Terminal differentiation is the major route of HSC loss during chronic infection

2016 ◽  
Vol 44 (9) ◽  
pp. S44
Author(s):  
Katherine King ◽  
Katie Matatall ◽  
Siyi Chen ◽  
Mira Jeong ◽  
Deqiang Sun ◽  
...  
2016 ◽  
Vol 3 (suppl_1) ◽  
Author(s):  
Katherine King ◽  
Katie Matatall ◽  
Siyi Chen ◽  
Mira Jeong ◽  
Deqiang Sun ◽  
...  

Cell Reports ◽  
2016 ◽  
Vol 17 (10) ◽  
pp. 2584-2595 ◽  
Author(s):  
Katie A. Matatall ◽  
Mira Jeong ◽  
Siyi Chen ◽  
Deqiang Sun ◽  
Fengju Chen ◽  
...  

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. SCI-32-SCI-32 ◽  
Author(s):  
Katherine Y. King

Abstract Infections and chronic inflammatory conditions affect blood production and are frequently associated with peripheral cytopenias. Common examples include tuberculosis and atypical mycobacterial infections, HIV, hepatitis C, rheumatoid arthritis, and inflammatory bowel disease. Low blood counts amplify morbidity and mortality in each of these conditions. Recent studies have contributed to a better understanding of the pathophysiologic conditions linking inflammation and bone marrow suppression. While studies to investigate the effects of chronic infection on human hematopoietic stem and progenitor cells (HSPCs) are limited in number and scope, we and others have used animal models to evaluate the effects of infection and inflammation on HSPCs. During persistent Mycobacterium avium infection of mice, hematopoietic stem cells (HSCs) exited from quiescence and differentiated at a high rate. The total number of HSCs decreased by approximately 95% in a period of 4 months, even in the absence of widespread myelofibrosis, and the animals developed pancytopenia. Physiologic changes during infection, including loss of quiescence, diminished self-renewal, and increased differentiation, could largely be recapitulated by administration of the inflammatory cytokine interferon gamma (IFNg) alone. Indeed, studies from our lab and others collectively indicate that inflammatory cytokines are key drivers of hematopoietic changes in chronic infections and inflammatory conditions. Cytokines including interferon alpha, IFNg, IL1, IL6 and TNFalpha and pathogen-associated molecular patterns such as LPS have all been shown to increase HSC division and reduce their self-renewal. In our system, the major mechanism of stem cell loss was through excessive terminal differentiation; however, the presence of multiple stressors may also increase replication stress and propensity for apoptosis in these cells. Recent studies further indicate that inflammatory signaling may alter the number and function of cells that are produced by HSPCs, through selective activation of HSPC subtypes or epigenetic reprogramming. Collectively, the effects of inflammatory signaling on HSPC self-renewal and differentiation have important clinical implications in a number of disease states. Excessive inflammatory signaling is associated with acquired aplastic anemia and myelodysplastic syndrome; therefore, identifying the molecular mechanisms by which inflammatory cytokines such as IFNg drive terminal differentiation and HSC loss may yield strategies to modify disease progression for these syndromes. Recent work showed that inflammation critically drives Tet2-associated clonal hematopoiesis; the role of inflammation in clonal hematopoiesis and malignant transformation more broadly remains an active area of investigation. Finally, the concept that inflammatory signaling may alter the lineage output of HSPCs, either through selective or directive differentiation, may aid in the understanding of immune responses to infectious diseases, with potential therapeutic implications. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2406-2406
Author(s):  
Katie A Matatall ◽  
Mira Jeong ◽  
Sun Deqiang ◽  
Claudine Salire ◽  
Katherine Y. King

Abstract Background: While inflammation is necessary to fight infection and repair damaged tissue, excessive inflammation can cause bone marrow suppression and promote cancer. In an extreme example, high levels of the inflammatory cytokine interferon gamma (IFNg) deplete hematopoietic stem cells (HSCs), resulting in aplastic anemia. Patients with this dangerous disease are pancytopenic and therefore at high risk of death from infection. Pancytopenia also occurs to a lesser extent in other inflammatory conditions such as chronic infections (tuberculosis, HIV), and autoimmune diseases (hemophagocytic histiocytosis). However, the mechanism by which HSCs are damaged by IFNg remains poorly understood. We used a mouse model of Mycobacterium avium infection to study the effects of sustained IFNg exposure on primitive hematopoiesis. In prior work, we found, surprisingly, that IFNg promotes division of quiescent HSCs. We hypothesized that cell division might lead to loss of HSCs through terminal differentiation, displacement, or activation of p53-dependent apoptosis pathways. Objective: We sought to determine whether prolonged IFNg stimulation would lead experimentally to exhaustion of the HSC compartment, and to determine the mechanism of inflammation-mediated HSC loss. Methods: We conducted repeated monthly infection of C57Bl/6 WT mice with 2 x 106 cfu M. avium, thereby generating a sustained chronic IFNg response. We characterized the blood and bone marrow of treated mice by histology, flow cytometry, colony forming assays, and bone marrow transplant. Results: Mice infected with M. avium became anemic and leukopenic after 6 months of repeated infection. High IFNg levels were sustained in the mice, with evidence of IFNg production by T cells and NK cells in the bone marrow. The number of committed hematopoietic progenitors gradually decreased and HSCs were depleted in the bone marrow by four months following initial infection, without evidence of extensive myelofibrosis. The marrow was hypercellular with a significant increase in granulocytes. Meanwhile, the myeloid differentiation capacity of the marrow was reduced, consistent with terminal differentiation of myeloid-biased HSCs, as we have previously described. Despite an overall reduction in HSC number, the HSCs that remained in chronically infected animals mostly retained their self-renewal potential, with subtle self-renewal defects evident only after two rounds of transplantation. Homing of HSCs from infected animals was not impaired, but ex vivo culture and apoptosis assays indicated that HSCs from chronically infected animals had reduced colony forming ability and were more prone to cell death upon secondary stress. These findings were recapitulated by introduction of recombinant IFNg alone. RNAseq profiling of HSCs from infected and control animals reflected increased proliferation and differentiation during infection, consistent with the above findings. Conclusions: We have established a novel mouse model of bone marrow failure related to chronic IFNg stimulation. We demonstrate that chronic infection can deplete the HSC pool by promoting HSC differentiation and lowering the threshold for apoptosis. These mechanisms may drive marrow suppression in patients with aplastic anemia, hemophagocytic histiocytosis (also associated with high IFNg levels), and patients with marrow failure associated with chronic infection. Furthermore, since a reduction in HSC number results in depletion of clonal heterogeneity, our findings have significant implications regarding the mechanism by which chronic inflammation can contribute to the emergence of clonal hematopoiesis and hematologic malignancies with age. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 732-732
Author(s):  
Katie A Matatall ◽  
Mira Jeong ◽  
Fengju Chen ◽  
Siyi Chen ◽  
Deqiang Sun ◽  
...  

Abstract Chronic infections including tuberculosis, hepatitis C, and HIV are estimated to affect over a third of the world's population and are associated with significant health implications including bone marrow suppression and an increased risk for cancer (Jian et al., 2014; Ramos-Casals et al., 2003; Scadden et al., 1989). Pancytopenia, a suppression of blood counts across multiple lineages, can affect as many as 12% of people with miliary tuberculosis and increases risk of death from the infection (Achi et al., 2013). However little is known about the mechanisms by which infections affect hematopoietic stem cells (HSCs). Here we show that chronic infection depletes HSCs and we identify terminal differentiation as the major route of HSC loss. Furthermore, we define Batf2 as a potent new mediator of inflammation-induced differentiation. Using an established model of chronic infection, we conducted repeated monthly infection of C57Bl/6 WT mice with 2 x 106 cfu Mycobacterium avium, generating a sustained chronic IFNg response. Mice became pancytopenic after 4-6 months of repeated infection. The number of HSCs in the bone marrow was depleted to just 5% of the starting number by 4 months following initial infection, without evidence of extensive myelofibrosis. Poor engraftment upon transplant of whole bone marrow (WBM) from mice repeatedly infected with M. avium confirmed that functional HSCs were lost. When WBM from Ifngr1-deficient mice infected with M. aviumwas transplanted, a similar engraftment defect was not seen, suggesting that HSC loss is IFNg-dependent. In addition to loss of HSCs, secondary HSC transplants revealed a defect in HSC self-renewal capacity following repeated M. aviuminfection. Mathematical modeling demonstrated that the rate of HSC loss after chronic infection must increase by 57% from steady state to account for the observed decay in total HSCs. In order to define the mechanism of HSC loss during infection, we conducted mobilization and apoptosis assays after infection or IFNg treatment but found no evidence of increased displacement or death. Next we performed RNAseq profiling of HSCs from infected and control animals. GSEA analysis reflected increased myeloid differentiation rather than apoptosis during infection, consistent with our previous findings. We confirmed that IFNg treatment alone promotes myeloid differentiation of human CD34+ progenitors in vitro, with a reciprocal decrease in the persistence of CD34+CD38- HSPCs. Out of 4 transcription factors among the 151 differentially regulated genes identified by RNAseq, Batf2 stood out as one whose role in HSC biology has not been studied. We confirmed that Batf2 is induced in murine HSCs during infection, and that BATF2 is upregulated in human CD34+ hematopoietic progenitor cells following IFNg treatment. Consistent with a role in differentiation, retroviral overexpression of Batf2 in Sca1+ murine progenitor cells resulted in increased myeloid production upon transplant. Further, knockout of BATF2 in human CD34+ progenitors using CRISPR-Cas9 gene editing resulted in impaired myeloid differentiation in response to IFNg in vitro. Our studies demonstrate that chronic infection depletes the HSC pool by promoting HSC differentiation via an IFNg-dependent mechanism, and we identify the transcription factor Batf2 as a key player in infection-induced myeloid differentiation. These findings may provide a therapeutic opportunity to protect or restore hematopoiesis in patients with aplastic anemia, hemophagocytic histiocytosis (also associated with high IFNg levels), and chronic infections. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Dale E. Bockman ◽  
L. Y. Frank Wu ◽  
Alexander R. Lawton ◽  
Max D. Cooper

B-lymphocytes normally synthesize small amounts of immunoglobulin, some of which is incorporated into the cell membrane where it serves as receptor of antigen. These cells, on contact with specific antigen, proliferate and differentiate to plasma cells which synthesize and secrete large quantities of immunoglobulin. The two stages of differentiation of this cell line (generation of B-lymphocytes and antigen-driven maturation to plasma cells) are clearly separable during ontogeny and in some immune deficiency diseases. The present report describes morphologic aberrations of B-lymphocytes in two diseases in which second stage differentiation is defective.


Author(s):  
S. Trachtenberg ◽  
P.M. Steinert ◽  
B.L. Trus ◽  
A.C. Steven

During terminal differentiation of vertebrate epidermis, certain specific keratin intermediate filament (KIF) proteins are produced. Keratinization of the epidermis involves cell death and disruption of the cytoplasm, leaving a network of KIF embedded in an amorphous matrix which forms the outer horny layer known as the stratum corneum. Eventually these cells are shed (desquamation). Normally, the processes of differentiation, keratinization, and desquamation are regulated in an orderly manner. In psoriasis, a chronic skin disease, a hyperkeratotic stratum corneum is produced, resulting in abnormal desquamation of unusually large scales. In this disease, the normal KIF proteins are diminished in amount or absent, and other proteins more typical of proliferative epidermal cells are present. There is also evidence of proteolytic degradation of the KIF.


Author(s):  
Karvita B. Ahluwalia ◽  
Nidhi Sharma

It is common knowledge that apparently similar tumors often show different responses to therapy. This experience has generated the idea that histologically similar tumors could have biologically distinct behaviour. The development of effective therapy therefore, has the explicit challenge of understanding biological behaviour of a tumor. The question is which parameters in a tumor could relate to its biological behaviour ? It is now recognised that the development of malignancy requires an alteration in the program of terminal differentiation in addition to aberrant growth control. In this study therefore, ultrastructural markers that relate to defective terminal differentiation and possibly invasive potential of cells have been identified in human oral leukoplakias, erythroleukoplakias and squamous cell carcinomas of the tongue.


2000 ◽  
Vol 15 (12) ◽  
pp. H29-H29
Author(s):  
Vera D. Yoewono ◽  
E. Krinuhoni ◽  
W Marwoto ◽  
S.O. Sri Widodo

Sign in / Sign up

Export Citation Format

Share Document