Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 295
Filter
Add more filters

Publication year range
1.
Cell ; 183(3): 752-770.e22, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33125891

ABSTRACT

A greater understanding of hematopoietic stem cell (HSC) regulation is required for dissecting protective versus detrimental immunity to pathogens that cause chronic infections such as Mycobacterium tuberculosis (Mtb). We have shown that systemic administration of Bacille Calmette-Guérin (BCG) or ß-glucan reprograms HSCs in the bone marrow (BM) via a type II interferon (IFN-II) or interleukin-1 (IL1) response, respectively, which confers protective trained immunity against Mtb. Here, we demonstrate that, unlike BCG or ß-glucan, Mtb reprograms HSCs via an IFN-I response that suppresses myelopoiesis and impairs development of protective trained immunity to Mtb. Mechanistically, IFN-I signaling dysregulates iron metabolism, depolarizes mitochondrial membrane potential, and induces cell death specifically in myeloid progenitors. Additionally, activation of the IFN-I/iron axis in HSCs impairs trained immunity to Mtb infection. These results identify an unanticipated immune evasion strategy of Mtb in the BM that controls the magnitude and intrinsic anti-microbial capacity of innate immunity to infection.


Subject(s)
Hematopoietic Stem Cells/microbiology , Immunity , Mycobacterium tuberculosis/physiology , Myelopoiesis , Animals , Bone Marrow Cells/metabolism , Cell Proliferation , Disease Susceptibility , Homeostasis , Interferon Type I/metabolism , Iron/metabolism , Kinetics , Lung/microbiology , Lung/pathology , Macrophages/immunology , Mice, Inbred C57BL , Myeloid Cells/metabolism , Necrosis , Signal Transduction , Transcription, Genetic , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/pathology
2.
Blood ; 139(1): 44-58, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34525198

ABSTRACT

Aging is associated with impaired hematopoietic and immune function caused in part by decreased fitness in the hematopoietic stem cell (HSC) population and an increased myeloid differentiation bias. The reasons for this aging-associated HSC impairment are incompletely understood. Here we demonstrate that older specific pathogen free (SPF) wild-type (WT) mice in contrast to young SPF mice produce more interleukin-1a and interleukin-1b (IL-1a/b) in steady-state bone marrow (BM), with most of the IL-1a/b being derived from myeloid BM cells. Furthermore, blood from steady-state older SPF WT mice contains higher levels of microbe-associated molecular patterns, specifically TLR4 and TLR8 ligands. In addition, BM myeloid cells from older mice produce more IL-1b in vitro, and older mice show higher and more durable IL-1a/b responses upon stimulation with lipopolysaccharide in vivo. To test whether HSC aging is driven by IL-1a/b, we evaluated HSCs from IL-1 receptor 1 (IL-1R1) knockout (KO) mice. Indeed, older HSCs from IL-1R1KO mice show significantly mitigated aging-associated inflammatory signatures. Moreover, HSCs from older IL-1R1KO and from germ-free mice maintain unbiased lymphomyeloid hematopoietic differentiation upon transplantation, thus resembling this functionality of young HSCs. Importantly, in vivo antibiotic suppression of microbiota or pharmacologic blockade of IL-1 signaling in older WT mice was similarly sufficient to reverse myeloid-biased output of their HSC populations. Collectively, our data define the microbiome/IL-1/IL-1R1 axis as a key, self-sustaining and also therapeutically partially reversible driver of HSC inflammaging.


Subject(s)
Hematopoietic Stem Cells/metabolism , Inflammation/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Microbiota , Aging , Animals , Cellular Senescence , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/microbiology , Inflammation/microbiology , Mice , Mice, Knockout
3.
Blood ; 136(10): 1144-1154, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32438398

ABSTRACT

Hematopoiesis is a system that provides red blood cells (RBCs), leukocytes, and platelets, which are essential for oxygen transport, biodefense, and hemostasis; its balance thus affects the outcome of various disorders. Here, we report that stem cell antigen-1 (Sca-1), a cell surface marker commonly used for the identification of multipotent hematopoietic progenitors (Lin-Sca-1+c-Kit+ cells; LSKs), is not suitable for the analysis of hematopoietic responses under biological stresses with interferon production. Lin-Sca-1-c-Kit+ cells (LKs), downstream progenitors of LSKs, acquire Sca-1 expression upon inflammation, which makes it impossible to distinguish between LSKs and LKs. As an alternative and stable marker even under such stresses, we identified CD86 by screening 180 surface markers. The analysis of infection/inflammation-triggered hematopoiesis on the basis of CD86 expression newly revealed urgent erythropoiesis producing stress-resistant RBCs and intact reconstitution capacity of LSKs, which could not be detected by conventional Sca-1-based analysis.


Subject(s)
B7-2 Antigen/metabolism , Bacterial Infections/complications , Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells/pathology , Inflammation/complications , Animals , Antigens, Ly/metabolism , Bacteria/metabolism , Cells, Cultured , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/microbiology , Lipopolysaccharides , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proto-Oncogene Proteins c-kit/metabolism
4.
Scand J Immunol ; 92(5): e12957, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32767789

ABSTRACT

Bone marrow haematopoietic stem and progenitor cells (HSPCs) express pattern recognition receptors such as Toll-like receptors (TLRs) to sense microbial products and activation of these innate immune receptors induces cytokine expression and redirects bone marrow haematopoiesis towards the increased production of myeloid cells. Secreted cytokines by HSPCs in response to TLR ligands can act in an autocrine or paracrine manner to regulate haematopoiesis. Moreover, tonic activation of HSPCs by microbiota-derived compounds might educate HSPCs to produce superior myeloid cells equipped with innate memory responses to combat pathogens. While haematopoietic stem cell activation through TLRs meets the increased demand for blood leucocytes to protect the host against infection, persistent exposure to inflammatory cytokines or microbial products might impair their function and even induce malignant transformation. This review highlights the potential outcomes of HSPCs in response to TLR ligands.


Subject(s)
Bone Marrow Cells/immunology , Hematopoietic Stem Cells/immunology , Microbiota/immunology , Myeloid Cells/immunology , Receptors, Pattern Recognition/immunology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/microbiology , Cytokines/immunology , Cytokines/metabolism , Hematopoiesis/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/microbiology , Humans , Myeloid Cells/metabolism , Receptors, Pattern Recognition/metabolism , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism
5.
J Infect Dis ; 219(5): 685-694, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30376080

ABSTRACT

Latent tuberculosis has been recognized for over a century, but discovery of new niches, where Mycobacterium tuberculosis resides, continues. We evaluated literature on M.tuberculosis locations during latency, highlighting that mesenchymal and hematopoietic stem cells harbor organisms in sensitized asymptomatic individuals.


Subject(s)
Hematopoietic Stem Cells/microbiology , Latent Tuberculosis/microbiology , Latent Tuberculosis/pathology , Mesenchymal Stem Cells/microbiology , Mycobacterium tuberculosis/isolation & purification , Phagocytes/microbiology , Humans , Mycobacterium tuberculosis/growth & development
6.
J Infect Dis ; 217(10): 1667-1671, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29471332

ABSTRACT

Persistence of Mycobacterium tuberculosis within human bone marrow stem cells has been identified as a potential bacterial niche during latent tuberculosis. Using a murine model of tuberculosis, we show here that bone marrow stem and progenitor cells containing M. tuberculosis propagated tuberculosis when transferred to naive mice, given that both transferred cells and recipient mice were unable to express inducible nitric oxide synthase, which mediates killing of intracellular bacteria via nitric oxide. Our findings suggest that bone marrow stem and progenitor cells containing M. tuberculosis propagate hallmarks of disease if nitric oxide-mediated killing of bacteria is defective.


Subject(s)
Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/microbiology , Mycobacterium tuberculosis/pathogenicity , Nitric Oxide Synthase Type II/metabolism , Stem Cells/metabolism , Stem Cells/microbiology , Tuberculosis/metabolism , Animals , Disease Models, Animal , Hematopoietic Stem Cell Transplantation/methods , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Tuberculosis/microbiology
7.
Cell Microbiol ; 18(10): 1339-48, 2016 10.
Article in English | MEDLINE | ID: mdl-26990252

ABSTRACT

Haemolytic anaemia is one of the characteristics of life-threatening extraintestinal complications in humans during infection with enterohaemorrhagic Escherichia coli (EHEC). Shiga toxins (Stxs) of EHEC preferentially damage microvascular endothelial cells of the kidney and the brain, whereby occluded small blood vessels may elicit anaemia through mechanical erythrocyte disruption. Here we show for the first time that Stx2a, the major virulence factor of EHEC, is also capable of direct targeting developing human erythrocytes. We employed an ex vivo erythropoiesis model using mobilized CD34(+) haematopoietic stem/progenitor cells from human blood and monitored expression of Stx receptors and Stx2a-mediated cellular injury of developing erythrocytes. CD34(+) haematopoietic stem/progenitor cells were negative for Stx2a receptors and resistant towards the toxin. Expression of Stx2a-binding glycosphingolipids and toxin sensitivity was apparent immediately after initiation of erythropoietic differentiation, peaked for basophilic and polychromatic erythroblast stages and declined during maturation into orthochromatic erythroblasts and reticulocytes, which became highly refractory to Stx2a. The observed Stx-mediated toxicity towards erythroblasts during the course of erythropoiesis might contribute, although speculative at this stage of research, to the anaemia caused by Stx-producing pathogens.


Subject(s)
Enterohemorrhagic Escherichia coli/physiology , Hematopoietic Stem Cells/physiology , Shiga Toxin/pharmacology , Cell Survival , Cells, Cultured , Erythrocytes/microbiology , Erythrocytes/physiology , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Humans
8.
J Immunol ; 193(3): 1064-70, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24973456

ABSTRACT

Both commensal bacteria and infiltrating inflammatory cells play essential roles in the pathogenesis of inflammatory bowel disease. The molecular mechanisms whereby these pathogenic factors are regulated during the disease are not fully understood. We report in this article that a member of the TNF-α-induced protein 8 (TNFAIP8) family called TIPE2 (TNFAIP8-like 2) plays a crucial role in regulating commensal bacteria dissemination and inflammatory cell function in experimental colitis induced by dextran sodium sulfate (DSS). Following DSS treatment, TIPE2-deficient mice, or chimeric mice that are deficient in TIPE2 only in their hematopoietic cells, lost less body weight and survived longer than wild-type controls. Consistent with this clinical observation, TIPE2-deficient mice exhibited significantly less severe colitis and colonic damage. This was associated with a marked reduction in the colonic expression of inflammatory cytokines, such as TNF-α, IL-6, and IL-12. Importantly, the ameliorated DSS-induced colitis in TIPE2(-/-) mice also was associated with reduced local dissemination of commensal bacteria and a weaker systemic inflammatory response. Combined with our previous report that TIPE2 is a negative regulator of antibacterial immunity, these results indicate that TIPE2 promotes colitis by inhibiting mucosal immunity to commensal bacteria.


Subject(s)
Colitis/genetics , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/physiology , Animals , Bacteria/genetics , Colitis/chemically induced , Colitis/pathology , Colon/drug effects , Colon/microbiology , Colon/pathology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Hematopoietic Stem Cells/pathology , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiation Chimera , Sulfates/toxicity
9.
Transfus Apher Sci ; 55(1): 125-8, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27184293

ABSTRACT

INTRODUCTION: Microbial contamination can be a marker for faulty process and is assumed to play an important role in the collection of hematopoietic progenitor cell (HPC) and infusion procedure. We aimed to determine the microbial contamination rates and evaluate the success of hematopoietic cell transplantation (HCT) in patients who received contaminated products. PATIENTS-METHODS: We analyzed microbial contamination records of HPC grafts between 2012 and 2015, retrospectively. Contamination rates of autologous donors were evaluated for at three steps: at the end of mobilization, following processing with dimethyl sulfoxide, and just before stem cell infusion. Grafts of allogeneic donors were assessed only before HCT. RESULT: A total of 445 mobilization procedures were carried out on 333 (167 autologous and 166 allogeneic) donors. The microbiological contamination of peripheral blood (323/333 donations) and bone marrow (10/333 donations) products were analyzed. Bacterial contamination was detected in 18 of 1552 (1.15 %) culture bottles of 333 donors. During the study period 248 patients underwent HCT and among these patients microbial contamination rate on sample basis was 1.3 % (16/1212). Microbial contamination detected in nine patients (7 autologous; 2 allogeneic). In 8 of 9 patients, a febrile neutropenic attack was observed. The median day for the neutropenic fever was 4 days (0-9). None of the patients died within the post-transplant 30 days who received contaminated products. CONCLUSION: The use of contaminated products with antibiotic prophylaxis may be safe in terms of the first day of fever, duration of fever, neutrophil, platelet engraftment and duration of hospitalization.


Subject(s)
Hematologic Neoplasms/microbiology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/microbiology , Adolescent , Adult , Aged , Allografts , Autografts , Female , Humans , Male , Middle Aged
10.
Blood ; 122(10): 1770-8, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23869087

ABSTRACT

During bacterial infection, hematopoietic stem and progenitor cells (HSPCs) differentiate into polymorphonuclear leukocytes (PMNs) in the bone marrow. We reported that HSPCs recruited to Staphylococcus aureus-infected skin wounds in mice undergo granulopoiesis, whereas other authors have demonstrated their differentiation in vitro after Toll-like receptor 2 (TLR2)/MyD88 stimulation. Here, we examined this pathway in HSPC trafficking and granulopoiesis within S aureus-infected wounds. Lineage- HSPCs from TLR2- or MyD88-deficient mice injected into infected wounds of wild-type (WT) mice exhibited impaired granulopoiesis. However, HSPCs from WT mice produced similar numbers of PMNs whether transferred into wounds of TLR2-, MyD88-deficient, or WT mice. Prostaglandin E2 (PGE2), which stimulates HSPC survival and proliferation, was produced by HSPCs after TLR2 stimulation, suggesting that TLR2/MyD88 activation promotes granulopoiesis in part by production and autocrine activity of PGE2. Pretreatment of TLR2- or MyD88-deficient HSPCs with PGE2 rescued granulocytic differentiation in vivo. Finally, we demonstrate that bone marrow-derived lin-/Sca-1+/c-kit+ cells produced PGE2 and underwent granulopoiesis after TLR2 stimulation. lin-/Sca-1+/c-kit+ cells deficient in TLR2 or MyD88 produced PMNs after PGE2 treatment when transferred into uninfected wounds. We conclude that granulopoiesis in S aureus-infected wounds is induced by TLR2/MyD88 activation of HSPCs through a mechanism that involves autocrine production and activity of PGE2.


Subject(s)
Dinoprostone/metabolism , Hematopoietic Stem Cells/immunology , Leukopoiesis , Myeloid Differentiation Factor 88/metabolism , Staphylococcus aureus/immunology , Toll-Like Receptor 2/metabolism , Wounds and Injuries/microbiology , Animals , Bone Marrow/pathology , Cell Count , Cell Differentiation , Cell Proliferation , Cell Separation , Dinoprostone/biosynthesis , Female , Granulocytes/immunology , Granulocytes/pathology , Hematopoietic Stem Cells/microbiology , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Signal Transduction/immunology , Skin/immunology , Skin/microbiology , Skin/pathology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Toll-Like Receptor 2/deficiency , Wounds and Injuries/immunology
11.
Vox Sang ; 108(3): 314-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25471358

ABSTRACT

In order to generate standardized conditions for the microbiological control of HPCs, the PEI recommended defined steps for validation that will lead to extensive validation as shown in this study, where a possible validation principle for the microbiological control of allogeneic SCPs is presented. Although it could be demonstrated that automated culture improves microbial safety of cellular products, the requirement for extensive validation studies needs to be considered.


Subject(s)
Cell Culture Techniques/standards , Guidelines as Topic , Hematopoietic Stem Cells/microbiology , Cell Culture Techniques/methods , Cells, Cultured , Germany , Humans
12.
J Immunol ; 190(9): 4725-35, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23526822

ABSTRACT

Hematopoietic stem and progenitor cell (HSPC) phenotype and function can change in response to infectious challenge. These changes can be mediated by cytokines, IFNs, and pathogen-associated molecules, via TLR, and are thought to promote tailored immune responses for particular pathogens. In this study, we investigated the signals that activate HSPCs during ehrlichiosis, a disease characterized by profound hematopoietic dysfunction in both humans and mice. In a mouse model of ehrlichiosis, we observed that infection-induced proliferation of bone marrow HSPCs was dependent on IFN-γ signaling and was partially dependent on MyD88. However, MyD88 was not required in HSPCs for their expansion during infection, because similar frequencies of MyD88-deficient and wild-type HSPCs proliferated in mixed bone marrow chimeric mice. MyD88-deficient mice exhibited low serum and bone marrow concentration of IFN-γ compared with wild-type mice. We next identified CD4 T cells as the primary cells producing IFN-γ in the bone marrow and demonstrated a nonredundant role for CD4-derived IFN-γ in increased HSPCs. Using mixed bone marrow chimeric mice, we identified a requirement for MyD88 in CD4 T cells for increased T-bet expression, optimal IFN-γ production, and CD4 T cell proliferation. Our data demonstrate an essential role for CD4 T cells in mediating HSPC activation in response to bacterial infection and illustrate a novel role for MyD88 signaling in CD4 T cells in this process. These findings further support the idea that IFN-γ production is essential for HSPC activation and hematopoietic responses to infection.


Subject(s)
Bacterial Infections/metabolism , CD4-Positive T-Lymphocytes/metabolism , Hematopoietic Stem Cells/metabolism , Interferon-gamma/biosynthesis , Myeloid Differentiation Factor 88/metabolism , Animals , Bacterial Infections/immunology , Bacterial Infections/microbiology , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow/microbiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Cell Proliferation , Ehrlichia/immunology , Ehrlichiosis/immunology , Ehrlichiosis/metabolism , Ehrlichiosis/microbiology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Myeloid Differentiation Factor 88/immunology , Signal Transduction/immunology
14.
Cell Microbiol ; 15(7): 1143-53, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23279268

ABSTRACT

Toll-like receptors (TLRs) are expressed by haematopoietic stem and progenitor cells (HSPCs), and may play a role in haematopoiesis in response to pathogens during infection. We have previously demonstrated that (i) inactivated yeasts of Candida albicans induce in vitro differentiation of HSPCs towards the myeloid lineage, and (ii) soluble TLR agonists induce in vivo their differentiation towards macrophages. In this work, using an in vivo model of HSPCs transplantation, we report for the first time that HSPCs sense C. albicans in vivo and subsequently are directed to produce macrophages by a TLR2-dependent signalling. Purified lineage-negative cells (Lin(-)) from bone marrow of C57BL/6 mice (CD45.2 alloantigen) were transplanted into B6Ly5.1 mice (CD45.1 alloantigen), which were then injected with viable or inactivated C. albicans yeasts. Transplanted cells were detected in the spleen and in the bone marrow of recipient mice, and they differentiate preferentially to macrophages, both in response to infection or in response to inactivated yeasts. The generation of macrophages was dependent on TLR2 but independent of TLR4, as transplanted Lin(-) cells from TLR2(-/-) mice did not give rise to macrophages, whereas Lin(-) cells from TLR4(-/-) mice generated macrophages similarly to control cells. Interestingly, the absence of TLR2, or in a minor extent TLR4, gives Lin(-) cells an advantage in transplantation assays, as increases the percentage of transplanted recovered cells. Our results indicatethat TLR-mediated recognition of C. albicans by HSPCs may help replace and/or increase cells that constitute the first line of defence against the fungus, and suggest that TLR-mediated signalling may lead to reprogramming early progenitors to rapidly replenishing the innate immune system and generate the most necessary mature cells to deal with the pathogen.


Subject(s)
Candida albicans/immunology , Cell Differentiation , Hematopoietic Stem Cells/physiology , Macrophages/immunology , Signal Transduction , Toll-Like Receptor 2/metabolism , Animals , Bone Marrow/immunology , Bone Marrow/microbiology , Cells, Cultured , Hematopoietic Stem Cells/microbiology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Spleen/immunology , Spleen/microbiology
15.
Vox Sang ; 106(4): 285-96, 2014 May.
Article in English | MEDLINE | ID: mdl-24697216

ABSTRACT

Bacterial safety of cellular preparations, especially haematopoietic progenitor cells (HPCs), as well as advanced therapy medicinal products (ATMPs) derived from stem cells of various origins, present a challenge for physicians, manufacturers and regulators. The article describes the background and practical issues in this area and illustrates why sterility of these products cannot currently be guaranteed. Advantages and limitations of approaches both for classical sterility testing and for microbiological control using automated culture systems are discussed. The review considers novel approaches for growth-based rapid microbiological control with high sensitivity and faster availability of results, as well as new methods for rapid bacterial detection in cellular preparations enabling meaningful information about product contamination within one to two hours. Generally, however, these direct rapid methods are less sensitive and have greater sampling error compared with the growth-based methods. Opportunities for pyrogen testing of cell therapeutics are also discussed. There is an urgent need for development of novel principles and methods applicable to bacterial safety of cellular therapeutics. We also need a major shift in approach from the traditional view of sterility evaluation (identify anything and everything) to a new thinking about how to find what is clinically relevant within the time frame available for the special clinical circumstances in which these products are used. The review concludes with recommendations for optimization of microbiological control of cellular preparations, focusing on HPCs.


Subject(s)
Bacterial Infections/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cells/microbiology , Animals , Bacterial Infections/prevention & control , Cells, Cultured , Disinfection , Humans
16.
Gut Microbes ; 16(1): 2350784, 2024.
Article in English | MEDLINE | ID: mdl-38727219

ABSTRACT

The gut microbiota constitutes a vast ecological system within the human body, forming a mutually interdependent entity with the host. In recent years, advancements in molecular biology technologies have provided a clearer understanding of the role of the gut microbiota. They not only influence the local immune status and metabolic functions of the host's intestinal tract but also impact the functional transformation of hematopoietic stem cells (HSCs) through the gut-blood axis. In this review, we will discuss the role of the gut microbiota in influencing hematopoiesis. We analyze the interactions between HSCs and other cellular components, with a particular emphasis on the direct functional regulation of HSCs by the gut microbiota and their indirect influence through cellular components in the bone marrow microenvironment. Additionally, we propose potential control targets for signaling pathways triggered by the gut microbiota to regulate hematopoietic function, filling crucial knowledge gaps in the development of this research field.


Subject(s)
Gastrointestinal Microbiome , Hematopoiesis , Hematopoietic Stem Cells , Hematopoiesis/physiology , Gastrointestinal Microbiome/physiology , Humans , Hematopoietic Stem Cells/microbiology , Animals , Signal Transduction , Bacteria/metabolism , Bacteria/classification , Bacteria/genetics , Gastrointestinal Tract/microbiology , Bone Marrow/microbiology , Bone Marrow/physiology
17.
Transfusion ; 53(11): 2690-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23461309

ABSTRACT

BACKGROUND: Microbial contamination of hematopoietic progenitor cells (HPCs) and other regenerative cells used in transplantation and regenerative medicine can occur during collection and after in vitro manipulation, including purging, cryopreservation, thawing, and infusion. STUDY DESIGN AND METHODS: Microbiologic culture findings on consecutive HPCs and other cell preparations at a single institution derived from peripheral blood, marrow, cord blood, and mesenchymal stromal cells during all phases of manipulation were retrospectively examined from 2005 through 2011. Results were classified as confirmed positive, false positive, and indeterminate. RESULTS: During the 6-year surveillance period, 365 patients underwent 912 procedures involving HPC or other cell-based transfusion. True positive microbial contamination was found in five of 663 (0.8%) peripheral blood and two of 34 (5.9%) marrow preparations (p = 0.04), while no contamination was found in 118 preparations from other sources. True-positive microbial contaminants included coagulase-negative staphylococci in autologous HPC products derived from peripheral blood from two patients with asymptomatic central venous catheter infections at time of apheresis and Propionibacterium acnes in one apheresis and two marrow products. Organism loads were low in all cases (≤500 colony-forming units/mL), and no adverse sequelae occurred in four patients that received contaminated products. CONCLUSION: The incidence of microbial contamination of progenitor cell products in our institution over a 6-year period was low (0.8% overall), with contaminants originating from infected central venous catheters or from skin flora. All contaminants were bacterial species of low virulence, present in low titers and, if transfused, did not result in adverse reactions.


Subject(s)
Bacteria/isolation & purification , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/microbiology , Regenerative Medicine , Blood Component Removal , Humans , Retrospective Studies
18.
Transfusion ; 53(11): 2659-66, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23438051

ABSTRACT

BACKGROUND: AABB Standards require monitoring of hematopoietic progenitor cell (HPC) products for microbial contamination. To date, there is no automated blood culture system cleared by the Food and Drug Administration for this application. Our objective was to validate the VersaTREK system (TREK Diagnostic Systems) for sterility testing of apheresis HPC products. STUDY DESIGN AND METHODS: Four aerobic bacteria (Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus mitis, and Bacillus cereus), five anaerobic bacteria (Fusobacterium necrophorum, Clostridium perfringens, Bacteroides fragilis, Prevotella loescheii, and Propionibacterium acnes), and one fungus (Candida albicans) were spiked into apheresis HPC products at concentrations of 10, 10(2) , 10(3) , and 10(4) colony-forming units (CFUs)/mL. Aerobic and anaerobic bottles were incubated until positive or for up to 5 days. DNA was simultaneously extracted for polymerase chain reaction amplification of 16S ribosomal RNA (rRNA) gene. RESULTS: All aerobic bacteria grew in both bottles at all concentrations tested within 24 hours, and the time to positivity (TTP) was significantly shorter with aerobic bottles. C. albicans grew in the aerobic media at all concentrations within 30 hours. Anaerobes grew in the anaerobic bottle at all concentrations within 5 days. No bacteria were detected by using 16S rRNA gene amplification at 10(4) CFUs/mL. CONCLUSION: Compared to culture, 16S rRNA gene amplification of HPCs does not improve sensitivity or turnaround time for HPC sterility testing. The VersaTREK system is a reliable tool for detecting microbial contamination of apheresis HPC products with a limit of detection of less than or equal to 10 CFUs/mL. Inclusion of both the aerobic and the anaerobic culture bottles achieves the shortest TTP for all species tested.


Subject(s)
Blood Component Removal/standards , Hematopoietic Stem Cells/microbiology , Sterilization , Bacteria, Aerobic/growth & development , Bacteria, Anaerobic/growth & development , Humans , Polymerase Chain Reaction , Prevotella/isolation & purification , RNA, Ribosomal, 16S/genetics , Reproducibility of Results
19.
J Immunol ; 186(2): 1032-43, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21149601

ABSTRACT

Although microbial infections can alter steady-state hematopoiesis, the mechanisms that drive such changes are not well understood. We addressed a role for IFN-γ signaling in infection-induced bone marrow suppression and anemia in a murine model of human monocytic ehrlichiosis, an emerging tick-borne disease. Within the bone marrow of Ehrlichia muris-infected C57BL/6 mice, we observed a reduction in myeloid progenitor cells, as defined both phenotypically and functionally. Infected mice exhibited a concomitant increase in developing myeloid cells within the bone marrow, an increase in the frequency of circulating monocytes, and an increase in splenic myeloid cells. The infection-induced changes in progenitor cell phenotype were critically dependent on IFN-γ, but not IFN-α, signaling. In mice deficient in the IFN-γ signaling pathway, we observed an increase in myeloid progenitor cells and CDllb(lo)Gr1(lo) promyelocytic cells within the bone marrow, as well as reduced frequencies of mature granulocytes and monocytes. Furthermore, E. muris-infected IFN-γR-deficient mice did not exhibit anemia or an increase in circulating monocytes, and they succumbed to infection. Gene transcription studies revealed that IFN-γR-deficient CDllb(lo)Gr1(lo) promyelocytes from E. muris-infected mice exhibited significantly reduced expression of irf-1 and irf-8, both key transcription factors that regulate the differentiation of granulocytes and monocytes. Finally, using mixed bone marrow chimeric mice, we show that IFN-γ-dependent infection-induced myelopoiesis occurs via the direct effect of the cytokine on developing myeloid cells. We propose that, in addition to its many other known roles, IFN-γ acts to control infection by directly promoting the differentiation of myeloid cells that contribute to host defense.


Subject(s)
Ehrlichiosis/immunology , Ehrlichiosis/metabolism , Interferon-gamma/physiology , Intracellular Fluid/microbiology , Myeloid Cells/immunology , Myeloid Cells/microbiology , Myelopoiesis/immunology , Signal Transduction/immunology , Animals , Blood Cell Count , Cell Differentiation/immunology , Cells, Cultured , Ehrlichia/immunology , Ehrlichia/pathogenicity , Ehrlichiosis/pathology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Hematopoietic Stem Cells/pathology , Humans , Immunophenotyping , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/pathology
20.
Transfus Apher Sci ; 48(3): 403-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23664302

ABSTRACT

INTRODUCTION: Microbial screening for contamination is a part of hematopoietic progenitor cell (HPC) collection and infusion procedure. We aimed to find out our microbial contamination rates during collection, processing and infusion steps of HPC products. We also evaluated the clinical course of patients who received contaminated HPC products. PATIENTS-METHODS: We retrospectively analyzed microbial contamination records of HPC grafts between 2010 and 2012. HPC products of autologous donors were evaluated for contamination at three steps: at the end of mobilization, following processing with DMSO and just before stem cell infusion. Grafts of allogeneic donors were assessed only before HPC transplantation (HCT). Microbiological analysis of HPC samples were performed with an automated system (BacT/Alert®). RESULT: During the study period a total of 492 mobilization procedures were performed on 329 (214 autologous and 115 allogeneic) donors. Bacterial contamination has been detected in 103 of 1630 samples (6%). Ninety-seven out of 1162 blood samples (8%) from 265 patients who were treated with HCT were contaminated. Forty-six patients (41 autologous and 5 allogeneic) were transplanted with contaminated HPC products. During HCT 42 patients experienced febrile neutropenic attack and 34 of them had positive blood culture results. In none of these 34 patients the isolated pathogens were the same organisms with those found in the final contaminated stem cell product before stem cell infusion. None of the patients who received contaminated products died because of sepsis within the posttransplant 30days. There was no significant difference between patients who received contaminated and non-contaminated products in terms of the first day of fever, duration of fever, engraftment kinetics and duration of hospitalization. CONCLUSION: Our results suggest that microbial contamination of HPC products is an issue to be prevented, although it may not have a major impact on the general success of HCT.


Subject(s)
Bacteria/isolation & purification , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/microbiology , Adolescent , Adult , Aged , Aged, 80 and over , Automation , Child , Female , Humans , Male , Middle Aged , Retrospective Studies , Specimen Handling/methods , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL