63 results on '"Sieweke MH"'
Search Results
2. Isolation, Ex Vivo Expansion, and Lentiviral Transduction of Alveolar Macrophages.
- Author
-
Busch CJ, Subramanian S, Linares J, Favret J, Yuda RAA, and Sieweke MH
- Subjects
- Animals, Mice, Thorax, Macrophages, Alveolar, Macrophages
- Abstract
Alveolar macrophages (AM) are resident macrophages of the lung and play important roles in the maintenance of tissue homeostasis as well as host defense. Here, we describe how they can be harvested from murine lungs, expanded in vitro, and transduced with lentiviral vectors., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
3. M-CSF directs myeloid and NK cell differentiation to protect from CMV after hematopoietic cell transplantation.
- Author
-
Kandalla PK, Subburayalu J, Cocita C, de Laval B, Tomasello E, Iacono J, Nitsche J, Canali MM, Cathou W, Bessou G, Mossadegh-Keller N, Huber C, Mouchiroud G, Bourette RP, Grasset MF, Bornhäuser M, Sarrazin S, Dalod M, and Sieweke MH
- Subjects
- Humans, Mice, Animals, Cytomegalovirus, Macrophage Colony-Stimulating Factor, Hematopoiesis, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Cell Differentiation, Hematopoietic Stem Cell Transplantation methods, Cytomegalovirus Infections prevention & control
- Abstract
Therapies reconstituting autologous antiviral immunocompetence may represent an important prophylaxis and treatment for immunosuppressed individuals. Following hematopoietic cell transplantation (HCT), patients are susceptible to Herpesviridae including cytomegalovirus (CMV). We show in a murine model of HCT that macrophage colony-stimulating factor (M-CSF) promoted rapid antiviral activity and protection from viremia caused by murine CMV. M-CSF given at transplantation stimulated sequential myeloid and natural killer (NK) cell differentiation culminating in increased NK cell numbers, production of granzyme B and interferon-γ. This depended upon M-CSF-induced myelopoiesis leading to IL15Rα-mediated presentation of IL-15 on monocytes, augmented by type I interferons from plasmacytoid dendritic cells. Demonstrating relevance to human HCT, M-CSF induced myelomonocytic IL15Rα expression and numbers of functional NK cells in G-CSF-mobilized hematopoietic stem and progenitor cells. Together, M-CSF-induced myelopoiesis triggered an integrated differentiation of myeloid and NK cells to protect HCT recipients from CMV. Thus, our results identify a rationale for the therapeutic use of M-CSF to rapidly reconstitute antiviral activity in immunocompromised individuals, which may provide a general paradigm to boost innate antiviral immunocompetence using host-directed therapies., (© 2023 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
4. [The dance between Brucella and hematopoietic stem cells].
- Author
-
Arce-Gorvel V, Hysenaj L, de Laval B, Sieweke MH, Sarrazin S, and Gorvel JP
- Subjects
- Humans, Hematopoietic Stem Cells, Brucella
- Published
- 2023
- Full Text
- View/download PDF
5. CD150-dependent hematopoietic stem cell sensing of Brucella instructs myeloid commitment.
- Author
-
Hysenaj L, de Laval B, Arce-Gorvel V, Bosilkovski M, González-Espinoza G, Debroas G, Sieweke MH, Sarrazin S, and Gorvel JP
- Subjects
- Hematopoietic Stem Cells metabolism, Bone Marrow, Bone Marrow Cells, Membrane Proteins metabolism, Cell Differentiation, Brucella
- Abstract
So far, hematopoietic stem cells (HSC) are considered the source of mature immune cells, the latter being the only ones capable of mounting an immune response. Recent evidence shows HSC can also directly sense cytokines released upon infection/inflammation and pathogen-associated molecular pattern interaction while keeping a long-term memory of previously encountered signals. Direct sensing of danger signals by HSC induces early myeloid commitment, increases myeloid effector cell numbers, and contributes to an efficient immune response. Here, by using specific genetic tools on both the host and pathogen sides, we show that HSC can directly sense B. abortus pathogenic bacteria within the bone marrow via the interaction of the cell surface protein CD150 with the bacterial outer membrane protein Omp25, inducing efficient functional commitment of HSC to the myeloid lineage. This is the first demonstration of direct recognition of a live pathogen by HSC via CD150, which attests to a very early contribution of HSC to immune response., (© 2023 Hysenaj et al.)
- Published
- 2023
- Full Text
- View/download PDF
6. Trained immunity and epigenetic memory in long-term self-renewing hematopoietic cells.
- Author
-
Johansson A, Lin DS, Mercier FE, Yamashita M, Divangahi M, and Sieweke MH
- Subjects
- Epigenetic Memory, Macrophages, Immunologic Memory genetics, Immunity, Innate genetics, Trained Immunity
- Abstract
Immunologic memory is a feature typically ascribed to the adaptive arm of the immune system. However, recent studies have demonstrated that hematopoietic stem cells (HSCs) and innate immune cells such as monocytes and macrophages can gain epigenetic signatures to enhance their response in the context of reinfection. This suggests the presence of long-term memory, a phenomenon referred to as trained immunity. Trained immunity in HSCs can occur via changes in the epigenetic landscape and enhanced chromatin accessibility in lineage-specific genes, as well as through metabolic alterations. These changes can lead to a skewing in lineage bias, particularly enhanced myelopoiesis and the generation of epigenetically modified innate immune cells that provide better protection against pathogens on secondary infection. Here, we summarize recent advancements in trained immunity and epigenetic memory formation in HSCs and self-renewing alveolar macrophages, which was the focus of the Spring 2022 International Society for Experimental Hematology (ISEH) webinar., (Copyright © 2023 ISEH -- Society for Hematology and Stem Cells. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
7. Network analysis of large-scale ImmGen and Tabula Muris datasets highlights metabolic diversity of tissue mononuclear phagocytes.
- Author
-
Gainullina A, Mogilenko DA, Huang LH, Todorov H, Narang V, Kim KW, Yng LS, Kent A, Jia B, Seddu K, Krchma K, Wu J, Crozat K, Tomasello E, Dress R, See P, Scott C, Gibbings S, Bajpai G, Desai JV, Maier B, This S, Wang P, Aguilar SV, Poupel L, Dussaud S, Zhou TA, Angeli V, Blander JM, Choi K, Dalod M, Dzhagalov I, Gautier EL, Jakubzick C, Lavine K, Lionakis MS, Paidassi H, Sieweke MH, Ginhoux F, Guilliams M, Benoist C, Merad M, Randolph GJ, Sergushichev A, and Artyomov MN
- Subjects
- Animals, Mice, Phagocytes, Single-Cell Analysis
- Abstract
The diversity of mononuclear phagocyte (MNP) subpopulations across tissues is one of the key physiological characteristics of the immune system. Here, we focus on understanding the metabolic variability of MNPs through metabolic network analysis applied to three large-scale transcriptional datasets: we introduce (1) an ImmGen MNP open-source dataset of 337 samples across 26 tissues; (2) a myeloid subset of ImmGen Phase I dataset (202 MNP samples); and (3) a myeloid mouse single-cell RNA sequencing (scRNA-seq) dataset (51,364 cells) assembled based on Tabula Muris Senis. To analyze such large-scale datasets, we develop a network-based computational approach, genes and metabolites (GAM) clustering, for unbiased identification of the key metabolic subnetworks based on transcriptional profiles. We define 9 metabolic subnetworks that encapsulate the metabolic differences within MNP from 38 different tissues. Obtained modules reveal that cholesterol synthesis appears particularly active within the migratory dendritic cells, while glutathione synthesis is essential for cysteinyl leukotriene production by peritoneal and lung macrophages., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
8. C/EBPβ-Dependent Epigenetic Memory Induces Trained Immunity in Hematopoietic Stem Cells.
- Author
-
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, and Sieweke MH
- Published
- 2023
- Full Text
- View/download PDF
9. Deciphering the heterogeneity of the Lyve1 + perivascular macrophages in the mouse brain.
- Author
-
Siret C, van Lessen M, Bavais J, Jeong HW, Reddy Samawar SK, Kapupara K, Wang S, Simic M, de Fabritus L, Tchoghandjian A, Fallet M, Huang H, Sarrazin S, Sieweke MH, Stumm R, Sorokin L, Adams RH, Schulte-Merker S, Kiefer F, and van de Pavert SA
- Subjects
- Animals, Mice, Leukocyte Count, Flow Cytometry, Brain, Macrophages, Phagocytes
- Abstract
Perivascular macrophages (pvMs) are associated with cerebral vasculature and mediate brain drainage and immune regulation. Here, using reporter mouse models, whole brain and section immunofluorescence, flow cytometry, and single cell RNA sequencing, besides the Lyve1
+ F4/80+ CD206+ CX3CR1+ pvMs, we identify a CX3CR1- pvM population that shares phagocytic functions and location. Furthermore, the brain parenchyma vasculature mostly hosts Lyve1+ MHCII- pvMs with low to intermediate CD45 expression. Using the double Cx3cr1GFP x Cx3cr1-Cre;RosatdT reporter mice for finer mapping of the lineages, we establish that CD45low CX3CR1- pvMs are derived from CX3CR1+ precursors and require PU.1 during their ontogeny. In parallel, results from the Cxcr4-CreErt2;Rosa26tdT lineage tracing model support a bone marrow-independent replenishment of all Lyve1+ pvMs in the adult mouse brain. Lastly, flow cytometry and 3D immunofluorescence analysis uncover increased percentage of pvMs following photothrombotic induced stroke. Our results thus show that the parenchymal pvM population is more heterogenous than previously described, and includes a CD45low and CX3CR1- pvM population., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
10. Autofluorescence identifies highly phagocytic tissue-resident macrophages in mouse and human skin and cutaneous squamous cell carcinoma.
- Author
-
Bourdely P, Petti L, Khou S, Meghraoui-Kheddar A, Elaldi R, Cazareth J, Mossadegh-Keller N, Boyer J, Sieweke MH, Poissonnet G, Sudaka A, Braud VM, and Anjuère F
- Subjects
- Adult, Humans, Animals, Mice, Phagocytosis, Macrophages pathology, Monocytes, Carcinoma, Squamous Cell pathology, Skin Neoplasms pathology
- Abstract
Macrophages from human and mouse skin share phenotypic and functional features, but remain to be characterized in pathological skin conditions. Skin-resident macrophages are known to derive from embryonic precursors or from adult hematopoiesis. In this report, we investigated the origins, phenotypes and functions of macrophage subsets in mouse and human skin and in cutaneous squamous cell carcinoma (cSCC) using the spectral flow cytometry technology that enables cell autofluorescence to be considered as a full-fledged parameter. Autofluorescence identifies macrophage subsets expressing the CD206 mannose receptor in human peri-tumoral skin and cSCC. In mouse, all AF
+ macrophages express the CD206 marker, a subset of which also displaying the TIM-4 marker. While TIM-4- CD206+ AF+ macrophages can differentiate from bone-marrow monocytes and infiltrate skin and tumor, TIM-4 identifies exclusively a skin-resident AF+ macrophage subset that can derive from prenatal hematopoiesis which is absent in tumor core. In mouse and human, AF+ macrophages from perilesional skin and cSCC are highly phagocytic cells contrary to their AF- counterpart, thus identifying autofluorescence as a bona fide marker for phagocytosis. Our data bring to light autofluorescence as a functional marker characterizing subsets of phagocytic macrophages in skin and cSCC. Autofluorescence can thus be considered as an attractive marker of function of macrophage subsets in pathological context., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Bourdely, Petti, Khou, Meghraoui-Kheddar, Elaldi, Cazareth, Mossadegh-Keller, Boyer, Sieweke, Poissonnet, Sudaka, Braud and Anjuère.)- Published
- 2022
- Full Text
- View/download PDF
11. Long-term culture-expanded alveolar macrophages restore their full epigenetic identity after transfer in vivo.
- Author
-
Subramanian S, Busch CJ, Molawi K, Geirsdottir L, Maurizio J, Vargas Aguilar S, Belahbib H, Gimenez G, Yuda RAA, Burkon M, Favret J, Gholamhosseinian Najjar S, de Laval B, Kandalla PK, Sarrazin S, Alexopoulou L, and Sieweke MH
- Subjects
- Animals, Chromatin metabolism, Epigenesis, Genetic, Epigenomics, Mice, Lung metabolism, Macrophages, Alveolar metabolism
- Abstract
Alveolar macrophages (AMs) are lung tissue-resident macrophages that can be expanded in culture, but it is unknown to what extent culture affects their in vivo identity. Here we show that mouse long-term ex vivo expanded AMs (exAMs) maintained a core AM gene expression program, but showed culture adaptations related to adhesion, metabolism and proliferation. Upon transplantation into the lung, exAMs reacquired full transcriptional and epigenetic AM identity, even after several months in culture and could self-maintain long-term in the alveolar niche. Changes in open chromatin regions observed in culture were fully reversible in transplanted exAMs and resulted in a gene expression profile indistinguishable from resident AMs. Our results indicate that long-term proliferation of AMs in culture did not compromise cellular identity in vivo. The robustness of exAM identity provides new opportunities for mechanistic analysis and highlights the therapeutic potential of exAMs., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
12. Author Correction: Trained immunity, tolerance, priming and differentiation: distinct immunological processes.
- Author
-
Divangahi M, Aaby P, Khader SA, Barreiro LB, Bekkering S, Chavakis T, van Crevel R, Curtis N, DiNardo AR, Dominguez-Andres J, Duivenvoorden R, Fanucchi S, Fayad Z, Fuchs E, Hamon M, Jeffrey KL, Khan N, Joosten LAB, Kaufmann E, Latz E, Matarese G, van der Meer JWM, Mhlanga M, Moorlag SJCFM, Mulder WJM, Naik S, Novakovic B, O'Neill L, Ochando J, Ozato K, Riksen NP, Sauerwein R, Sherwood ER, Schlitzer A, Schultze JL, Sieweke MH, Benn CS, Stunnenberg H, Sun J, van de Veerdonk FL, Weis S, Williams DL, Xavier R, and Netea MG
- Published
- 2021
- Full Text
- View/download PDF
13. TLR7 Signaling Drives the Development of Sjögren's Syndrome.
- Author
-
Wang Y, Roussel-Queval A, Chasson L, Hanna Kazazian N, Marcadet L, Nezos A, Sieweke MH, Mavragani C, and Alexopoulou L
- Subjects
- Adult, Aged, Animals, Chemokines genetics, Cytokines genetics, Female, Humans, Male, Mice, Mice, Inbred C57BL, Middle Aged, Pneumonia etiology, Signal Transduction physiology, Sjogren's Syndrome immunology, Sjogren's Syndrome etiology, Toll-Like Receptor 7 physiology
- Abstract
Sjögren's syndrome (SS) is a chronic systemic autoimmune disease that affects predominately salivary and lacrimal glands. SS can occur alone or in combination with another autoimmune disease like systemic lupus erythematosus (SLE). Here we report that TLR7 signaling drives the development of SS since TLR8-deficient (TLR8ko) mice that develop lupus due to increased TLR7 signaling by dendritic cells, also develop an age-dependent secondary pathology similar to associated SS. The SS phenotype in TLR8ko mice is manifested by sialadenitis, increased anti-SSA and anti-SSB autoantibody production, immune complex deposition and increased cytokine production in salivary glands, as well as lung inflammation. Moreover, ectopic lymphoid structures characterized by B/T aggregates, formation of high endothelial venules and the presence of dendritic cells are formed in the salivary glands of TLR8ko mice. Interestingly, all these phenotypes are abrogated in double TLR7/8-deficient mice, suggesting that the SS phenotype in TLR8-deficient mice is TLR7-dependent. In addition, evaluation of TLR7 and inflammatory markers in the salivary glands of primary SS patients revealed significantly increased TLR7 expression levels compared to healthy individuals, that were positively correlated to TNF , LT-α , CXCL13 and CXCR5 expression. These findings establish an important role of TLR7 signaling for local and systemic SS disease manifestations, and inhibition of such will likely have therapeutic value., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Wang, Roussel-Queval, Chasson, Hanna Kazazian, Marcadet, Nezos, Sieweke, Mavragani and Alexopoulou.)
- Published
- 2021
- Full Text
- View/download PDF
14. Trained immunity, tolerance, priming and differentiation: distinct immunological processes.
- Author
-
Divangahi M, Aaby P, Khader SA, Barreiro LB, Bekkering S, Chavakis T, van Crevel R, Curtis N, DiNardo AR, Dominguez-Andres J, Duivenvoorden R, Fanucchi S, Fayad Z, Fuchs E, Hamon M, Jeffrey KL, Khan N, Joosten LAB, Kaufmann E, Latz E, Matarese G, van der Meer JWM, Mhlanga M, Moorlag SJCFM, Mulder WJM, Naik S, Novakovic B, O'Neill L, Ochando J, Ozato K, Riksen NP, Sauerwein R, Sherwood ER, Schlitzer A, Schultze JL, Sieweke MH, Benn CS, Stunnenberg H, Sun J, van de Veerdonk FL, Weis S, Williams DL, Xavier R, and Netea MG
- Subjects
- Animals, BCG Vaccine immunology, Cell Differentiation, Humans, Vaccination, Adaptive Immunity immunology, Immune Tolerance immunology, Immunity, Innate immunology, Immunologic Memory immunology
- Published
- 2021
- Full Text
- View/download PDF
15. C/EBPβ-Dependent Epigenetic Memory Induces Trained Immunity in Hematopoietic Stem Cells.
- Author
-
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, and Sieweke MH
- Published
- 2020
- Full Text
- View/download PDF
16. C/EBPβ-Dependent Epigenetic Memory Induces Trained Immunity in Hematopoietic Stem Cells.
- Author
-
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, and Sieweke MH
- Subjects
- Epigenomics, Humans, Myelopoiesis, CCAAT-Enhancer-Binding Protein-beta genetics, Epigenesis, Genetic, Hematopoietic Stem Cells immunology, Immunity, Innate
- 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., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
17. Tissue-resident macrophages in omentum promote metastatic spread of ovarian cancer.
- Author
-
Etzerodt A, Moulin M, Doktor TK, Delfini M, Mossadegh-Keller N, Bajenoff M, Sieweke MH, Moestrup SK, Auphan-Anezin N, and Lawrence T
- Subjects
- Animals, Antigens, CD genetics, Antigens, CD metabolism, Antigens, Differentiation, Myelomonocytic genetics, Antigens, Differentiation, Myelomonocytic metabolism, Disease Models, Animal, Disease Progression, Female, Gene Expression Profiling, Membrane Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Phenotype, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism, Transcriptome, Macrophages metabolism, Omentum metabolism, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Peritoneal Neoplasms metabolism, Peritoneal Neoplasms secondary
- Abstract
Experimental and clinical evidence suggests that tumor-associated macrophages (TAMs) play important roles in cancer progression. Here, we have characterized the ontogeny and function of TAM subsets in a mouse model of metastatic ovarian cancer that is representative for visceral peritoneal metastasis. We show that the omentum is a critical premetastatic niche for development of invasive disease in this model and define a unique subset of CD163+ Tim4+ resident omental macrophages responsible for metastatic spread of ovarian cancer cells. Transcriptomic analysis showed that resident CD163+ Tim4+ omental macrophages were phenotypically distinct and maintained their resident identity during tumor growth. Selective depletion of CD163+ Tim4+ macrophages in omentum using genetic and pharmacological tools prevented tumor progression and metastatic spread of disease. These studies describe a specific role for tissue-resident macrophages in the invasive progression of metastatic ovarian cancer. The molecular pathways of cross-talk between tissue-resident macrophages and disseminated cancer cells may represent new targets to prevent metastasis and disease recurrence., Competing Interests: Disclosures: Dr. Etzerodt reported personal fees from Stipe Therapeutics outside the submitted work. No other disclosures were reported., (© 2020 Etzerodt et al.)
- Published
- 2020
- Full Text
- View/download PDF
18. Bhlhe40 and Bhlhe41 transcription factors regulate alveolar macrophage self-renewal and identity.
- Author
-
Rauschmeier R, Gustafsson C, Reinhardt A, A-Gonzalez N, Tortola L, Cansever D, Subramanian S, Taneja R, Rossner MJ, Sieweke MH, Greter M, Månsson R, Busslinger M, and Kreslavsky T
- Subjects
- Acetylation, Animals, Basic Helix-Loop-Helix Transcription Factors metabolism, Cell Differentiation, Cell Proliferation, Cell Self Renewal, Cell Survival, Down-Regulation, Gene Knockdown Techniques, Histones metabolism, Homeodomain Proteins metabolism, Macrophages, Alveolar metabolism, Mice, Organ Specificity, Phenotype, Sequence Analysis, RNA, Basic Helix-Loop-Helix Transcription Factors genetics, Gene Expression Profiling methods, Homeodomain Proteins genetics, Macrophages, Alveolar cytology
- Abstract
Tissues in multicellular organisms are populated by resident macrophages, which perform both generic and tissue-specific functions. The latter are induced by signals from the microenvironment and rely on unique tissue-specific molecular programs requiring the combinatorial action of tissue-specific and broadly expressed transcriptional regulators. Here, we identify the transcription factors Bhlhe40 and Bhlhe41 as novel regulators of alveolar macrophages (AMs)-a population that provides the first line of immune defense and executes homeostatic functions in lung alveoli. In the absence of these factors, AMs exhibited decreased proliferation that resulted in a severe disadvantage of knockout AMs in a competitive setting. Gene expression analyses revealed a broad cell-intrinsic footprint of Bhlhe40/Bhlhe41 deficiency manifested by a downregulation of AM signature genes and induction of signature genes of other macrophage lineages. Genome-wide characterization of Bhlhe40 DNA binding suggested that these transcription factors directly repress the expression of lineage-inappropriate genes in AMs. Taken together, these results identify Bhlhe40 and Bhlhe41 as key regulators of AM self-renewal and guardians of their identity., (© 2019 The Authors.)
- Published
- 2019
- Full Text
- View/download PDF
19. Isolation and Long-term Cultivation of Mouse Alveolar Macrophages.
- Author
-
Busch CJ, Favret J, Geirsdóttir L, Molawi K, and Sieweke MH
- Abstract
Alveolar macrophages (AM) are tissue-resident macrophages that colonize the lung around birth and can self-maintain long-term in an adult organism without contribution of monocytes. AM are located in the pulmonary alveoli and can be harvested by washing the lungs using the method of bronchoalveolar lavage (BAL). Here, we compared different conditions of BAL to obtain high yields of murine AM for in vitro culture and expansion of AM. In addition, we describe specific culture conditions, under which AM proliferate long-term in liquid culture in the presence of granulocyte-macrophage colony-stimulating factor. This method can be used to obtain large numbers of AM for in vivo transplantation or for in vitro experiments with primary mouse macrophages., Competing Interests: Competing interests The authors declare no competing financial interests.
- Published
- 2019
- Full Text
- View/download PDF
20. Characterization of Mouse Adult Testicular Macrophage Populations by Immunofluorescence Imaging and Flow Cytometry.
- Author
-
Mossadegh-Keller N and Sieweke MH
- Abstract
Testicular macrophages (tMΦ) are the most abundant immune cells residing in the testis, an immune-privileged organ. TMΦ are known to exhibit different functions, such as protecting spermatozoa from auto-immune attack by producing immunosuppressive cytokines and trophic roles in supporting spermatogenesis and male sex hormone production. They also contribute to fetal testicular development. Recently, we characterized two distinct tMΦ populations based on their morphology, localization, cell surface markers, and gene expression profiling. Here, we focus and describe in detail the phenotypical distinction of these two tMΦ populations by fluorescence-activated cell sorting (FACS) using multicolor panel antibodies combining with high-resolution immunofluorescence (IF) imaging. These two techniques enable to classify two tMΦ populations: interstitial tMΦ and peritubular tMΦ., Competing Interests: Competing interests The authors declare no competing financial interests.
- Published
- 2019
- Full Text
- View/download PDF
21. Publisher Correction: c-Maf controls immune responses by regulating disease-specific gene networks and repressing IL-2 in CD4 + T cells.
- Author
-
Gabryšová L, Alvarez-Martinez M, Luisier R, Cox LS, Sodenkamp J, Hosking C, Pérez-Mazliah D, Whicher C, Kannan Y, Potempa K, Wu X, Bhaw L, Wende H, Sieweke MH, Elgar G, Wilson M, Briscoe J, Metzis V, Langhorne J, Luscombe NM, and O'Garra A
- Abstract
In the version of this article initially published, the Supplementary Data file was an incorrect version. The correct version is now provided. The error has been corrected in the HTML and PDF version of the article.
- Published
- 2019
- Full Text
- View/download PDF
22. DNA Damage Signaling Instructs Polyploid Macrophage Fate in Granulomas.
- Author
-
Herrtwich L, Nanda I, Evangelou K, Nikolova T, Horn V, Sagar, Erny D, Stefanowski J, Rogell L, Klein C, Gharun K, Follo M, Seidl M, Kremer B, Münke N, Senges J, Fliegauf M, Aschman T, Pfeifer D, Sarrazin S, Sieweke MH, Wagner D, Dierks C, Haaf T, Ness T, Zaiss MM, Voll RE, Deshmukh SD, Prinz M, Goldmann T, Hölscher C, Hauser AE, Lopez-Contreras AJ, Grün D, Gorgoulis V, Diefenbach A, Henneke P, and Triantafyllopoulou A
- Published
- 2018
- Full Text
- View/download PDF
23. Testicular macrophages: Guardians of fertility.
- Author
-
Mossadegh-Keller N and Sieweke MH
- Subjects
- Animals, Humans, Immunity immunology, Macrophages metabolism, Male, Models, Immunological, Spermatogenesis immunology, Testis cytology, Testis metabolism, Testosterone metabolism, Fertility immunology, Macrophages immunology, Testis immunology, Testosterone immunology
- Abstract
Macrophages are innate immune cells present in essentially every organ of the body with dedicated tissue specific functions. We will present in this review the unique properties and functions of macrophage populations residing in the testis, an immune-privileged organ. Testicular macrophages (tMΦ) could be seen as guardians of fertility due to their immunosuppressive functions protecting spermatogenesis from auto immune-attack. They exhibit testis specific functions with essential roles in normal testis homeostasis and fetal testicular development. Recently, two distinct testicular macrophage populations have been characterized based on different localization, morphology, gene expression profiles, developmental origin and postnatal development. We will discuss the importance of these two testicular macrophage populations for organ specific functions such as testosterone production and spermatogenesis, as well as their role in establishing immuno-privilege highlighting the contributions of macrophages to male fertility., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
24. c-Maf controls immune responses by regulating disease-specific gene networks and repressing IL-2 in CD4 + T cells.
- Author
-
Gabryšová L, Alvarez-Martinez M, Luisier R, Cox LS, Sodenkamp J, Hosking C, Pérez-Mazliah D, Whicher C, Kannan Y, Potempa K, Wu X, Bhaw L, Wende H, Sieweke MH, Elgar G, Wilson M, Briscoe J, Metzis V, Langhorne J, Luscombe NM, and O'Garra A
- Subjects
- Animals, Interleukin-2 immunology, Mice, CD4-Positive T-Lymphocytes immunology, Gene Expression Regulation immunology, Gene Regulatory Networks immunology, Interleukin-2 biosynthesis, Proto-Oncogene Proteins c-maf immunology
- Abstract
The transcription factor c-Maf induces the anti-inflammatory cytokine IL-10 in CD4
+ T cells in vitro. However, the global effects of c-Maf on diverse immune responses in vivo are unknown. Here we found that c-Maf regulated IL-10 production in CD4+ T cells in disease models involving the TH 1 subset of helper T cells (malaria), TH 2 cells (allergy) and TH 17 cells (autoimmunity) in vivo. Although mice with c-Maf deficiency targeted to T cells showed greater pathology in TH 1 and TH 2 responses, TH 17 cell-mediated pathology was reduced in this context, with an accompanying decrease in TH 17 cells and increase in Foxp3+ regulatory T cells. Bivariate genomic footprinting elucidated the c-Maf transcription-factor network, including enhanced activity of NFAT; this led to the identification and validation of c-Maf as a negative regulator of IL-2. The decreased expression of the gene encoding the transcription factor RORγt (Rorc) that resulted from c-Maf deficiency was dependent on IL-2, which explained the in vivo observations. Thus, c-Maf is a positive and negative regulator of the expression of cytokine-encoding genes, with context-specific effects that allow each immune response to occur in a controlled yet effective manner.- Published
- 2018
- Full Text
- View/download PDF
25. Trained macrophages support hygiene hypothesis.
- Author
-
de Laval B and Sieweke MH
- Subjects
- Humans, Hygiene Hypothesis, Macrophages, Macrophages, Alveolar, Monocytes, Asthma, Hypersensitivity
- Published
- 2017
- Full Text
- View/download PDF
26. Developmental origin and maintenance of distinct testicular macrophage populations.
- Author
-
Mossadegh-Keller N, Gentek R, Gimenez G, Bigot S, Mailfert S, and Sieweke MH
- Subjects
- Animals, Cell Proliferation physiology, Flow Cytometry, Gene Expression Profiling, Immunity, Cellular physiology, Macrophages cytology, Macrophages metabolism, Male, Mice, Mice, Inbred C57BL, Real-Time Polymerase Chain Reaction, Spermatogenesis physiology, Stem Cells physiology, Testis immunology, Testis metabolism, Macrophages physiology, Testis cytology
- Abstract
Testicular macrophages (tMφ) are the principal immune cells of the mammalian testis. Beyond classical immune functions, they have been shown to be important for organogenesis, spermatogenesis, and male hormone production. In the adult testis, two different macrophage populations have been identified based on their distinct tissue localization and morphology, but their developmental origin and mode of homeostatic maintenance are unknown. In this study, we use genetic lineage-tracing models and adoptive transfer protocols to address this question. We show that embryonic progenitors give rise to the interstitial macrophage population, whereas peritubular macrophages are exclusively seeded postnatally in the prepuberty period from bone marrow (BM)-derived progenitors. As the proliferative capacity of interstitial macrophages declines, BM progenitors also contribute to this population. Once established, both the peritubular and interstitial macrophage populations exhibit a long life span and a low turnover in the steady state. Our observations identify distinct developmental pathways for two different tMφ populations that have important implications for the further dissection of their distinct roles in organ homeostasis and testicular function., (© 2017 Mossadegh-Keller et al.)
- Published
- 2017
- Full Text
- View/download PDF
27. SIRT1 regulates macrophage self-renewal.
- Author
-
Imperatore F, Maurizio J, Vargas Aguilar S, Busch CJ, Favret J, Kowenz-Leutz E, Cathou W, Gentek R, Perrin P, Leutz A, Berruyer C, and Sieweke MH
- Subjects
- Animals, Cell Cycle, Gene Expression, Gene Knockdown Techniques, Gene Knockout Techniques, Mice, Sirtuin 1 genetics, Cell Proliferation, Cell Self Renewal, Macrophages physiology, Sirtuin 1 metabolism
- Abstract
Mature differentiated macrophages can self-maintain by local proliferation in tissues and can be extensively expanded in culture under specific conditions, but the mechanisms of this phenomenon remain only partially defined. Here, we show that SIRT1, an evolutionary conserved regulator of life span, positively affects macrophage self-renewal ability in vitro and in vivo Overexpression of SIRT1 during bone marrow-derived macrophage differentiation increased their proliferative capacity. Conversely, decrease of SIRT1 expression by shRNA inactivation, CRISPR/Cas9 mediated deletion and pharmacological inhibition restricted macrophage self-renewal in culture. Furthermore, pharmacological SIRT1 inhibition in vivo reduced steady state and cytokine-induced proliferation of alveolar and peritoneal macrophages. Mechanistically, SIRT1 inhibition negatively regulated G1/S transition, cell cycle progression and a network of self-renewal genes. This included inhibition of E2F1 and Myc and concomitant activation of FoxO1, SIRT1 targets mediating cell cycle progression and stress response, respectively. Our findings indicate that SIRT1 is a key regulator of macrophage self-renewal that integrates cell cycle and longevity pathways. This suggests that macrophage self-renewal might be a relevant parameter of ageing., (© 2017 The Authors.)
- Published
- 2017
- Full Text
- View/download PDF
28. DNA Damage Signaling Instructs Polyploid Macrophage Fate in Granulomas.
- Author
-
Herrtwich L, Nanda I, Evangelou K, Nikolova T, Horn V, Sagar, Erny D, Stefanowski J, Rogell L, Klein C, Gharun K, Follo M, Seidl M, Kremer B, Münke N, Senges J, Fliegauf M, Aschman T, Pfeifer D, Sarrazin S, Sieweke MH, Wagner D, Dierks C, Haaf T, Ness T, Zaiss MM, Voll RE, Deshmukh SD, Prinz M, Goldmann T, Hölscher C, Hauser AE, Lopez-Contreras AJ, Grün D, Gorgoulis V, Diefenbach A, Henneke P, and Triantafyllopoulou A
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins metabolism, Cell Differentiation, Cell Proliferation, Humans, Inflammation immunology, Lipoproteins immunology, Mice, Mice, Inbred C57BL, Mitosis, Proto-Oncogene Proteins c-myc metabolism, Toll-Like Receptor 2, DNA Damage, Granuloma immunology, Macrophages immunology, Mycobacterium tuberculosis immunology
- Abstract
Granulomas are immune cell aggregates formed in response to persistent inflammatory stimuli. Granuloma macrophage subsets are diverse and carry varying copy numbers of their genomic information. The molecular programs that control the differentiation of such macrophage populations in response to a chronic stimulus, though critical for disease outcome, have not been defined. Here, we delineate a macrophage differentiation pathway by which a persistent Toll-like receptor (TLR) 2 signal instructs polyploid macrophage fate by inducing replication stress and activating the DNA damage response. Polyploid granuloma-resident macrophages formed via modified cell divisions and mitotic defects and not, as previously thought, by cell-to-cell fusion. TLR2 signaling promoted macrophage polyploidy and suppressed genomic instability by regulating Myc and ATR. We propose that, in the presence of persistent inflammatory stimuli, pathways previously linked to oncogene-initiated carcinogenesis instruct a long-lived granuloma-resident macrophage differentiation program that regulates granulomatous tissue remodeling., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
29. Efficient CRISPR-mediated mutagenesis in primary immune cells using CrispRGold and a C57BL/6 Cas9 transgenic mouse line.
- Author
-
Chu VT, Graf R, Wirtz T, Weber T, Favret J, Li X, Petsch K, Tran NT, Sieweke MH, Berek C, Kühn R, and Rajewsky K
- Subjects
- Animals, Cell Differentiation genetics, Cells, Cultured, Lymphocyte Activation genetics, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Plasma Cells metabolism, Reproducibility of Results, B-Lymphocytes metabolism, CRISPR-Cas Systems, Gene Editing methods, Macrophages metabolism, Mutagenesis, T-Lymphocytes metabolism
- Abstract
Applying clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 (Cas9)-mediated mutagenesis to primary mouse immune cells, we used high-fidelity single guide RNAs (sgRNAs) designed with an sgRNA design tool (CrispRGold) to target genes in primary B cells, T cells, and macrophages isolated from a Cas9 transgenic mouse line. Using this system, we achieved an average knockout efficiency of 80% in B cells. On this basis, we established a robust small-scale CRISPR-mediated screen in these cells and identified genes essential for B-cell activation and plasma cell differentiation. This screening system does not require deep sequencing and may serve as a precedent for the application of CRISPR/Cas9 to primary mouse cells., Competing Interests: The authors declare no conflict of interest.
- Published
- 2016
- Full Text
- View/download PDF
30. M-CSF improves protection against bacterial and fungal infections after hematopoietic stem/progenitor cell transplantation.
- Author
-
Kandalla PK, Sarrazin S, Molawi K, Berruyer C, Redelberger D, Favel A, Bordi C, de Bentzmann S, and Sieweke MH
- Subjects
- Animals, Aspergillosis blood, Aspergillosis microbiology, Aspergillus drug effects, Aspergillus physiology, Blood Glucose metabolism, Blood Platelets drug effects, Blood Platelets metabolism, Cell Differentiation drug effects, Cell Lineage drug effects, Cell Self Renewal drug effects, Hematopoietic Stem Cells drug effects, Humans, Macrophage Colony-Stimulating Factor pharmacology, Mice, Inbred C57BL, Myelopoiesis drug effects, Pseudomonas Infections blood, Pseudomonas Infections microbiology, Pseudomonas aeruginosa drug effects, Pseudomonas aeruginosa physiology, Aspergillosis drug therapy, Aspergillosis prevention & control, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells cytology, Macrophage Colony-Stimulating Factor therapeutic use, Pseudomonas Infections drug therapy, Pseudomonas Infections prevention & control
- Abstract
Myeloablative treatment preceding hematopoietic stem cell (HSC) and progenitor cell (HS/PC) transplantation results in severe myeloid cytopenia and susceptibility to infections in the lag period before hematopoietic recovery. We have previously shown that macrophage colony-stimulating factor (CSF-1; M-CSF) directly instructed myeloid commitment in HSCs. In this study, we tested whether this effect had therapeutic benefit in improving protection against pathogens after HS/PC transplantation. M-CSF treatment resulted in an increased production of mature myeloid donor cells and an increased survival of recipient mice infected with lethal doses of clinically relevant opportunistic pathogens, namely the bacteria Pseudomonas aeruginosa and the fungus Aspergillus fumigatus M-CSF treatment during engraftment or after infection efficiently protected from these pathogens as early as 3 days after transplantation and was effective as a single dose. It was more efficient than granulocyte CSF (G-CSF), a common treatment of severe neutropenia, which showed no protective effect under the tested conditions. M-CSF treatment showed no adverse effect on long-term lineage contribution or stem cell activity and, unlike G-CSF, did not impede recovery of HS/PCs, thrombocyte numbers, or glucose metabolism. These results encourage potential clinical applications of M-CSF to prevent severe infections after HS/PC transplantation., (© 2016 Kandalla et al.)
- Published
- 2016
- Full Text
- View/download PDF
31. Microglia development follows a stepwise program to regulate brain homeostasis.
- Author
-
Matcovitch-Natan O, Winter DR, Giladi A, Vargas Aguilar S, Spinrad A, Sarrazin S, Ben-Yehuda H, David E, Zelada González F, Perrin P, Keren-Shaul H, Gury M, Lara-Astaiso D, Thaiss CA, Cohen M, Bahar Halpern K, Baruch K, Deczkowska A, Lorenzo-Vivas E, Itzkovitz S, Elinav E, Sieweke MH, Schwartz M, and Amit I
- Subjects
- Animals, Blood-Brain Barrier embryology, Blood-Brain Barrier immunology, Brain immunology, Chromatin metabolism, Epigenesis, Genetic, Female, Gene Expression Profiling, Gene Knockout Techniques, Histone Code, Homeostasis genetics, Immunity genetics, MafB Transcription Factor genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Microglia immunology, Myeloid Cells cytology, Neurogenesis genetics, Single-Cell Analysis, Brain embryology, Homeostasis physiology, Microglia cytology, Neurogenesis immunology
- Abstract
Microglia, the resident myeloid cells of the central nervous system, play important roles in life-long brain maintenance and in pathology. Despite their importance, their regulatory dynamics during brain development have not been fully elucidated. Using genome-wide chromatin and expression profiling coupled with single-cell transcriptomic analysis throughout development, we found that microglia undergo three temporal stages of development in synchrony with the brain--early, pre-, and adult microglia--which are under distinct regulatory circuits. Knockout of the gene encoding the adult microglia transcription factor MAFB and environmental perturbations, such as those affecting the microbiome or prenatal immune activation, led to disruption of developmental genes and immune response pathways. Together, our work identifies a stepwise microglia developmental program integrating immune response pathways that may be associated with several neurodevelopmental disorders., (Copyright © 2016, American Association for the Advancement of Science.)
- Published
- 2016
- Full Text
- View/download PDF
32. Eosinophils and mast cells: a lineage apart.
- Author
-
Sarrazin S and Sieweke MH
- Subjects
- Humans, Eosinophils, Mast Cells
- Published
- 2016
- Full Text
- View/download PDF
33. Lineage-specific enhancers activate self-renewal genes in macrophages and embryonic stem cells.
- Author
-
Soucie EL, Weng Z, Geirsdóttir L, Molawi K, Maurizio J, Fenouil R, Mossadegh-Keller N, Gimenez G, VanHille L, Beniazza M, Favret J, Berruyer C, Perrin P, Hacohen N, Andrau JC, Ferrier P, Dubreuil P, Sidow A, and Sieweke MH
- Subjects
- Animals, Cell Proliferation, Cells, Cultured, Down-Regulation, Gene Regulatory Networks, MafB Transcription Factor metabolism, Mice, Proto-Oncogene Proteins c-maf metabolism, Single-Cell Analysis, Transcriptional Activation, Cell Differentiation genetics, Cell Lineage genetics, Embryonic Stem Cells cytology, Enhancer Elements, Genetic physiology, Gene Expression Regulation, Macrophages cytology
- Abstract
Differentiated macrophages can self-renew in tissues and expand long term in culture, but the gene regulatory mechanisms that accomplish self-renewal in the differentiated state have remained unknown. Here we show that in mice, the transcription factors MafB and c-Maf repress a macrophage-specific enhancer repertoire associated with a gene network that controls self-renewal. Single-cell analysis revealed that, in vivo, proliferating resident macrophages can access this network by transient down-regulation of Maf transcription factors. The network also controls embryonic stem cell self-renewal but is associated with distinct embryonic stem cell-specific enhancers. This indicates that distinct lineage-specific enhancer platforms regulate a shared network of genes that control self-renewal potential in both stem and mature cells., (Copyright © 2016, American Association for the Advancement of Science.)
- Published
- 2016
- Full Text
- View/download PDF
34. Molecular profiling of CD8 T cells in autochthonous melanoma identifies Maf as driver of exhaustion.
- Author
-
Giordano M, Henin C, Maurizio J, Imbratta C, Bourdely P, Buferne M, Baitsch L, Vanhille L, Sieweke MH, Speiser DE, Auphan-Anezin N, Schmitt-Verhulst AM, and Verdeil G
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, DNA Primers genetics, Flow Cytometry, Gene Expression Profiling, Homeodomain Proteins genetics, Interleukin-6 metabolism, Luciferases, Mice, Mice, Transgenic, Proto-Oncogene Proteins c-maf genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Transforming Growth Factor beta metabolism, CD8-Positive T-Lymphocytes cytology, Cell Differentiation immunology, Melanoma metabolism, Proto-Oncogene Proteins c-maf metabolism, Tumor Microenvironment physiology
- Abstract
T cells infiltrating neoplasms express surface molecules typical of chronically virus-stimulated T cells, often termed "exhausted" T cells. We compared the transcriptome of "exhausted" CD8 T cells infiltrating autochthonous melanomas to those of naïve and acutely stimulated CD8 T cells. Despite strong similarities between transcriptional signatures of tumor- and virus-induced exhausted CD8 T cells, notable differences appeared. Among transcriptional regulators, Nr4a2 and Maf were highly overexpressed in tumor-exhausted T cells and significantly upregulated in CD8 T cells from human melanoma metastases. Transduction of murine tumor-specific CD8 T cells to express Maf partially reproduced the transcriptional program associated with tumor-induced exhaustion. Upon adoptive transfer, the transduced cells showed normal homeostasis but failed to accumulate in tumor-bearing hosts and developed defective anti-tumor effector responses. We further identified TGFβ and IL-6 as main inducers of Maf expression in CD8 T cells and showed that Maf-deleted tumor-specific CD8 T cells were much more potent to restrain tumor growth in vivo. Therefore, the melanoma microenvironment contributes to skewing of CD8 T cell differentiation programs, in part by TGFβ/IL-6-mediated induction of Maf., (© 2015 The Authors.)
- Published
- 2015
- Full Text
- View/download PDF
35. Monocytes compensate Kupffer cell loss during bacterial infection.
- Author
-
Molawi K and Sieweke MH
- Subjects
- Animals, Kupffer Cells physiology, Listeria monocytogenes immunology, Listeriosis immunology, Liver pathology, Monocytes immunology
- Abstract
Liver Kupffer cells (KCs) are self-maintained tissue-resident macrophages. In this issue of Immunity, Blériot et al. demonstrate that bacterial infection leads to KC necroptosis and quantitative replacement by monocyte-derived macrophages that contribute to antibacterial immunity and restoration of tissue integrity., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
36. Waddington's valleys and Captain Cook's islands.
- Author
-
Sieweke MH
- Abstract
Somatic reprogramming has relied heavily on theoretical models that view differentiation in terms of developmental branch point decisions. Recent studies in Cell now reveal a dominant role of the microenvironment in shaping epigenetic identity of macrophages, thus providing support for alternative models of cell fate acquisition., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
37. Tissue macrophage identity and self-renewal.
- Author
-
Gentek R, Molawi K, and Sieweke MH
- Subjects
- Animals, Cell Differentiation, Cell Proliferation, Humans, Macrophages immunology, Organ Specificity, Macrophages cytology, Macrophages metabolism, Phenotype
- Abstract
Macrophages are cellular components of the innate immune system that reside in virtually all tissues and contribute to immunity, repair, and homeostasis. The traditional view that all tissue-resident macrophages derive from the bone marrow through circulating monocyte intermediates has dramatically shifted recently with the observation that macrophages from embryonic progenitors can persist into adulthood and self-maintain by local proliferation. In several tissues, however, monocytes also contribute to the resident macrophage population, on which the local environment can impose tissue-specific macrophage functions. These observations have raised important questions: What determines resident macrophage identity and function, ontogeny or environment? How is macrophage proliferation regulated? In this review, we summarize the current knowledge about the identity, proliferation, and turnover of tissue-resident macrophages and how they differ from freshly recruited short-lived monocyte-derived cells. We examine whether macrophage proliferation can be qualified as self-renewal of mature differentiated cells and whether the concepts and molecular pathways are comparable to self-renewal mechanisms in stem cells. Finally, we discuss how improved understanding of macrophage identity and self-renewal could be exploited for therapeutic intervention of macrophage-mediated pathologies by selectively targeting freshly recruited or resident macrophages., (© 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
38. Progressive replacement of embryo-derived cardiac macrophages with age.
- Author
-
Molawi K, Wolf Y, Kandalla PK, Favret J, Hagemeyer N, Frenzel K, Pinto AR, Klapproth K, Henri S, Malissen B, Rodewald HR, Rosenthal NA, Bajenoff M, Prinz M, Jung S, and Sieweke MH
- Subjects
- Age Factors, Animals, Animals, Newborn, Antigens, Surface metabolism, Cell Differentiation, Cell Proliferation, Female, Immunophenotyping, Mice, Mice, Transgenic, Monocytes cytology, Monocytes metabolism, Phenotype, Macrophages cytology, Macrophages metabolism, Myocardium cytology
- Abstract
Cardiac macrophages (cMΦ) are critical for early postnatal heart regeneration and fibrotic repair in the adult heart, but their origins and cellular dynamics during postnatal development have not been well characterized. Tissue macrophages can be derived from embryonic progenitors or from monocytes during inflammation. We report that within the first weeks after birth, the embryo-derived population of resident CX3CR1(+) cMΦ diversifies into MHCII(+) and MHCII(-) cells. Genetic fate mapping demonstrated that cMΦ derived from CX3CR1(+) embryonic progenitors persisted into adulthood but the initially high contribution to resident cMΦ declined after birth. Consistent with this, the early significant proliferation rate of resident cMΦ decreased with age upon diversification into subpopulations. Bone marrow (BM) reconstitution experiments showed monocyte-dependent quantitative replacement of all cMΦ populations. Furthermore, parabiotic mice and BM chimeras of nonirradiated recipient mice revealed a slow but significant donor contribution to cMΦ. Together, our observations indicate that in the heart, embryo-derived cMΦ show declining self-renewal with age and are progressively substituted by monocyte-derived macrophages, even in the absence of inflammation., (© 2014 Molawi et al.)
- Published
- 2014
- Full Text
- View/download PDF
39. Design of a bZip transcription factor with homo/heterodimer-induced DNA-binding preference.
- Author
-
Pogenberg V, Consani Textor L, Vanhille L, Holton SJ, Sieweke MH, and Wilmanns M
- Subjects
- Amino Acid Sequence, Animals, Basic-Leucine Zipper Transcription Factors chemistry, Basic-Leucine Zipper Transcription Factors metabolism, Binding Sites, DNA metabolism, MafB Transcription Factor genetics, Mice, Models, Molecular, Molecular Sequence Data, Mutation, Protein Conformation, Protein Engineering methods, Protein Multimerization, Proto-Oncogene Proteins c-fos chemistry, Proto-Oncogene Proteins c-fos genetics, Proto-Oncogene Proteins c-fos metabolism, MafB Transcription Factor chemistry, MafB Transcription Factor metabolism
- Abstract
The ability of basic leucine zipper transcription factors for homo- or heterodimerization provides a paradigm for combinatorial control of eukaryotic gene expression. It has been unclear, however, how facultative dimerization results in alternative DNA-binding repertoires on distinct regulatory elements. To unravel the molecular basis of such coupled preferences, we determined two high-resolution structures of the transcription factor MafB as a homodimer and as a heterodimer with c-Fos bound to variants of the Maf-recognition element. The structures revealed several unexpected and dimer-specific coiled-coil-heptad interactions. Based on these findings, we have engineered two MafB mutants with opposite dimerization preferences. One of them showed a strong preference for MafB/c-Fos heterodimerization and enabled selection of heterodimer-favoring over homodimer-specific Maf-recognition element variants. Our data provide a concept for transcription factor design to selectively activate dimer-specific pathways and binding repertoires., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
40. Beyond stem cells: self-renewal of differentiated macrophages.
- Author
-
Sieweke MH and Allen JE
- Subjects
- Animals, Cell Proliferation, Cytokines metabolism, Embryonic Stem Cells cytology, Humans, Mice, Monocytes cytology, Rats, Signal Transduction, Cell Differentiation, Macrophages cytology, Stem Cells cytology
- Abstract
In many mammalian tissues, mature differentiated cells are replaced by self-renewing stem cells, either continuously during homeostasis or in response to challenge and injury. For example, hematopoietic stem cells generate all mature blood cells, including monocytes, which have long been thought to be the major source of tissue macrophages. Recently, however, major macrophage populations were found to be derived from embryonic progenitors and to renew independently of hematopoietic stem cells. This process may not require progenitors, as mature macrophages can proliferate in response to specific stimuli indefinitely and without transformation or loss of functional differentiation. These findings suggest that macrophages are mature differentiated cells that may have a self-renewal potential similar to that of stem cells.
- Published
- 2013
- Full Text
- View/download PDF
41. M-CSF instructs myeloid lineage fate in single haematopoietic stem cells.
- Author
-
Mossadegh-Keller N, Sarrazin S, Kandalla PK, Espinosa L, Stanley ER, Nutt SL, Moore J, and Sieweke MH
- Subjects
- Animals, Cell Proliferation drug effects, Cell Survival drug effects, Granulocyte-Macrophage Colony-Stimulating Factor pharmacology, Mice, Mice, Inbred C57BL, Promoter Regions, Genetic genetics, Proto-Oncogene Proteins biosynthesis, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Single-Cell Analysis, Trans-Activators biosynthesis, Trans-Activators genetics, Trans-Activators metabolism, Cell Differentiation drug effects, Cell Lineage drug effects, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells drug effects, Macrophage Colony-Stimulating Factor pharmacology, Myeloid Cells cytology, Myeloid Cells drug effects
- Abstract
Under stress conditions such as infection or inflammation the body rapidly needs to generate new blood cells that are adapted to the challenge. Haematopoietic cytokines are known to increase output of specific mature cells by affecting survival, expansion and differentiation of lineage-committed progenitors, but it has been debated whether long-term haematopoietic stem cells (HSCs) are susceptible to direct lineage-specifying effects of cytokines. Although genetic changes in transcription factor balance can sensitize HSCs to cytokine instruction, the initiation of HSC commitment is generally thought to be triggered by stochastic fluctuation in cell-intrinsic regulators such as lineage-specific transcription factors, leaving cytokines to ensure survival and proliferation of the progeny cells. Here we show that macrophage colony-stimulating factor (M-CSF, also called CSF1), a myeloid cytokine released during infection and inflammation, can directly induce the myeloid master regulator PU.1 and instruct myeloid cell-fate change in mouse HSCs, independently of selective survival or proliferation. Video imaging and single-cell gene expression analysis revealed that stimulation of highly purified HSCs with M-CSF in culture resulted in activation of the PU.1 promoter and an increased number of PU.1(+) cells with myeloid gene signature and differentiation potential. In vivo, high systemic levels of M-CSF directly stimulated M-CSF-receptor-dependent activation of endogenous PU.1 protein in single HSCs and induced a PU.1-dependent myeloid differentiation preference. Our data demonstrate that lineage-specific cytokines can act directly on HSCs in vitro and in vivo to instruct a change of cell identity. This fundamentally changes the current view of how HSCs respond to environmental challenge and implicates stress-induced cytokines as direct instructors of HSC fate.
- Published
- 2013
- Full Text
- View/download PDF
42. Transcriptional control of macrophage identity, self-renewal, and function.
- Author
-
Molawi K and Sieweke MH
- Subjects
- Animals, Cell Differentiation, Humans, Macrophages metabolism, Myeloid Progenitor Cells metabolism, Transcription Factors metabolism, Gene Expression Regulation, Macrophages cytology, Macrophages immunology, Transcription, Genetic
- Abstract
Macrophages not only are prominent effector cells of the immune system that are critical in inflammation and innate immune responses but also fulfill important functions in tissue homeostasis. Transcription factors can define macrophage identity and control their numbers and functions through the induction and maintenance of specific transcriptional programs. Here, we review the mechanisms employed by lineage-specific transcription factors to shape macrophage identity during the development from hematopoietic stem and progenitor cells. We also present current insight into how specific transcription factors control macrophage numbers, by regulating coordinated proliferation and differentiation of myeloid progenitor cells and self-renewal of mature macrophages. We finally discuss how functional specialization of mature macrophages in response to environmental stimuli can be induced through synergistic activity of lineage- and stimulus-specific transcription factors that plug into preexisting transcriptional programs. Understanding the mechanisms that define macrophage identity, numbers, and functions will provide important insights into the differential properties of macrophage populations under various physiological and pathological conditions., (© 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
43. Development of monocytes, macrophages, and dendritic cells.
- Author
-
Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, and Ley K
- Subjects
- Animals, Cell Lineage, Cell Proliferation, Cytokines metabolism, Dendritic Cells cytology, Dendritic Cells immunology, Homeostasis, Humans, Inflammation immunology, Macrophages cytology, Macrophages immunology, Mice, Monocytes cytology, Monocytes immunology, Myeloid Progenitor Cells cytology, Myeloid Progenitor Cells physiology, Phagocytosis, Transcription, Genetic, Dendritic Cells physiology, Macrophages physiology, Monocytes physiology, Myelopoiesis
- Abstract
Monocytes and macrophages are critical effectors and regulators of inflammation and the innate immune response, the immediate arm of the immune system. Dendritic cells initiate and regulate the highly pathogen-specific adaptive immune responses and are central to the development of immunologic memory and tolerance. Recent in vivo experimental approaches in the mouse have unveiled new aspects of the developmental and lineage relationships among these cell populations. Despite this, the origin and differentiation cues for many tissue macrophages, monocytes, and dendritic cell subsets in mice, and the corresponding cell populations in humans, remain to be elucidated.
- Published
- 2010
- Full Text
- View/download PDF
44. Creating a blood line from human skin.
- Author
-
Sieweke MH
- Subjects
- Cell Culture Techniques methods, Cell Differentiation, Cell Lineage, Fibroblasts cytology, Humans, Blood Cells cytology, Cell Line, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Skin cytology
- Abstract
A recent study has generated blood cell progenitors with therapeutic potential by direct lineage conversion of human fibroblasts, thus circumventing reprogramming to pluripotent stem cells.
- Published
- 2010
- Full Text
- View/download PDF
45. MafB/c-Maf deficiency enables self-renewal of differentiated functional macrophages.
- Author
-
Aziz A, Soucie E, Sarrazin S, and Sieweke MH
- Subjects
- Animals, Cell Differentiation, Cell Transformation, Neoplastic, Cells, Cultured, Kruppel-Like Factor 4, Kruppel-Like Transcription Factors genetics, Kruppel-Like Transcription Factors physiology, Macrophage Colony-Stimulating Factor metabolism, Macrophage Colony-Stimulating Factor pharmacology, Macrophages cytology, Macrophages transplantation, MafB Transcription Factor genetics, MafB Transcription Factor physiology, Mice, Mice, Knockout, Monocytes cytology, Monocytes physiology, Myeloid Progenitor Cells cytology, Myeloid Progenitor Cells physiology, Phagocytosis, Proto-Oncogene Proteins c-maf genetics, Proto-Oncogene Proteins c-maf physiology, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc physiology, Up-Regulation, Cell Proliferation, Macrophages physiology, MafB Transcription Factor deficiency, Proto-Oncogene Proteins c-maf deficiency
- Abstract
In metazoan organisms, terminal differentiation is generally tightly linked to cell cycle exit, whereas the undifferentiated state of pluripotent stem cells is associated with unlimited self-renewal. Here, we report that combined deficiency for the transcription factors MafB and c-Maf enables extended expansion of mature monocytes and macrophages in culture without loss of differentiated phenotype and function. Upon transplantation, the expanded cells are nontumorigenic and contribute to functional macrophage populations in vivo. Small hairpin RNA inactivation shows that continuous proliferation of MafB/c-Maf deficient macrophages requires concomitant up-regulation of two pluripotent stem cell-inducing factors, KLF4 and c-Myc. Our results indicate that MafB/c-MafB deficiency renders self-renewal compatible with terminal differentiation. It thus appears possible to amplify functional differentiated cells without malignant transformation or stem cell intermediates.
- Published
- 2009
- Full Text
- View/download PDF
46. MafB restricts M-CSF-dependent myeloid commitment divisions of hematopoietic stem cells.
- Author
-
Sarrazin S, Mossadegh-Keller N, Fukao T, Aziz A, Mourcin F, Vanhille L, Kelly Modis L, Kastner P, Chan S, Duprez E, Otto C, and Sieweke MH
- Subjects
- Animals, Cell Culture Techniques, Cell Differentiation, Hematopoietic Stem Cells metabolism, Mice, Mice, Inbred C57BL, Proto-Oncogene Proteins metabolism, Receptor, Macrophage Colony-Stimulating Factor metabolism, Trans-Activators metabolism, Cell Lineage, Hematopoietic Stem Cells cytology, Macrophage Colony-Stimulating Factor metabolism, MafB Transcription Factor metabolism, Myeloid Cells cytology
- Abstract
While hematopoietic stem cell (HSC) self-renewal is well studied, it remains unknown whether distinct control mechanisms enable HSC divisions that generate progeny cells with specific lineage bias. Here, we report that the monocytic transcription factor MafB specifically restricts the ability of M-CSF to instruct myeloid commitment divisions in HSCs. MafB deficiency specifically enhanced sensitivity to M-CSF and caused activation of the myeloid master-regulator PU.1 in HSCs in vivo. Single-cell analysis revealed that reduced MafB levels enabled M-CSF to instruct divisions producing asymmetric daughter pairs with one PU.1(+) cell. As a consequence, MafB(-/-) HSCs showed a PU.1 and M-CSF receptor-dependent competitive repopulation advantage specifically in the myelomonocytic, but not T lymphoid or erythroid, compartment. Lineage-biased repopulation advantage was progressive, maintained long term, and serially transplantable. Together, this indicates that an integrated transcription factor/cytokine circuit can control the rate of specific HSC commitment divisions without compromising other lineages or self-renewal.
- Published
- 2009
- Full Text
- View/download PDF
47. Blood monocytes: development, heterogeneity, and relationship with dendritic cells.
- Author
-
Auffray C, Sieweke MH, and Geissmann F
- Subjects
- Animals, Cell Differentiation immunology, Cytokines immunology, Cytokines metabolism, Dendritic Cells metabolism, Homeostasis immunology, Humans, Inflammation metabolism, Macrophage Colony-Stimulating Factor immunology, Macrophage Colony-Stimulating Factor metabolism, Macrophages metabolism, Mice, Monocytes metabolism, Receptor, Macrophage Colony-Stimulating Factor immunology, Receptor, Macrophage Colony-Stimulating Factor metabolism, Transcription Factors immunology, Transcription Factors metabolism, Dendritic Cells immunology, Inflammation immunology, Macrophages immunology, Monocytes immunology
- Abstract
Monocytes are circulating blood leukocytes that play important roles in the inflammatory response, which is essential for the innate response to pathogens. But inflammation and monocytes are also involved in the pathogenesis of inflammatory diseases, including atherosclerosis. In adult mice, monocytes originate in the bone marrow in a Csf-1R (MCSF-R, CD115)-dependent manner from a hematopoietic precursor common for monocytes and several subsets of macrophages and dendritic cells (DCs). Monocyte heterogeneity has long been recognized, but in recent years investigators have identified three functional subsets of human monocytes and two subsets of mouse monocytes that exert specific roles in homeostasis and inflammation in vivo, reminiscent of those of the previously described classically and alternatively activated macrophages. Functional characterization of monocytes is in progress in humans and rodents and will provide a better understanding of the pathophysiology of inflammation.
- Published
- 2009
- Full Text
- View/download PDF
48. Regulation of the transcription factor Ets-1 by DNA-mediated homo-dimerization.
- Author
-
Lamber EP, Vanhille L, Textor LC, Kachalova GS, Sieweke MH, and Wilmanns M
- Subjects
- Cell Line, Crystallography, X-Ray, Dimerization, Models, Molecular, Regulatory Elements, Transcriptional, Transcriptional Activation, DNA metabolism, Matrix Metalloproteinase 3 genetics, Promoter Regions, Genetic, Proto-Oncogene Protein c-ets-1 chemistry, Proto-Oncogene Protein c-ets-1 metabolism
- Abstract
The function of the Ets-1 transcription factor is regulated by two regions that flank its DNA-binding domain. A previously established mechanism for auto-inhibition of monomeric Ets-1 on DNA response elements with a single ETS-binding site, however, has not been observed for the stromelysin-1 promoter containing two palindromic ETS-binding sites. We present the structure of Ets-1 on this promoter element, revealing a ternary complex in which protein homo-dimerization is mediated by the specific arrangement of the two ETS-binding sites. In this complex, the N-terminal-flanking region is required for ternary protein-DNA assembly. Ets-1 variants, in which residues from this region are mutated, loose the ability for DNA-mediated dimerization and stromelysin-1 promoter transactivation. Thus, our data unravel the molecular basis for relief of auto-inhibition and the ability of Ets-1 to function as a facultative dimeric transcription factor on this site. Our findings may also explain previous data of Ets-1 function in the context of heterologous transcription factors, thus providing a molecular model that could also be valid for Ets-1 regulation by hetero-oligomeric assembly.
- Published
- 2008
- Full Text
- View/download PDF
49. SUMO modification regulates MafB-driven macrophage differentiation by enabling Myb-dependent transcriptional repression.
- Author
-
Tillmanns S, Otto C, Jaffray E, Du Roure C, Bakri Y, Vanhille L, Sarrazin S, Hay RT, and Sieweke MH
- Subjects
- Animals, Cell Line, Cell Proliferation, Chickens, Humans, Mice, Models, Biological, Myeloid Cells cytology, Protein Binding, Stem Cells cytology, Transcriptional Activation genetics, Cell Differentiation, Macrophages cytology, MafB Transcription Factor metabolism, Oncogene Proteins v-myb metabolism, Repressor Proteins metabolism, Small Ubiquitin-Related Modifier Proteins metabolism, Transcription, Genetic
- Abstract
During the execution of differentiation programs, lineage-specific transcription factors are in competition with antagonistic factors that drive progenitor proliferation. Thus, the myeloid transcription factor MafB promotes macrophage differentiation of myeloid progenitors, but a constitutively active Myb transcription factor (v-Myb) can maintain proliferation and block differentiation. Little is known, however, about the regulatory mechanisms that control such competing activities. Here we report that the small ubiquitin-like protein SUMO-1 can modify MafB in vitro and in vivo on lysines 32 and 297. The absence of MafB SUMO modification increased MafB-driven transactivation and macrophage differentiation potential but inhibited cell cycle progression and myeloid progenitor growth. Furthermore, we observed that direct repression of MafB transactivation by v-Myb was strictly dependent on MafB SUMO modification. Consequently, a SUMOylation-deficient MafB K32R K297R (K32,297R) mutant could specify macrophage fate even after activation of inducible Myb alleles and resist their differentiation-inhibiting activity. Our findings suggest that SUMO modification of MafB affects the balance between myeloid progenitor expansion and terminal macrophage differentiation by controlling MafB transactivation capacity and susceptibility to Myb repression. SUMO modification of lineage-specific transcription factors may thus modulate transcription factor antagonism to control tissue homeostasis in the hematopoietic system.
- Published
- 2007
- Full Text
- View/download PDF
50. Development of macrophages with altered actin organization in the absence of MafB.
- Author
-
Aziz A, Vanhille L, Mohideen P, Kelly LM, Otto C, Bakri Y, Mossadegh N, Sarrazin S, and Sieweke MH
- Subjects
- Animals, Animals, Newborn, Cell Differentiation, Embryo, Mammalian cytology, Fetus cytology, Flow Cytometry, Gene Expression Profiling, Gene Expression Regulation, Hematopoietic System cytology, Liver cytology, Mice, Mice, Inbred C57BL, Proto-Oncogene Proteins c-maf genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Spleen cytology, Whole-Body Irradiation, Actins metabolism, Macrophages cytology, MafB Transcription Factor deficiency
- Abstract
In the hematopoietic system the bZip transcription factor MafB is selectively expressed at high levels in monocytes and macrophages and promotes macrophage differentiation in myeloid progenitors, whereas a dominant-negative allele can inhibit this process. To analyze the requirement of MafB for macrophage development, we generated MafB-deficient mice and, due to their neonatal lethal phenotype, analyzed macrophage differentiation in vitro, in the embryo, and in reconstituted mice. Surprisingly we observed in vitro differentiation of macrophages from E14.5 fetal liver (FL) cells and E18.5 splenocytes. Furthermore we found normal numbers of F4/80(+)/Mac-1(+) macrophages and monocytes in fetal liver, spleen, and blood as well as in bone marrow, spleen, and peritoneum of adult MafB(-/-) FL reconstituted mice. MafB(-/-) macrophages showed intact basic macrophage functions such as phagocytosis of latex beads or Listeria monocytogenes and nitric oxide production in response to lipopolysaccharide. By contrast, MafB(-/-) macrophages expressed increased levels of multiple genes involved in actin organization. Consistent with this, phalloidin staining revealed an altered morphology involving increased numbers of branched protrusions of MafB(-/-) macrophages in response to macrophage colony-stimulating factor. Together these data point to an unexpected redundancy of MafB function in macrophage differentiation and a previously unknown role in actin-dependent macrophage morphology.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.