model synthesized from the aggregate literature is that HIF in lymphocytes generally serves to modulate function in a manner dependent on the molecular context framed by other TFs and signals.
Collapse
Affiliation(s)
- Mark Boothby
- Departments of Pathology, Microbiology, Immunology (Molecular Pathogenesis Division), Vanderbilt University Medical Center, Nashville, TN, USA.
- Medicine (Rheumatology and Immunology Division), Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Inflammation, Immunity (VI4), Nashville, TN, USA.
| | - Sung Hoon Cho
- Departments of Pathology, Microbiology, Immunology (Molecular Pathogenesis Division), Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Inflammation, Immunity (VI4), Nashville, TN, USA
| |
Collapse
|
5
|
Sharma P, Sri Swetha Victoria V, Praneeth Kumar P, Karmakar S, Swetha M, Reddy A. Cross-talk between insulin resistance and nitrogen species in hypoxia leads to deterioration of tissue and homeostasis. Int Immunopharmacol 2023; 122:110472. [PMID: 37392570 DOI: 10.1016/j.intimp.2023.110472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/19/2023] [Accepted: 06/07/2023] [Indexed: 07/03/2023]
Abstract
Hypoxia has been linked with insulin resistance as it produces changes in the metabolism of the cell; in which the adipocytes impede the insulin receptor tyrosine, phosphorylation, directing at decreased levels of transport of glucose. At this juncture, we are focusing on cross-talk between insulin resistance and nitrogen species in hypoxia, leading to the deterioration of tissue and homeostasis. Physiological levels of nitric oxide play a very crucial role in acting as a priority effector and signaling molecule, arbitrating the body's responses to hypoxia. Both ROS and RNS are associated with a reduction in IRS1 phosphorylation in tyrosine, which leads to reduced levels of IRS1 content and insulin response, which further leads to insulin resistance. Cellular hypoxia is a trigger to inflammatory mediators which signal tissue impairment and initiate survival requirements. But, hypoxia-mediated inflammation act as a protective role by an immune response and promotes wound healing during infection. In this review, we abridge the crosstalk between the inflammation and highlight the dysregulation in physiological consequences due to diabetes mellitus. Finally, we review various treatments available for its related physiological complications.
Collapse
Affiliation(s)
- Priyanshy Sharma
- Animal Cell Culture Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nādu, India
| | - V Sri Swetha Victoria
- Animal Cell Culture Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nādu, India
| | - P Praneeth Kumar
- Animal Cell Culture Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nādu, India
| | - Sarbani Karmakar
- Animal Cell Culture Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nādu, India
| | - Mudduluru Swetha
- Animal Cell Culture Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nādu, India
| | - Amala Reddy
- Animal Cell Culture Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nādu, India.
| |
Collapse
|
6
|
Maurya DK, Sharma D, Sandur SK. Hypoxia induces dichotomous and reversible attenuation of T cell responses through reactive oxygen species-dependent phenotype redistribution and delay in lymphoblast proliferation. Free Radic Res 2023; 57:1-13. [PMID: 36947008 DOI: 10.1080/10715762.2023.2178918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
As T cells transit between blood, lymphoid organs, and peripheral tissues, they experience varied levels of oxygen/hypoxia in inflamed tissues, skin, intestinal lining, and secondary lymphoid organs. Critical illness among COVID-19 patients is also associated with transient hypoxia and attenuation of T cell responses. Hypoxia is the fulcrum of altered metabolism, impaired functions, and cessation of growth of a subset of T cells. However, the restoration of normal T cell functions following transient hypoxia and kinetics of their phenotype-redistribution is not completely understood. Here, we sought to understand kinetics and reversibility of dichotomous T cell responses under sustained and transient hypoxia. We found that a subset of activated T cells accumulated as lymphoblasts under hypoxia. Further, T cells showed the normal expression of activation markers CD25 and CD69 and inflammatory cytokine secretion but a subset exhibited delayed cell proliferation under hypoxia. Increased levels of reactive oxygen species (ROS) in cytosol and mitochondria were seen during dichotomous and reversible attenuation of T cell response under hypoxia. Cell cycle analysis revealed maximum levels of cytosolic and mitochondrial ROS in dividing T cells (in S, G2, or M phase). Hypoxic T cells also showed specific attenuation of activation induced memory phenotype conversion without affecting naïve and activated T cells. Hypoxia-related attenuation of T cell proliferation was also found to be reversible in an allogeneic leukocyte specific mixed lymphocyte reaction assay. In summary, our results show that hypoxia induces a reversible delay in proliferation of a subset of T cells which is associated with obliteration of memory phenotype and specific increase in cytosolic/mitochondrial ROS levels in actively dividing subpopulation. Thus, the transient reoxygenation of hypoxic patients may restore normal T cell responses.
Collapse
Affiliation(s)
- Dharmendra Kumar Maurya
- Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Santosh Kumar Sandur
- Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
7
|
Park J, Hsueh PC, Li Z, Ho PC. Microenvironment-driven metabolic adaptations guiding CD8 + T cell anti-tumor immunity. Immunity 2023; 56:32-42. [PMID: 36630916 DOI: 10.1016/j.immuni.2022.12.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023]
Abstract
The metabolic stress occurring in the tumor microenvironment (TME) hampers T cell anti-tumor immunity by disturbing T cell metabolic and epigenetic programs. Recent studies are making headway toward identifying strategies to unleash T cell activities by targeting T cell metabolism. Furthermore, efforts have been made to improve the efficacy of immune checkpoint blockade and adoptive cell transfer therapies. However, distinct treatment outcomes across different cancers raise the question of whether our understanding of the features of CD8+ T cells within the TME are universal, regardless of their tissue of origin. Here, we review the common and distinct environmental factors affecting CD8+ T cells across tumors. Moreover, we discuss how distinct tissue-specific niches are interpreted by CD8+ T cells based on studies on tissue-resident memory T (Trm) cells and how these insights can pave the way for a better understanding of the metabolic regulation of CD8+ T cell differentiation and anti-tumor immunity.
Collapse
Affiliation(s)
- Jaeoh Park
- Department of Fundamental Oncology, University of Lausanne, 1066 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland.
| | - Pei-Chun Hsueh
- Department of Fundamental Oncology, University of Lausanne, 1066 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland.
| | - Zhiyu Li
- Department of Fundamental Oncology, University of Lausanne, 1066 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland; Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, 1066 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland.
| |
Collapse
|
8
|
Simonetti G, Angeli D, Petracci E, Fonzi E, Vedovato S, Sperotto A, Padella A, Ghetti M, Ferrari A, Robustelli V, Di Liddo R, Conconi MT, Papayannidis C, Cerchione C, Rondoni M, Astolfi A, Ottaviani E, Martinelli G, Gottardi M. Adrenomedullin Expression Characterizes Leukemia Stem Cells and Associates With an Inflammatory Signature in Acute Myeloid Leukemia. Front Oncol 2021; 11:684396. [PMID: 34150648 PMCID: PMC8208888 DOI: 10.3389/fonc.2021.684396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
Adrenomedullin (ADM) is a hypotensive and vasodilator peptide belonging to the calcitonin gene-related peptide family. It is secreted in vitro by endothelial cells and vascular smooth muscle cells, and is significantly upregulated by a number of stimuli. Moreover, ADM participates in the regulation of hematopoietic compartment, solid tumors and leukemias, such as acute myeloid leukemia (AML). To better characterize ADM involvement in AML pathogenesis, we investigated its expression during human hematopoiesis and in leukemic subsets, based on a morphological, cytogenetic and molecular characterization and in T cells from AML patients. In hematopoietic stem/progenitor cells and T lymphocytes from healthy subjects, ADM transcript was barely detectable. It was expressed at low levels by megakaryocytes and erythroblasts, while higher levels were measured in neutrophils, monocytes and plasma cells. Moreover, cells populating the hematopoietic niche, including mesenchymal stem cells, showed to express ADM. ADM was overexpressed in AML cells versus normal CD34+ cells and in the subset of leukemia compared with hematopoietic stem cells. In parallel, we detected a significant variation of ADM expression among cytogenetic subgroups, measuring the highest levels in inv(16)/t(16;16) or complex karyotype AML. According to the mutational status of AML-related genes, the analysis showed a lower expression of ADM in FLT3-ITD, NPM1-mutated AML and FLT3-ITD/NPM1-mutated cases compared with wild-type ones. Moreover, ADM expression had a negative impact on overall survival within the favorable risk class, while showing a potential positive impact within the subgroup receiving a not-intensive treatment. The expression of 135 genes involved in leukemogenesis, regulation of cell proliferation, ferroptosis, protection from apoptosis, HIF-1α signaling, JAK-STAT pathway, immune and inflammatory responses was correlated with ADM levels in the bone marrow cells of at least two AML cohorts. Moreover, ADM was upregulated in CD4+ T and CD8+ T cells from AML patients compared with healthy controls and some ADM co-expressed genes participate in a signature of immune tolerance that characterizes CD4+ T cells from leukemic patients. Overall, our study shows that ADM expression in AML associates with a stem cell phenotype, inflammatory signatures and genes related to immunosuppression, all factors that contribute to therapy resistance and disease relapse.
Collapse
Affiliation(s)
- Giorgia Simonetti
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Davide Angeli
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Elisabetta Petracci
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Eugenio Fonzi
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Susanna Vedovato
- Department of Clinical and Experimental Medicine, University of Padova, Padua, Italy
| | - Alessandra Sperotto
- Hematology and Transplant Center Unit, Dipartimento di Area Medica (DAME), Udine University Hospital, Udine, Italy
| | - Antonella Padella
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Martina Ghetti
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Anna Ferrari
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Valentina Robustelli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Rosa Di Liddo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Maria Teresa Conconi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Cristina Papayannidis
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Michela Rondoni
- Hematology Unit & Romagna Transplant Network, Ravenna Hospital, Ravenna, Italy
| | - Annalisa Astolfi
- “Giorgio Prodi” Cancer Research Center, University of Bologna, Bologna, Italy
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Emanuela Ottaviani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Giovanni Martinelli
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Michele Gottardi
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV, IRCCS, Padua, Italy
| |
Collapse
|
9
|
Qiu J, Wu B, Goodman SB, Berry GJ, Goronzy JJ, Weyand CM. Metabolic Control of Autoimmunity and Tissue Inflammation in Rheumatoid Arthritis. Front Immunol 2021; 12:652771. [PMID: 33868292 PMCID: PMC8050350 DOI: 10.3389/fimmu.2021.652771] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022] Open
Abstract
Like other autoimmune diseases, rheumatoid arthritis (RA) develops in distinct stages, with each phase of disease linked to immune cell dysfunction. HLA class II genes confer the strongest genetic risk to develop RA. They encode for molecules essential in the activation and differentiation of T cells, placing T cells upstream in the immunopathology. In Phase 1 of the RA disease process, T cells lose a fundamental function, their ability to be self-tolerant, and provide help for autoantibody-producing B cells. Phase 2 begins many years later, when mis-differentiated T cells gain tissue-invasive effector functions, enter the joint, promote non-resolving inflammation, and give rise to clinically relevant arthritis. In Phase 3 of the RA disease process, abnormal innate immune functions are added to adaptive autoimmunity, converting synovial inflammation into a tissue-destructive process that erodes cartilage and bone. Emerging data have implicated metabolic mis-regulation as a fundamental pathogenic pathway in all phases of RA. Early in their life cycle, RA T cells fail to repair mitochondrial DNA, resulting in a malfunctioning metabolic machinery. Mitochondrial insufficiency is aggravated by the mis-trafficking of the energy sensor AMPK away from the lysosomal surface. The metabolic signature of RA T cells is characterized by the shunting of glucose toward the pentose phosphate pathway and toward biosynthetic activity. During the intermediate and terminal phase of RA-imposed tissue inflammation, tissue-residing macrophages, T cells, B cells and stromal cells are chronically activated and under high metabolic stress, creating a microenvironment poor in oxygen and glucose, but rich in metabolic intermediates, such as lactate. By sensing tissue lactate, synovial T cells lose their mobility and are trapped in the tissue niche. The linkage of defective DNA repair, misbalanced metabolic pathways, autoimmunity, and tissue inflammation in RA encourages metabolic interference as a novel treatment strategy during both the early stages of tolerance breakdown and the late stages of tissue inflammation. Defining and targeting metabolic abnormalities provides a new paradigm to treat, or even prevent, the cellular defects underlying autoimmune disease.
Collapse
Affiliation(s)
- Jingtao Qiu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Bowen Wu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Stuart B Goodman
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Jorg J Goronzy
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Cornelia M Weyand
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
10
|
Chen Y, Gaber T. Hypoxia/HIF Modulates Immune Responses. Biomedicines 2021; 9:biomedicines9030260. [PMID: 33808042 PMCID: PMC8000289 DOI: 10.3390/biomedicines9030260] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 02/07/2023] Open
Abstract
Oxygen availability varies throughout the human body in health and disease. Under physiological conditions, oxygen availability drops from the lungs over the blood stream towards the different tissues into the cells and the mitochondrial cavities leading to physiological low oxygen conditions or physiological hypoxia in all organs including primary lymphoid organs. Moreover, immune cells travel throughout the body searching for damaged cells and foreign antigens facing a variety of oxygen levels. Consequently, physiological hypoxia impacts immune cell function finally controlling innate and adaptive immune response mainly by transcriptional regulation via hypoxia-inducible factors (HIFs). Under pathophysiological conditions such as found in inflammation, injury, infection, ischemia and cancer, severe hypoxia can alter immune cells leading to dysfunctional immune response finally leading to tissue damage, cancer progression and autoimmunity. Here we summarize the effects of physiological and pathophysiological hypoxia on innate and adaptive immune activity, we provide an overview on the control of immune response by cellular hypoxia-induced pathways with focus on the role of HIFs and discuss the opportunity to target hypoxia-sensitive pathways for the treatment of cancer and autoimmunity.
Collapse
Affiliation(s)
- Yuling Chen
- Charité—Universitätsmedizin Berlin, Corporate Ember of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Charitéplatz 1, 10117 Berlin, Germany;
| | - Timo Gaber
- Charité—Universitätsmedizin Berlin, Corporate Ember of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Charitéplatz 1, 10117 Berlin, Germany;
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-513364
| |
Collapse
|
11
|
Saragovi A, Abramovich I, Omar I, Arbib E, Toker O, Gottlieb E, Berger M. Systemic hypoxia inhibits T cell response by limiting mitobiogenesis via matrix substrate-level phosphorylation arrest. eLife 2020; 9:56612. [PMID: 33226340 PMCID: PMC7728436 DOI: 10.7554/elife.56612] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 11/21/2020] [Indexed: 11/30/2022] Open
Abstract
Systemic oxygen restriction (SOR) is prevalent in numerous clinical conditions, including chronic obstructive pulmonary disease (COPD), and is associated with increased susceptibility to viral infections. However, the influence of SOR on T cell immunity remains uncharacterized. Here we show the detrimental effect of hypoxia on mitochondrial-biogenesis in activated mouse CD8+ T cells. We find that low oxygen level diminishes CD8+ T cell anti-viral response in vivo. We reveal that respiratory restriction inhibits ATP-dependent matrix processes that are critical for mitochondrial-biogenesis. This respiratory restriction-mediated effect could be rescued by TCA cycle re-stimulation, which yielded increased mitochondrial matrix-localized ATP via substrate-level phosphorylation. Finally, we demonstrate that the hypoxia-arrested CD8+ T cell anti-viral response could be rescued in vivo through brief exposure to atmospheric oxygen pressure. Overall, these findings elucidate the detrimental effect of hypoxia on mitochondrial-biogenesis in activated CD8+ T cells, and suggest a new approach for reducing viral infections in COPD.
Collapse
Affiliation(s)
- Amijai Saragovi
- The Lautenberg center for Immunology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University Medical School, Jerusalem, Israel
| | - Ifat Abramovich
- The Ruth and Bruce Rappaport, Faculty of Medicine, Technion - Israel Institute of Technology, Jerusalem, Israel
| | - Ibrahim Omar
- The Lautenberg center for Immunology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University Medical School, Jerusalem, Israel
| | - Eliran Arbib
- The Lautenberg center for Immunology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University Medical School, Jerusalem, Israel
| | - Ori Toker
- Faculty of Medicine, Hebrew University of Jerusalem; The Allergy and Immunology Unit, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Eyal Gottlieb
- The Ruth and Bruce Rappaport, Faculty of Medicine, Technion - Israel Institute of Technology, Jerusalem, Israel
| | - Michael Berger
- The Lautenberg center for Immunology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University Medical School, Jerusalem, Israel
| |
Collapse
|
12
|
Iranparast S, Tayebi S, Ahmadpour F, Yousefi B. Tumor-Induced Metabolism and T Cells Located in Tumor Environment. Curr Cancer Drug Targets 2020; 20:741-756. [PMID: 32691710 DOI: 10.2174/1568009620666200720010647] [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: 02/17/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022]
Abstract
Several subtypes of T cells are located in a tumor environment, each of which supplies their energy using different metabolic mechanisms. Since the cancer cells require high levels of glucose, the conditions of food poverty in the tumor environment can cause inactivation of immune cells, especially the T-effector cells, due to the need for glucose in the early stages of these cells activity. Different signaling pathways, such as PI3K-AKt-mTOR, MAPK, HIF-1α, etc., are activated or inactivated by the amount and type of energy source or oxygen levels that determine the fate of T cells in a cancerous environment. This review describes the metabolites in the tumor environment and their effects on the function of T cells. It also explains the signaling pathway of T cells in the tumor and normal conditions, due to the level of access to available metabolites and subtypes of T cells in the tumor environment.
Collapse
Affiliation(s)
- Sara Iranparast
- Department of Immunology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran,Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sanaz Tayebi
- Department of Immunology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Ahmadpour
- Department of Biochemistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
13
|
Guo X, Chen G. Hypoxia-Inducible Factor Is Critical for Pathogenesis and Regulation of Immune Cell Functions in Rheumatoid Arthritis. Front Immunol 2020; 11:1668. [PMID: 32849577 PMCID: PMC7399093 DOI: 10.3389/fimmu.2020.01668] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease with characteristics of synovial inflammation, pannus formation, cartilage destruction, and bone erosion. Further, the inflammation is linked to increased oxygen consumption, resulting in hypoxia within the inflammatory area. Hypoxia-inducible factor (HIF) was reported to be associated with adaptation to the hypoxic microenvironment in the RA synovium. Here, we have briefly summarized the structure and expression of HIF. Moreover, the function of HIF in inflammation, angiogenesis, cartilage damage, and immune cells of RA has been discussed.
Collapse
Affiliation(s)
- Xin Guo
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Guangjie Chen
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| |
Collapse
|
14
|
Zhuang X, Pedroza-Pacheco I, Nawroth I, Kliszczak AE, Magri A, Paes W, Rubio CO, Yang H, Ashcroft M, Mole D, Balfe P, Borrow P, McKeating JA. Hypoxic microenvironment shapes HIV-1 replication and latency. Commun Biol 2020; 3:376. [PMID: 32665623 PMCID: PMC7360605 DOI: 10.1038/s42003-020-1103-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 06/17/2020] [Indexed: 12/14/2022] Open
Abstract
Viral replication is defined by the cellular microenvironment and one key factor is local oxygen tension, where hypoxia inducible factors (HIFs) regulate the cellular response to oxygen. Human immunodeficiency virus (HIV) infected cells within secondary lymphoid tissues exist in a low-oxygen or hypoxic environment in vivo. However, the majority of studies on HIV replication and latency are performed under laboratory conditions where HIFs are inactive. We show a role for HIF-2α in restricting HIV transcription via direct binding to the viral promoter. Hypoxia reduced tumor necrosis factor or histone deacetylase inhibitor, Romidepsin, mediated reactivation of HIV and inhibiting HIF signaling-pathways reversed this phenotype. Our data support a model where the low-oxygen environment of the lymph node may suppress HIV replication and promote latency. We identify a mechanism that may contribute to the limited efficacy of latency reversing agents in reactivating HIV and suggest new strategies to control latent HIV-1.
Collapse
Affiliation(s)
- Xiaodong Zhuang
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | | | - Isabel Nawroth
- Institute of Immunity and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Anna E Kliszczak
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Andrea Magri
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Wayne Paes
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | | | - Hongbing Yang
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Margaret Ashcroft
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH, UK
| | - David Mole
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Peter Balfe
- Institute of Immunity and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Persephone Borrow
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Jane A McKeating
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK.
| |
Collapse
|
15
|
Hypoxia and Macrophages Act in Concert Towards a Beneficial Outcome in Colon Cancer. Cancers (Basel) 2020; 12:cancers12040818. [PMID: 32231135 PMCID: PMC7226541 DOI: 10.3390/cancers12040818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 12/21/2022] Open
Abstract
In colon cancer, the prognostic value of macrophages is controversial, and it is still unknown how hypoxia modulates macrophage-cancer cell crosstalk. To unravel this, co-cultures of human primary macrophages and colon cancer cells were performed at 20% and 1% O2, followed by characterization of both cellular components. Different colon cancer patient cohorts were analyzed for hypoxia and immune markers, and their association with patient overall survival was established. A positive correlation between HIF1A and CD68 in colon cancer patients was identified but, unexpectedly, in cases with higher macrophage infiltration, HIF1A expression was associated with a better prognosis, in contrast to breast, gastric, and lung cancers. Under hypoxia, co-cultures' secretome indicated a shift towards a pro-inflammatory phenotype. These alterations occurred along with increased macrophage phagocytic activity and decreased SIRPα expression. Cancer cells were more invasive and exhibited higher CD47 expression. We hypothesize that the better prognosis associated with HIF1AHighCD68High tumors could occur due to macrophagic pro-inflammatory pressure. Indeed, we found that tumors HIF1AHighCD68High expressed increased levels of CD8A, which is positively correlated with HIF1A. In conclusion, we show that in colon cancer, hypoxia drives macrophages into a pro-inflammatory phenotype, concomitant with increased infiltration of anti-tumor immune cells, favoring better disease outcome.
Collapse
|
16
|
Hilchey SP, Palshikar MG, Emo JA, Li D, Garigen J, Wang J, Mendelson ES, Cipolla V, Thakar J, Zand MS. Cyclosporine a directly affects human and mouse b cell migration in vitro by disrupting a hIF-1 αdependent, o 2 sensing, molecular switch. BMC Immunol 2020; 21:13. [PMID: 32183695 PMCID: PMC7079363 DOI: 10.1186/s12865-020-0342-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 02/27/2020] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Hypoxia is a potent molecular signal for cellular metabolism, mitochondrial function, and migration. Conditions of low oxygen tension trigger regulatory cascades mediated via the highly conserved HIF-1 α post-translational modification system. In the adaptive immune response, B cells (Bc) are activated and differentiate under hypoxic conditions within lymph node germinal centers, and subsequently migrate to other compartments. During migration, they traverse through changing oxygen levels, ranging from 1-5% in the lymph node to 5-13% in the peripheral blood. Interestingly, the calcineurin inhibitor cyclosporine A is known to stimulate prolyl hydroxylase activity, resulting in HIF-1 α destabilization and may alter Bc responses directly. Over 60% of patients taking calcineurin immunosuppressant medications have hypo-gammaglobulinemia and poor vaccine responses, putting them at high risk of infection with significantly increased morbidity and mortality. RESULTS We demonstrate that O 2 tension is a previously unrecognized Bc regulatory switch, altering CXCR4 and CXCR5 chemokine receptor signaling in activated Bc through HIF-1 α expression, and controlling critical aspects of Bc migration. Our data demonstrate that calcineurin inhibition hinders this O 2 regulatory switch in primary human Bc. CONCLUSION This previously unrecognized effect of calcineurin inhibition directly on human Bc has significant and direct clinical implications.
Collapse
Affiliation(s)
- Shannon P Hilchey
- University of Rochester Medical CenterDivision of Nephrology, 601 Elmwood Ave., Rochester, 14642 NY USA
| | - Mukta G Palshikar
- University of RochesterBiophysics, Structural, and Computational Biology Program, 601 Elmwood Ave. - Box 675, Rochester, 14642 NY USA
| | - Jason A Emo
- University of Rochester Medical CenterDivision of Nephrology, 601 Elmwood Ave., Rochester, 14642 NY USA
| | - Dongmei Li
- University of RochesterClinical and Translational Science Institute, 265 Crittenden Blvd., Rochester, 14642 NY USA
| | - Jessica Garigen
- University of RochesterClinical and Translational Science Institute, 265 Crittenden Blvd., Rochester, 14642 NY USA
| | - Jiong Wang
- University of Rochester Medical CenterDivision of Nephrology, 601 Elmwood Ave., Rochester, 14642 NY USA
| | - Eric S Mendelson
- University of Rochester Medical CenterDivision of Nephrology, 601 Elmwood Ave., Rochester, 14642 NY USA
| | - Valentina Cipolla
- University of Rochester Medical CenterDivision of Nephrology, 601 Elmwood Ave., Rochester, 14642 NY USA
| | - Juilee Thakar
- University of RochesterDepartment of Microbiology and Immunology, 601 Elmwood Ave - Box 672, Rochester, 14642 NY USA
- University of RochesterDepartment of Biostatistics and Computational Biology, 265 Crittenden Blvd., Rochester, 14642 NY USA
| | - Martin S Zand
- University of Rochester Medical CenterDivision of Nephrology, 601 Elmwood Ave., Rochester, 14642 NY USA
- University of RochesterClinical and Translational Science Institute, 265 Crittenden Blvd., Rochester, 14642 NY USA
| |
Collapse
|
17
|
Hirota K. Basic Biology of Hypoxic Responses Mediated by the Transcription Factor HIFs and its Implication for Medicine. Biomedicines 2020; 8:biomedicines8020032. [PMID: 32069878 PMCID: PMC7168341 DOI: 10.3390/biomedicines8020032] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 02/08/2020] [Accepted: 02/12/2020] [Indexed: 12/19/2022] Open
Abstract
Oxygen (O2) is essential for human life. Molecular oxygen is vital for the production of adenosine triphosphate (ATP) in human cells. O2 deficiency leads to a reduction in the energy levels that are required to maintain biological functions. O2 acts as the final acceptor of electrons during oxidative phosphorylation, a series of ATP synthesis reactions that occur in conjunction with the electron transport system in mitochondria. Persistent O2 deficiency may cause death due to malfunctioning biological processes. The above account summarizes the classic view of oxygen. However, this classic view has been reviewed over the last two decades. Although O2 is essential for life, higher organisms such as mammals are unable to biosynthesize molecular O2 in the body. Because the multiple organs of higher organisms are constantly exposed to the risk of “O2 deficiency,” living organisms have evolved elaborate strategies to respond to hypoxia. In this review, I will describe the system that governs oxygen homeostasis in the living body from the point-of-view of the transcription factor hypoxia-inducible factor (HIF).
Collapse
Affiliation(s)
- Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
18
|
Krzywinska E, Stockmann C. Hypoxia, Metabolism and Immune Cell Function. Biomedicines 2018; 6:E56. [PMID: 29762526 PMCID: PMC6027519 DOI: 10.3390/biomedicines6020056] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a hallmark of inflamed, infected or damaged tissue, and the adaptation to inadequate tissue oxygenation is regulated by hypoxia-inducible factors (HIFs). HIFs are key mediators of the cellular response to hypoxia, but they are also associated with pathological stress such as inflammation, bacteriological infection or cancer. In addition, HIFs are central regulators of many innate and adaptive immunological functions, including migration, antigen presentation, production of cytokines and antimicrobial peptides, phagocytosis as well as cellular metabolic reprogramming. A characteristic feature of immune cells is their ability to infiltrate and operate in tissues with low level of nutrients and oxygen. The objective of this article is to discuss the role of HIFs in the function of innate and adaptive immune cells in hypoxia, with a focus on how hypoxia modulates immunometabolism.
Collapse
Affiliation(s)
- Ewelina Krzywinska
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center, Unit 970, 56 Rue Leblanc, 75015 Paris, France.
| | - Christian Stockmann
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris Cardiovascular Research Center, Unit 970, 56 Rue Leblanc, 75015 Paris, France.
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| |
Collapse
|
19
|
Gropper Y, Feferman T, Shalit T, Salame TM, Porat Z, Shakhar G. Culturing CTLs under Hypoxic Conditions Enhances Their Cytolysis and Improves Their Anti-tumor Function. Cell Rep 2018; 20:2547-2555. [PMID: 28903036 DOI: 10.1016/j.celrep.2017.08.071] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/06/2017] [Accepted: 08/22/2017] [Indexed: 01/14/2023] Open
Abstract
Cytotoxic T lymphocytes (CTLs) used in immunotherapy are typically cultured under atmospheric O2 pressure but encounter hypoxic conditions inside tumors. Activating CTLs under hypoxic conditions has been shown to improve their cytotoxicity in vitro, but the mechanism employed and the implications for immunotherapy remain unknown. We activated and cultured OT-I CD8 T cells at either 1% or 20% O2. Hypoxic CTLs survived, as well as normoxic ones, in vitro but killed OVA-expressing B16 melanoma cells more efficiently. Hypoxic CTLs contained similar numbers of cytolytic granules and released them as efficiently but packaged more granzyme-B in each granule without producing more perforin. We imaged CTL distribution and motility inside B16-OVA tumors using confocal and intravital 2-photon microscopy and observed no obvious differences. However, mice treated with hypoxic CTLs exhibited better tumor regression and survived longer. Thus, hypoxic CTLs may perform better in tumor immunotherapy because of higher intrinsic cytotoxicity rather than improved migration inside tumors.
Collapse
Affiliation(s)
- Yael Gropper
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Tali Feferman
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Tali Shalit
- The Mantoux Bioinformatics Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Tomer-Meir Salame
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Ziv Porat
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Guy Shakhar
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
20
|
Wang JS, Chen YC, Chen WL, Lin CP. Effects of normoxic and hypoxic exercise regimens on lymphocyte apoptosis induced by oxidative stress in sedentary males. Eur J Appl Physiol 2017; 117:2445-2455. [PMID: 28988307 DOI: 10.1007/s00421-017-3731-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 09/28/2017] [Indexed: 02/04/2023]
Abstract
PURPOSE Oxidative stress-induced lymphocyte apoptosis is linked to hypoxemic individuals suffering from cardiopulmonary disorders or exposed to hypoxic environments. What kind of the exercise strategy under hypoxic condition improves exercise performance and simultaneously minimizes lymphocyte dysfunction caused by oxidative stress has not yet been established. This study elucidates how various exercises regimens with/without hypoxia affect lymphocyte apoptosis induced by oxidative stress. METHODS A total of 60 sedentary males were randomly divided into five groups. Each group (n = 12) received one of the five interventions: hypoxic-absolute exercise (HAT, 50%W max under 15%O2), hypoxic-relative exercise (HRT, 50% heart rate reserve under 15%O2), normoxic exercise (NT, 50%W max under 21%O2), hypoxic control (HC, resting under 15%O2), or normoxic control (NC, resting under 21%O2) for 30 min/day, 5 days/week for 4 weeks. RESULTS Before the intervention, the graded exercise test (GXT, progressive exercise up to VO2max) decreased the surface thiol level on lymphocytes and subsequently augmented the extents of H2O2-induced mitochondria transmembrane potential (MTP) diminishing, caspase 3/8/9 activations, and phosphotidyl serine (PS) exposure in lymphocytes. However, 4 weeks of NT, HRT, or HAT reduced the extents of surface thiol decreasing on lymphocytes and H2O2-induced MTP diminishing, caspase 3/8/9 activations, and PS exposure in lymphocytes following GXT. Moreover, the HAT group exhibited greater improvements in pulmonary ventilation and VO2max than either NT or HRT group did. CONCLUSIONS Exercise training with/without hypoxic exposure effectively alleviates lymphocyte apoptosis induced by oxidative stress following strenuous exercise. However, the HAT is superior to the NT or HRT for enhancing aerobic capacity.
Collapse
Affiliation(s)
- Jong-Shyan Wang
- Healthy Aging Research Center, Graduate Institute of Rehabilitation Science, Medical College, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 333, Taiwan. .,Department of Physical Medicine and Rehabilitation, Heart Failure Center, Chang Gung Memorial Hospital, Keelung, Taiwan. .,Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan.
| | - Yi-Ching Chen
- Department of Rehabilitation Science, Jenteh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Wan-Ling Chen
- Healthy Aging Research Center, Graduate Institute of Rehabilitation Science, Medical College, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 333, Taiwan
| | - Chin-Pu Lin
- Healthy Aging Research Center, Graduate Institute of Rehabilitation Science, Medical College, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 333, Taiwan
| |
Collapse
|
21
|
Abstract
There has been dramatic success in treating patients with adoptive transfer of autologous T cells genetically modified to express a chimeric antigen receptor redirecting them to the antigen CD19. Despite this success, the application of chimeric antigen receptor T-cell therapy in solid malignancies has encountered many challenges that need to be overcome if similar success across other cancers is to become a reality. These challenges can be classified into 6 categories: the heterogeneity of tumor cell clones and tumor-associated antigen expression; poor T-cell trafficking into the tumor site; poor T-cell survival and persistence; the presence of suppressive immune cells; the secretion of suppressive soluble factors in the tumor microenvironment; and the upregulation of T-cell intrinsic inhibitory pathways. We outline specific representative hurdles in each of these categories and summarize the progress made in understanding them and developing strategies to overcome them.
Collapse
|
22
|
Andrejeva G, Rathmell JC. Similarities and Distinctions of Cancer and Immune Metabolism in Inflammation and Tumors. Cell Metab 2017; 26:49-70. [PMID: 28683294 PMCID: PMC5555084 DOI: 10.1016/j.cmet.2017.06.004] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/22/2017] [Accepted: 06/09/2017] [Indexed: 12/20/2022]
Abstract
It has been appreciated for nearly 100 years that cancer cells are metabolically distinct from resting tissues. More recently understood is that this metabolic phenotype is not unique to cancer cells but instead reflects characteristics of proliferating cells. Similar metabolic transitions also occur in the immune system as cells transition from resting state to stimulated effectors. A key finding in immune metabolism is that the metabolic programs of different cell subsets are distinctly associated with immunological function. Further, interruption of those metabolic pathways can shift immune cell fate to modulate immunity. These studies have identified numerous metabolic similarities between cancer and immune cells but also critical differences that may be exploited and that affect treatment of cancer and immunological diseases.
Collapse
Affiliation(s)
- Gabriela Andrejeva
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center and Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Jeffrey C Rathmell
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center and Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
23
|
Sukumar M, Kishton RJ, Restifo NP. Metabolic reprograming of anti-tumor immunity. Curr Opin Immunol 2017; 46:14-22. [PMID: 28412583 PMCID: PMC6327315 DOI: 10.1016/j.coi.2017.03.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/11/2017] [Indexed: 12/18/2022]
Abstract
Immunotherapies designed to trigger T cell destruction of tumor cells can result in sustained and complete responses in patients whose cancers were resistant to available treatment options. Evidence suggests that powering the T cell response - how T cells generate energy - plays an important role in their effectiveness. Furthermore the metabolism of T cells can be modulated to improve their anti-cancer activities. In this review, we will discuss the key metabolic properties of anti-cancer T cells, along with potential strategies to enhance immunotherapy through the modulation of T cell metabolism.
Collapse
Affiliation(s)
- Madhusudhanan Sukumar
- Center for Cell-Based Therapy, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Surgery Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Rigel J Kishton
- Center for Cell-Based Therapy, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Surgery Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nicholas P Restifo
- Center for Cell-Based Therapy, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Surgery Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
24
|
Vuillefroy de Silly R, Dietrich PY, Walker PR. Hypoxia and antitumor CD8 + T cells: An incompatible alliance? Oncoimmunology 2016; 5:e1232236. [PMID: 28123871 PMCID: PMC5214994 DOI: 10.1080/2162402x.2016.1232236] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 12/27/2022] Open
Abstract
T Lymphocytes face pathologically low O2 tensions within the tumor bed at which they will have to function in order to impact on the malignancy. Recent studies highlighting the importance of O2 and hypoxia-inducible factors for CD8+ T-cell function and fate must now be integrated into tumor immunology concepts if immunotherapies are to progress. Here, we discuss, reinterpret, and reconcile the many apparent contradictions in these data and we propose that O2 is a master regulator of the CD8+ T-cell response. Certain T cell functions are enhanced, others suppressed, but on balance, hypoxia is globally detrimental to the antitumor response.
Collapse
Affiliation(s)
- Romain Vuillefroy de Silly
- Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Ludwig Center for Cancer Research of the University of Lausanne, Biopôle III, Epalinges, Switzerland
| | | | - Paul R Walker
- Geneva University Hospitals and University of Geneva , Geneva, Switzerland
| |
Collapse
|
25
|
Quiñonez-Flores CM, González-Chávez SA, Pacheco-Tena C. Hypoxia and its implications in rheumatoid arthritis. J Biomed Sci 2016; 23:62. [PMID: 27549205 PMCID: PMC4994473 DOI: 10.1186/s12929-016-0281-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 08/09/2016] [Indexed: 02/07/2023] Open
Abstract
Alterations in tissue oxygen pressure contribute to a number of diseases, including rheumatoid arthritis (RA). Low partial pressure of oxygen, a condition known as hypoxia, is a relevant feature in RA since it is involved in angiogenesis, inflammation, apoptosis, cartilage degradation, energy metabolism, and oxidative damage. Therefore, alterations in hypoxia-related signaling pathways are considered potential mechanisms of disease pathogenesis. The objective of this review is to highlight and update our current knowledge of the role of hypoxia in the pathogenesis of RA. We describe the experimental evidence that RA synovial tissue exists in a hypoxic state, as well as the origin and involvement of synovial hypoxia in different aspects of the pathogenic process.
Collapse
Affiliation(s)
- Celia María Quiñonez-Flores
- Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Circuito No.1, Nuevo Campus Universitario, Chihuahua, C.P. 31240 México
- Facultad de Ciencias de la Cultura Física, Universidad Autónoma de Chihuahua, Circuito No.1, Nuevo Campus Universitario, Chihuahua, C.P. 31240 México
| | - Susana Aideé González-Chávez
- Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Circuito No.1, Nuevo Campus Universitario, Chihuahua, C.P. 31240 México
- Facultad de Ciencias de la Cultura Física, Universidad Autónoma de Chihuahua, Circuito No.1, Nuevo Campus Universitario, Chihuahua, C.P. 31240 México
| | - César Pacheco-Tena
- Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Circuito No.1, Nuevo Campus Universitario, Chihuahua, C.P. 31240 México
| |
Collapse
|
26
|
Aggressiveness Niche: Can It Be the Foster Ground for Cancer Metastasis Precursors? Stem Cells Int 2016; 2016:4829106. [PMID: 27493669 PMCID: PMC4963571 DOI: 10.1155/2016/4829106] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/15/2016] [Indexed: 12/26/2022] Open
Abstract
The relationship between tumor initiation and tumor progression can follow a linear projection in which all tumor cells are equally endowed with the ability to progress into metastasis. Alternatively, not all tumor cells are equal genetically and/or epigenetically, and only few cells are induced to become metastatic tumor cells. The location of these cells within the tumor can also impact the fate of these cells. The most inner core of a tumor where an elevated pressure of adverse conditions forms, such as necrosis-induced inflammation and hypoxia-induced immunosuppressive environment, seems to be the most fertile ground to generate such tumor cells with metastatic potential. Here we will call this necrotic/hypoxic core the “aggressiveness niche” and will present data to support its involvement in generating these metastatic precursors. Within this niche, interaction of hypoxia-surviving cells with the inflammatory microenvironment influenced by newly recruited mesenchymal stromal cells (MSCs), tumor-associated macrophages (TAMs), and other types of cells and the establishment of bidirectional interactions between them elevate the aggressiveness of these tumor cells. Additionally, immune evasion properties induced in these cells most likely contribute in the formation and maintenance of such aggressiveness niche.
Collapse
|
27
|
Chou TF, Chuang YT, Hsieh WC, Chang PY, Liu HY, Mo ST, Hsu TS, Miaw SC, Chen RH, Kimchi A, Lai MZ. Tumour suppressor death-associated protein kinase targets cytoplasmic HIF-1α for Th17 suppression. Nat Commun 2016; 7:11904. [PMID: 27312851 PMCID: PMC4915028 DOI: 10.1038/ncomms11904] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 05/11/2016] [Indexed: 01/01/2023] Open
Abstract
Death-associated protein kinase (DAPK) is a tumour suppressor. Here we show that DAPK also inhibits T helper 17 (Th17) and prevents Th17-mediated pathology in a mouse model of autoimmunity. We demonstrate that DAPK specifically downregulates hypoxia-inducible factor 1α (HIF-1α). In contrast to the predominant nuclear localization of HIF-1α in many cell types, HIF-1α is located in both the cytoplasm and nucleus in T cells, allowing for a cytosolic DAPK–HIF-1α interaction. DAPK also binds prolyl hydroxylase domain protein 2 (PHD2) and increases HIF-1α-PHD2 association. DAPK thereby promotes the proline hydroxylation and proteasome degradation of HIF-1α. Consequently, DAPK deficiency leads to excess HIF-1α accumulation, enhanced IL-17 expression and exacerbated experimental autoimmune encephalomyelitis. Additional knockout of HIF-1α restores the normal differentiation of Dapk−/− Th17 cells and prevents experimental autoimmune encephalomyelitis development. Our results reveal a mechanism involving DAPK-mediated degradation of cytoplasmic HIF-1α, and suggest that raising DAPK levels could be used for treatment of Th17-associated inflammatory diseases. HIF-1α is critical for Th17 differentiation. Here the authors show that DAPK (Death-Associated Protein Kinase) inhibits Th17 differentiation and immunopathology in a mouse model of multiple sclerosis by promoting HIF1-α binding to its negative regulator PHD2.
Collapse
Affiliation(s)
- Ting-Fang Chou
- Graduate Institute of Life Sciences, National Defense Medical College, Taipei 11490, Taiwan.,Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Ya-Ting Chuang
- Department of Medical Research, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Wan-Chen Hsieh
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Pei-Yun Chang
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Hsin-Yu Liu
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Shu-Ting Mo
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Tzu-Sheng Hsu
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Shi-Chuen Miaw
- Institute of Immunology, National Taiwan University, Taipei 10057, Taiwan
| | - Ruey-Hwa Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan.,Institute of Molecular Medicine, National Taiwan University, Taipei 10057, Taiwan
| | - Adi Kimchi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ming-Zong Lai
- Graduate Institute of Life Sciences, National Defense Medical College, Taipei 11490, Taiwan.,Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan.,Institute of Immunology, National Taiwan University, Taipei 10057, Taiwan
| |
Collapse
|
28
|
Hypoxia, mitochondrial dysfunction and synovial invasiveness in rheumatoid arthritis. Nat Rev Rheumatol 2016; 12:385-97. [DOI: 10.1038/nrrheum.2016.69] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Kitsunai H, Makino Y, Sakagami H, Mizumoto K, Yanagimachi T, Atageldiyeva K, Takeda Y, Fujita Y, Abiko A, Takiyama Y, Haneda M. High glucose induces platelet-derived growth factor-C via carbohydrate response element-binding protein in glomerular mesangial cells. Physiol Rep 2016; 4:4/6/e12730. [PMID: 27033449 PMCID: PMC4814887 DOI: 10.14814/phy2.12730] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 02/12/2016] [Indexed: 01/29/2023] Open
Abstract
Persistent high concentration of glucose causes cellular stress and damage in diabetes via derangement of gene expressions. We previously reported high glucose activates hypoxia‐inducible factor‐1α and downstream gene expression in mesangial cells, leading to an extracellular matrix expansion in the glomeruli. A glucose‐responsive transcription factor carbohydrate response element‐binding protein (ChREBP) is a key mediator for such perturbation of gene regulation. To provide insight into glucose‐mediated gene regulation in mesangial cells, we performed chromatin immunoprecipitation followed by DNA microarray analysis and identified platelet‐derived growth factor‐C (PDGF‐C) as a novel target gene of ChREBP. In streptozotocin‐induced diabetic mice, glomerular cells showed a significant increase in PDGF‐C expression; the ratio of PDGF‐C‐positive cells to the total number glomerular cells demonstrated more than threefold increase when compared with control animals. In cultured human mesangial cells, high glucose enhanced expression of PDGF‐C protein by 1.9‐fold. Knock‐down of ChREBP abrogated this induction response. Upregulated PDGF‐C contributed to the production of type IV and type VI collagen, possibly via an autocrine mechanism. Interestingly, urinary PDGF‐C levels in diabetic model mice were significantly elevated in a fashion similar to urinary albumin. Taken together, we hypothesize that a high glucose‐mediated induction of PDGF‐C via ChREBP in mesangial cells contributes to the development of glomerular mesangial expansion in diabetes, which may provide a platform for novel predictive and therapeutic strategies for diabetic nephropathy.
Collapse
Affiliation(s)
- Hiroya Kitsunai
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Yuichi Makino
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Hidemitsu Sakagami
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Katsutoshi Mizumoto
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Tsuyoshi Yanagimachi
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Kuralay Atageldiyeva
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Yasutaka Takeda
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Yukihiro Fujita
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Atsuko Abiko
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Yumi Takiyama
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Masakazu Haneda
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
30
|
Kouidhi S, Noman MZ, Kieda C, Elgaaied AB, Chouaib S. Intrinsic and Tumor Microenvironment-Induced Metabolism Adaptations of T Cells and Impact on Their Differentiation and Function. Front Immunol 2016; 7:114. [PMID: 27066006 PMCID: PMC4810024 DOI: 10.3389/fimmu.2016.00114] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/14/2016] [Indexed: 01/09/2023] Open
Abstract
It is well recognized that the immune system and metabolism are highly integrated. In this context, multilevel interactions between metabolic system and T lymphocyte signaling and fate exist. This review will discuss different potential cell metabolism pathways involved in shaping T lymphocyte function and differentiation. We will also provide a general framework for understanding how tumor microenvironmental metabolism, associated with hypoxic stress, interferes with T-cell priming and expansion. How T-cell metabolism drives T-cell-mediated immunity and how the manipulation of metabolic programing for therapeutic purposes will be also discussed.
Collapse
Affiliation(s)
- Soumaya Kouidhi
- Laboratory BVBGR, LR11ES31, ISBST, Higher Institute of Biotechnology of Sidi Thabet, University of Manouba, Tunis, Tunisia; Laboratory of Genetics, Immunology and Human Pathology, Faculty of Sciences of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Muhammad Zaeem Noman
- Laboratory «Integrative Tumor Immunology and Genetic Oncology» Equipe Labellisée LIGUE 2015, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1186, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Claudine Kieda
- Centre de Biophysique Moléculaire, CNRS UPR 4301 , Orléans , France
| | - Amel Benammar Elgaaied
- Laboratory of Genetics, Immunology and Human Pathology, Faculty of Sciences of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Salem Chouaib
- Laboratory «Integrative Tumor Immunology and Genetic Oncology» Equipe Labellisée LIGUE 2015, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1186, Villejuif, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
31
|
Hirota K. Involvement of hypoxia-inducible factors in the dysregulation of oxygen homeostasis in sepsis. Cardiovasc Hematol Disord Drug Targets 2015; 15:29-40. [PMID: 25567333 PMCID: PMC4435091 DOI: 10.2174/1871529x15666150108115553] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 09/20/2014] [Accepted: 10/10/2014] [Indexed: 12/14/2022]
Abstract
Sepsis is a state of infection with serious systemic manifestations, and if severe enough, can be associated with multiple organ dysfunction and systemic hypotension, which can cause tissues to be hypoxic. Inflammation, as part of the multifaceted biological response to injurious stimuli, such as pathogens or damaged tissues and cells, underlies these biological processes. Prolonged and persistent inflammation, also known as chronic inflammation, results in progressive alteration in the various types of cells at the site of inflammation and is characterized by the simultaneous destruction and healing of tissue during the process. Tissue hypoxia during inflammation is not just a simple bystander process, but can considerably affect the development or attenuation of inflammation by causing the regulation of hypoxia-dependent gene expression. Indeed, the study of transcriptionally regulated tissue adaptation to hypoxia requires intense investigation to help control hypoxia-induced inflammation and organ failure. In this review, I have described the pathophysiology of sepsis with respect to oxygen metabolism and expression of hypoxia-inducible factor 1.
Collapse
Affiliation(s)
- Kiichi Hirota
- Department of Anesthesiology, Kansai Medical University, 2-3-1 Shin-Machi, Hirakata, Osaka 573-1191, Japan.
| |
Collapse
|
32
|
Roth KJ, Copple BL. Role of Hypoxia-Inducible Factors in the Development of Liver Fibrosis. Cell Mol Gastroenterol Hepatol 2015; 1:589-597. [PMID: 28210703 PMCID: PMC5301877 DOI: 10.1016/j.jcmgh.2015.09.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/16/2015] [Indexed: 02/08/2023]
Abstract
Liver fibrosis remains a significant clinical problem in the United States and throughout the world. Although important advances in the understanding of this disease have been made, no effective pharmacologic agents have been developed that directly prevent or reverse the fibrotic process. Many of the successes in liver fibrosis treatment have been targeted toward treating the cause of fibrosis, such as the development of new antivirals that eradicate hepatitis virus. For many patients, however, this is not feasible, so a liver transplant remains the only viable option. Thus, there is a critical need to identify new therapeutic targets that will slow or reverse the progression of fibrosis in such patients. Research over the last 16 years has identified hypoxia-inducible factors (HIFs) as key transcription factors that drive many aspects of liver fibrosis, making them potential targets of therapy. In this review, we discuss the latest work on HIFs and liver fibrosis, including the cell-specific functions of these transcription factors in the development of liver fibrosis.
Collapse
Key Words
- BDL, bile duct ligation
- CCl4, carbon tetrachloride
- Ccr, C-C chemokine receptor
- FGF, fibroblast growth factor
- HGF, hepatocyte growth factor
- HIFs, hypoxia-inducible factors
- HSC, hepatic stellate cell
- Hepatic Stellate Cells
- Hypoxia-Inducible Factors
- Jmjd, Jumonji domain-containing
- Kupffer Cells
- Liver Fibrosis
- PAI-1, plasminogen activator inhibitor-1
- PDGF, platelet-derived growth factor
- Rgs, regulator of G-protein signaling
- TGF-β, transforming growth factor β
- VEGF, vascular endothelial growth factor
- α-SMA, α-smooth muscle actin
Collapse
Affiliation(s)
| | - Bryan L. Copple
- Correspondence Address correspondence to: Bryan L. Copple, PhD, Department of Pharmacology and Toxicology, Michigan State University, 1355 Bogue Street, B403 Life Sciences Building, East Lansing, Michigan 48824.Department of Pharmacology and ToxicologyMichigan State University1355 Bogue Street, B403 Life Sciences BuildingEast LansingMichigan 48824
| |
Collapse
|
33
|
de Aquino MTP, Malhotra A, Mishra MK, Shanker A. Challenges and future perspectives of T cell immunotherapy in cancer. Immunol Lett 2015; 166:117-33. [PMID: 26096822 PMCID: PMC4499494 DOI: 10.1016/j.imlet.2015.05.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 05/10/2015] [Accepted: 05/27/2015] [Indexed: 12/15/2022]
Abstract
Since the formulation of the tumour immunosurveillance theory, considerable focus has been on enhancing the effectiveness of host antitumour immunity, particularly with respect to T cells. A cancer evades or alters the host immune response by various ways to ensure its development and survival. These include modifications of the immune cell metabolism and T cell signalling. An inhibitory cytokine milieu in the tumour microenvironment also leads to immune suppression and tumour progression within a host. This review traces the development in the field and attempts to summarize the hurdles that the approach of adoptive T cell immunotherapy against cancer faces, and discusses the conditions that must be improved to allow effective eradication of cancer.
Collapse
Affiliation(s)
- Maria Teresa P de Aquino
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Anshu Malhotra
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Manoj K Mishra
- Department of Biological Sciences, Alabama State University, Montgomery, AL 36101, USA
| | - Anil Shanker
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; Tumor-Host Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
34
|
Transcription regulates HIF-1α expression in CD4(+) T cells. Immunol Cell Biol 2015; 94:109-13. [PMID: 26150319 DOI: 10.1038/icb.2015.64] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 06/03/2015] [Indexed: 12/11/2022]
Abstract
The transcription factor hypoxia inducible factor-1α (HIF-1α) mediates the metabolic adaptation of cells to hypoxia and T-helper cell fate. However, HIF-1α regulation in CD4(+) T cells (T cells) remains elusive. Here we observed that depletion of oxygen (O2⩽2%) alone was not sufficient to induce HIF-1α expression in T cells. However, when hypoxic T cells were stimulated, HIF-1α was expressed and this was dependent on nuclear factor-κB- and nuclear factor of activated T cell (NFAT)-mediated transcriptional upregulation of Hif-1α mRNA. HIF-1α upregulation could be blocked by drugs inhibiting NF-κB, NFAT or mammalian target of rapamycin precluding CD4(+) T-cell stimulation or translation in T cells, as well as by blocking transcription. CD3, CD28, phorbol-12-myristat-13-acetat (PMA) or ionomycin-stimulated T cells did not express HIF-1α under normoxic conditions. In conclusion, regulation of HIF-1α expression in CD4(+) T cells in hypoxia gravely relies on its transcriptional upregulation and subsequent enhanced protein stabilization.
Collapse
|
35
|
Vuillefroy de Silly R, Ducimetière L, Yacoub Maroun C, Dietrich PY, Derouazi M, Walker PR. Phenotypic switch of CD8(+) T cells reactivated under hypoxia toward IL-10 secreting, poorly proliferative effector cells. Eur J Immunol 2015; 45:2263-75. [PMID: 25929785 PMCID: PMC7163737 DOI: 10.1002/eji.201445284] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 04/13/2015] [Accepted: 04/29/2015] [Indexed: 11/10/2022]
Abstract
CD8(+) T cells controlling pathogens or tumors must function at sites where oxygen tension is frequently low, and never as high as under atmospheric culture conditions. However, T-cell function in vivo is generally analyzed indirectly, or is extrapolated from in vitro studies under nonphysiologic oxygen tensions. In this study, we delineate the role of physiologic and pathologic oxygen tension in vitro during reactivation and differentiation of tumor-specific CD8(+) T cells. Using CD8(+) T cells from pmel-1 mice, we observed that the generation of CTLs under 5% O2, which corresponds to physioxia in lymph nodes, gave rise to a higher effector signature than those generated under atmospheric oxygen fractions (21% O2). Hypoxia (1% O2) did not modify cytotoxicity, but decreasing O2 tensions during CTL and CD8(+) tumor-infiltrating lymphocyte reactivation dose-dependently decreased proliferation, induced secretion of the immunosuppressive cytokine IL-10, and upregulated the expression of CD137 (4-1BB) and CD25. Overall, our data indicate that oxygen tension is a key regulator of CD8(+) T-cell function and fate and suggest that IL-10 release may be an unanticipated component of CD8(+) T cell-mediated immune responses in most in vivo microenvironments.
Collapse
Affiliation(s)
| | - Laura Ducimetière
- Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | | | | | - Madiha Derouazi
- Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Paul R Walker
- Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| |
Collapse
|
36
|
Liu C, Chapman NM, Karmaus PWF, Zeng H, Chi H. mTOR and metabolic regulation of conventional and regulatory T cells. J Leukoc Biol 2015; 97:837-847. [PMID: 25714803 DOI: 10.1189/jlb.2ri0814-408r] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 12/12/2022] Open
Abstract
mTOR signaling links bioenergetic and biosynthetic metabolism to immune responses. mTOR is activated by diverse upstream stimuli, including immune signals, growth factors, and nutrients. Recent studies highlight crucial roles of mTOR signaling in immune functions mediated by conventional T cells and Tregs In this review, we discuss the regulation of mTOR signaling in T cells and the functional impacts of mTOR and metabolic pathways on T cell-mediated immune responses, with a particular focus on the differentiation and function of Tregs.
Collapse
Affiliation(s)
- Chaohong Liu
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Nicole M Chapman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Peer W F Karmaus
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Hu Zeng
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
37
|
Hsiao HW, Hsu TS, Liu WH, Hsieh WC, Chou TF, Wu YJ, Jiang ST, Lai MZ. Deltex1 antagonizes HIF-1α and sustains the stability of regulatory T cells in vivo. Nat Commun 2015; 6:6353. [PMID: 25695215 PMCID: PMC4346631 DOI: 10.1038/ncomms7353] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 01/22/2015] [Indexed: 12/21/2022] Open
Abstract
Application of regulatory T cells (Tregs) in transplantation, autoimmunity and allergy has been extensively explored, but how Foxp3 and Treg stability is regulated in vivo is incompletely understood. Here, we identify a requirement for Deltex1 (DTX1), a contributor to T-cell anergy and Foxp3 protein level maintenance in vivo. Dtx1(-/-) Tregs are as effective as WT Tregs in the inhibition of CD4(+)CD25(-) T-cell activation in vitro. However, the suppressive ability of Dtx1(-/-) Tregs is greatly impaired in vivo. We find that Foxp3 expression is diminished when Dtx1(-/-) Tregs are co-transferred with effector T cells in vivo. DTX1 promotes the degradation of HIF-1α. Knockout of HIF-1α restores the Foxp3 stability and rescues the defective suppressive activity in Dtx1(-/-) Treg cells in vivo. Our results suggest that DTX1 exerts another level of control on Treg stability in vivo by sustaining the expression of Foxp3 protein in Tregs.
Collapse
Affiliation(s)
- Huey-Wen Hsiao
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
| | - Tzu-Sheng Hsu
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
- Graduate Institute of Immunology, National Taiwan University, Taipei 10057, Taiwan, R.O.C
| | - Wen-Hsien Liu
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
| | - Wan-Chen Hsieh
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
| | - Ting-Fang Chou
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
| | - Yu-Jung Wu
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
- Graduate Institute of Immunology, National Taiwan University, Taipei 10057, Taiwan, R.O.C
| | - Si-Tse Jiang
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan 74147, Taiwan, R.O.C
| | - Ming-Zong Lai
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
- Graduate Institute of Immunology, National Taiwan University, Taipei 10057, Taiwan, R.O.C
| |
Collapse
|
38
|
Mockler MB, Conroy MJ, Lysaght J. Targeting T cell immunometabolism for cancer immunotherapy; understanding the impact of the tumor microenvironment. Front Oncol 2014; 4:107. [PMID: 24904823 PMCID: PMC4032940 DOI: 10.3389/fonc.2014.00107] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 04/28/2014] [Indexed: 12/20/2022] Open
Abstract
The immune system has a key role to play in controlling cancer initiation and progression. T cell activation, which is central to anti-tumor immune responses, coincides with changes in cellular metabolism. Naïve T cells predominantly require an ATP generating metabolic profile, whereas proliferating effector T cells require anabolic metabolic profiles that promote rapid growth and proliferation. Furthermore, specific T cell subsets require distinct energetic and biosynthetic pathways to match their functional requirements. The often hostile tumor microenvironment can affect T cell immune responses by altering the resulting cellular metabolism. Tailoring immune responses by manipulating cellular metabolic pathways may provide an exciting new option for cancer immunotherapy. T cell responses might also be skewed via metabolic manipulation to treat the complications of obesity-associated inflammation, which is a rapidly growing global health problem and a major risk factor for many malignancies. In this review, the diverse metabolic requirements of T cells in anti-tumor immunity are discussed, as well as the profound influence of the tumor microenvironment and the possible avenues for manipulation to enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Mary B Mockler
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin , Dublin , Ireland
| | - Melissa J Conroy
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin , Dublin , Ireland
| | - Joanne Lysaght
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin , Dublin , Ireland
| |
Collapse
|
39
|
|
40
|
Tajdini M, Mirbagheri SA, Nikooie R, Ostovaneh MR, Ghoreyshi Hefzabad SM, Garg SK, Hosseini SMR. Tissue hypoxia in pathogenesis of ulcerative colitis: should we change all our beliefs? Scand J Gastroenterol 2013; 48:1487-8. [PMID: 24134784 DOI: 10.3109/00365521.2013.845798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Masih Tajdini
- Department of Gastroenterology, Amira'lam Hospital, Faculty of Medicine, Tehran University of Medical Science , Tehran , Iran
| | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW Activation of the immune system only occurs when stimulated cells generate sufficient energy to support their growth and proliferation. Moreover, efficient HIV-1 infection requires that CD4(+) T cells meet the energy demands involved in completing the different steps of the virus life cycle. In this review, we highlight recent studies revealing the importance of nutrient fuels, nucleotide metabolism and the oxygen microenvironment in regulating HIV-1 infection, T-cell differentiation and the generation of HIV-1-specific immune responses. RECENT FINDINGS Glucose uptake via the Glut1 glucose transporter is required for efficient HIV-1 infection of CD4(+) lymphocytes. Other nutrients can also be used as sources of energy and their utilization conditions the differentiation of CD4(+) T cells to distinct effector fates. The conversion of ATP to adenosine inhibits HIV-specific effector cells and the hydrolysis of dNTPs by SAMHD1 restricts infection. Furthermore, oxygen concentration modulates metabolic status, thereby altering T-cell differentiation and potential to mediate a specific immune response. SUMMARY The availability and use of energy resources in fluctuating environments regulate T-cell function and susceptibility to HIV-1 infection. Identification of the targets coordinating the selected metabolic pathways will advance new strategic avenues for controlling HIV-1 disease progression.
Collapse
|
42
|
Abstract
A current view of the inflammatory bowel diseases (IBDs) includes the luminal triggering of innate immune disease in a genetically susceptible host. Given the unique anatomy and complex environment of the intestine, local microenvironmental cues likely contribute significantly to both disease progression and resolution in IBD. Compartmentalized tissue and microbe populations within the intestine result in significant metabolic shifts within these tissue microenvironments. During active inflammatory disease, metabolic demands often exceed supply, resulting in localized areas of metabolic stress and diminished oxygen delivery (hypoxia). There is much recent interest in harnessing these microenvironmental changes to the benefit of the tissue, including targeting these pathways for therapy of IBD. Here, we review the current understanding of metabolic microenvironments within the intestine in IBD, with discussion of the advantages and disadvantages of targeting these pathways to treat patients with IBD.
Collapse
|
43
|
McNamee EN, Korns Johnson D, Homann D, Clambey ET. Hypoxia and hypoxia-inducible factors as regulators of T cell development, differentiation, and function. Immunol Res 2013; 55:58-70. [PMID: 22961658 DOI: 10.1007/s12026-012-8349-8] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oxygen is a molecule that is central to cellular respiration and viability, yet there are multiple physiologic and pathological contexts in which cells experience conditions of insufficient oxygen availability, a state known as hypoxia. Given the metabolic challenges of a low oxygen environment, hypoxia elicits a range of adaptive responses at the cellular, tissue, and systemic level to promote continued survival and function. Within this context, T lymphocytes are a highly migratory cell type of the adaptive immune system that frequently encounters a wide range of oxygen tensions in both health and disease. It is now clear that oxygen availability regulates T cell differentiation and function, a response orchestrated in large part by the hypoxia-inducible factor transcription factors. Here, we discuss the physiologic scope of hypoxia and hypoxic signaling, the contribution of these pathways in regulating T cell biology, and current gaps in our understanding. Finally, we discuss how emerging therapies that modulate the hypoxic response may offer new modalities to alter T cell function and the outcome of acute and chronic pathologies.
Collapse
Affiliation(s)
- Eóin N McNamee
- Mucosal Inflammation Program, Department of Anesthesiology, School of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | |
Collapse
|
44
|
Rullé S, Ah Kioon MD, Asensio C, Mussard J, Ea HK, Boissier MC, Lioté F, Falgarone G. Adrenomedullin, a neuropeptide with immunoregulatory properties induces semi-mature tolerogenic dendritic cells. Immunology 2012; 136:252-64. [PMID: 22348691 DOI: 10.1111/j.1365-2567.2012.03577.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DC) play a pivotal role in tolerance. Adrenomedullin (AM), a neuropeptide with anti-apoptotic and anti-inflammatory effects, may decrease T helper type 1 effector cells and induce regulatory T (Treg) cells. The aim of this study was to evaluate AM effects on murine dendritic cell (DC) maturation and functions. Bone marrow-derived DC were produced and stimulated with CpG motifs, lipopolysaccharide or AM for 24 hr. Then, DC maturation and expression of AM and AM receptors were evaluated. Compared with lipopolysaccharide-stimulated or CpG-stimulated DC, AM-stimulated DC had lower levels of co-stimulatory molecule expression and pro-inflammatory cytokine release. The AM induced high levels of interferon-γ but not of interleukin-10. Importantly, AM inhibited lipopolysaccharide-induced maturation of DC. However, allogeneic T-cell stimulation and endocytic capacity of AM-stimulated DC were comparable to those of semi-mature and mature DC. Moreover, DC expressed AM and its receptors at a basal level, and AM receptor expression increased with DC maturation. The AM stimulation induced indoleamine 2,3-dioxygenase (IDO) expression, promoting Treg cell expansion. For the first time, we describe the DC maturation phenotype by a neuropeptide (AM). We have demonstrated that AM and its receptors are expressed in DC and that exogenous AM can modify the DC phenotype and functions and can induce a semi-mature DC phenotype with IDO expression. These results indicate close interactions among immune system regulation mechanisms and calcitonin-like peptides.
Collapse
Affiliation(s)
- Sandrine Rullé
- Sorbonne Paris Cité, Université Paris 13, Bobigny, France
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Liverani E, McLeod JD, Paul C. Adrenomedullin receptors on human T cells are glucocorticoid-sensitive. Int Immunopharmacol 2012; 14:75-81. [PMID: 22732685 DOI: 10.1016/j.intimp.2012.06.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 05/15/2012] [Accepted: 06/12/2012] [Indexed: 12/12/2022]
Abstract
Adrenomedullin (AM) is a novel vasodilatatory peptide which acts primarily through the calcitonin receptor-like receptor (CLR) in combination with either receptor-activity-modifying-protein (RAMP) 2 or 3 (forming receptors, AM(1) and AM(2) respectively). AM plays an important role during inflammation, with its expression increasing following cytokine treatment, promoting macrophage action in situ and high expression by T cells during hypoxic conditions. Examination of T cell AM receptor expression has previously been incomplete, hence we here consider the presentation of AM receptors and their responsiveness to AM and glucocorticoids (GC). AM receptor expression was examined by PCR and flow cytometry in primary human T cells, revealing that RAMP2, 3 and CLR are physiologically expressed in unstimulated T cells, both intracellularly and on the cell surface. PHA stimulation decreased receptor proteins, significantly so for CLR and RAMP3. Incubation with AM elicited limited receptor alterations however, GC treatment (10(-6) M; 24 h) markedly affected cell surface expression, significantly increasing receptor components in unstimulated cells and significantly decreasing the same in stimulated T cells. Our findings indicate that human T cells utilize both AM(1) and AM(2) receptors, which are GC-sensitive in an activation-state dependent manner.
Collapse
Affiliation(s)
- Elisabetta Liverani
- Faculty of Health and Life Sciences, Centre for Research in Biosciences, University of the West of England, Frenchay, Bristol, UK.
| | | | | |
Collapse
|
46
|
Bader HL, Hsu T. Systemic VHL gene functions and the VHL disease. FEBS Lett 2012; 586:1562-9. [PMID: 22673568 DOI: 10.1016/j.febslet.2012.04.032] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 04/16/2012] [Accepted: 04/17/2012] [Indexed: 12/19/2022]
Abstract
The von Hippel-Lindau tumor suppressor gene (VHL) is best known as an E3 ubiquitin ligase that negatively regulates the hypoxia inducible factor (HIF). VHL mutations are the genetic defects underlying several human diseases including polycythemia, familial VHL tumor syndrome and sporadic renal cell carcinoma. VHL mutations can lead to cell-autonomous phenotypes in the tumor cells. However, non-tumor cell-autonomous functions of VHL have also been noted. VHL tumor-derived cytokines can promote inflammation and induce mobilization of endothelial progenitor cells. Up-regulation of HIF caused by VHL loss-of-function mutants, including heterozygotes, has been shown to increase the activities of hematopoietic stem cells, endothelial cells and myeloid cells. As such, systemic functions of VHL likely play important roles in the development of VHL disease.
Collapse
Affiliation(s)
- Hannah L Bader
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | | |
Collapse
|
47
|
Hypoxia--a key regulator of angiogenesis and inflammation in rheumatoid arthritis. Nat Rev Rheumatol 2012; 8:153-62. [PMID: 22293762 DOI: 10.1038/nrrheum.2011.205] [Citation(s) in RCA: 237] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The importance of inflammation in rheumatoid arthritis (RA) is well understood. This knowledge has resulted in the development of anti-inflammatory therapies--either broadly acting (such as steroids) or more specific approaches (such as antibodies against TNF)--with biologic therapies (including TNF inhibitors) revolutionizing the treatment of RA. However, what is less well appreciated in RA are the links between inflammation, blood-vessel formation (angiogenesis) and cellular responses to changes in oxygen tension. Inadequate oxygenation, termed hypoxia, is thought to drive the increase in synovial angiogenesis that occurs in RA, through expression of hypoxia-inducible molecules, including vascular endothelial growth factor (VEGF). This process promotes further infiltration of inflammatory cells and production of inflammatory mediators, perpetuating synovitis. This Review highlights the molecular pathways activated by hypoxia, and how these pathways might interact with inflammatory signaling to promote and maintain synovitis in RA, with a particular focus on the response of macrophages to hypoxia in the context of RA. Successful treatment of RA, for example with anti-TNF antibodies, reduces levels of proangiogenic factors, including VEGF, and leads to normalization of the vasculature. These processes emphasise the close links between hypoxia, angiogenesis and inflammation in this disease and supports the concept that angiogenesis blockade could be of therapeutic benefit in RA.
Collapse
|
48
|
Abstract
Cell cycle entry is commonly considered to positively regulate HIV-1 infection of CD4 T cells, raising the question as to how quiescent lymphocytes, representing a large portion of the viral reservoir, are infected in vivo. Factors such as the homeostatic cytokine IL-7 have been shown to render quiescent T cells permissive to HIV-1 infection, presumably by transiently stimulating their entry into the cell cycle. However, we show here that at physiological oxygen (O(2)) levels (2-5% O(2) tension in lymphoid organs), IL-7 stimulation generates an environment permissive to HIV-1 infection, despite a significantly attenuated level of cell cycle entry. We identify the IL-7-induced increase in Glut1 expression, resulting in augmented glucose uptake, as a key factor in rendering these T lymphocytes susceptible to HIV-1 infection. HIV-1 infection of human T cells is abrogated either by impairment of Glut1 signal transduction or by siRNA-mediated Glut1 down-regulation. Consistent with this, we show that the susceptibility of human thymocyte subsets to HIV-1 infection correlates with Glut1 expression; single-round infection is markedly higher in the Glut1-expressing double-positive thymocyte population than in any of the Glut1-negative subsets. Thus, our studies reveal the Glut1-mediated metabolic pathway as a critical regulator of HIV-1 infection in human CD4 T cells and thymocytes.
Collapse
|
49
|
When Cells Suffocate: Autophagy in Cancer and Immune Cells under Low Oxygen. Int J Cell Biol 2011; 2011:470597. [PMID: 22190938 PMCID: PMC3235465 DOI: 10.1155/2011/470597] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 08/14/2011] [Indexed: 11/18/2022] Open
Abstract
Hypoxia is a signature feature of growing tumors. This cellular state creates an inhospitable condition that impedes the growth and function of all cells within the immediate and surrounding tumor microenvironment. To adapt to hypoxia, cells activate autophagy and undergo a metabolic shift increasing the cellular dependency on anaerobic metabolism. Autophagy upregulation in cancer cells liberates nutrients, decreases the buildup of reactive oxygen species, and aids in the clearance of misfolded proteins. Together, these features impart a survival advantage for cancer cells in the tumor microenvironment. This observation has led to intense research efforts focused on developing autophagy-modulating drugs for cancer patient treatment. However, other cells that infiltrate the tumor environment such as immune cells also encounter hypoxia likely resulting in hypoxia-induced autophagy. In light of the fact that autophagy is crucial for immune cell proliferation as well as their effector functions such as antigen presentation and T cell-mediated killing of tumor cells, anticancer treatment strategies based on autophagy modulation will need to consider the impact of autophagy on the immune system.
Collapse
|
50
|
Colgan SP, Eltzschig HK. Adenosine and hypoxia-inducible factor signaling in intestinal injury and recovery. Annu Rev Physiol 2011; 74:153-75. [PMID: 21942704 DOI: 10.1146/annurev-physiol-020911-153230] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The gastrointestinal mucosa has proven to be an interesting tissue in which to investigate disease-related metabolism. In this review, we outline some of the evidence that implicates hypoxia-mediated adenosine signaling as an important signature within both healthy and diseased mucosa. Studies derived from cultured cell systems, animal models, and human patients have revealed that hypoxia is a significant component of the inflammatory microenvironment. These studies have revealed a prominent role for hypoxia-induced factor (HIF) and hypoxia signaling at several steps along the adenine nucleotide metabolism and adenosine receptor signaling pathways. Likewise, studies to date in animal models of intestinal inflammation have demonstrated an almost uniformly beneficial influence of HIF stabilization on disease outcomes. Ongoing studies to define potential similarities with and differences between innate and adaptive immune responses will continue to teach us important lessons about the complexity of the gastrointestinal tract. Such information has provided new insights into disease pathogenesis and, importantly, will provide insights into new therapeutic targets.
Collapse
Affiliation(s)
- Sean P Colgan
- Departments of Medicine and Anesthesiology and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045, USA.
| | | |
Collapse
|