scholarly journals Obesity-Induced Inflammation Cooperates with Loss of DNA Methyltransferase 3A to Develop Early-Onset of Leukemia

2021 ◽  
Vol 4 (1) ◽  
Author(s):  
Taruni Pandhiri ◽  
Santhosh Kumar Pasupuleti ◽  
Baskar Ramdas ◽  
Rahul Kanumuri ◽  
Reuben Kapur

Obesity is an increasing epidemic disease world-wide responsible for enhancing the risk for developing Type 2 diabetes mellitus (T2DM) as well as cancer. However, it is unclear if and how obesity contributes to the transformation of pre-leukemic stem and progenitors (pre-LHSC/Ps) into full-blown leukemia such as acute myeloid leukemia (AML) or severe form of myeloproliferative neoplasm (MPN). We hypothesized that obesity induced chronic inflammation might be responsible for clonal selection of pre-LHSC/Ps bearing pre-leukemic mutations such as DNA methyltransferase 3A (DNMT3A) and for promoting the progression of early-onset MPN towards severe forms of AML/leukemia. To test this hypothesis, we genetically crossed pre-leukemic Dnmt3a+/-;Mx-Cre+ mice with leptin deficient obese (LepOb/Ob) mice to obtain Ob/Ob;Dnmt3a+/-;Mx-Cre+ compound mutant mice. Further, the Dnmt3a gene was deleted by giving the PolyIC and the deletion was confirmed through PCR. After 12 days of post-PolyIC the myeloid cells (neutrophils and monocytes) were expanded in Ob/Ob;Dnmt3a+/-;Mx-Cre+ mice compared to Dnmt3a+/-;Mx-Cre+, Dnmt3a+/-;Mx-Cre-, Ob/Ob and WT mice. We have harvested and analyzed all these mice after 26 days of post-PolyIC. Interestingly, Ob/Ob;Dnmt3a+/-;Mx-Cre+ mice showed increased BM cellularity, both the frequency of lineage negative, Sca-1+ and c-KIT+ (LSK) cells, short-term hematopoietic stem cells (ST-HSCs; LSK/CD48+/CD150-), granulocyte macrophage progenitor (GMPs; LSK/CD16+/CD34+), and reduction in LT-HSCs (LT-HSCs; LSK/CD48-/CD150+) compared to other groups. Flow cytometry analysis of PB, BM and spleen from Ob/Ob;Dnmt3a+/-;Mx-Cre+ mice demonstrated a significant increase in the frequency of mature myeloid cells (Gr-1+/Mac-1+) and a profound reduction in B220+ B cells compared to other groups. Remarkably, these mice also showed splenomegaly, elevated heart size and early signs of AML blasts as reflected by the presence of c-KIT+/CD11b+ double positive cells in the BM, consistent with severe MPN/AML development. Taken together, these results demonstrate that obesity induced inflammation cooperates with pre-leukemic Dnmt3a+/- mutation to induce an early-onset of severe MPN/AML like disease.       

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1094-1094
Author(s):  
Santhosh Kumar Pasupuleti ◽  
Baskar Ramdas ◽  
Sarah S. Burns ◽  
Ramesh Kumar ◽  
Taruni Reddy Pandhiri ◽  
...  

Abstract Obesity is an increasing epidemic world-wide responsible for enhancing the risk for developing Type 2 diabetes mellitus (T2DM), cardiovascular disease (CVD) and cancer. However, it is unclear if and how obesity contributes to the transformation of pre-leukemic stem and progenitors (pre-LHSC/Ps) into full-blown leukemia such as acute myeloid leukemia (AML) or severe form of myeloproliferative neoplasm (MPN) or CVD. We hypothesized that obesity induced chronic inflammation might be responsible for clonal selection of pre-LHSC/Ps bearing pre-leukemic clonal hematopoiesis of indeterminate potential (CHIP) mutations such as DNMT3A, TET2, ASXL1, and JAK2 and for promoting the progression of early-onset MPN, AML/leukemia and CVD. To study the linkage between obesity and CHIP in humans, we first examined the UK biobank. After exclusions, the final study cohort included 47,466 unrelated participants free of T2DM at baseline and having valid CHIP measurements. The mean (SD) age at enrollment was 56.5 (8.0), 45.0% were male, 43.9% never smoked, and 82.6% self-reported as European decedents. At baseline, the mean (SD) body mass index (BMI) was 27.3 (4.7) kg/m 2, with 43.0% overweight and 23.6% obese, and the overall mean (SD) waist-to-hip ratio (WHR) was 0.87 (0.09). CHIP was present among 5.8% of the study population the most common mutations on the DNMT3A (3.7%) and TET2 (1.0%) genes; large CHIP clone defined as CHIP mutation with variant allele fraction >10% was present among 2.4% of the study population. Individuals with CHIP mutations on average had higher WHR. The presence of CHIP mutation was associated with a 0.0028 increase of WHR (p=0.03). Furthermore, CHIP prevalence increased with higher WHR: the percentage of participants with CHIP was 4.93%, 5.75%, 6.56% in the lowest, middle, and highest WHR quintiles respectively, signifying that dysfunctional metabolism may accelerate expansion of clonal hematopoiesis (CH). To better define the mechanism of obesity driven CH, we utilized several novel mouse models bearing Tet2 -/-, Dnmt3a +/-, Asxl1 +/- and Jak2 +/- mutations to mimic the human pre-LHSC/Ps condition and obesity, in the form of leptin deficient Lep Ob/Ob (Ob/Ob) mutation, which induces obesity and T2DM. We show that both the compound mutant (Tet2 -/-;Ob/Ob, Dnmt3a +/-;Ob/Ob, Asxl1 +/-;Ob/Ob and Jak2 +/-;Ob/Ob) and CHIP mutant bone marrow (BM; Tet2 -/-, Dnmt3a +/-, Asxl1 +/- and Jak2 +/-) transplanted into Ob/Ob mice develop rapid growth of mature myeloid cells and HSC/Ps leading to severe form of MPN/AML as well as CVD. This was associated with upregulation of pro-inflammatory cytokines such as IL-1β, IL-6 and TNF-α. Flow cytometry analysis of LSK and progenitor cells isolated from Tet2 -/-;Ob/Ob mice revealed an up-regulation of intracellular Ca2+ levels. We hypothesized that up-regulated Ca2+ signaling in Tet2 -/-; Ob/Ob HSC/Ps promotes aberrant signaling leading to an early-onset of severe MPN/AML. We performed a competitive transplantation experiment using, Tet2 -/-: Boy/J BM cells (1:1 ratio) into Ob/Ob and WT recipients. After 8 weeks post transplantation, we investigated the role of Ca2+ blockers in driving CH in Ob/Ob recipients using pharmacological inhibitors, either individually, or in combination, of metformin (100 mg/kg, orally), nifedipine (100 mg/kg, orally), MCC950 (30 mg/kg, orally) and anakinra (10 mg/kg, i.p). The combination treatment markedly reduced monocytes, neutrophils, WBC counts, and improved RBCs, hematocrits and platelets. The spleen and liver, heart, body weights and blood glucose levels were significantly reduced, along with a greater re-emergence of normal CD45.1 wild-type cells in the PB, BM, and spleen and a significant reduction in Tet2 mutant CD45.2 pre-LHSC/Ps and myeloid cells in the PB, BM, and spleen. Importantly, the frequency of leukemic blasts, LSK cells, ST-HSCs, LT-HSCs and granulocyte macrophage progenitors (GMP) were significantly reduced. Furthermore, the combination of drug treatment showed greater heart protective activity by reducing the atherosclerotic lesions in Ob/Ob recipients bearing CHIP by suppressing Ca2+ signaling. Taken together, these data suggest that obesity is highly associated with the presence of CHIP in humans and that targeting CHIP mutant cells with a combination of metformin/nifedipine/MCC950/anakinra is a safe and inexpensive way to rescue CH and its associated leukemic and cardiovascular defects. Disclosures Natarajan: Amgen: Research Funding; Apple: Consultancy, Research Funding; AstraZeneca: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Boston Scientific: Research Funding; Blackstone Life Sciences: Consultancy; Genentech: Consultancy; Foresite Labs: Consultancy.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2810-2810 ◽  
Author(s):  
Xiaoli Wang ◽  
Sonam Prakash ◽  
Min Lu ◽  
Yan Li ◽  
Attilio Orazi ◽  
...  

Abstract Abstract 2810 Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by abnormal trafficking of hematopoietic stem cells (HSC) and hematopoietic progenitor cells (HPC), resulting in their constitutive mobilization and the establishment of extramedullary hematopoiesis. The mechanism by which HSC/HPC preferentially migrate, reside, proliferate and differentiate in the spleen remains poorly understood. We phenotypically and functionally characterized PMF splenic CD34+ cells (PMFSC) (n=8). Greater numbers of CD34+ (5.9±1.9%), CD34+CD38- (3.2±1.6%), CD34+CD90+ (2.0±1.5%), CD34+Lin- (4.4±1.8%) cells and assayable HPCs (4513±240/106 mononuclear cells (MNC)) were detected in PMF spleens as compared with PMF peripheral blood (PB) (CD34+: 1.4±0.6%; CD34+CD38-: 0.8±0.6%; CD34+CD90+: 0.1±0%; CD34+Lin-: 0.8±0.4%; HPC: 2496±677/106 MNCs). In addition, the expression of CXCR4 by CD34+ cells present in the spleen (20.8±3.7%) was shown to be greater than that previously observed in PMF PB CD34+ cells (PMFPBC) (7.4±2.2%, P<0.05). We next examined PMF spleens for the presence of SCID repopulating cells by analyzing human cells in the marrow and spleen of NOD/SCID γcnull mice transplanted with PMFSC using flow cytometry. Five months following transplantation, 12.1% of the marrow cells were hCD45+ in mice receiving 2×105 PMFSC as compared to 3.4% in mice receiving same number of PMFPBC from the same patient. The percentage of hCD45+ cells in the marrow significantly increased to 40.6% in mice receiving 2×106 PMFSC, while 2.7% hCD45+ cells were detected in the marrow of mice receiving similar number of PMFPBC. Surprisingly, 0.6% of the cells were hCD45+ in the spleens of mice receiving 2×104 PMFSC, while 33.9% of the cells were hCD45+ in the spleens of mice receiving 2×105 PMFSC. By contrast, as few as 0.1% hCD45+ cells were detected in the spleens of mice transplanted with equal numbers of PMFPBC. Moreover, the spleens of mice transplanted with PMFSC contained greater number of hCD34+ cells, while no hCD34+ cells were observed in the spleens of mice transplanted with PMFPBC. The human cells in the marrow of mice receiving PMFSCs were further found to be capable of differentiating into not only myeloid cells (CD33+, 8.7%; glycophorin A+, 1.1% and CD34+, 1.3%) but also B (2.3%) and T cells (2.9%). While, the human cells in the marrow of mice receiving similar numbers of PMFPBC were composed of myeloid cells (CD33+, 0.5%; glycophorin A+, 0.1% and CD34+, 0.1%) but very few CD19+ (0.1%) and no CD3+ cells. Moreover, both myeloid cells (CD33+, 14.5%; glycophorin A+, 3.9% and CD34+, 6.8%) and CD19+ (6.7%) and CD3+ cells (23.1%) were detected in the spleens of mice receiving PMFSC. The engraftment of human cells in the spleen and marrow of mice receiving PMFSC and the distinct differentiative potential of PMFSC in mice were further confirmed by immunohistochemical analysis. Greater numbers of hCD45+ cells were observed in the marrow and spleen of mice receiving PMFSC and the degree of human cell engraftment was consistent with the number of PMFSC transplanted. Moreover, the white pulps of the spleens of mice receiving PMFSC were composed primarily of human lymphocytes. Both human granulocytic cells and human lymphocytes were observed in red pulps of the spleens of mice transplanted with PMFSC. In addition, following the infusion of 5×105 PMFSC and PMFPBC, reduced numbers of PMFSC (90.3±16.8/106 BMCs) were detected in the marrows of these mice as compared with G-CSF mobilized PB (mPB) CD34+ cells (196±19/106 BMCs; P<0.05), and the number of PMFSC that homed to the marrow of the mice was lower than PMFPBC (101.7±8.8/106 BMCs; P=0.09). By contrast, similar numbers of PMFSC and PMFPBC and mPB CD34+ cells were detected in the spleens of these mice. Recently the use of JAK2 inhibitors has been shown to dramatically reduce the size of the spleen in patients with PMF. PMFSC and PMFPBC were treated with a JAK2 inhibitor, AZD1480. Similar reductions in the number of total cells, CD34+ cells and assayable HPCs were observed as compared with cells exposed to cytokines alone. These findings suggest that multipotent HSCs are present in the spleen but not the PB of PMF patients and that PMFSC have a distinct differentiative program and migratory behavior that distinguishes them from PMF and normal PB CD34+ cells. In addition treatment with JAK2 inhibitors does not appear to lead to a reduction of splenomegly by preferentially inhibiting splenic HPCs. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 ◽  
Author(s):  
Zhi-Chun Ding ◽  
Nada S. Aboelella ◽  
Locke Bryan ◽  
Huidong Shi ◽  
Gang Zhou

Cyclophosphamide (CTX) is a major component of the chemotherapy conditioning regimens used in the clinic to prepare cancer patients for hematopoietic stem cell transplantation or adoptive T cell therapy. Previous studies have shown that CTX given at nonmyeloablative doses in mice and patients leads to expansion of myeloid cells within which the monocytic subset exhibits immunosuppressive activity. However, the ontogeny and gene expression signature of these CTX-induced monocytes are not well-defined. Here, we report that the expansion of myeloid cells is a default process intrinsic to hematopoietic recovery after chemotherapy. During this process, the monocytes repopulated in mice acquire immunosuppressive activity, which can persist long after cessation of chemotherapy. Moreover, monocytes acquire a gene signature characteristic of neutrophil precursors, marked by increased proliferative capability and elevated expressions of multiple primary and secondary granules. We provide evidence that CTX-induced myeloid cell expansion is regulated by DNA methyltransferase 1 (Dnmt1) and dependent on chemotherapy-induced microbial translocation. These findings help advance our understanding of the differentiation, heterogeneity, and function of myeloid cells repopulating after chemotherapy.


Cells ◽  
2021 ◽  
Vol 10 (5) ◽  
pp. 968
Author(s):  
Tyler J. Wildes ◽  
Bayli DiVita Dean ◽  
Catherine T. Flores

Our understanding of the relationship between the immune system and cancers has undergone significant discovery recently. Immunotherapy with T cell therapies and checkpoint blockade has meaningfully changed the oncology landscape. While remarkable clinical advances in adaptive immunity are occurring, modulation of innate immunity has proven more difficult. The myeloid compartment, including macrophages, neutrophils, and dendritic cells, has a significant impact on the persistence or elimination of tumors. Myeloid cells, specifically in the tumor microenvironment, have direct contact with tumor tissue and coordinate with tumor-reactive T cells to either stimulate or antagonize cancer immunity. However, the myeloid compartment comprises a broad array of cells in various stages of development. In addition, hematopoietic stem and progenitor cells at various stages of myelopoiesis in distant sites undergo significant modulation by tumors. Understanding how tumors exert their influence on myeloid progenitors is critical to making clinically meaningful improvements in these pathways. Therefore, this review will cover recent developments in our understanding of how solid tumors modulate myelopoiesis to promote the formation of pro-tumor immature myeloid cells. Then, it will cover some of the potential avenues for capitalizing on these mechanisms to generate antitumor immunity.


Biomolecules ◽  
2021 ◽  
Vol 11 (1) ◽  
pp. 122
Author(s):  
Mariarita Spampinato ◽  
Cesarina Giallongo ◽  
Alessandra Romano ◽  
Lucia Longhitano ◽  
Enrico La Spina ◽  
...  

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by hematopoietic stem-cell-derived clonal proliferation, leading to bone marrow (BM) fibrosis. Hematopoiesis alterations are closely associated with modifications of the BM microenvironment, characterized by defective interactions between vascular and endosteal niches. As such, neoangiogenesis, megakaryocytes hyperplasia and extensive bone marrow fibrosis, followed by osteosclerosis and bone damage, are the most relevant consequences of PMF. Moreover, bone tissue deposition, together with progressive fibrosis, represents crucial mechanisms of disabilities in patients. Although the underlying mechanisms of bone damage observed in PMF are still unclear, the involvement of cytokines, growth factors and bone marrow microenvironment resident cells have been linked to disease progression. Herein, we focused on the role of megakaryocytes and their alterations, associated with cytokines and chemokines release, in modulating functions of most of the bone marrow cell populations and in creating a complex network where impaired signaling strongly contributes to progression and disabilities.


Sign in / Sign up

Export Citation Format

Share Document