151
|
Horev Y, Salameh R, Nassar M, Capucha T, Saba Y, Barel O, Zubeidat K, Matanes D, Leibovich A, Heyman O, Eli-Berchoer L, Hanhan S, Betser-Cohen G, Shapiro H, Elinav E, Bercovier H, Wilensky A, Hovav AH. Niche rather than origin dysregulates mucosal Langerhans cells development in aged mice. Mucosal Immunol 2020; 13:767-776. [PMID: 32457449 DOI: 10.1038/s41385-020-0301-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/02/2020] [Accepted: 04/19/2020] [Indexed: 02/04/2023]
Abstract
Unlike epidermal Langerhans cells (LCs) that originate from embryonic precursors and are self-renewed locally, mucosal LCs arise and are replaced by circulating bone marrow (BM) precursors throughout life. While the unique lifecycle of epidermal LCs is associated with an age-dependent decrease in their numbers, whether and how aging has an impact on mucosal LCs remains unclear. Focusing on gingival LCs we found that mucosal LCs are reduced with age but exhibit altered morphology with that observed in aged epidermal LCs. The reduction of gingival but not epidermal LCs in aged mice was microbiota-dependent; nevertheless, the impact of the microbiota on gingival LCs was indirect. We next compared the ability of young and aged BM precursors to differentiate to mucosal LCs. Mixed BM chimeras, as well as differentiation cultures, demonstrated that aged BM has intact if not superior capacity to differentiate into LCs than young BM. This was in line with the higher percentages of mucosal LC precursors, pre-DCs, and monocytes, detected in aged BM. These findings suggest that while aging is associated with reduced LC numbers, the niche rather than the origin controls this process in mucosal barriers.
Collapse
Affiliation(s)
- Yael Horev
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.,Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Rana Salameh
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Maria Nassar
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Tal Capucha
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Yasmin Saba
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Or Barel
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Khaled Zubeidat
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Daniela Matanes
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Amit Leibovich
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Oded Heyman
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Luba Eli-Berchoer
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Salem Hanhan
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Gili Betser-Cohen
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Hagit Shapiro
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Herve Bercovier
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Asaf Wilensky
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel.
| | - Avi-Hai Hovav
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.
| |
Collapse
|
152
|
Renga G, Bellet MM, Pariano M, Gargaro M, Stincardini C, D'Onofrio F, Mosci P, Brancorsini S, Bartoli A, Goldstein AL, Garaci E, Romani L, Costantini C. Thymosin α1 protects from CTLA-4 intestinal immunopathology. Life Sci Alliance 2020; 3:3/10/e202000662. [PMID: 32817121 PMCID: PMC7441522 DOI: 10.26508/lsa.202000662] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 12/21/2022] Open
Abstract
This study demonstrates that Tα1 protects mice from anti–CTLA-4–induced colitis and sustains its antitumor activity, thus suggesting that Tα1 may be used in combination protocols. The advent of immune checkpoint inhibitors has represented a major boost in cancer therapy, but safety concerns are increasingly being recognized. Indeed, although beneficial at the tumor site, unlocking a safeguard mechanism of the immune response may trigger autoimmune-like effects at the periphery, thus making the safety of immune checkpoint inhibitors a research priority. Herein, we demonstrate that thymosin α1 (Tα1), an endogenous peptide with immunomodulatory activities, can protect mice from intestinal toxicity in a murine model of immune checkpoint inhibitor–induced colitis. Specifically, Tα1 efficiently prevented immune adverse pathology in the gut by promoting the indoleamine 2,3-dioxygenase (IDO) 1–dependent tolerogenic immune pathway. Notably, Tα1 did not induce IDO1 in the tumor microenvironment, but rather modulated the infiltration of T-cell subsets by inverting the ratio between CD8+ and Treg cells, an effect that may depend on Tα1 ability to regulate the differentiation and chemokine expression profile of DCs. Thus, through distinct mechanisms that are contingent upon the context, Tα1 represents a plausible candidate to improve the safety/efficacy profile of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marco Gargaro
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Fiorella D'Onofrio
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Paolo Mosci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Andrea Bartoli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Allan L Goldstein
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Enrico Garaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, Rome, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
153
|
Carpenter RS, Marbourg JM, Brennan FH, Mifflin KA, Hall JCE, Jiang RR, Mo XM, Karunasiri M, Burke MH, Dorrance AM, Popovich PG. Spinal cord injury causes chronic bone marrow failure. Nat Commun 2020; 11:3702. [PMID: 32710081 PMCID: PMC7382469 DOI: 10.1038/s41467-020-17564-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 07/01/2020] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) causes immune dysfunction, increasing the risk of infectious morbidity and mortality. Since bone marrow hematopoiesis is essential for proper immune function, we hypothesize that SCI disrupts bone marrow hematopoiesis. Indeed, SCI causes excessive proliferation of bone marrow hematopoietic stem and progenitor cells (HSPC), but these cells cannot leave the bone marrow, even after challenging the host with a potent inflammatory stimulus. Sequestration of HSPCs in bone marrow after SCI is linked to aberrant chemotactic signaling that can be reversed by post-injury injections of Plerixafor (AMD3100), a small molecule inhibitor of CXCR4. Even though Plerixafor liberates HSPCs and mature immune cells from bone marrow, competitive repopulation assays show that the intrinsic long-term functional capacity of HSPCs is still impaired in SCI mice. Together, our data suggest that SCI causes an acquired bone marrow failure syndrome that may contribute to chronic immune dysfunction.
Collapse
Affiliation(s)
- Randall S Carpenter
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Jessica M Marbourg
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Faith H Brennan
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Katherine A Mifflin
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Jodie C E Hall
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Roselyn R Jiang
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Xiaokui M Mo
- Center for Biostatistics and Bioinformatics, The Ohio State University, Columbus, OH, USA
| | - Malith Karunasiri
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Matthew H Burke
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Adrienne M Dorrance
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA.
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
154
|
Seyfried AN, Maloney JM, MacNamara KC. Macrophages Orchestrate Hematopoietic Programs and Regulate HSC Function During Inflammatory Stress. Front Immunol 2020; 11:1499. [PMID: 32849512 PMCID: PMC7396643 DOI: 10.3389/fimmu.2020.01499] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
The bone marrow contains distinct cell types that work in coordination to generate blood and immune cells, and it is the primary residence of hematopoietic stem cells (HSCs) and more committed multipotent progenitors (MPPs). Even at homeostasis the bone marrow is a dynamic environment where billions of cells are generated daily to replenish short-lived immune cells and produce the blood factors and cells essential for hemostasis and oxygenation. In response to injury or infection, the marrow rapidly adapts to produce specific cell types that are in high demand revealing key insight to the inflammatory nature of "demand-adapted" hematopoiesis. Here we focus on the role that resident and monocyte-derived macrophages play in driving these hematopoietic programs and how macrophages impact HSCs and downstream MPPs. Macrophages are exquisite sensors of inflammation and possess the capacity to adapt to the environment, both promoting and restraining inflammation. Thus, macrophages hold great potential for manipulating hematopoietic output and as potential therapeutic targets in a variety of disease states where macrophage dysfunction contributes to or is necessary for disease. We highlight essential features of bone marrow macrophages and discuss open questions regarding macrophage function, their role in orchestrating demand-adapted hematopoiesis, and mechanisms whereby they regulate HSC function.
Collapse
Affiliation(s)
- Allison N Seyfried
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Jackson M Maloney
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Katherine C MacNamara
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| |
Collapse
|
155
|
Mitroulis I, Kalafati L, Bornhäuser M, Hajishengallis G, Chavakis T. Regulation of the Bone Marrow Niche by Inflammation. Front Immunol 2020; 11:1540. [PMID: 32849521 PMCID: PMC7396603 DOI: 10.3389/fimmu.2020.01540] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 06/11/2020] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem cells (HSC) reside in the bone marrow (BM) within a specialized micro-environment, the HSC niche, which comprises several cellular constituents. These include cells of mesenchymal origin, endothelial cells and HSC progeny, such as megakaryocytes and macrophages. The BM niche and its cell populations ensure the functional preservation of HSCs. During infection or systemic inflammation, HSCs adapt to and respond directly to inflammatory stimuli, such as pathogen-derived signals and elicited cytokines, in a process termed emergency myelopoiesis, which includes HSC activation, expansion, and enhanced myeloid differentiation. The cell populations of the niche participate in the regulation of emergency myelopoiesis, in part through secretion of paracrine factors in response to pro-inflammatory stimuli, thereby indirectly affecting HSC function. Here, we review the crosstalk between HSCs and cell populations in the BM niche, specifically focusing on the adaptation of the HSC niche to inflammation and how this inflammatory adaptation may, in turn, regulate emergency myelopoiesis.
Collapse
Affiliation(s)
- Ioannis Mitroulis
- First Department of Internal Medicine, Department of Haematology and Laboratory of Molecular Hematology, Democritus University of Thrace, Alexandroupolis, Greece.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lydia Kalafati
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
| | - Martin Bornhäuser
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
| | - George Hajishengallis
- Laboratory of Innate Immunity and Inflammation, Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
| |
Collapse
|
156
|
Piras F, Kajaste-Rudnitski A. Antiviral immunity and nucleic acid sensing in haematopoietic stem cell gene engineering. Gene Ther 2020; 28:16-28. [PMID: 32661282 PMCID: PMC7357672 DOI: 10.1038/s41434-020-0175-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023]
Abstract
The low gene manipulation efficiency of human hematopoietic stem and progenitor cells (HSPC) remains a major hurdle for sustainable and broad clinical application of innovative therapies for a wide range of disorders. Given that all current and emerging gene transfer and editing technologies are bound to expose HSPC to exogenous nucleic acids and most often also to viral vectors, we reason that host antiviral factors and nucleic acid sensors play a pivotal role in the efficacy of HSPC genetic manipulation. Here, we review recent progress in our understanding of vector–host interactions and innate immunity in HSPC upon gene engineering and discuss how dissecting this crosstalk can guide the development of more stealth and efficient gene therapy approaches in the future.
Collapse
Affiliation(s)
- Francesco Piras
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
157
|
Abstract
PURPOSE OF REVIEW The innate immune system is essential in the protection against microbial infection and facilitating tissue repair mechanisms. During these stresses, the maintenance of innate immune cell numbers through stress-induced or emergency hematopoiesis is key for our survival. One major mechanism to recognize danger signals is through the activation of Toll-like receptors (TLRs) on the surface of hematopoietic cells, including hematopoietic stem cell (HSC) and hematopoietic progenitor cell (HPC), and nonhematopoietic cells, which recognize pathogen-derived or damaged-induced compounds and can influence the emergency hematopoietic response. This review explores how direct pathogen-sensing by HSC/HPC regulates hematopoiesis, and the positive and negative consequences of these signals. RECENT FINDINGS Recent studies have highlighted new roles for TLRs in regulating HSC and HPC differentiation to innate immune cells of both myeloid and lymphoid origin and augmenting HSC and HPC migration capabilities. Most interestingly, new insights as to how acute versus chronic stimulation of TLR signaling regulates HSC and HPC function has been explored. SUMMARY Recent evidence suggests that TLRs may play an important role in many inflammation-associated diseases. This suggests a possible use for TLR agonists or antagonists as potential therapeutics. Understanding the direct effects of TLR signaling by HSC and HPC may help regulate inflammatory/danger signal-driven emergency hematopoiesis.
Collapse
|
158
|
Yang X, Chen D, Long H, Zhu B. The mechanisms of pathological extramedullary hematopoiesis in diseases. Cell Mol Life Sci 2020; 77:2723-2738. [PMID: 31974657 PMCID: PMC11104806 DOI: 10.1007/s00018-020-03450-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/24/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023]
Abstract
Extramedullary hematopoiesis (EMH) is the expansion and differentiation of hematopoietic stem and progenitor cells outside of the bone marrow. In postnatal life, as a compensatory mechanism for ineffective hematopoiesis of the bone marrow, pathological EMH is triggered by hematopoietic disorders, insufficient hematopoietic compensation, and other pathological stress conditions, such as infection, advanced tumors, anemia, and metabolic stress. Pathological EMH has been reported in many organs, and the sites of pathological EMH may be related to reactivation of the embryonic hematopoietic structure in these organs. As a double-edged sword (blood and immune cell supplementation as well as clinical complications), pathological EMH has been widely studied in recent years. In particular, pathological EMH induced by late-stage tumors contributes to tumor immunosuppression. Thus, a deeper understanding of the mechanism of pathological EMH may be conducive to the development of therapies against the pathological processes that induce EMH. This article reviews the recent progress of research on the cellular and molecular mechanisms of pathological EMH in specific diseases.
Collapse
Affiliation(s)
- Xinxin Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Degao Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
159
|
Perez de Acha O, Rossi M, Gorospe M. Circular RNAs in Blood Malignancies. Front Mol Biosci 2020; 7:109. [PMID: 32676504 PMCID: PMC7333357 DOI: 10.3389/fmolb.2020.00109] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/08/2020] [Indexed: 12/30/2022] Open
Abstract
Circular (circ)RNAs influence a wide range of biological processes at least in part by interacting with proteins and microRNAs. CircRNAs expressed in the hematopoietic compartment have been increasingly recognized as modulators of physiological and pathological features of hematopoetic stem cell (HSC)-derived populations. In particular, several circRNAs were found to enhance or suppress tumor progression in blood malignancies such as leukemias and lymphomas. Moreover, numerous circRNAs have been proposed to help confer resistance to the conventional treatments used in hematopoietic cancers. Here, we review the most important circRNAs described thus far in acute myeloid leukemia (AML), chronic myeloid leukemia (CML), acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), lymphomas, and multiple myeloma (MM). We discuss the usefulness of circRNAs as diagnostic and prognostic markers and their potential value as therapeutic targets.
Collapse
Affiliation(s)
- Olivia Perez de Acha
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Martina Rossi
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
160
|
Charles-Messance H, Sheedy FJ. Train to Lose: Innate Immune Memory in Metaflammation. Mol Nutr Food Res 2020; 65:e1900480. [PMID: 32529783 DOI: 10.1002/mnfr.201900480] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 06/02/2020] [Indexed: 01/21/2023]
Abstract
Westernized diets and lifestyle are linked to the development of metabolic syndrome, characterized by obesity, type 2 diabetes, and increased cardiovascular disease risk. Systemic low-grade inflammation is a common feature of chronic metabolic disorders and is believed to promote disease progression. Therefore, modulating inflammation is a commonly explored strategy to prevent obesity-associated co-morbidities. In this review, how current knowledge on the recently described concept of innate immune memory could underline metaflammation in the context of metabolic syndrome is explored. It is hoped that these insights provide a new perspective to address the question of innate immune activation during disease progression.
Collapse
Affiliation(s)
- Hugo Charles-Messance
- Macrophage Homeostasis Research Group, School of Biochemistry and Immunology, Trinity College, Dublin, D02 R590, Ireland.,Trinity Biomedical Sciences Institute, Trinity College, Dublin, D02 R590, Ireland
| | - Frederick J Sheedy
- Macrophage Homeostasis Research Group, School of Biochemistry and Immunology, Trinity College, Dublin, D02 R590, Ireland.,Trinity Biomedical Sciences Institute, Trinity College, Dublin, D02 R590, Ireland
| |
Collapse
|
161
|
Pucella JN, Upadhaya S, Reizis B. The Source and Dynamics of Adult Hematopoiesis: Insights from Lineage Tracing. Annu Rev Cell Dev Biol 2020; 36:529-550. [PMID: 32580566 DOI: 10.1146/annurev-cellbio-020520-114601] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The generation of all blood cell lineages (hematopoiesis) is sustained throughout the entire life span of adult mammals. Studies using cell transplantation identified the self-renewing, multipotent hematopoietic stem cells (HSCs) as the source of hematopoiesis in adoptive hosts and delineated a hierarchy of HSC-derived progenitors that ultimately yield mature blood cells. However, much less is known about adult hematopoiesis as it occurs in native hosts, i.e., without transplantation. Here we review recent advances in our understanding of native hematopoiesis, focusing in particular on the application of genetic lineage tracing in mice. The emerging evidence has established HSCs as the major source of native hematopoiesis, helped to define the kinetics of HSC differentiation, and begun exploring native hematopoiesis in stress conditions such as aging and inflammation. Major outstanding questions about native hematopoiesis still remain, such as its clonal composition, the nature of lineage commitment, and the dynamics of the process in humans.
Collapse
Affiliation(s)
- Joseph N Pucella
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; , ,
| | - Samik Upadhaya
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; , ,
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; , ,
| |
Collapse
|
162
|
Dectin-1 Stimulation of Hematopoietic Stem and Progenitor Cells Occurs In Vivo and Promotes Differentiation Toward Trained Macrophages via an Indirect Cell-Autonomous Mechanism. mBio 2020; 11:mBio.00781-20. [PMID: 32576672 PMCID: PMC7315119 DOI: 10.1128/mbio.00781-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Invasive candidiasis is an increasingly frequent cause of serious and often fatal infections. Understanding host defense is essential to design novel therapeutic strategies to boost immune protection against Candida albicans. In this article, we delve into two new concepts that have arisen over the last years: (i) the delivery of myelopoiesis-inducing signals by microbial components directly sensed by hematopoietic stem and progenitor cells (HSPCs) and (ii) the concept of “trained innate immunity” that may also apply to HSPCs. We demonstrate that dectin-1 ligation in vivo activates HSPCs and induces their differentiation to trained macrophages by a cell-autonomous indirect mechanism. This points to new mechanisms by which pathogen detection by HSPCs may modulate hematopoiesis in real time to generate myeloid cells better prepared to deal with the infection. Manipulation of this process may help to boost the innate immune response during candidiasis. Toll-like receptor (TLR) agonists drive hematopoietic stem and progenitor cells (HSPCs) to differentiate along the myeloid lineage. In this study, we used an HSPC transplantation model to investigate the possible direct interaction of β-glucan and its receptor (dectin-1) on HSPCs in vivo. Purified HSPCs from bone marrow of B6Ly5.1 mice (CD45.1 alloantigen) were transplanted into dectin-1−/− mice (CD45.2 alloantigen), which were then injected with β-glucan (depleted zymosan). As recipient mouse cells do not recognize the dectin-1 agonist injected, interference by soluble mediators secreted by recipient cells is negligible. Transplanted HSPCs differentiated into macrophages in response to depleted zymosan in the spleens and bone marrow of recipient mice. Functionally, macrophages derived from HSPCs exposed to depleted zymosan in vivo produced higher levels of inflammatory cytokines (tumor necrosis factor alpha [TNF-α] and interleukin 6 [IL-6]). These results demonstrate that trained immune responses, already described for monocytes and macrophages, also take place in HSPCs. Using a similar in vivo model of HSPC transplantation, we demonstrated that inactivated yeasts of Candida albicans induce differentiation of HSPCs through a dectin-1- and MyD88-dependent pathway. Soluble factors produced following exposure of HSPCs to dectin-1 agonists acted in a paracrine manner to induce myeloid differentiation and to influence the function of macrophages derived from dectin-1-unresponsive or β-glucan-unexposed HSPCs. Finally, we demonstrated that an in vitro transient exposure of HSPCs to live C. albicans cells, prior to differentiation, is sufficient to induce a trained phenotype of the macrophages they produce in a dectin-1- and Toll-like receptor 2 (TLR2)-dependent manner.
Collapse
|
163
|
Paracatu LC, Schuettpelz LG. Contribution of Aberrant Toll Like Receptor Signaling to the Pathogenesis of Myelodysplastic Syndromes. Front Immunol 2020; 11:1236. [PMID: 32625214 PMCID: PMC7313547 DOI: 10.3389/fimmu.2020.01236] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Toll like receptors (TLRs) are a family of pattern recognition receptors that play a central role in the innate immune response. These receptors are expressed on a wide variety of immune and non-immune cells, and they help shape the immune response to infection and injury through the recognition of pathogen-associated molecular patterns (PAMPs) as well as endogenous damage-associated molecular patterns (DAMPs). Accumulating evidence suggests that, in addition to regulating mature effector immune cells, TLRs can influence the immune response from the level of the hematopoietic stem cell (HSC). HSCs express TLRs, and exposure to TLR ligands influences the cycling, differentiation, and function of HSCs, with chronic TLR stimulation leading to impairment of normal HSC repopulating activity. Moreover, enhanced TLR expression and signaling is associated with myelodysplastic syndromes (MDS), a heterogenous group of HSC disorders characterized by ineffective hematopoiesis and a high risk of transformation to acute leukemias. In this review, we will discuss the role of TLR signaling in the pathogenesis of MDS, focusing on the known direct and indirect effects of this type of signaling on HSCs, the mechanisms of TLR signaling upregulation in MDS, the changes in TLR expression with disease progression, and the therapeutic implications for modulating TLR signaling in the treatment of MDS.
Collapse
Affiliation(s)
- Luana Chiquetto Paracatu
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Laura G Schuettpelz
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
164
|
TLR2 and Dectin-1 Signaling in Mouse Hematopoietic Stem and Progenitor Cells Impacts the Ability of the Antigen Presenting Cells They Produce to Activate CD4 T Cells. Cells 2020; 9:cells9051317. [PMID: 32466296 PMCID: PMC7290964 DOI: 10.3390/cells9051317] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 12/25/2022] Open
Abstract
Microbial recognition by pattern recognition receptors (PRRs) expressed on hematopoietic stem and progenitor cells (HSPCs) not only activates myelopoiesis but also programs the function of the monocytes and macrophages they produce. For instance, changes in HSPC programming modify the ability of macrophages derived from them to produce inflammatory cytokines. While HSPCs exposed to a TLR2 agonist give rise to tolerized macrophages (lower proinflammatory cytokine production), HSPCs treated with Dectin-1 ligands produce trained macrophages (higher proinflammatory cytokine production). However, nothing is known about the impact of HSPC exposure to microbes on the function of antigen presenting cells (APCs). In this study we evaluated whether treatment of murine bone marrow HSPCs with a TLR2 or Dectin-1 ligand impacts the antigen presenting capacity of APCs derived from them in vitro. Following activation with microbial ligands or Candida albicans yeasts, APCs derived from TLR2/Dectin-1-programed HSPCs exhibit altered expression of MHCII (signal 1), co-stimulatory molecules (CD40, CD80 and CD86; signal 2) and cytokines (TNF-α, IL-6, IL-12 p40 and IL-2; signal 3). Moreover, APCs derived from TLR2/Dectin-1-programed HSPCs prime enhanced Th1 and Th17 responses, which are important for antifungal defense, in CD4 T cell cocultures. Overall, these results demonstrate for the first time that microbial detection by bone marrow HSPCs can modulate the adaptive immune response by inducing the production of APCs with an altered phenotype.
Collapse
|
165
|
Luo F, Yu S, Jin LH. The Posterior Signaling Center Is an Important Microenvironment for Homeostasis of the Drosophila Lymph Gland. Front Cell Dev Biol 2020; 8:382. [PMID: 32509789 PMCID: PMC7253591 DOI: 10.3389/fcell.2020.00382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/28/2020] [Indexed: 01/02/2023] Open
Abstract
Hematopoiesis is a necessary process for development and immune defense in Drosophila from the embryonic period to adulthood. There are two main stages in this process: the first stage occurs in the head mesoderm during the embryonic stage, and the second occurs in a specialized hematopoietic organ along the dorsal vessel, the lymph gland, during the larval stage. The lymph gland consists of paired lobes, each of which has distinct regions: the cortical zone (CZ), which contains mature hemocytes; the medullary zone (MZ), which contains hematopoietic progenitors; and the posterior signaling center (PSC), which specifically expresses the early B-cell factor (EBF) transcription factor Collier (Col) and the HOX factor Antennapedia (Antp) to form a microenvironment similar to that of the mammalian bone marrow hematopoietic stem cell niche. The PSC plays a key role in regulating hematopoietic progenitor differentiation. Moreover, the PSC contributes to the cellular immune response to wasp parasitism triggered by elevated ROS levels. Two recent studies have revealed that hematopoietic progenitor maintenance is directly regulated by Col expressed in the MZ and is independent of the PSC, challenging the traditional model. In this review, we summarize the regulatory networks of PSC cell proliferation, the controversy regarding PSC-mediated regulation of hematopoietic progenitor differentiation, and the wasp egg infection response. In addition, we discuss why the PSC is an ideal model for investigating mammalian hematopoietic stem cell niches and leukemia.
Collapse
Affiliation(s)
| | | | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| |
Collapse
|
166
|
Sjövall D, Staffas A. The origin of leukemia: Genetic alterations and inflammatory factors in the development of premalignant clonal hematopoiesis. Semin Hematol 2020; 57:7-12. [PMID: 32690142 DOI: 10.1053/j.seminhematol.2020.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 12/17/2022]
Abstract
Clonal hematopoiesis of indetermined potential (CHIP) is increasingly common with age and identified in more than 1 in 10 healthy individuals at the age of 70. Mutations in epigenetic and splicing factors are recurrent genetic events in CHIP, and experimental data suggest that microbial and inflammatory factors may contribute to the selective expansion of hematopoietic stem cells carrying these mutations. In parallel, CHIP is associated with an increased incidence of cardiovascular disease and studies in mice support a causal relationship where mutated hematopoietic cells contribute to inflammation and atherosclerotic plaque formation. Collectively, current clinical and experimental data suggest a complex network where genetic alterations and inflammatory factors contribute to the development of the early stages of hematological malignancy.
Collapse
Affiliation(s)
- Daniel Sjövall
- Sahlgrenska Cancer Center, University of Gothenburg, Sweden; Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Sweden
| | - Anna Staffas
- Sahlgrenska Cancer Center, University of Gothenburg, Sweden; Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Sweden; Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
167
|
de Laval B, Maurizio J, Kandalla PK, Brisou G, Simonnet L, Huber C, Gimenez G, Matcovitch-Natan O, Reinhardt S, David E, Mildner A, Leutz A, Nadel B, Bordi C, Amit I, Sarrazin S, Sieweke MH. C/EBPβ-Dependent Epigenetic Memory Induces Trained Immunity in Hematopoietic Stem Cells. Cell Stem Cell 2020; 26:657-674.e8. [PMID: 32169166 DOI: 10.1016/j.stem.2020.01.017] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 08/23/2019] [Accepted: 01/23/2020] [Indexed: 02/08/2023]
Abstract
Hematopoietic stem cells (HSCs) maintain life-long production of immune cells and can directly respond to infection, but sustained effects on the immune response remain unclear. We show that acute immune stimulation with lipopolysaccharide (LPS) induced only transient changes in HSC abundance, composition, progeny, and gene expression, but persistent alterations in accessibility of specific myeloid lineage enhancers occurred, which increased responsiveness of associated immune genes to secondary stimulation. Functionally, this was associated with increased myelopoiesis of pre-exposed HSCs and improved innate immunity against the gram-negative bacterium P. aeruginosa. The accessible myeloid enhancers were enriched for C/EBPβ targets, and C/EBPβ deletion erased the long-term inscription of LPS-induced epigenetic marks and gene expression. Thus, short-term immune signaling can induce C/EBPβ-dependent chromatin accessibility, resulting in HSC-trained immunity, during secondary infection. This establishes a mechanism for how infection history can be epigenetically inscribed in HSCs as an integral memory function of innate immunity.
Collapse
Affiliation(s)
| | - Julien Maurizio
- Aix Marseille University, CNRS, INSERM, CIML, 13009 Marseille, France; Inovarion, 75005 Paris, France
| | - Prashanth K Kandalla
- Aix Marseille University, CNRS, INSERM, CIML, 13009 Marseille, France; Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany
| | - Gabriel Brisou
- Aix Marseille University, CNRS, INSERM, CIML, 13009 Marseille, France
| | - Louise Simonnet
- Aix Marseille University, CNRS, INSERM, CIML, 13009 Marseille, France
| | - Caroline Huber
- Aix Marseille University, CNRS, INSERM, CIML, 13009 Marseille, France
| | - Gregory Gimenez
- Aix Marseille University, CNRS, INSERM, CIML, 13009 Marseille, France; Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), 13125 Berlin, Germany
| | | | - Susanne Reinhardt
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Alexander Mildner
- Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), 13125 Berlin, Germany
| | - Achim Leutz
- Institute of Biology, Humboldt University of Berlin, 10115 Berlin, Germany; Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), 13125 Berlin, Germany
| | - Bertrand Nadel
- Aix Marseille University, CNRS, INSERM, CIML, 13009 Marseille, France
| | - Christophe Bordi
- Institut de Microbiologie de la Méditerranée, CNRS, 13009 Marseille, France
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sandrine Sarrazin
- Aix Marseille University, CNRS, INSERM, CIML, 13009 Marseille, France.
| | - Michael H Sieweke
- Aix Marseille University, CNRS, INSERM, CIML, 13009 Marseille, France; Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany; Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), 13125 Berlin, Germany.
| |
Collapse
|
168
|
Ferluga J, Yasmin H, Al-Ahdal MN, Bhakta S, Kishore U. Natural and trained innate immunity against Mycobacterium tuberculosis. Immunobiology 2020; 225:151951. [PMID: 32423788 DOI: 10.1016/j.imbio.2020.151951] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/05/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) infection, remains a major global health emergency. It is estimated that one third of global population are affected, predominantly with latent granuloma form of the disease. Mtb co-evolved with humans, for its obligatory intra-macrophage phagosome habitat and slow replication, balanced against unique mycobacterial innate immunity, which appears to be highly complex. TB is transmitted via cough aerosol Mtb inhalation. Bovine TB attenuated Bacillus Calmette Guerin (BCG) live vaccine has been in practice for protection of young children from severe disseminated Mtb infection, but not sufficiently for their lungs, as obtained by trials in TB endemic community. To augment BCG vaccine-driven innate and adaptive immunity for neonates and better protection against adult pulmonary TB, a number of BCG pre-vaccination based, subset vaccine candidates have been tested via animal preclinical, followed by safe clinical trials. BCG also enhances innate macrophage trained immunity and memory, through primordial intracellular Toll-like receptors (TLRs) 7 and 9, which recognise distinct mycobacterial molecular pattern signature. This signature is transmitted by TLR signalling via nuclear factor-κB, for activating innate immune transcription and expression of gene profiling in a mycobacterial signature-specific manner. These are epigenetically imprinted in reprogramming of distinct chromatin areas for innate immune memory, to be recalled following lung reinfection. Unique TB innate immunity and its trained memory are considered independent from adaptive immune B and T cells. On the other hand, adaptive immunity is crucial in Mtb containment in granulomatous latency, supported by innate immune cell infiltration. In nearly 5-10 % of susceptible people, latent TB may be activated due to immune evasion by Mtb from intracellular phagosome within macrophage, perpetrating TB. However, BCG and new recombinant BCG vaccines have the capacity, as indicated in pre- and clinical trials, to overcome such Mtb evasion. Various strategies include pro-inflammatory-bactericidal type 1 polarisation (M1) phenotype of the infected macrophage, involving thrombospondin-TLR pathway. Saprophytic M. smegmatis-based recombinant vaccines are also promising candidates against TB. BCG vaccination of neonates/infants in TB endemic countries also reduced their pneumonia caused by various microbes independent of TB immunity. Here, we discuss host immune response against Mtb, its immune evasion strategies, and the important role innate immunity plays in the development of protection against TB.
Collapse
Affiliation(s)
- Janez Ferluga
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, United Kingdom
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Mohammed N Al-Ahdal
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Sanjib Bhakta
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, London WC1E 7HX, United Kingdom
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, United Kingdom.
| |
Collapse
|
169
|
Zhang X, Karatepe K, Chiewchengchol D, Zhu H, Guo R, Liu P, Yu H, Ren Q, Luo X, Cheng T, Ma F, Xu Y, Han M, Luo HR. Bacteria-Induced Acute Inflammation Does Not Reduce the Long-Term Reconstitution Capacity of Bone Marrow Hematopoietic Stem Cells. Front Immunol 2020; 11:626. [PMID: 32373117 PMCID: PMC7179742 DOI: 10.3389/fimmu.2020.00626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/19/2020] [Indexed: 12/04/2022] Open
Abstract
Pathogen-initiated chronic inflammation or autoimmune diseases accelerate proliferation and promote differentiation of hematopoietic stem cells (HSCs) but simultaneously reduce reconstitution capacity. Nevertheless, the effect of acute infection and inflammation on functional HSCs is still largely unknown. Here we found that acute infection elicited by heat-inactivated Escherichia coli (HIEC) expanded bone marrow lineage-negative (Lin)− stem-cell antigen 1 (Sca-1)+cKit+ (LSK) cell population, leading to reduced frequency of functional HSCs in LSK population. However, the total number of BM phenotypic HSCs (Flk2−CD48−CD150+ LSK cells) was not altered in HIEC-challenged mice. Additionally, the reconstitution capacity of the total BM between infected and uninfected mice was similar by both the competitive repopulation assay and measurement of functional HSCs by limiting dilution. Thus, occasionally occurring acute inflammation, which is critical for host defenses, is unlikely to affect HSC self-renewal and maintenance of long-term reconstitution capacity. During acute bacterial infection and inflammation, the hematopoietic system can replenish hematopoietic cells consumed in the innate inflammatory response by accelerating hematopoietic stem and progenitor cell proliferation, but preserving functional HSCs in the BM.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Pathology, Harvard Stem Cell Institute (HSCI), Harvard Medical School, Boston, MA, United States.,Department of Lab Medicine, The Stem Cell Program, Children's Hospital Boston, Boston, MA, United States.,Dana-Farber/Harvard Cancer Center, Boston, MA, United States.,The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Kutay Karatepe
- Department of Pathology, Harvard Stem Cell Institute (HSCI), Harvard Medical School, Boston, MA, United States.,Department of Lab Medicine, The Stem Cell Program, Children's Hospital Boston, Boston, MA, United States.,Dana-Farber/Harvard Cancer Center, Boston, MA, United States
| | - Direkrit Chiewchengchol
- Department of Pathology, Harvard Stem Cell Institute (HSCI), Harvard Medical School, Boston, MA, United States.,Department of Lab Medicine, The Stem Cell Program, Children's Hospital Boston, Boston, MA, United States.,Dana-Farber/Harvard Cancer Center, Boston, MA, United States
| | - Haiyan Zhu
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Rongxia Guo
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Peng Liu
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hongbo Yu
- Department of Pathology and Laboratory Medicine, VA Boston Healthcare System, West Roxbury, MA, United States
| | - Qian Ren
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiao Luo
- Department of Pathology, Harvard Stem Cell Institute (HSCI), Harvard Medical School, Boston, MA, United States.,Department of Lab Medicine, The Stem Cell Program, Children's Hospital Boston, Boston, MA, United States.,Dana-Farber/Harvard Cancer Center, Boston, MA, United States
| | - Tao Cheng
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Fengxia Ma
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yuanfu Xu
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Mingzhe Han
- Department of Hematopoietic Stem Cell Transplantation, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hongbo R Luo
- Department of Pathology, Harvard Stem Cell Institute (HSCI), Harvard Medical School, Boston, MA, United States.,Department of Lab Medicine, The Stem Cell Program, Children's Hospital Boston, Boston, MA, United States.,Dana-Farber/Harvard Cancer Center, Boston, MA, United States
| |
Collapse
|
170
|
Jaeger-Ruckstuhl CA, Hinterbrandner M, Höpner S, Correnti CE, Lüthi U, Friedli O, Freigang S, Al Sayed MF, Bührer ED, Amrein MA, Schürch CM, Radpour R, Riether C, Ochsenbein AF. TNIK signaling imprints CD8 + T cell memory formation early after priming. Nat Commun 2020; 11:1632. [PMID: 32242021 PMCID: PMC7118140 DOI: 10.1038/s41467-020-15413-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 03/04/2020] [Indexed: 01/15/2023] Open
Abstract
Co-stimulatory signals, cytokines and transcription factors regulate the balance between effector and memory cell differentiation during T cell activation. Here, we analyse the role of the TRAF2-/NCK-interacting kinase (TNIK), a signaling molecule downstream of the tumor necrosis factor superfamily receptors such as CD27, in the regulation of CD8+ T cell fate during acute infection with lymphocytic choriomeningitis virus. Priming of CD8+ T cells induces a TNIK-dependent nuclear translocation of β-catenin with consecutive Wnt pathway activation. TNIK-deficiency during T cell activation results in enhanced differentiation towards effector cells, glycolysis and apoptosis. TNIK signaling enriches for memory precursors by favouring symmetric over asymmetric cell division. This enlarges the pool of memory CD8+ T cells and increases their capacity to expand after re-infection in serial re-transplantation experiments. These findings reveal that TNIK is an important regulator of effector and memory T cell differentiation and induces a population of stem cell-like memory T cells. Coordinate expression of multiple factors play critical roles in the regulation between effector and memory CD8+ T cell differentiation. Here the authors show upon acute viral infection TNIK is critically required as a regulator of effector and memory T cell differentiation.
Collapse
Affiliation(s)
- Carla A Jaeger-Ruckstuhl
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, 3010, Switzerland.,Department of BioMedical Research (DBMR), University of Bern, Bern, 3008, Switzerland.,Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, 3012, Switzerland.,Program in Immunology, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, WA, 98109, USA
| | - Magdalena Hinterbrandner
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, 3010, Switzerland.,Department of BioMedical Research (DBMR), University of Bern, Bern, 3008, Switzerland.,Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, 3012, Switzerland
| | - Sabine Höpner
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, 3010, Switzerland.,Department of BioMedical Research (DBMR), University of Bern, Bern, 3008, Switzerland
| | - Colin E Correnti
- Clinical Research Division, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, WA, 98109, USA
| | - Ursina Lüthi
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, 3010, Switzerland.,Department of BioMedical Research (DBMR), University of Bern, Bern, 3008, Switzerland
| | - Olivier Friedli
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, 3012, Switzerland.,Institute of Pathology, University of Bern, Bern, 3008, Switzerland
| | - Stefan Freigang
- Institute of Pathology, University of Bern, Bern, 3008, Switzerland
| | - Mohamad F Al Sayed
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, 3010, Switzerland.,Department of BioMedical Research (DBMR), University of Bern, Bern, 3008, Switzerland.,Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, 3012, Switzerland
| | - Elias D Bührer
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, 3010, Switzerland.,Department of BioMedical Research (DBMR), University of Bern, Bern, 3008, Switzerland.,Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, 3012, Switzerland
| | - Michael A Amrein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, 3010, Switzerland.,Department of BioMedical Research (DBMR), University of Bern, Bern, 3008, Switzerland.,Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, 3012, Switzerland
| | - Christian M Schürch
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, 3010, Switzerland.,Department of BioMedical Research (DBMR), University of Bern, Bern, 3008, Switzerland.,Institute of Pathology, University of Bern, Bern, 3008, Switzerland
| | - Ramin Radpour
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, 3010, Switzerland.,Department of BioMedical Research (DBMR), University of Bern, Bern, 3008, Switzerland
| | - Carsten Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, 3010, Switzerland.,Department of BioMedical Research (DBMR), University of Bern, Bern, 3008, Switzerland
| | - Adrian F Ochsenbein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, 3010, Switzerland. .,Department of BioMedical Research (DBMR), University of Bern, Bern, 3008, Switzerland.
| |
Collapse
|
171
|
Rosales C. Neutrophils at the crossroads of innate and adaptive immunity. J Leukoc Biol 2020; 108:377-396. [DOI: 10.1002/jlb.4mir0220-574rr] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 02/17/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Carlos Rosales
- Departamento de Inmunología Instituto de Investigaciones Biomédicas Universidad Nacional Autónoma de México Mexico City Mexico
| |
Collapse
|
172
|
Bieber K, Autenrieth SE. Dendritic cell development in infection. Mol Immunol 2020; 121:111-117. [PMID: 32199210 DOI: 10.1016/j.molimm.2020.02.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/03/2020] [Accepted: 02/20/2020] [Indexed: 01/21/2023]
Abstract
The immune system protects from infections primarily by detecting and eliminating invading pathogens. This is predominantly mediated by innate immune cells like neutrophils, monocytes and dendritic cells (DCs) expressing specific receptors recognizing pathogen-associated molecular patterns. DC activation by pathogens leads to the initiation of antigen-specific adaptive immune responses, thereby bridging the innate and adaptive immune systems. However, various pathogens have evolved immune evasion strategies to ensure their survival. In this review, we highlight recent findings on how various microorganisms or their structural features affect or modulate DC development and whether this has any consequences for a protective immune response.
Collapse
Affiliation(s)
- Kristin Bieber
- Department of Internal Medicine II, University of Tübingen, Germany
| | | |
Collapse
|
173
|
Stress Erythropoiesis is a Key Inflammatory Response. Cells 2020; 9:cells9030634. [PMID: 32155728 PMCID: PMC7140438 DOI: 10.3390/cells9030634] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 12/21/2022] Open
Abstract
Bone marrow medullary erythropoiesis is primarily homeostatic. It produces new erythrocytes at a constant rate, which is balanced by the turnover of senescent erythrocytes by macrophages in the spleen. Despite the enormous capacity of the bone marrow to produce erythrocytes, there are times when it is unable to keep pace with erythroid demand. At these times stress erythropoiesis predominates. Stress erythropoiesis generates a large bolus of new erythrocytes to maintain homeostasis until steady state erythropoiesis can resume. In this review, we outline the mechanistic differences between stress erythropoiesis and steady state erythropoiesis and show that their responses to inflammation are complementary. We propose a new hypothesis that stress erythropoiesis is induced by inflammation and plays a key role in maintaining erythroid homeostasis during inflammatory responses.
Collapse
|
174
|
Courties G, Frodermann V, Honold L, Zheng Y, Herisson F, Schloss MJ, Sun Y, Presumey J, Severe N, Engblom C, Hulsmans M, Cremer S, Rohde D, Pittet MJ, Scadden DT, Swirski FK, Kim DE, Moskowitz MA, Nahrendorf M. Glucocorticoids Regulate Bone Marrow B Lymphopoiesis After Stroke. Circ Res 2020; 124:1372-1385. [PMID: 30782088 DOI: 10.1161/circresaha.118.314518] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE After a stroke, patients frequently experience altered systemic immunity resulting in peripheral immunosuppression and higher susceptibility to infections, which is at least partly attributed to lymphopenia. The mechanisms that profoundly change the systemic leukocyte repertoire after stroke are incompletely understood. Emerging evidence indicates that stroke alters hematopoietic output of the bone marrow. OBJECTIVE To explore the mechanisms that lead to defects of B lymphopoiesis after ischemic stroke. METHODS AND RESULTS We here report that ischemic stroke triggers brain-bone marrow communication via hormonal long-range signals that regulate hematopoietic B lineage decisions. Bone marrow fluorescence-activated cell sorter analyses and serial intravital microscopy indicate that transient middle cerebral artery occlusion in mice arrests B-cell development beginning at the pro-B-cell stage. This phenotype was not rescued in Myd88-/- and TLR4-/- mice with disrupted TLR (Toll-like receptor) signaling or after blockage of peripheral sympathetic nerves. Mechanistically, we identified stroke-induced glucocorticoid release as the main instigator of B lymphopoiesis defects. B-cell lineage-specific deletion of the GR (glucocorticoid receptor) in CD19-Cre loxP Nr3c1 mice attenuated lymphocytopenia after transient middle cerebral artery. In 20 patients with acute stroke, increased cortisol levels inversely correlated with blood lymphocyte numbers. CONCLUSIONS Our data demonstrate that the hypothalamic-pituitary-adrenal axis mediates B lymphopoiesis defects after ischemic stroke.
Collapse
Affiliation(s)
- Gabriel Courties
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Vanessa Frodermann
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Lisa Honold
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Yi Zheng
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Fanny Herisson
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Maximilian J Schloss
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Yuan Sun
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Jessy Presumey
- Massachusetts General Hospital and Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Pediatrics (J.P.), Harvard Medical School, Boston
| | - Nicolas Severe
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.).,Harvard Stem Cell Institute, Cambridge, MA (N.S., D.T.S.).,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (N.S., D.T.S.)
| | - Camilla Engblom
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Maarten Hulsmans
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Sebastian Cremer
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - David Rohde
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Mikael J Pittet
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.).,Harvard Stem Cell Institute, Cambridge, MA (N.S., D.T.S.).,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (N.S., D.T.S.)
| | - Filip K Swirski
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Dong-Eog Kim
- Molecular Imaging and Neurovascular Research Laboratory, Department of Neurology, Dongguk University College of Medicine, Goyang, South Korea (D.-E.K.)
| | - Michael A Moskowitz
- Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown (M.A.M.)
| | - Matthias Nahrendorf
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston.,Cardiovascular Research Center (M.N.), Harvard Medical School, Boston
| |
Collapse
|
175
|
Apostol AC, Jensen KDC, Beaudin AE. Training the Fetal Immune System Through Maternal Inflammation-A Layered Hygiene Hypothesis. Front Immunol 2020; 11:123. [PMID: 32117273 PMCID: PMC7026678 DOI: 10.3389/fimmu.2020.00123] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/17/2020] [Indexed: 12/14/2022] Open
Abstract
Over the last century, the alarming surge in allergy and autoimmune disease has led to the hypothesis that decreasing exposure to microbes, which has accompanied industrialization and modern life in the Western world, has fundamentally altered the immune response. In its current iteration, the “hygiene hypothesis” suggests that reduced microbial exposures during early life restricts the production and differentiation of immune cells suited for immune regulation. Although it is now well-appreciated that the increase in hypersensitivity disorders represents a “perfect storm” of many contributing factors, we argue here that two important considerations have rarely been explored. First, the window of microbial exposure that impacts immune development is not limited to early childhood, but likely extends into the womb. Second, restricted microbial interactions by an expectant mother will bias the fetal immune system toward hypersensitivity. Here, we extend this discussion to hypothesize that the cell types sensing microbial exposures include fetal hematopoietic stem cells, which drive long-lasting changes to immunity.
Collapse
Affiliation(s)
- April C Apostol
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| | - Kirk D C Jensen
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| | - Anna E Beaudin
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| |
Collapse
|
176
|
Bone marrow CX3CR1+ mononuclear cells relay a systemic microbiota signal to control hematopoietic progenitors in mice. Blood 2020; 134:1312-1322. [PMID: 31387916 DOI: 10.1182/blood.2019000495] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/15/2019] [Indexed: 12/16/2022] Open
Abstract
The microbiota regulate hematopoiesis in the bone marrow (BM); however, the detailed mechanisms remain largely unknown. In this study, we explored how microbiota-derived molecules (MDMs) were transferred to the BM and sensed by the local immune cells to control hematopoiesis under steady-state conditions. We reveal that MDMs, including bacterial DNA (bDNA), reach the BM via systemic blood circulation and are captured by CX3CR1+ mononuclear cells (MNCs). CX3CR1+ MNCs sense MDMs via endolysosomal Toll-like receptors (TLRs) to produce inflammatory cytokines, which control the basal expansion of hematopoietic progenitors, but not hematopoietic stem cells, and their differentiation potential toward myeloid lineages. CX3CR1+ MNCs colocate with hematopoietic progenitors at the perivascular region, and the depletion of CX3CR1+ MNCs impedes bDNA influx into the BM. Moreover, the abrogation of TLR pathways in CX3CR1+ MNCs abolished the microbiota effect on hematopoiesis. These studies demonstrate that systemic MDMs control BM hematopoiesis by producing CX3CR1+ MNC-mediated cytokines in the steady-state.
Collapse
|
177
|
Cyclosporine H Improves the Multi-Vector Lentiviral Transduction of Murine Haematopoietic Progenitors and Stem Cells. Sci Rep 2020; 10:1812. [PMID: 32020016 PMCID: PMC7000727 DOI: 10.1038/s41598-020-58724-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/13/2020] [Indexed: 01/14/2023] Open
Abstract
Haematopoietic stem cells (HSCs) have the potential for lifetime production of blood and immune cells. The introduction of transgenes into HSCs is important for basic research, as well as for multiple clinical applications, because HSC transplantation is an already established procedure. Recently, a major advancement has been reported in the use of cyclosporine H (CsH), which can significantly enhance the lentivirus (LV) transduction of human haematopoietic stem and progenitor cells (HSPCs). In this study, we employed CsH for LV transduction of murine HSCs and defined haematopoietic progenitors, confirming previous findings in more specific subsets of primitive haematopoietic cells. Our data confirm increased efficiencies, in agreement with the published data. We further experimented with the transduction with the simultaneous use of several vectors. The use of CsH yielded an even more robust increase in rates of multi-vector infection than the increase for a single-vector. CsH was reported to reduce the innate resistance mechanism against LV infection. We indeed found that additional pretreatment could increase the efficiency of transduction, in agreement with the originally reported results. Our data also suggest that CsH does not reduce the efficiency of transplantation into immune-competent hosts or the differentiation of HSCs while enhancing stable long-term expression in vivo. This new additive will surely help many studies in animal models and might be very useful for the development of novel HSC gene therapy approaches.
Collapse
|
178
|
Guo SS, Li BX, Zou DB, Yang SJ, Sheng LX, Ouyang GF, Mu QT, Huang H. Tip of the iceberg: roles of circRNAs in hematological malignancies. Am J Cancer Res 2020; 10:367-382. [PMID: 32195014 PMCID: PMC7061755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 06/10/2023] Open
Abstract
Circular RNAs (circRNAs) are a new class of covalently closed RNA molecules whose 3'- and 5'-ends are linked by a back-splicing event. Emerging evidence has shown that circRNAs play a vital role in the occurrence and development of many diseases and are promising biomarkers and therapeutic targets. However, knowledge of circRNAs in hematological malignancies is limited. In this review, the biogenesis, categories, characteristics, and functions of circRNAs are summarized, especially the roles of circRNAs in hematopoiesis and hematological malignancies.
Collapse
Affiliation(s)
- Shan-Shan Guo
- Ningbo Hospital, School of Medicine, Zhejiang UniversityNingbo, Zhejiang, PR China
| | - Bi-Xia Li
- Ningbo University School of MedicineNingbo, Zhejiang, PR China
| | - Duo-Bing Zou
- Laboratory of Stem Cell Transplantation, Ningbo Hospital, School of Medicine, Zhejiang UniversityNingbo, Zhejiang, PR China
| | - Shu-Jun Yang
- Laboratory of Stem Cell Transplantation, Ningbo Hospital, School of Medicine, Zhejiang UniversityNingbo, Zhejiang, PR China
- Department of Hematology, Ningbo Hospital, School of Medicine, Zhejiang UniversityNingbo, Zhejiang, PR China
| | - Li-Xia Sheng
- Department of Hematology, Ningbo Hospital, School of Medicine, Zhejiang UniversityNingbo, Zhejiang, PR China
| | - Gui-Fang Ouyang
- Department of Hematology, Ningbo Hospital, School of Medicine, Zhejiang UniversityNingbo, Zhejiang, PR China
| | - Qi-Tian Mu
- Laboratory of Stem Cell Transplantation, Ningbo Hospital, School of Medicine, Zhejiang UniversityNingbo, Zhejiang, PR China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, PR China
| |
Collapse
|
179
|
Garcia MM, Goicoechea C, Molina-Álvarez M, Pascual D. Toll-like receptor 4: A promising crossroads in the diagnosis and treatment of several pathologies. Eur J Pharmacol 2020; 874:172975. [PMID: 32017939 DOI: 10.1016/j.ejphar.2020.172975] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 12/20/2019] [Accepted: 01/29/2020] [Indexed: 12/26/2022]
Abstract
Toll-like receptor 4 (TLR4) is expressed in a wide variety of cells and is the central component of the mammalian innate immune system. Since its discovery in 1997, TLR4 has been assigned an ever-increasing number of functions that extend from pathogen recognition to tissue damage identification and promotion of the intrinsic "damage repair response" in pain, intestinal, respiratory and vascular disorders. Precisely, the finding of conserved sequence homology among species along with the molecular and functional characterisation of the TLR4 gene enabled researchers to envisage a common operating system in the activation of innate immunity and the initiation of plastic changes at the onset of chronic pain. Malfunctioning in other conditions was conceived in parallel. In this respect, "pivot" proteins and pathway redundancy are not just evolutionary leftovers but essential for normal functioning or cell survival. Indeed, at present, TLR4 single nucleotide polymorphisms (SNP) and their association with certain dysfunctions and diseases are being confirmed in different pools of patients. However, despite its ability to trigger pathogen infection or alternatively tissue injury communications to immune system, TLR4 targeting might not be considered a panacea. This review article represents a compilation of what we know about TLR4 from clinics and basic research on the 20th anniversary of its discovery. Understanding how to fine-tune the interaction between TLR4 and its specific ligands may lead in the next decades to the development of promising new treatments, reducing polypharmacy and probably having an impact on drug use in numerous pathologies.
Collapse
Affiliation(s)
- Miguel M Garcia
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Universidad Rey Juan Carlos, Avda, Atenas S/n, 28922, Alcorcón, Spain
| | - Carlos Goicoechea
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Universidad Rey Juan Carlos, Avda, Atenas S/n, 28922, Alcorcón, Spain
| | - Miguel Molina-Álvarez
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Universidad Rey Juan Carlos, Avda, Atenas S/n, 28922, Alcorcón, Spain
| | - David Pascual
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Universidad Rey Juan Carlos, Avda, Atenas S/n, 28922, Alcorcón, Spain.
| |
Collapse
|
180
|
Abstract
PURPOSE OF REVIEW We reviewed recent progress on the role of sclerostin (SOST) and its effects on the immune system in order to summarize the current state of knowledge in osteoimmunology, in regard to hematopoiesis, lymphopoiesis, and inflammation. RECENT FINDINGS Changes in sclerostin levels affect distinct niches within the bone marrow that support hematopoietic stem cells and B cell development. Sclerostin's regulation of adipogenesis could also be important for immune cell maintenance with age. Surprisingly, B cell development in the bone marrow is influenced by Sost produced by mesenchymal stem cells and osteoblasts, but not by osteocytes. Additionally, extramedullary hematopoiesis in the spleen and increased pro-inflammatory cytokine levels in the bone marrow are observed in global Sost-/- mice. In addition to changes in bone marrow density, sclerostin depletion affects B lymphopoiesis and myelopoiesis, as well as other changes within the bone marrow cavity that could affect hematopoiesis. It is therefore important to monitor for hematopoietic changes in patients receiving sclerostin-depleting therapies.
Collapse
Affiliation(s)
- Cristine Donham
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, 95343, USA
| | - Jennifer O Manilay
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, 95343, USA.
- Dept. of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, 5200, Lake Road Merced, North, CA, 95343, USA.
| |
Collapse
|
181
|
Histone lysine demethylase KDM5B maintains chronic myeloid leukemia via multiple epigenetic actions. Exp Hematol 2020; 82:53-65. [PMID: 32007477 DOI: 10.1016/j.exphem.2020.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 11/23/2022]
Abstract
The histone lysine demethylase KDM5 family is implicated in normal development and stem cell maintenance by epigenetic modulation of histone methylation status. Deregulation of the KDM5 family has been reported in various types of cancers, including hematological malignancies. However, their transcriptional regulatory roles in the context of leukemia remain unclear. Here, we find that KDM5B is strongly expressed in normal CD34+ hematopoietic stem/progenitor cells and chronic myeloid leukemia (CML) cells. Knockdown of KDM5B in K562 CML cells reduced leukemia colony-forming potential. Transcriptome profiling of KDM5B knockdown K562 cells revealed the deregulation of genes involved in myeloid differentiation and Toll-like receptor signaling. Through the integration of transcriptome and ChIP-seq profiling data, we show that KDM5B is enriched at the binding sites of the GATA and AP-1 transcription factor families, suggesting their collaborations in the regulation of transcription. Even though the binding of KDM5B substantially overlapped with H3K4me1 or H3K4me3 mark at gene promoters, only a small subset of the KDM5B targets showed differential expression in association with the histone demethylation activity. By characterizing the interacting proteins in K562 cells, we discovered that KDM5B recruits protein complexes involved in the mRNA processing machinery, implying an alternative epigenetic action mediated by KDM5B in gene regulation. Our study highlights the oncogenic functions of KDM5B in CML cells and suggests that KDM5B is vital to the transcriptional regulation via multiple epigenetic mechanisms.
Collapse
|
182
|
Defective negative regulation of Toll-like receptor signaling leads to excessive TNF-α in myeloproliferative neoplasm. Blood Adv 2020; 3:122-131. [PMID: 30647074 PMCID: PMC6341195 DOI: 10.1182/bloodadvances.2018026450] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022] Open
Abstract
Patients with myeloproliferative neoplasms (MPN) have high levels of inflammatory cytokines, some of which drive many of the debilitating constitutional symptoms associated with the disease and may also promote expansion of the neoplastic clone. We report here that monocytes from patients with MPN have defective negative regulation of Toll-like receptor (TLR) signaling that leads to unrestrained production of the inflammatory cytokine tumor necrosis factor α (TNF-α) after TLR activation. Specifically, monocytes of patients with MPN are insensitive to the anti-inflammatory cytokine interleukin 10 (IL-10) that negatively regulates TLR-induced TNF-α production. This inability to respond to IL-10 is a not a direct consequence of JAK2 V617F , as the phenotype of persistent TNF-α production is a feature of JAK2 V617F and wild-type monocytes alike from JAK2 V617F -positive patients. Moreover, persistent TNF-α production was also discovered in the unaffected identical twin of a patient with MPN, suggesting it could be an intrinsic feature of those predisposed to acquire MPN. This work implicates sustained TLR signaling as not only a contributor to the chronic inflammatory state of MPN patients but also a potential predisposition to acquire MPN.
Collapse
|
183
|
Benova A, Tencerova M. Obesity-Induced Changes in Bone Marrow Homeostasis. Front Endocrinol (Lausanne) 2020; 11:294. [PMID: 32477271 PMCID: PMC7235195 DOI: 10.3389/fendo.2020.00294] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/20/2020] [Indexed: 12/24/2022] Open
Abstract
Obesity is characterized by low-grade inflammation, which is accompanied by increased accumulation of immune cells in peripheral tissues including adipose tissue (AT), skeletal muscle, liver and pancreas, thereby impairing their primary metabolic functions in the regulation of glucose homeostasis. Obesity has also shown to have a detrimental effect on bone homeostasis by altering bone marrow and hematopoietic stem cell differentiation and thus impairing bone integrity and immune cell properties. The origin of immune cells arises in the bone marrow, which has been shown to be affected with the obesogenic condition via increased cellularity and shifting differentiation and function of hematopoietic and bone marrow mesenchymal stem cells in favor of myeloid progenitors and increased bone marrow adiposity. These obesity-induced changes in the bone marrow microenvironment lead to dramatic bone marrow remodeling and compromising immune cell functions, which in turn affect systemic inflammatory conditions and regulation of whole-body metabolism. However, there is limited information on the inflammatory secretory factors creating the bone marrow microenvironment and how these factors changed during metabolic complications. This review summarizes recent findings on inflammatory and cellular changes in the bone marrow in relation to obesity and further discuss whether dietary intervention or physical activity may have beneficial effects on the bone marrow microenvironment and whole-body metabolism.
Collapse
|
184
|
Batsivari A, Haltalli MLR, Passaro D, Pospori C, Lo Celso C, Bonnet D. Dynamic responses of the haematopoietic stem cell niche to diverse stresses. Nat Cell Biol 2020; 22:7-17. [PMID: 31907409 DOI: 10.1038/s41556-019-0444-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/27/2019] [Indexed: 01/01/2023]
Abstract
Adult haematopoietic stem cells (HSCs) mainly reside in the bone marrow, where stromal and haematopoietic cells regulate their function. The steady state HSC niche has been extensively studied. In this Review, we focus on how bone marrow microenvironment components respond to different insults including inflammation, malignant haematopoiesis and chemotherapy. We highlight common and unique patterns among multiple cell types and their environment and discuss current limitations in our understanding of this complex and dynamic tissue.
Collapse
Affiliation(s)
- Antoniana Batsivari
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute , London, UK
| | - Myriam Luydmila Rachelle Haltalli
- Department of Life Sciences, Imperial College London, South Kensington campus, London, UK
- Lo Celso Laboratory, The Francis Crick Institute, London, UK
| | - Diana Passaro
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute , London, UK
| | - Constandina Pospori
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute , London, UK
- Department of Life Sciences, Imperial College London, South Kensington campus, London, UK
- Lo Celso Laboratory, The Francis Crick Institute, London, UK
| | - Cristina Lo Celso
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute , London, UK.
- Department of Life Sciences, Imperial College London, South Kensington campus, London, UK.
- Lo Celso Laboratory, The Francis Crick Institute, London, UK.
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute , London, UK.
| |
Collapse
|
185
|
Alkie TN, Yitbarek A, Hodgins DC, Kulkarni RR, Taha-Abdelaziz K, Sharif S. Development of innate immunity in chicken embryos and newly hatched chicks: a disease control perspective. Avian Pathol 2019; 48:288-310. [PMID: 31063007 DOI: 10.1080/03079457.2019.1607966] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Newly hatched chickens are confronted by a wide array of pathogenic microbes because their adaptive immune defences have limited capabilities to control these pathogens. In such circumstances, and within this age group, innate responses provide a degree of protection. Moreover, as the adaptive immune system is relatively naïve to foreign antigens, synergy with innate defences is critical. This review presents knowledge on the ontogeny of innate immunity in chickens pre-hatch and early post-hatch and provides insights into possible interventions to modulate innate responses early in the life of the bird. As in other vertebrate species, the chicken innate immune system which include cellular mediators, cytokine and chemokine repertoires and molecules involved in antigen detection, develop early in life. Comparison of innate immune systems in newly hatched chickens and mature birds has revealed differences in magnitude and quality, but responses in younger chickens can be boosted using innate immune system modulators. Functional expression of pattern recognition receptors and several defence molecules by innate immune system cells of embryos and newly hatched chicks suggests that innate responses can be modulated at this stage of development to combat pathogens. Improved understanding of innate immune system ontogeny and functionality in chickens is critical for the implementation of sound and safe interventions to provide long-term protection against pathogens. Next-generation tools for studying genetic and epigenetic regulation of genes, functional metagenomics and gene knockouts can be used in the future to explore and dissect the contributions of signalling pathways of innate immunity and to devise more efficacious disease control strategies.
Collapse
Affiliation(s)
- Tamiru N Alkie
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada
| | - Alexander Yitbarek
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada
| | - Douglas C Hodgins
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada
| | - Raveendra R Kulkarni
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada
| | - Khaled Taha-Abdelaziz
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada.,b Pathology Department, Faculty of Veterinary Medicine , Beni-Suef University , Beni-Suef , Egypt
| | - Shayan Sharif
- a Department of Pathobiology, Ontario Veterinary College , University of Guelph , Guelph , ON , Canada
| |
Collapse
|
186
|
Mann M, Mehta A, de Boer CG, Kowalczyk MS, Lee K, Haldeman P, Rogel N, Knecht AR, Farouq D, Regev A, Baltimore D. Heterogeneous Responses of Hematopoietic Stem Cells to Inflammatory Stimuli Are Altered with Age. Cell Rep 2019; 25:2992-3005.e5. [PMID: 30540934 DOI: 10.1016/j.celrep.2018.11.056] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 10/05/2018] [Accepted: 11/13/2018] [Indexed: 12/30/2022] Open
Abstract
Long-term hematopoietic stem cells (LT-HSCs) maintain hematopoietic output throughout an animal's lifespan. However, with age, the balance is disrupted, and LT-HSCs produce a myeloid-biased output, resulting in poor immune responses to infectious challenge and the development of myeloid leukemias. Here, we show that young and aged LT-HSCs respond differently to inflammatory stress, such that aged LT-HSCs produce a cell-intrinsic, myeloid-biased expression program. Using single-cell RNA sequencing (scRNA-seq), we identify a myeloid-biased subset within the LT-HSC population (mLT-HSCs) that is prevalent among aged LT-HSCs. We identify CD61 as a marker of mLT-HSCs and show that CD61-high LT-HSCs are uniquely primed to respond to acute inflammatory challenge. We predict that several transcription factors regulate the mLT-HSCs gene program and show that Klf5, Ikzf1, and Stat3 play an important role in age-related inflammatory myeloid bias. We have therefore identified and isolated an LT-HSC subset that regulates myeloid versus lymphoid balance under inflammatory challenge and with age.
Collapse
Affiliation(s)
- Mati Mann
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Arnav Mehta
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; David Geffen School of Medicine, UCLA, Los Angeles, CA 90025, USA
| | - Carl G de Boer
- Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | | | - Kevin Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Pearce Haldeman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Noga Rogel
- Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Abigail R Knecht
- Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Daneyal Farouq
- Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Koch Institute of Integrative Cancer Biology, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02140, USA.
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
187
|
D'Atri LP, Rodríguez CS, Miguel CP, Pozner RG, Ortiz Wilczyñski JM, Negrotto S, Carrera-Silva EA, Heller PG, Schattner M. Activation of toll-like receptors 2 and 4 on CD34 + cells increases human megakaryo/thrombopoiesis induced by thrombopoietin. J Thromb Haemost 2019; 17:2196-2210. [PMID: 31397069 DOI: 10.1111/jth.14605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/07/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Platelet Toll-like receptor (TLR)2/4 are key players in amplifying the host immune response; however, their role in human megakaryo/thrombopoiesis has not yet been defined. OBJECTIVES We evaluated whether Pam3CSK4 or lipopolysaccharide (LPS), TLR2/4 ligands respectively, modulate human megakaryocyte development and platelet production. METHODS CD34+ cells from human umbilical cord were stimulated with LPS or Pam3CSK4 with or without thrombopoietin (TPO). RESULTS CD34+ cells and megakaryocytes express TLR2 and TLR4 at both RNA and protein level; however, direct stimulation of CD34+ cells with LPS or Pam3CSK4 had no effect on cell growth. Interestingly, both TLR ligands markedly increased TPO-induced CD34+ cell proliferation, megakaryocyte number and maturity, proplatelet and platelet production when added at day 0. In contrast, this synergism was not observed when TLR agonists were added 7 days after TPO addition. Interleukin-6 (IL-6) release was observed upon CD34+ or megakaryocyte stimulation with LPS or Pam3CSK4 but not with TPO and this effect was potentiated in combination with TPO. The increased proliferation and IL-6 production induced by TPO + LPS or Pam3CSK4 were suppressed by TLR2/4 or IL-6 neutralizing antibodies, as well as by PI3K/AKT and nuclear factor-κB inhibitors. Additionally, increased proplatelet and platelet production were associated with enhanced nuclear translocation of nuclear factor-E2. Finally, the supernatants of CD34+ cells stimulated with TPO+LPS-induced CFU-M colonies. CONCLUSIONS Our data suggest that the activation of TLR2 and TLR4 in CD34+ cells and megakaryocytes in the presence of TPO may contribute to warrant platelet provision during infection episodes by an autocrine IL-6 loop triggered by PI3K/NF-κB axes.
Collapse
Affiliation(s)
- Lina Paola D'Atri
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Camila Sofía Rodríguez
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Carolina Paula Miguel
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Roberto Gabriel Pozner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Juan Manuel Ortiz Wilczyñski
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Soledad Negrotto
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Eugenio Antonio Carrera-Silva
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Paula Graciela Heller
- Institute of Medical Research Dr. Alfredo Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
- Department of Hematology Research, National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Institute of Medical Research (IDIM), Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| |
Collapse
|
188
|
Abstract
Genetic defects that accumulate in haematopoietic stem cells (HSCs) are thought to be responsible for age-related changes in haematopoiesis that include a decline in lymphopoiesis and skewing towards the myeloid lineage. This HSC-centric view is based largely on studies showing that HSCs from aged mice exhibit these lineage biases following transplantation into irradiated young recipient mice. In this Opinion article, we make the case that the reliance on this approach has led to inaccurate conclusions regarding the effects of ageing on blood-forming stem cells; we suggest instead that changes in the environment contribute to haematopoietic system ageing. We propose that a complete understanding of how ageing affects haematopoiesis depends on the analysis of blood cell production in unperturbed mice. We describe how this can be achieved using in situ fate mapping. This approach indicates that changes in downstream progenitors, in addition to any HSC defects, may explain the reduced lymphopoiesis and sustained myelopoiesis that occur during ageing.
Collapse
|
189
|
Toll-like receptor 2 expression on c-kit + cells tracks the emergence of embryonic definitive hematopoietic progenitors. Nat Commun 2019; 10:5176. [PMID: 31729371 PMCID: PMC6858454 DOI: 10.1038/s41467-019-13150-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 10/21/2019] [Indexed: 12/27/2022] Open
Abstract
Hematopoiesis in mammalian embryos proceeds through three successive waves of hematopoietic progenitors. Since their emergence spatially and temporally overlap and phenotypic markers are often shared, the specifics regarding their origin, development, lineage restriction and mutual relationships have not been fully determined. The identification of wave-specific markers would aid to resolve these uncertainties. Here, we show that toll-like receptors (TLRs) are expressed during early mouse embryogenesis. We provide phenotypic and functional evidence that the expression of TLR2 on E7.5 c-kit+ cells marks the emergence of precursors of erythro-myeloid progenitors (EMPs) and provides resolution for separate tracking of EMPs from primitive progenitors. Using in vivo fate mapping, we show that at E8.5 the Tlr2 locus is already active in emerging EMPs and in progenitors of adult hematopoietic stem cells (HSC). Together, this data demonstrates that the activation of the Tlr2 locus tracks the earliest events in the process of EMP and HSC specification. There is limited knowledge of markers to identify various waves of murine embryonic hematopoiesis. Here, the authors show that the expression of toll-like receptor 2 (TLR2) on E7.5 c-kit+ cells marks the emergence of erythro-myeloid progenitor precursors and that the Tlr2 locus is active in E8.5 precursors of adult HSCs.
Collapse
|
190
|
Neutrophil Maturation and Survival Is Controlled by IFN-Dependent Regulation of NAMPT Signaling. Int J Mol Sci 2019; 20:ijms20225584. [PMID: 31717318 PMCID: PMC6888478 DOI: 10.3390/ijms20225584] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/01/2019] [Accepted: 11/06/2019] [Indexed: 12/16/2022] Open
Abstract
Granulocyte-colony stimulating factor (G-CSF)/nicotinamide phosphoribosyltransferase (NAMPT) signaling has been shown to be crucial for the modulation of neutrophil development and functionality. As this signaling pathway is significantly suppressed by type I interferons (IFNs), we aimed to study how the regulation of neutrophil differentiation and phenotype is altered in IFN-deficient mice during granulopoiesis. The composition of bone marrow granulocyte progenitors and their Nampt expression were assessed in bone marrow of type I IFN receptor knockout (Ifnar1-/-) mice and compared to wild-type animals. The impact of NAMPT inhibition on the proliferation, survival, and differentiation of murine bone marrow progenitors, as well as of murine 32D and human HL-60 neutrophil-like cell lines, was estimated. The progressive increase of Nampt expression during neutrophil progenitor maturation could be observed, and it was more prominent in IFN-deficient animals. Altered composition of bone marrow progenitors in these mice correlated with the dysregulation of apoptosis and altered differentiation of these cells. We observed that NAMPT is vitally important for survival of early progenitors, while at later stages it delays the differentiation of neutrophils, with moderate effect on their survival. This study shows that IFN-deficiency leads to the elevated NAMPT expression in the bone marrow, which in turn modulates neutrophil development and differentiation, even in the absence of tumor-derived stimuli.
Collapse
|
191
|
Interleukin-6 signaling regulates hematopoietic stem cell emergence. Exp Mol Med 2019; 51:1-12. [PMID: 31649245 PMCID: PMC6813302 DOI: 10.1038/s12276-019-0320-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 04/19/2019] [Accepted: 05/30/2019] [Indexed: 12/20/2022] Open
Abstract
Hematopoietic stem cells (HSCs) produce all lineages of mature blood cells for the lifetime of an organism. In vertebrates, HSCs derive from the transition of the hemogenic endothelium (HE) in the floor of the embryonic dorsal aorta. Most recently, a series of proinflammatory factors, such as tumor necrosis factor-α, interferon-γ, and Toll-like receptor 4, have been confirmed to play a key role in HSC specification. However, the full complement of necessary signaling inputs remains unknown to date. Here, we show that interleukin-6R (IL6R) via IL6 is required and sufficient for HSC generation. We found that Notch activates IL6R by regulating its expression in the HE and in HSCs. The secretion of IL6 mainly originates from HSC-independent myeloid cells, but not from HSCs and their adjacent vascular endothelial cells. In addition, blocking IL6 signaling does not affect vascular development or the production of primitive erythrocytes. Taken together, our results uncover a previously obscure relationship between IL6 signaling and HSC production and provide new insights into HSC regeneration using proinflammatory factors in vitro. A molecule that triggers inflammation, interleukin-6 (IL6), is crucial for development of blood stem cells, known as hematopoetic stem cells (HSCs). HSCs can differentiate into any type of blood or immune cell, and have enormous therapeutic potential. Although some of the signaling molecules that trigger their development are known, HSCs have yet to be grown under laboratory conditions, so knowledge gaps remain. Based on clues that IL6 might be one of the missing signals, He Huang at the Zhejiang University School of Medicine, Hangzhou, China, and coworkers investigated how IL6 affects HSC development in zebrafish, a common model of development. Zebrafish embryos lacking IL6 were deficient in HSCs, but showed normal blood vessel growth. Treating with IL6 restored HSC production. These results represent another step toward being able to grow HSCs for transplantation and therapy.
Collapse
|
192
|
Vandoorne K, Rohde D, Kim HY, Courties G, Wojtkiewicz G, Honold L, Hoyer FF, Frodermann V, Nayar R, Herisson F, Jung Y, Désogère PA, Vinegoni C, Caravan P, Weissleder R, Sosnovik DE, Lin CP, Swirski FK, Nahrendorf M. Imaging the Vascular Bone Marrow Niche During Inflammatory Stress. Circ Res 2019; 123:415-427. [PMID: 29980569 DOI: 10.1161/circresaha.118.313302] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Inflammatory stress induced by exposure to bacterial lipopolysaccharide causes hematopoietic stem cell expansion in the bone marrow niche, generating a cellular immune response. As an integral component of the hematopoietic stem cell niche, the bone marrow vasculature regulates the production and release of blood leukocytes, which protect the host against infection but also fuel inflammatory diseases. OBJECTIVE We aimed to develop imaging tools to explore vascular changes in the bone marrow niche during acute inflammation. METHODS AND RESULTS Using the TLR (Toll-like receptor) ligand lipopolysaccharide as a prototypical danger signal, we applied multiparametric, multimodality and multiscale imaging to characterize how the bone marrow vasculature adapts when hematopoiesis boosts leukocyte supply. In response to lipopolysaccharide, ex vivo flow cytometry and histology showed vascular changes to the bone marrow niche. Specifically, proliferating endothelial cells gave rise to new vasculature in the bone marrow during hypoxic conditions. We studied these vascular changes with complementary intravital microscopy and positron emission tomography/magnetic resonance imaging. Fluorescence and positron emission tomography integrin αVβ3 imaging signal increased during lipopolysaccharide-induced vascular remodeling. Vascular leakiness, quantified by albumin-based in vivo microscopy and magnetic resonance imaging, rose when neutrophils departed and hematopoietic stem and progenitor cells proliferated more vigorously. CONCLUSIONS Introducing a tool set to image bone marrow either with cellular resolution or noninvasively within the entire skeleton, this work sheds light on angiogenic responses that accompany emergency hematopoiesis. Understanding and monitoring bone marrow vasculature may provide a key to unlock therapeutic targets regulating systemic inflammation.
Collapse
Affiliation(s)
- Katrien Vandoorne
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - David Rohde
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Hye-Yeong Kim
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | | | - Gregory Wojtkiewicz
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Lisa Honold
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Friedrich Felix Hoyer
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Vanessa Frodermann
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Ribhu Nayar
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Fanny Herisson
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Yookyung Jung
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.).,Wellman Center for Photomedicine (Y.J., C.P.L.)
| | - Pauline A Désogère
- Massachusetts General Hospital and Harvard Medical School, Boston; Department of Radiology, Martinos Center for Biomedical Imaging (P.A.D., P.C., D.E.S.)
| | - Claudio Vinegoni
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Peter Caravan
- Massachusetts General Hospital and Harvard Medical School, Boston; Department of Radiology, Martinos Center for Biomedical Imaging (P.A.D., P.C., D.E.S.)
| | - Ralph Weissleder
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.).,Massachusetts General Hospital and Harvard Medical School, Charlestown; and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - David E Sosnovik
- Massachusetts General Hospital and Harvard Medical School, Boston; Department of Radiology, Martinos Center for Biomedical Imaging (P.A.D., P.C., D.E.S.).,Cardiovascular Research Center (D.E.S., M.N.)
| | - Charles P Lin
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.).,Wellman Center for Photomedicine (Y.J., C.P.L.)
| | - Filip K Swirski
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Matthias Nahrendorf
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.).,Cardiovascular Research Center (D.E.S., M.N.)
| |
Collapse
|
193
|
"Hierarchy" and "Holacracy"; A Paradigm of the Hematopoietic System. Cells 2019; 8:cells8101138. [PMID: 31554248 PMCID: PMC6830102 DOI: 10.3390/cells8101138] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023] Open
Abstract
The mammalian hematopoietic system has long been viewed as a hierarchical paradigm in which a small number of hematopoietic stem cells (HSCs) are located at the apex. HSCs were traditionally thought to be homogeneous and quiescent in a homeostatic state. However, recent observations, through extramedullary hematopoiesis and clonal assays, have cast doubt on the validity of the conventional interpretation. A key issue is understanding the characteristics of HSCs from different viewpoints, including dynamic physics and social network theory. The aim of this literature review is to propose a new paradigm of our hematopoietic system, in which individual HSCs are actively involved.
Collapse
|
194
|
Mei M, Wang Y, Li Z, Zhang M. Role of circular RNA in hematological malignancies. Oncol Lett 2019; 18:4385-4392. [PMID: 31611947 PMCID: PMC6781753 DOI: 10.3892/ol.2019.10836] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 08/13/2019] [Indexed: 12/18/2022] Open
Abstract
Compared with linear RNA, circular RNAs (circRNAs) form a covalently closed circular continuous loop and are highly conserved, stable and tissue-specific. In recent years, circRNAs received considerable attention in the diagnosis, classification, treatment and prognosis of hematological tumors. circRNAs function as microRNA sponges and competitive endogenous RNAs that play an essential role in the translation, regulation and interaction of proteins. The present review discussed the fundamental properties and functions of circRNAs and the latest advancements in the context of circRNAs in the clinical research of hematological malignancies, namely acute and chronic myeloid leukemia, and chronic lymphocytic leukemia. circRNAs show potential in the diagnosis and prognosis of various diseases and can be used as therapeutic targets and biomarkers for disease.
Collapse
Affiliation(s)
- Mei Mei
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yingjun Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
195
|
Bennett LF, Liao C, Quickel MD, Yeoh BS, Vijay-Kumar M, Hankey-Giblin P, Prabhu KS, Paulson RF. Inflammation induces stress erythropoiesis through heme-dependent activation of SPI-C. Sci Signal 2019; 12:12/598/eaap7336. [PMID: 31506384 DOI: 10.1126/scisignal.aap7336] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Inflammation alters bone marrow hematopoiesis to favor the production of innate immune effector cells at the expense of lymphoid cells and erythrocytes. Furthermore, proinflammatory cytokines inhibit steady-state erythropoiesis, which leads to the development of anemia in diseases with chronic inflammation. Acute anemia or hypoxic stress induces stress erythropoiesis, which generates a wave of new erythrocytes to maintain erythroid homeostasis until steady-state erythropoiesis can resume. Although hypoxia-dependent signaling is a key component of stress erythropoiesis, we found that inflammation also induced stress erythropoiesis in the absence of hypoxia. Using a mouse model of sterile inflammation, we demonstrated that signaling through Toll-like receptors (TLRs) paradoxically increased the phagocytosis of erythrocytes (erythrophagocytosis) by macrophages in the spleen, which enabled expression of the heme-responsive gene encoding the transcription factor SPI-C. Increased amounts of SPI-C coupled with TLR signaling promoted the expression of Gdf15 and Bmp4, both of which encode ligands that initiate the expansion of stress erythroid progenitors (SEPs) in the spleen. Furthermore, despite their inhibition of steady-state erythropoiesis in the bone marrow, the proinflammatory cytokines TNF-α and IL-1β promoted the expansion and differentiation of SEPs in the spleen. These data suggest that inflammatory signals induce stress erythropoiesis to maintain erythroid homeostasis when inflammation inhibits steady-state erythropoiesis.
Collapse
Affiliation(s)
- Laura F Bennett
- Intercollege Graduate Program in Genetics, Penn State University, University Park, PA 16802, USA.,Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA 16802, USA
| | - Chang Liao
- Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA 16802, USA.,Graduate Program in Pathobiology, Penn State University, University Park, PA 16802, USA.,Center for Molecular Immunology and Infectious Disease, Penn State University, University Park, PA 16802, USA
| | - Michael D Quickel
- Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA 16802, USA.,Graduate Program in Pathobiology, Penn State University, University Park, PA 16802, USA.,Clinical and Translational Science Institute, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Beng San Yeoh
- Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA 16802, USA
| | - Matam Vijay-Kumar
- Center for Molecular Immunology and Infectious Disease, Penn State University, University Park, PA 16802, USA.,Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA 16802, USA.,Department of Nutritional Sciences, Penn State University, University Park, PA 16802, USA
| | - Pamela Hankey-Giblin
- Intercollege Graduate Program in Genetics, Penn State University, University Park, PA 16802, USA.,Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA 16802, USA.,Graduate Program in Pathobiology, Penn State University, University Park, PA 16802, USA.,Center for Molecular Immunology and Infectious Disease, Penn State University, University Park, PA 16802, USA
| | - K Sandeep Prabhu
- Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA 16802, USA.,Graduate Program in Pathobiology, Penn State University, University Park, PA 16802, USA.,Center for Molecular Immunology and Infectious Disease, Penn State University, University Park, PA 16802, USA.,Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA 16802, USA
| | - Robert F Paulson
- Intercollege Graduate Program in Genetics, Penn State University, University Park, PA 16802, USA. .,Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA 16802, USA.,Center for Molecular Immunology and Infectious Disease, Penn State University, University Park, PA 16802, USA.,Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA 16802, USA
| |
Collapse
|
196
|
Cuartero S, Innes AJ, Merkenschlager M. Towards a Better Understanding of Cohesin Mutations in AML. Front Oncol 2019; 9:867. [PMID: 31552185 PMCID: PMC6746210 DOI: 10.3389/fonc.2019.00867] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022] Open
Abstract
Classical driver mutations in acute myeloid leukemia (AML) typically affect regulators of cell proliferation, differentiation, and survival. The selective advantage of increased proliferation, improved survival, and reduced differentiation on leukemia progression is immediately obvious. Recent large-scale sequencing efforts have uncovered numerous novel AML-associated mutations. Interestingly, a substantial fraction of the most frequently mutated genes encode general regulators of transcription and chromatin state. Understanding the selective advantage conferred by these mutations remains a major challenge. A striking example are mutations in genes of the cohesin complex, a major regulator of three-dimensional genome organization. Several landmark studies have shown that cohesin mutations perturb the balance between self-renewal and differentiation of hematopoietic stem and progenitor cells (HSPC). Emerging data now begin to uncover the molecular mechanisms that underpin this phenotype. Among these mechanisms is a role for cohesin in the control of inflammatory responses in HSPCs and myeloid cells. Inflammatory signals limit HSPC self-renewal and drive HSPC differentiation. Consistent with this, cohesin mutations promote resistance to inflammatory signals, and may provide a selective advantage for AML progression. In this review, we discuss recent progress in understanding cohesin mutations in AML, and speculate whether vulnerabilities associated with these mutations could be exploited therapeutically.
Collapse
Affiliation(s)
- Sergi Cuartero
- Faculty of Medicine, MRC London Institute of Medical Sciences, Institute of Clinical Sciences, Imperial College London, London, United Kingdom.,Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
| | - Andrew J Innes
- Faculty of Medicine, MRC London Institute of Medical Sciences, Institute of Clinical Sciences, Imperial College London, London, United Kingdom.,Faculty of Medicine, Centre for Haematology, Imperial College London, London, United Kingdom
| | - Matthias Merkenschlager
- Faculty of Medicine, MRC London Institute of Medical Sciences, Institute of Clinical Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
197
|
Khader SA, Divangahi M, Hanekom W, Hill PC, Maeurer M, Makar KW, Mayer-Barber KD, Mhlanga MM, Nemes E, Schlesinger LS, van Crevel R, Vankayalapati R(K, Xavier RJ, Netea MG. Targeting innate immunity for tuberculosis vaccination. J Clin Invest 2019; 129:3482-3491. [PMID: 31478909 PMCID: PMC6715374 DOI: 10.1172/jci128877] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Vaccine development against tuberculosis (TB) is based on the induction of adaptive immune responses endowed with long-term memory against mycobacterial antigens. Memory B and T cells initiate a rapid and robust immune response upon encounter with Mycobacterium tuberculosis, thus achieving long-lasting protection against infection. Recent studies have shown, however, that innate immune cell populations such as myeloid cells and NK cells also undergo functional adaptation after infection or vaccination, a de facto innate immune memory that is also termed trained immunity. Experimental and epidemiological data have shown that induction of trained immunity contributes to the beneficial heterologous effects of vaccines such as bacille Calmette-Guérin (BCG), the licensed TB vaccine. Moreover, increasing evidence argues that trained immunity also contributes to the anti-TB effects of BCG vaccination. An interaction among immunological signals, metabolic rewiring, and epigenetic reprogramming underlies the molecular mechanisms mediating trained immunity in myeloid cells and their bone marrow progenitors. Future studies are warranted to explore the untapped potential of trained immunity to develop a future generation of TB vaccines that would combine innate and adaptive immune memory induction.
Collapse
Affiliation(s)
- Shabaana A. Khader
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Maziar Divangahi
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, and Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Willem Hanekom
- Bill & Melinda Gates Foundation, Seattle, Washington, USA
| | - Philip C. Hill
- Centre for International Health, Department of Preventive and Social Medicine, University of Otago Medical School, Dunedin, New Zealand
| | - Markus Maeurer
- Department of Oncology/Haematology, Krankenhaus Nordwest (KHNW), Frankfurt, Germany
- ImmunoSurgery Unit, Champalimaud Foundation, Lisbon, Portugal
| | - Karen W. Makar
- Bill & Melinda Gates Foundation, Seattle, Washington, USA
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Musa M. Mhlanga
- Division of Chemical Systems & Synthetic Biology, Institute for Infectious Disease & Molecular Medicine (IDM), Faculty of Health Sciences, Department of Integrative Biomedical Sciences, and
| | - Elisa Nemes
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | | | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Raman (Krishna) Vankayalapati
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Ramnik J. Xavier
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Computational and Integrative Biology and
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | | |
Collapse
|
198
|
Balistreri CR, Garagnani P, Madonna R, Vaiserman A, Melino G. Developmental programming of adult haematopoiesis system. Ageing Res Rev 2019; 54:100918. [PMID: 31226498 DOI: 10.1016/j.arr.2019.100918] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/15/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022]
Abstract
The Barker hypothesis of 'foetal origin of adult diseases' has led to emphasize the concept of 'developmental programming', based on the crucial role of epigenetic factors. Accordingly, it has been demonstrated that parental adversity (before conception and during pregnancy) and foetal factors (i.e., hypoxia, malnutrition and placental insufficiency) permanently modify the physiological systems of the progeny, predisposing them to premature ageing and chronic disease during adulthood. Thus, an altered functionality of the endocrine, immune, nervous and cardiovascular systems is observed in the progeny. However, it remains to be understood whether the haematopoietic system itself also represents a portrait of foetal programming. Here, we provide evidence, reporting and discussing related theories, and results of studies described in the literature. In addition, we have outlined our opinions and suggest how it is possible to intervene to correct foetal mal-programming. Some pro-health interventions and recommendations are proposed, with the hope of guarantee the health of future generations and trying to combat the continuous increase in age-related diseases in human populations.
Collapse
|
199
|
Guillamot M, Ouazia D, Dolgalev I, Yeung ST, Kourtis N, Dai Y, Corrigan K, Zea-Redondo L, Saraf A, Florens L, Washburn MP, Tikhonova AN, Malumbres M, Gong Y, Tsirigos A, Park C, Barbieri C, Khanna KM, Busino L, Aifantis I. The E3 ubiquitin ligase SPOP controls resolution of systemic inflammation by triggering MYD88 degradation. Nat Immunol 2019; 20:1196-1207. [PMID: 31406379 PMCID: PMC7376385 DOI: 10.1038/s41590-019-0454-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 06/26/2019] [Indexed: 01/25/2023]
Abstract
The response to systemic infection and injury requires the rapid adaptation of hematopoietic stem cells (HSCs), which proliferate and divert their differentiation toward the myeloid lineage. Significant interest has emerged in understanding the signals that trigger the emergency hematopoietic program. However, the mechanisms that halt this response of HSCs, which is critical to restore homeostasis, remain unknown. Here we reveal that the E3 ubiquitin ligase Speckle-type BTB-POZ protein (SPOP) restrains the inflammatory activation of HSCs. In the absence of Spop, systemic inflammation proceeded in an unresolved manner, and the sustained response in the HSCs resulted in a lethal phenotype reminiscent of hyper-inflammatory syndrome or sepsis. Our proteomic studies decipher that SPOP restricted inflammation by ubiquitinating the innate signal transducer myeloid differentiation primary response protein 88 (MYD88). These findings unearth an HSC-intrinsic post-translational mechanism that is essential for reestablishing homeostasis after emergency hematopoiesis.
Collapse
Affiliation(s)
- Maria Guillamot
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA.,These authors contributed equally: Maria Guillamot, Dahmane Ouazia.,Correspondence and requests for materials should be addressed to M.G., L.B. or I.A., ; ;
| | - Dahmane Ouazia
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,These authors contributed equally: Maria Guillamot, Dahmane Ouazia
| | - Igor Dolgalev
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA.,Applied Bioinformatics Laboratories, Office of Science & Research, NYU School of Medicine, New York, NY, USA
| | - Stephen T. Yeung
- Department of Microbiology, NYU School of Medicine, New York, NY, USA
| | - Nikos Kourtis
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Yuling Dai
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Kate Corrigan
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Luna Zea-Redondo
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Anita Saraf
- The Stowers Institute of Medical Research, Kansas City, MO, USA
| | | | - Michael P. Washburn
- The Stowers Institute of Medical Research, Kansas City, MO, USA.,Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Anastasia N. Tikhonova
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Marina Malumbres
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Yixiao Gong
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Aristotelis Tsirigos
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA.,Applied Bioinformatics Laboratories, Office of Science & Research, NYU School of Medicine, New York, NY, USA
| | - Christopher Park
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Christopher Barbieri
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.,Department of Urology, Weill Cornell Medicine, New York, NY, USA
| | - Kamal M. Khanna
- Department of Microbiology, NYU School of Medicine, New York, NY, USA
| | - Luca Busino
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,These authors contributed equally: Maria Guillamot, Dahmane Ouazia.,These authors jointly supervised this work: Luca Busino and Iannis Aifantis.,Correspondence and requests for materials should be addressed to M.G., L.B. or I.A., ; ;
| | - Iannis Aifantis
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA.,These authors contributed equally: Maria Guillamot, Dahmane Ouazia.,These authors jointly supervised this work: Luca Busino and Iannis Aifantis.,Correspondence and requests for materials should be addressed to M.G., L.B. or I.A., ; ;
| |
Collapse
|
200
|
Hematopoietic stem cell response to acute thrombocytopenia requires signaling through distinct receptor tyrosine kinases. Blood 2019; 134:1046-1058. [PMID: 31434705 DOI: 10.1182/blood.2019000721] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/14/2019] [Indexed: 12/13/2022] Open
Abstract
Although bone marrow niche cells are essential for hematopoietic stem cell (HSC) maintenance, their interaction in response to stress is not well defined. Here, we used a mouse model of acute thrombocytopenia to investigate the cross talk between HSCs and niche cells during restoration of the thrombocyte pool. This process required membrane-localized stem cell factor (m-SCF) in megakaryocytes, which was regulated, in turn, by vascular endothelial growth factor A (VEGF-A) and platelet-derived growth factor-BB (PDGF-BB). HSCs and multipotent progenitors type 2 (MPP2), but not MPP3/4, were subsequently activated by a dual-receptor tyrosine kinase (RTK)-dependent signaling event, m-SCF/c-Kit and VEGF-A/vascular endothelial growth factor receptor 2 (VEGFR-2), contributing to their selective and early proliferation. Our findings describe a dynamic network of signals in response to the acute loss of a single blood cell type and reveal the important role of 3 RTKs and their ligands in orchestrating the selective activation of hematopoietic stem and progenitor cells (HSPCs) in thrombocytopenia.
Collapse
|