42 results on '"Trowbridge JJ"'
Search Results
2. In vivo models of subclonal oncogenesis and dependency in hematopoietic malignancy.
- Author
-
Bowman RL, Dunbar AJ, Mishra T, Xiao W, Waarts MR, Maestre IF, Eisman SE, Cai L, Mowla S, Shah N, Youn A, Bennett L, Fontenard S, Gounder S, Gandhi A, Bowman M, O'Connor K, Zaroogian Z, Sánchez-Vela P, Martinez Benitez AR, Werewski M, Park Y, Csete IS, Krishnan A, Lee D, Boorady N, Potts CR, Jenkins MT, Cai SF, Carroll MP, Meyer SE, Miles LA, Ferrell PB Jr, Trowbridge JJ, and Levine RL
- Subjects
- Animals, Humans, Mice, Isocitrate Dehydrogenase genetics, DNA (Cytosine-5-)-Methyltransferases genetics, DNA (Cytosine-5-)-Methyltransferases metabolism, Hematologic Neoplasms genetics, Hematologic Neoplasms pathology, Cell Transformation, Neoplastic genetics, Nuclear Proteins genetics, Nuclear Proteins metabolism, Mutagenesis, Mutation, Nucleophosmin, DNA Methyltransferase 3A genetics, Carcinogenesis genetics, fms-Like Tyrosine Kinase 3 genetics
- Abstract
Cancer evolution is a multifaceted process leading to dysregulation of cellular expansion and differentiation through somatic mutations and epigenetic dysfunction. Clonal expansion and evolution is driven by cell-intrinsic and -extrinsic selective pressures, which can be captured with increasing resolution by single-cell and bulk DNA sequencing. Despite the extensive genomic alterations revealed in profiling studies, there remain limited experimental systems to model and perturb evolutionary processes. Here, we integrate multi-recombinase tools for reversible, sequential mutagenesis from premalignancy to leukemia. We demonstrate that inducible Flt3 mutations differentially cooperate with Dnmt3a, Idh2, and Npm1 mutant alleles, and that changing the order of mutations influences cellular and transcriptional landscapes. We next use a generalizable, reversible approach to demonstrate that mutation reversion results in rapid leukemic regression with distinct differentiation patterns depending upon co-occurring mutations. These studies provide a path to experimentally model sequential mutagenesis, investigate mechanisms of transformation and probe oncogenic dependency in disease evolution., Competing Interests: Declaration of interests R.L.L. is on the Supervisory board of Qiagen, a co-founder/board member at Ajax, and is a scientific advisor to Mission Bio, Syndax, Zentalis, Auron, Prelude, and C4 Therapeutics; for each of these entities he receives equity. He has received research support from Calico, Zentalis and Ajax, and has consulted for Incyte, Astra Zeneca and Janssen. A.J.D. is on an advisory board for Morphosys and a consultant for RayThera. S.F.C. is a consultant for Daiichi-Sankyo and Ursamin. He was previously a consultant for Dava Oncology and held equity interest in Imago Biosciences, none of which are directly related to the content of this paper. R.L.B. and L.A.M. have received honoraria from Mission Bio and are members of the Speakers Bureau for Mission Bio. M.P.C. has consulted for Janssen Pharmaceuticals. J.J.T. held a sponsored research project with H3 Biomedicine. P.B.F. has received research funding from Novartis. No other authors report competing interests. R.L.B., A.J.D., L.A.M., and R.L.L. have a pending patent on this work PCT/US2023/066910., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Oncostatin M receptor-dependent signaling assessed by RNA sequencing in mouse hematopoietic stem cells.
- Author
-
Schwartz LS, Saxl RL, Stearns T, Telpoukhovskaia M, and Trowbridge JJ
- Subjects
- Animals, Mice, Oncostatin M Receptor beta Subunit genetics, Sequence Analysis, RNA, Receptors, Oncostatin M genetics, RNA-Seq, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells drug effects, Signal Transduction, Oncostatin M pharmacology
- Abstract
Oncostatin M (OSM) is a member of the interleukin-6 (IL-6) family of cytokines and has been found to have anti-inflammatory and pro-inflammatory properties in various cellular and disease contexts. OSM signals through two receptor complexes, one of which includes OSMRβ. Here, we investigated OSM-OSMRβ signaling in adult mouse hematopoietic stem cells (HSCs) using the conditional Osmr
fl/fl mouse model B6;129-Osmrtm1.1Nat /J. We crossed Osmrfl/fl mice to interferon-inducible Mx1-Cre, which is robustly induced in adult HSCs. From these mice, we isolated HSCs by flow cytometry, stimulated with recombinant OSM or vehicle for 1 hour, and assessed gene expression changes in control versus Osmr knockout HSCs by RNA-seq. This data may be utilized to investigate OSMRβ -dependent and -independent OSM signaling as well as the transcriptional effects of an IL-6 family cytokine on mouse HSCs to further define its anti-inflammatory versus pro-inflammatory properties., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
4. Variation in mesenchymal KITL/SCF and IGF1 expression in middle age underlies steady-state hematopoietic stem cell aging.
- Author
-
Young KA, Telpoukhovskaia MA, Hofmann J, Mistry JJ, Kokkaliaris KD, and Trowbridge JJ
- Subjects
- Animals, Mice, Female, Aging metabolism, Aging genetics, Mice, Inbred C57BL, Mesenchymal Stem Cells metabolism, Mesenchymal Stem Cells cytology, Transcriptome, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells cytology, Insulin-Like Growth Factor I metabolism, Insulin-Like Growth Factor I genetics, Stem Cell Factor metabolism, Stem Cell Factor genetics, Cellular Senescence
- Abstract
Abstract: Intrinsic molecular programs and extrinsic factors including proinflammatory molecules are understood to regulate hematopoietic aging. This is based on foundational studies using genetic perturbation to evaluate causality. However, individual organisms exhibit natural variation in the hematopoietic aging phenotypes and the molecular basis of this heterogeneity is poorly understood. Here, we generated individual single-cell transcriptomic profiles of hematopoietic and nonhematopoietic cell types in 5 young adult and 9 middle-aged C57BL/6J female mice, providing a web-accessible transcriptomic resource for the field. Among all assessed cell types, hematopoietic stem cells (HSCs) exhibited the greatest phenotypic variation in expansion among individual middle-aged mice. We computationally pooled samples to define modules representing the molecular signatures of middle-aged HSCs and interrogated, which extrinsic regulatory cell types and factors would predict the variance in these signatures between individual middle-aged mice. Decline in signaling mediated by adiponectin, kit ligand (KITL) and insulin-like growth factor 1 (IGF1) from mesenchymal stromal cells (MSCs) was predicted to have the greatest transcriptional impact on middle-aged HSCs, as opposed to signaling mediated by endothelial cells or mature hematopoietic cell types. In individual middle-aged mice, lower expression of Kitl and Igf1 in MSCs was highly correlated with reduced lymphoid lineage commitment of HSCs and increased signatures of differentiation-inactive HSCs. These signatures were independent of expression of aging-associated proinflammatory cytokines including interleukin-1β (IL-1β), IL-6, tumor necrosis factor α and RANTES. In sum, we find that Kitl and Igf1 expression are coregulated and variable between individual mice at the middle age and expression of these factors is predictive of HSC activation and lymphoid commitment independently of inflammation., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
5. Mesenchymal Stromal Cell Senescence Induced by Dnmt3a -Mutant Hematopoietic Cells is a Targetable Mechanism Driving Clonal Hematopoiesis and Initiation of Hematologic Malignancy.
- Author
-
Mistry JJ, Young KA, Colom Díaz PA, Maestre IF, Levine RL, and Trowbridge JJ
- Abstract
Clonal hematopoiesis (CH) can predispose to blood cancers due to enhanced fitness of mutant hematopoietic stem and progenitor cells (HSPCs), but the mechanisms driving this progression are not understood. We hypothesized that malignant progression is related to microenvironment-remodelling properties of CH-mutant HSPCs. Single-cell transcriptomic profiling of the bone marrow microenvironment in Dnmt3a
R878H/+ mice revealed signatures of cellular senescence in mesenchymal stromal cells (MSCs). Dnmt3aR878H/+ HSPCs caused MSCs to upregulate the senescence markers SA-β-gal, BCL-2, BCL-xL, Cdkn1a (p21) and Cdkn2a (p16), ex vivo and in vivo . This effect was cell contact-independent and can be replicated by IL-6 or TNFα, which are produced by Dnmt3aR878H/+ HSPCs. Depletion of senescent MSCs in vivo reduced the fitness of Dnmt3aR878H/+ hematopoietic cells and the progression of CH to myeloid neoplasms using a sequentially inducible Dnmt3a ; Npm1 -mutant model. Thus, Dnmt3a -mutant HSPCs reprogram their microenvironment via senescence induction, creating a self-reinforcing niche favoring fitness and malignant progression., Statement of Significance: Mesenchymal stromal cell senescence induced by Dnmt3a -mutant hematopoietic stem and progenitor cells drives clonal hematopoiesis and initiation of hematologic malignancy.- Published
- 2024
- Full Text
- View/download PDF
6. Metformin reduces the clonal fitness of Dnmt3a R878H hematopoietic stem and progenitor cells by reversing their aberrant metabolic and epigenetic state.
- Author
-
Hosseini M, Voisin V, Chegini A, Varesi A, Cathelin S, Ayyathan DM, Liu ACH, Yang Y, Wang V, Maher A, Grignano E, Reisz JA, D'Alessandro A, Young K, Wu Y, Fiumara M, Ferrari S, Naldini L, Gaiti F, Pai S, Schimmer AD, Bader GD, Dick JE, Xie SZ, Trowbridge JJ, and Chan SM
- Abstract
Clonal hematopoiesis (CH) arises when a hematopoietic stem cell (HSC) acquires a mutation that confers a competitive advantage over wild-type (WT) HSCs, resulting in its clonal expansion. Individuals with CH are at an increased risk of developing hematologic neoplasms and a range of age-related inflammatory illnesses
1-3 . Therapeutic interventions that suppress the expansion of mutant HSCs have the potential to prevent these CH-related illnesses; however, such interventions have not yet been identified. The most common CH driver mutations are in the DNA methyltransferase 3 alpha ( DNMT3A ) gene with arginine 882 (R882) being a mutation hotspot. Here we show that murine hematopoietic stem and progenitor cells (HSPCs) carrying the Dnmt3aR878H/+ mutation, which is equivalent to human DNMT3AR882H/+ , have increased mitochondrial respiration compared with WT cells and are dependent on this metabolic reprogramming for their competitive advantage. Importantly, treatment with metformin, an oral anti-diabetic drug with inhibitory activity against complex I in the electron transport chain (ETC), reduced the fitness of Dnmt3aR878H/+ HSCs. Through a multi-omics approach, we discovered that metformin acts by enhancing the methylation potential in Dnmt3aR878H/+ HSPCs and reversing their aberrant DNA CpG methylation and histone H3K27 trimethylation (H3K27me3) profiles. Metformin also reduced the fitness of human DNMT3AR882H HSPCs generated by prime editing. Our findings provide preclinical rationale for investigating metformin as a preventive intervention against illnesses associated with DNMT3AR882 mutation-driven CH in humans., Competing Interests: Conflicts of Interest S.M.C. has received research funding from the Centre for Oncology and Immunology in Hong Kong, Celgene/BMS, AbbVie Pharmaceuticals, Agios Pharmaceuticals, and Servier Laboratories. F.G. serves as a consultant for S2 Genomics Inc. A.D.S. has received research funding from Takeda Pharmaceuticals, BMS and Medivir AB, and consulting fees/honorarium from Takeda, Novartis, Jazz, and Otsuka Pharmaceuticals. A.D.S. is named on a patent application for the use of DNT cells to treat AML. A.D.S. is a member of the Medical and Scientific Advisory Board of the Leukemia and Lymphoma Society of Canada. A.D.S. holds the Ronald N. Buick Chair in Oncology Research. J.E.D. has received research funding from Celgene/BMS, and has patents licensed to Trillium Therapeutics/Pfizer.- Published
- 2024
- Full Text
- View/download PDF
7. Transcriptional and functional consequences of Oncostatin M signaling on young Dnmt3a-mutant hematopoietic stem cells.
- Author
-
Schwartz LS, Young KA, Stearns TM, Boyer N, Mujica KD, and Trowbridge JJ
- Subjects
- Animals, Mice, Anti-Inflammatory Agents, Hematopoiesis genetics, Oncostatin M genetics, Bone Marrow, Hematopoietic Stem Cells
- Abstract
Age-associated clonal hematopoiesis (CH) occurs due to somatic mutations accrued in hematopoietic stem cells (HSCs) that confer a selective growth advantage in the context of aging. The mechanisms by which CH-mutant HSCs gain this advantage with aging are not comprehensively understood. Using unbiased transcriptomic approaches, we identified Oncostatin M (OSM) signaling as a candidate contributor to age-related Dnmt3a-mutant CH. We found that Dnmt3a-mutant HSCs from young adult mice (3-6 months old) subjected to acute OSM stimulation do not demonstrate altered proliferation, apoptosis, hematopoietic engraftment, or myeloid differentiation. Dnmt3a-mutant HSCs from young mice do transcriptionally upregulate an inflammatory cytokine network in response to acute in vitro OSM stimulation as evidenced by significant upregulation of the genes encoding IL-6, IL-1β, and TNFα. OSM-stimulated Dnmt3a-mutant HSCs also demonstrate upregulation of the anti-inflammatory genes Socs3, Atf3, and Nr4a1. In the context of an aged bone marrow (BM) microenvironment, Dnmt3a-mutant HSCs upregulate proinflammatory genes but not the anti-inflammatory genes Socs3, Atf3, and Nr4a1. The results from our studies suggest that aging may exhaust the regulatory mechanisms that HSCs employ to resolve inflammatory states in response to factors such as OSM., Competing Interests: Conflict of Interest Disclosure J.J.T. has received research support from H3 Biomedicine, Inc., and patent royalties from Fate Therapeutics. All other authors declare no competing interests., (Copyright © 2023 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Author Correction: Clonal hematopoiesis, aging and Alzheimer's disease.
- Author
-
Sánchez Vela P, Trowbridge JJ, and Levine RL
- Published
- 2024
- Full Text
- View/download PDF
9. Characterization of an Osmr Conditional Knockout Mouse Model.
- Author
-
Schwartz LS, Saxl RL, Stearns T, and Trowbridge JJ
- Abstract
Oncostatin M (OSM) is a member of the interleukin-6 (IL-6) family of cytokines and has been found to have distinct anti-inflammatory and pro-inflammatory properties in various cellular and disease contexts. OSM signals through two receptor complexes, one of which includes OSMRβ. To investigate OSM-OSMRβ signaling in adult hematopoiesis, we utilized the readily available conditional Osmr
fl/fl mouse model B6;129- Osmrtm1.1Nat /J, which is poorly characterized in the literature. This model contains loxP sites flanking exon 2 of the Osmr gene. We crossed Osmrfl/fl mice to interferon-inducible Mx1 -Cre, which is robustly induced in adult hematopoietic cells. We observed complete recombination of the Osmrfl allele and loss of exon 2 in hematopoietic (bone marrow) as well as non-hematopoietic (liver, lung, kidney) tissues. Using a TaqMan assay with probes downstream of exon 2, Osmr transcript was lower in the kidney but equivalent in bone marrow, lung, and liver from Osmrfl/fl Mx1 -Cre versus Mx1 -Cre control mice, suggesting that transcript is being produced despite loss of this exon. Western blots show that liver cells from Osmrfl/fl Mx1 -Cre mice had complete loss of OSMR protein, while bone marrow, kidney, and lung cells had reduced OSMR protein at varying levels. RNA-seq analysis of a subpopulation of bone marrow cells (hematopoietic stem cells) finds that some OSM-stimulated genes, but not all, are suppressed in Osmrfl/fl Mx1 -Cre cells. Together, our data suggest that the B6;129- Osmrtm1.1Nat /J model should be utilized with caution as loss of Osmr exon 2 has variable and tissue-dependent impact on mRNA and protein expression., Competing Interests: CONFLICT OF INTEREST DISCLOSURE J.J.T. has previously received research support from H3 Biomedicine, Inc., and patent royalties from Fate Therapeutics.- Published
- 2023
- Full Text
- View/download PDF
10. New insight into the causes, consequences, and correction of hematopoietic stem cell aging.
- Author
-
Mansell E, Lin DS, Loughran SJ, Milsom MD, and Trowbridge JJ
- Subjects
- Humans, Aged, Hematopoietic Stem Cells metabolism, Cellular Senescence genetics, Aging pathology, Hematologic Diseases metabolism
- Abstract
Aging of hematopoietic stem cells (HSCs) is characterized by lineage bias, increased clonal expansion, and functional decrease. At the molecular level, aged HSCs typically display metabolic dysregulation, upregulation of inflammatory pathways, and downregulation of DNA repair pathways. Cellular aging of HSCs, driven by cell-intrinsic and cell-extrinsic factors, causes a predisposition to anemia, adaptive immune compromise, myelodys, plasia, and malignancy. Most hematologic diseases are strongly associated with age. But what is the biological foundation for decreased fitness with age? And are there therapeutic windows to resolve age-related hematopoietic decline? These questions were the focus of the International Society for Experimental Hematology (ISEH) New Investigator Committee Fall 2022 Webinar. This review touches on the latest insights from two leading laboratories into inflammatory- and niche-driven stem cell aging and includes speculation on strategies to prevent or correct age-related decline in HSC function., Competing Interests: Conflict of Interest Disclosure The authors do not have any conflicts of interest to declare in relation to this work, (Copyright © 2023 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. Hematopoietic stem cell aging and leukemia transformation.
- Author
-
Colom Díaz PA, Mistry JJ, and Trowbridge JJ
- Subjects
- Humans, Aged, Cellular Senescence, Aging, Hematopoietic Stem Cells physiology, Hematopoiesis physiology, Leukemia therapy, Hematologic Neoplasms therapy
- Abstract
With aging, hematopoietic stem cells (HSCs) have an impaired ability to regenerate, differentiate, and produce an entire repertoire of mature blood and immune cells. Owing to dysfunctional hematopoiesis, the incidence of hematologic malignancies increases among elderly individuals. Here, we provide an update on HSC-intrinsic and -extrinsic factors and processes that were recently discovered to contribute to the functional decline of HSCs during aging. In addition, we discuss the targets and timing of intervention approaches to maintain HSC function during aging and the extent to which these same targets may prevent or delay transformation to hematologic malignancies., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Oncostatin M is a Master Regulator of an Inflammatory Network in Dnmt3a -Mutant Hematopoietic Stem Cells.
- Author
-
Schwartz LS, Young KA, Stearns TM, Boyer N, Mujica KD, and Trowbridge JJ
- Abstract
Age-associated clonal hematopoiesis (CH) occurs due to somatic mutations accrued in hematopoietic stem cells (HSCs) that confer a selective advantage in the context of aging. The mechanisms by which CH-mutant HSCs gain this advantage with aging are not comprehensively understood. Using unbiased transcriptomic approaches, we identify Oncostatin M (OSM) signaling as a candidate contributor to aging-driven Dnmt3a -mutant CH. We find that Dnmt3a -mutant HSCs from young mice do not functionally respond to acute OSM stimulation with respect to proliferation, apoptosis, hematopoietic engraftment, or myeloid differentiation. However, young Dnmt3a -mutant HSCs transcriptionally upregulate an inflammatory cytokine network in response to acute OSM stimulation including genes encoding IL-6, IL-1β and TNFα. In addition, OSM-stimulated Dnmt3a -mutant HSCs upregulate the anti-inflammatory genes Socs3, Atf3 and Nr4a1 , creating a negative feedback loop limiting sustained activation of the inflammatory network. In the context of an aged bone marrow (BM) microenvironment with chronically elevated levels of OSM, Dnmt3a -mutant HSCs upregulate pro-inflammatory genes but do not upregulate Socs3, Atf3 and Nr4a1 . Together, our work suggests that chronic inflammation with aging exhausts the regulatory mechanisms in young CH-mutant HSCs that resolve inflammatory states, and that OSM is a master regulator of an inflammatory network that contributes to age-associated CH.
- Published
- 2023
- Full Text
- View/download PDF
13. Clonal hematopoiesis, aging and Alzheimer's disease.
- Author
-
Sánchez Vela P, Trowbridge JJ, and Levine RL
- Subjects
- Humans, Aging genetics, Clonal Hematopoiesis, Hematopoiesis genetics, Mutation genetics, Alzheimer Disease genetics, Cardiovascular Diseases
- Published
- 2023
- Full Text
- View/download PDF
14. Variation in histone configurations correlates with gene expression across nine inbred strains of mice.
- Author
-
Tyler AL, Spruce C, Kursawe R, Haber A, Ball RL, Pitman WA, Fine AD, Raghupathy N, Walker M, Philip VM, Baker CL, Mahoney JM, Churchill GA, Trowbridge JJ, Stitzel ML, Paigen K, Petkov PM, and Carter GW
- Subjects
- Humans, Mice, Animals, Promoter Regions, Genetic, Chromatin genetics, Epigenesis, Genetic, Histone Code, Mice, Inbred Strains, Gene Expression, Histones genetics, Histones metabolism, DNA Methylation
- Abstract
The Diversity Outbred (DO) mice and their inbred founders are widely used models of human disease. However, although the genetic diversity of these mice has been well documented, their epigenetic diversity has not. Epigenetic modifications, such as histone modifications and DNA methylation, are important regulators of gene expression and, as such, are a critical mechanistic link between genotype and phenotype. Therefore, creating a map of epigenetic modifications in the DO mice and their founders is an important step toward understanding mechanisms of gene regulation and the link to disease in this widely used resource. To this end, we performed a strain survey of epigenetic modifications in hepatocytes of the DO founders. We surveyed four histone modifications (H3K4me1, H3K4me3, H3K27me3, and H3K27ac), as well as DNA methylation. We used ChromHMM to identify 14 chromatin states, each of which represents a distinct combination of the four histone modifications. We found that the epigenetic landscape is highly variable across the DO founders and is associated with variation in gene expression across strains. We found that epigenetic state imputed into a population of DO mice recapitulated the association with gene expression seen in the founders, suggesting that both histone modifications and DNA methylation are highly heritable mechanisms of gene expression regulation. We illustrate how DO gene expression can be aligned with inbred epigenetic states to identify putative cis -regulatory regions. Finally, we provide a data resource that documents strain-specific variation in the chromatin state and DNA methylation in hepatocytes across nine widely used strains of laboratory mice., (© 2023 Tyler et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2023
- Full Text
- View/download PDF
15. Challenges and opportunities for modeling aging and cancer.
- Author
-
Anczuków O, Airhart S, Chuang JH, Coussens LM, Kuchel GA, Korstanje R, Li S, Lucido AL, McAllister SS, Politi K, Polyak K, Ratliff T, Ren G, Trowbridge JJ, Ucar D, and Palucka K
- Subjects
- Animals, Mice, Disease Models, Animal, Risk Factors, Aging, Neoplasms genetics
- Abstract
Age is among the main risk factors for cancer, and any cancer study in adults is faced with an aging tissue and organism. Yet, pre-clinical studies are carried out using young mice and are not able to address the impact of aging and associated comorbidities on disease biology and treatment outcomes. Here, we discuss the limitations of current mouse cancer models and suggest strategies for developing novel models to address these major gaps in knowledge and experimental approaches., Competing Interests: Declaration of interests K. Polyak serves on the scientific advisory boards of Novartis, Vividion Therapeutics, Ideya Biosciences, and Scorpion Therapeutics; holds equity options in Scorpion Therapeutics; has received honorarium from Astra-Zeneca, New Equilibrium Biosciences, and Roche in the past 12 months; and receives sponsored research funding from Novartis. K. Politi is coinventor on a patent for EGFRT790M mutation testing issued, licensed, and with royalties paid from Molecular Diagnostics/Memorial Sloan Kettering Cancer Center. She reports research funding to her institution from AstraZeneca, Roche/Genentech, Boehringer Ingelheim, and D2G Oncology, and consulting for AstraZeneca and Jannssen. K. Palucka is a cofounder of Guardian Bio and holds equity and receives research support from Guardian Bio. She is a member of the scientific advisory board and holds equity from Cue Biopharma. She received research support from Merck in the past. O.A. has received research support from Sanofi and Pacbio in the past. J.J.T. receives patent royalties from Fate Therapeutics. She has also received research support from H3 Biomedicine, Inc. L.M.C. reports consulting services for Cell Signaling Technologies, AbbVie, the Susan G. Komen Foundation, and Shasqi; has received reagent and/or research support from Cell Signaling Technologies, Syndax Pharmaceuticals, ZelBio, Inc., Hibercell, Inc., Acerta Pharma, Prospect Creek Foundation, the Susan G. Komen Foundation, and National Foundation for Cancer Research; and has participated in advisory boards for Syndax Pharmaceuticals, Carisma Therapeutics, Inc., CytomX Therapeutics, Inc., Kineta, Inc., Hibercell, Inc., Cell Signaling Technologies, Alkermes, Inc., Genenta Sciences, Pio Therapeutics, Pty., Ltd., PDX Pharmaceuticals, Inc., NextCure, Guardian Bio, the AstraZeneca Partner of Choice Network, the Lustgarten Foundation, and the NIH/NCI-Frederick National Laboratory Advisory Committee., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
16. Distinct Tumor Necrosis Factor Alpha Receptors Dictate Stem Cell Fitness versus Lineage Output in Dnmt3a-Mutant Clonal Hematopoiesis.
- Author
-
SanMiguel JM, Eudy E, Loberg MA, Young KA, Mistry JJ, Mujica KD, Schwartz LS, Stearns TM, Challen GA, and Trowbridge JJ
- Subjects
- Animals, Mice, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha metabolism, Hematopoiesis genetics, Hematopoietic Stem Cells metabolism, Cell Lineage genetics, Clonal Hematopoiesis, Receptors, Tumor Necrosis Factor, Type I genetics, Receptors, Tumor Necrosis Factor, Type I metabolism
- Abstract
Clonal hematopoiesis resulting from the enhanced fitness of mutant hematopoietic stem cells (HSC) associates with both favorable and unfavorable health outcomes related to the types of mature mutant blood cells produced, but how this lineage output is regulated is unclear. Using a mouse model of a clonal hematopoiesis-associated mutation, DNMT3AR882/+ (Dnmt3aR878H/+), we found that aging-induced TNFα signaling promoted the selective advantage of mutant HSCs and stimulated the production of mutant B lymphoid cells. The genetic loss of the TNFα receptor TNFR1 ablated the selective advantage of mutant HSCs without altering their lineage output, whereas the loss of TNFR2 resulted in the overproduction of mutant myeloid cells without altering HSC fitness. These results nominate TNFR1 as a target to reduce clonal hematopoiesis and the risk of associated diseases and support a model in which clone size and mature blood lineage production can be independently controlled to modulate favorable and unfavorable clonal hematopoiesis outcomes., Significance: Through the identification and dissection of TNFα signaling as a key driver of murine Dnmt3a-mutant hematopoiesis, we report the discovery that clone size and production of specific mature blood cell types can be independently regulated. See related commentary by Niño and Pietras, p. 2724. This article is highlighted in the In This Issue feature, p. 2711., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
17. Cell origin-dependent cooperativity of mutant Dnmt3a and Npm1 in clonal hematopoiesis and myeloid malignancy.
- Author
-
SanMiguel JM, Eudy E, Loberg MA, Miles LA, Stearns T, Mistry JJ, Rauh MJ, Levine RL, and Trowbridge JJ
- Subjects
- Animals, Chromatin, Clonal Hematopoiesis, DNA (Cytosine-5-)-Methyltransferases genetics, DNA Methyltransferase 3A, Mice, Nucleophosmin, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases therapeutic use, Leukemia, Myeloid, Acute drug therapy, Myeloproliferative Disorders pathology
- Abstract
In adult acute myeloid leukemia (AML), the acquisition of driver somatic mutations may be preceded by a benign state termed clonal hematopoiesis (CH). To develop therapeutic strategies to prevent leukemia development from CH, it is important to understand the mechanisms by which CH-driving and AML-driving mutations cooperate. Here, we use mice with inducible mutant alleles common in human CH (DNMT3AR882; mouse Dnmt3aR878H) and AML (NPM1c; mouse Npm1cA). We find that Dnmt3aR878H/+ hematopoietic stem cells (HSCs), but not multipotent progenitor cell (MPP) subsets, have reduced cytokine expression and proinflammatory transcriptional signatures and a functional competitive advantage over their wild-type counterparts. Dnmt3aR878H/+ HSCs are the most potent cell type transformed by Npm1cA, generating myeloid malignancies in which few additional cooperating somatic mutation events were detected. At a molecular level, Npm1cA, in cooperation with Dnmt3aR878H, acutely increased the accessibility of a distinct set of promoters in HSCs compared with MPP cells. These promoters were enriched for cell cycling, PI3K/AKT/mTOR signaling, stem cell signatures, and targets of transcription factors, including NFAT and the chromatin binding factor HMGB1, which have been implicated in human AML. These results demonstrate cooperativity between preexisting Dnmt3aR878H and Npm1cA at the chromatin level, where specific loci altered in accessibility by Npm1cA are dependent on cell context as well as Dnmt3a mutation status. These findings have implications for biological understanding and therapeutic intervention in the transformation from CH to AML., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
18. Actinomycin D Targets NPM1c-Primed Mitochondria to Restore PML-Driven Senescence in AML Therapy.
- Author
-
Wu HC, Rérolle D, Berthier C, Hleihel R, Sakamoto T, Quentin S, Benhenda S, Morganti C, Wu C, Conte L, Rimsky S, Sebert M, Clappier E, Souquere S, Gachet S, Soulier J, Durand S, Trowbridge JJ, Bénit P, Rustin P, El Hajj H, Raffoux E, Ades L, Itzykson R, Dombret H, Fenaux P, Espeli O, Kroemer G, Brunetti L, Mak TW, Lallemand-Breitenbach V, Bazarbachi A, Falini B, Ito K, Martelli MP, and de Thé H
- Subjects
- Dactinomycin pharmacology, Dactinomycin therapeutic use, Humans, Mitochondria metabolism, Nucleophosmin, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Nuclear Proteins genetics, Nuclear Proteins metabolism
- Abstract
Acute myeloid leukemia (AML) pathogenesis often involves a mutation in the NPM1 nucleolar chaperone, but the bases for its transforming properties and overall association with favorable therapeutic responses remain incompletely understood. Here we demonstrate that an oncogenic mutant form of NPM1 (NPM1c) impairs mitochondrial function. NPM1c also hampers formation of promyelocytic leukemia (PML) nuclear bodies (NB), which are regulators of mitochondrial fitness and key senescence effectors. Actinomycin D (ActD), an antibiotic with unambiguous clinical efficacy in relapsed/refractory NPM1c-AMLs, targets these primed mitochondria, releasing mitochondrial DNA, activating cyclic GMP-AMP synthase signaling, and boosting reactive oxygen species (ROS) production. The latter restore PML NB formation to drive TP53 activation and senescence of NPM1c-AML cells. In several models, dual targeting of mitochondria by venetoclax and ActD synergized to clear AML and prolong survival through targeting of PML. Our studies reveal an unexpected role for mitochondria downstream of NPM1c and implicate a mitochondrial/ROS/PML/TP53 senescence pathway as an effector of ActD-based therapies., Significance: ActD induces complete remissions in NPM1-mutant AMLs. We found that NPM1c affects mitochondrial biogenesis and PML NBs. ActD targets mitochondria, yielding ROS which enforce PML NB biogenesis and restore senescence. Dual targeting of mitochondria with ActD and venetoclax sharply potentiates their anti-AML activities in vivo. This article is highlighted in the In This Issue feature, p. 2945., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
19. Decline in IGF1 in the bone marrow microenvironment initiates hematopoietic stem cell aging.
- Author
-
Young K, Eudy E, Bell R, Loberg MA, Stearns T, Sharma D, Velten L, Haas S, Filippi MD, and Trowbridge JJ
- Subjects
- Aging, Animals, Cross-Sectional Studies, Hematopoiesis, Hematopoietic Stem Cells, Mice, Bone Marrow, Stem Cell Niche
- Abstract
Decline in hematopoietic stem cell (HSC) function with age underlies limited health span of our blood and immune systems. In order to preserve health into older age, it is necessary to understand the nature and timing of initiating events that cause HSC aging. By performing a cross-sectional study in mice, we discover that hallmarks of aging in HSCs and hematopoiesis begin to accumulate by middle age and that the bone marrow (BM) microenvironment at middle age induces and is indispensable for hematopoietic aging. Using unbiased approaches, we find that decreased levels of the longevity-associated molecule IGF1 in the local middle-aged BM microenvironment are a factor causing HSC aging. Direct stimulation of middle-aged HSCs with IGF1 rescues molecular and functional hallmarks of aging, including restored mitochondrial activity. Thus, although decline in IGF1 supports longevity, our work indicates that this also compromises HSC function and limits hematopoietic health span., Competing Interests: Declaration of interests J.J.T. holds a sponsored research project with H3 Biomedicine., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
20. Innate immune pathways and inflammation in hematopoietic aging, clonal hematopoiesis, and MDS.
- Author
-
Trowbridge JJ and Starczynowski DT
- Subjects
- Animals, Clonal Hematopoiesis immunology, Humans, Signal Transduction immunology, Aging immunology, Hematopoiesis immunology, Hematopoietic Stem Cells immunology, Immunity, Innate immunology, Inflammation immunology, Myelodysplastic Syndromes immunology
- Abstract
With a growing aged population, there is an imminent need to develop new therapeutic strategies to ameliorate disorders of hematopoietic aging, including clonal hematopoiesis and myelodysplastic syndrome (MDS). Cell-intrinsic dysregulation of innate immune- and inflammatory-related pathways as well as systemic inflammation have been implicated in hematopoietic defects associated with aging, clonal hematopoiesis, and MDS. Here, we review and discuss the role of dysregulated innate immune and inflammatory signaling that contribute to the competitive advantage and clonal dominance of preleukemic and MDS-derived hematopoietic cells. We also propose how emerging concepts will further reveal critical biology and novel therapeutic opportunities., Competing Interests: Disclosures: J.J. Trowbridge reported grants from H3 Biomedicine and non-financial support from Navitor Pharmaceuticals, Inc outside the submitted work; in addition, J.J. Trowbridge had a patent to US-7850960-B2 Methods for regulation of stem cells with royalties paid by Fate Therapeutics. D.T. Starczynowski reported personal fees from Kurome Therapeutics, Kymera Therapeutics, and Captor Therapeutics, and grants from Tolera Therapeutics outside the submitted work. Additionally, D.T. Starczynowski serves on the scientific advisory board and has equity in Kurome Therapeutics., (© 2021 Trowbridge and Starczynowski.)
- Published
- 2021
- Full Text
- View/download PDF
21. Hand in hand: intrinsic and extrinsic drivers of aging and clonal hematopoiesis.
- Author
-
SanMiguel JM, Young K, and Trowbridge JJ
- Subjects
- Aging genetics, Animals, Antineoplastic Agents pharmacology, Bone Marrow growth & development, Cellular Senescence genetics, Cellular Senescence physiology, Chromatin genetics, Chromatin ultrastructure, Clone Cells, DNA (Cytosine-5-)-Methyltransferases genetics, DNA (Cytosine-5-)-Methyltransferases physiology, DNA Damage, DNA Methylation, DNA Methyltransferase 3A, Feedback, Physiological, Female, Forecasting, Hematopoietic Stem Cells classification, Humans, Inflammation genetics, Male, Mice, Mutation, Myeloid Cells cytology, Selection, Genetic, Stem Cell Niche, Aging physiology, Hematopoiesis physiology, Hematopoietic Stem Cells cytology
- Abstract
Over the past 25 years, the importance of hematopoietic stem cell (HSC) aging in overall hematopoietic and immune system health span has been appreciated. Much work has been done in model organisms to understand the intrinsic dysregulation that occurs in HSCs during aging, with the goal of identifying modifiable mechanisms that represent the proverbial "fountain of youth." Much more recently, the discovery of somatic mutations that are found to provide a selective advantage to HSCs and accumulate in the hematopoietic system during aging, termed clonal hematopoiesis (CH), inspires revisiting many of these previously defined drivers of HSC aging in the context of these somatic mutations. To truly understand these processes and develop a holistic picture of HSC aging, ongoing and future studies must include investigation of the critical changes that occur in the HSC niche or bone marrow microenvironment with aging, as increasing evidence supports that these HSC-extrinsic alterations provide necessary inflammation, signaling pathway activation or repression, and other selective pressures to favor HSC aging-associated phenotypes and CH. Here, we provide our perspectives based on the past 8 years of our own laboratory's investigations into these mechanisms and chart a path for integrative studies that, in our opinion, will provide an ideal opportunity to discover HSC and hematopoietic health span-extending interventions. This path includes examining when and how aging-associated HSC-intrinsic and HSC-extrinsic changes accumulate over time in different individuals and developing new models to track and test relevant HSC-extrinsic changes, complementary to innovative HSC lineage tracing systems that have recently been developed., (Copyright © 2020 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
22. Heritable genetic background alters survival and phenotype of Mll-AF9-induced leukemias.
- Author
-
Young K, Loberg MA, Eudy E, Schwartz LS, Mujica KD, and Trowbridge JJ
- Subjects
- Animals, Carcinogenesis metabolism, Carcinogenesis pathology, Cell Lineage genetics, Disease Progression, Female, Gene Knock-In Techniques, Genetic Predisposition to Disease, Humans, Leukemia, Biphenotypic, Acute metabolism, Leukemia, Biphenotypic, Acute mortality, Leukemia, Biphenotypic, Acute pathology, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute pathology, Lymphocyte Count, Lymphocytes metabolism, Lymphocytes pathology, Male, Mice, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, Inbred Strains, Mice, Transgenic, Myeloid Cells metabolism, Myeloid Cells pathology, Myeloproliferative Disorders metabolism, Myeloproliferative Disorders mortality, Myeloproliferative Disorders pathology, Oncogene Proteins, Fusion metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Survival Analysis, Carcinogenesis genetics, Gene Expression Regulation, Leukemic, Leukemia, Biphenotypic, Acute genetics, Leukemia, Myeloid, Acute genetics, Myeloproliferative Disorders genetics, Oncogene Proteins, Fusion genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
The MLL-AF9 fusion protein occurring as a result of t(9;11) translocation gives rise to pediatric and adult acute leukemias of distinct lineages, including acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), and mixed-phenotype acute leukemia (MPAL). The mechanisms underlying how this same fusion protein results in diverse leukemia phenotypes among different individuals are not well understood. Given emerging evidence from genome-wide association studies that genetic risk factors contribute to MLL-rearranged leukemogenesis, here we tested the impact of genetic background on survival and phenotype of a well-characterized Mll-AF9 knockin mouse model. We crossed this model with five distinct inbred strains (129, A/J, C57BL/6, NOD, CAST) and tested their F1 hybrid progeny for dominant genetic effects on Mll-AF9 phenotypes. We discovered that genetic background altered peripheral blood composition, with Mll-AF9 CAST F1 having a significantly increased B-lymphocyte frequency, while the remainder of the strains exhibited myeloid-biased hematopoiesis, similar to the parental line. Genetic background also had an impact on overall survival, with Mll-AF9 A/J F1 and Mll-AF9 129 F1 having significantly shorter survival and Mll-AF9 CAST F1 having longer survival, compared with the parental line. Furthermore, we observed a range of hematologic malignancies, with Mll-AF9 A/J F1, Mll-AF9 129 F1, and Mll-AF9 B6 F1 developing exclusively myeloid cell malignancies (myeloproliferative disorder [MPD] and AML), whereas a subset of Mll-AF9 NOD F1 developed MPAL and Mll-AF9 CAST F1 developed ALL. This study provides a novel in vivo experimental model in which to evaluate the underlying mechanisms by which MLL-AF9 results in diverse leukemia phenotypes and provides definitive experimental evidence that genetic risk factors contribute to survival and phenotype of MLL-rearranged leukemogenesis., (Copyright © 2020 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
23. Aging-associated decrease in the histone acetyltransferase KAT6B is linked to altered hematopoietic stem cell differentiation.
- Author
-
Khokhar ES, Borikar S, Eudy E, Stearns T, Young K, and Trowbridge JJ
- Subjects
- Aging genetics, Aging pathology, Animals, Epigenesis, Genetic, Erythroid Cells pathology, Gene Expression Profiling, Gene Knockout Techniques, Histone Acetyltransferases genetics, Male, Mice, Mice, Transgenic, Myeloid Progenitor Cells pathology, Transcriptome, Aging metabolism, Cell Differentiation, Erythroid Cells enzymology, Gene Expression Regulation, Enzymologic, Histone Acetyltransferases biosynthesis, Myeloid Progenitor Cells enzymology
- Abstract
Aged hematopoietic stem cells (HSCs) undergo biased lineage priming and differentiation toward production of myeloid cells. A comprehensive understanding of gene regulatory mechanisms causing HSC aging is needed to devise new strategies to sustainably improve immune function in aged individuals. Here, a focused short hairpin RNA screen of epigenetic factors reveals that the histone acetyltransferase Kat6b regulates myeloid cell production from hematopoietic progenitor cells. Within the stem and progenitor cell compartment, Kat6b is highly expressed in long-term (LT)-HSCs and is significantly decreased with aging at the transcript and protein levels. Knockdown of Kat6b in young LT-HSCs causes skewed production of myeloid cells at the expense of erythroid cells both in vitro and in vivo. Transcriptome analysis identifies enrichment of aging and macrophage-associated gene signatures alongside reduced expression of self-renewal and multilineage priming signatures. Together, our work identifies KAT6B as a novel epigenetic regulator of hematopoietic differentiation and a target to improve aged immune function., (Copyright © 2020 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
24. Sequentially inducible mouse models reveal that Npm1 mutation causes malignant transformation of Dnmt3a-mutant clonal hematopoiesis.
- Author
-
Loberg MA, Bell RK, Goodwin LO, Eudy E, Miles LA, SanMiguel JM, Young K, Bergstrom DE, Levine RL, Schneider RK, and Trowbridge JJ
- Subjects
- Animals, Biomarkers, Tumor genetics, Cell Transformation, Neoplastic genetics, DNA (Cytosine-5-)-Methyltransferases physiology, DNA Methyltransferase 3A, Disease Progression, Female, Hematopoiesis, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Myeloid Progenitor Cells pathology, Myeloid Progenitor Cells transplantation, Myeloproliferative Disorders genetics, Nuclear Proteins physiology, Nucleophosmin, Cell Transformation, Neoplastic pathology, Clonal Evolution, DNA (Cytosine-5-)-Methyltransferases genetics, Disease Models, Animal, Leukemia, Myeloid, Acute etiology, Mutation, Myeloproliferative Disorders pathology, Nuclear Proteins genetics
- Abstract
Clonal hematopoiesis (CH) is a common aging-associated condition with increased risk of hematologic malignancy. Knowledge of the mechanisms driving evolution from CH to overt malignancy has been hampered by a lack of in vivo models that orthogonally activate mutant alleles. Here, we develop independently regulatable mutations in DNA methyltransferase 3A (Dnmt3a) and nucleophosmin 1 (Npm1), observed in human CH and AML, respectively. We find Dnmt3a mutation expands hematopoietic stem and multipotent progenitor cells (HSC/MPPs), modeling CH. Induction of mutant Npm1 after development of Dnmt3a-mutant CH causes progression to myeloproliferative disorder (MPD), and more aggressive MPD is observed with longer latency between mutations. MPDs uniformly progress to acute myeloid leukemia (AML) following transplant, accompanied by a decrease in HSC/MPPs and an increase in myeloid-restricted progenitors, the latter of which propagate AML in tertiary recipient mice. At a molecular level, progression of CH to MPD is accompanied by selection for mutations activating Ras/Raf/MAPK signaling. Progression to AML is characterized by additional oncogenic signaling mutations (Ptpn11, Pik3r1, Flt3) and/or mutations in epigenetic regulators (Hdac1, Idh1, Arid1a). Together, our study demonstrates that Npm1 mutation drives evolution of Dnmt3a-mutant CH to AML and rate of disease progression is accelerated with longer latency of CH.
- Published
- 2019
- Full Text
- View/download PDF
25. Context-specific tumor suppression by PHF6.
- Author
-
Trowbridge JJ
- Subjects
- Carrier Proteins, Humans, Repressor Proteins, Hematopoietic Stem Cell Transplantation, Hematopoietic System, Leukemia
- Abstract
Competing Interests: Conflict-of-interest disclosure: J.J.T. holds a patent licensed by and receives royalties from Fate Therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
26. The Mediator of Hematopoietic Stem Cell Homeostasis.
- Author
-
Borikar S and Trowbridge JJ
- Subjects
- Humans, Hematopoietic Stem Cells, Homeostasis
- Abstract
Mechanisms establishing and maintaining promoter-enhancer interactions in hematopoietic stem cells (HSCs) to maintain stem cell identity are not fully understood. In this issue of Cell Stem Cell, Aranda-Orgilles et al. (2016) describe a role for a member of the Mediator complex in maintaining HSC-specific enhancers and hematopoietic homeostasis., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
27. Progressive alterations in multipotent hematopoietic progenitors underlie lymphoid cell loss in aging.
- Author
-
Young K, Borikar S, Bell R, Kuffler L, Philip V, and Trowbridge JJ
- Subjects
- Animals, Cell Cycle genetics, Cell Differentiation, Cell Lineage genetics, Cells, Cultured, Cellular Senescence genetics, Female, Hematopoietic Stem Cells metabolism, Lymphocytes metabolism, Mice, Inbred C57BL, Multipotent Stem Cells metabolism, Myeloid Cells cytology, Single-Cell Analysis, Transcriptome genetics, Aging physiology, Hematopoietic Stem Cells cytology, Lymphocytes cytology, Multipotent Stem Cells cytology
- Abstract
Declining immune function with age is associated with reduced lymphoid output of hematopoietic stem cells (HSCs). Currently, there is poor understanding of changes with age in the heterogeneous multipotent progenitor (MPP) cell compartment, which is long lived and responsible for dynamically regulating output of mature hematopoietic cells. In this study, we observe an early and progressive loss of lymphoid-primed MPP cells (LMPP/MPP4) with aging, concomitant with expansion of HSCs. Transcriptome and in vitro functional analyses at the single-cell level reveal a concurrent increase in cycling of aging LMPP/MPP4 with loss of lymphoid priming and differentiation potential. Impaired lymphoid differentiation potential of aged LMPP/MPP4 is not rescued by transplantation into a young bone marrow microenvironment, demonstrating cell-autonomous changes in the MPP compartment with aging. These results pinpoint an age and cellular compartment to focus further interrogation of the drivers of lymphoid cell loss with aging., (© 2016 Young et al.)
- Published
- 2016
- Full Text
- View/download PDF
28. Open chromatin profiling as a novel strategy for identifying cancer cell of origin.
- Author
-
Young K and Trowbridge JJ
- Abstract
Tumor cell of origin is an important prognostic measure but is challenging to assess. We recently demonstrated in acute myeloid leukemia (AML) that the chromatin landscape serves as a biomarker of transformed cell of origin. Thus, open chromatin loci offer important prognostic information as well as targets for development of novel therapies in cancer treatment.
- Published
- 2016
- Full Text
- View/download PDF
29. Loss-of-function mutations in the C9ORF72 mouse ortholog cause fatal autoimmune disease.
- Author
-
Burberry A, Suzuki N, Wang JY, Moccia R, Mordes DA, Stewart MH, Suzuki-Uematsu S, Ghosh S, Singh A, Merkle FT, Koszka K, Li QZ, Zon L, Rossi DJ, Trowbridge JJ, Notarangelo LD, and Eggan K
- Subjects
- Animals, Autoimmune Diseases metabolism, Autoimmunity genetics, Autoimmunity physiology, CRISPR-Cas Systems genetics, CRISPR-Cas Systems physiology, Cytokines metabolism, Leukemia genetics, Leukemia metabolism, Mice, Mutation genetics, Splenomegaly genetics, Splenomegaly immunology, Thrombocytopenia genetics, Thrombocytopenia immunology, Autoimmune Diseases etiology, Autoimmune Diseases genetics, C9orf72 Protein genetics
- Abstract
C9ORF72 mutations are found in a significant fraction of patients suffering from amyotrophic lateral sclerosis and frontotemporal dementia, yet the function of the C9ORF72 gene product remains poorly understood. We show that mice harboring loss-of-function mutations in the ortholog of C9ORF72 develop splenomegaly, neutrophilia, thrombocytopenia, increased expression of inflammatory cytokines, and severe autoimmunity, ultimately leading to a high mortality rate. Transplantation of mutant mouse bone marrow into wild-type recipients was sufficient to recapitulate the phenotypes observed in the mutant animals, including autoimmunity and premature mortality. Reciprocally, transplantation of wild-type mouse bone marrow into mutant mice improved their phenotype. We conclude that C9ORF72 serves an important function within the hematopoietic system to restrict inflammation and the development of autoimmunity., (Copyright © 2016, American Association for the Advancement of Science.)
- Published
- 2016
- Full Text
- View/download PDF
30. Leukaemia cell of origin identified by chromatin landscape of bulk tumour cells.
- Author
-
George J, Uyar A, Young K, Kuffler L, Waldron-Francis K, Marquez E, Ucar D, and Trowbridge JJ
- Subjects
- Animals, Cell Transformation, Neoplastic, Epigenesis, Genetic, Female, HEK293 Cells, Hematopoietic Stem Cells metabolism, Humans, Leukemia, Myeloid, Acute metabolism, Mice, Inbred C57BL, Multipotent Stem Cells metabolism, Myeloid-Lymphoid Leukemia Protein genetics, Oncogene Proteins, Fusion genetics, Transcriptome, Chromatin Assembly and Disassembly, Leukemia, Myeloid, Acute etiology
- Abstract
The precise identity of a tumour's cell of origin can influence disease prognosis and outcome. Methods to reliably define tumour cell of origin from primary, bulk tumour cell samples has been a challenge. Here we use a well-defined model of MLL-rearranged acute myeloid leukaemia (AML) to demonstrate that transforming haematopoietic stem cells (HSCs) and multipotent progenitors results in more aggressive AML than transforming committed progenitor cells. Transcriptome profiling reveals a gene expression signature broadly distinguishing stem cell-derived versus progenitor cell-derived AML, including genes involved in immune escape, extravasation and small GTPase signal transduction. However, whole-genome profiling of open chromatin reveals precise and robust biomarkers reflecting each cell of origin tested, from bulk AML tumour cell sampling. We find that bulk AML tumour cells exhibit distinct open chromatin loci that reflect the transformed cell of origin and suggest that open chromatin patterns may be leveraged as prognostic signatures in human AML.
- Published
- 2016
- Full Text
- View/download PDF
31. Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A.
- Author
-
Xu J, Bauer DE, Kerenyi MA, Vo TD, Hou S, Hsu YJ, Yao H, Trowbridge JJ, Mandel G, and Orkin SH
- Subjects
- Animals, Carrier Proteins metabolism, Cell Line, Tumor, Chromatin metabolism, Chromatin Immunoprecipitation, Chromatography, Liquid, Erythroid Precursor Cells, Humans, Mice, Nuclear Proteins metabolism, Proteomics, RNA Interference, Real-Time Polymerase Chain Reaction, Repressor Proteins, Tandem Mass Spectrometry, beta-Globins metabolism, Carrier Proteins pharmacology, Co-Repressor Proteins metabolism, Fetal Hemoglobin metabolism, Gene Expression Regulation, Developmental physiology, Multiprotein Complexes metabolism, Nuclear Proteins pharmacology
- Abstract
Reactivation of fetal hemoglobin (HbF) in adults ameliorates the severity of the common β-globin disorders. The transcription factor BCL11A is a critical modulator of hemoglobin switching and HbF silencing, yet the molecular mechanism through which BCL11A coordinates the developmental switch is incompletely understood. Particularly, the identities of BCL11A cooperating protein complexes and their roles in HbF expression and erythroid development remain largely unknown. Here we determine the interacting partner proteins of BCL11A in erythroid cells by a proteomic screen. BCL11A is found within multiprotein complexes consisting of erythroid transcription factors, transcriptional corepressors, and chromatin-modifying enzymes. We show that the lysine-specific demethylase 1 and repressor element-1 silencing transcription factor corepressor 1 (LSD1/CoREST) histone demethylase complex interacts with BCL11A and is required for full developmental silencing of mouse embryonic β-like globin genes and human γ-globin genes in adult erythroid cells in vivo. In addition, LSD1 is essential for normal erythroid development. Furthermore, the DNA methyltransferase 1 (DNMT1) is identified as a BCL11A-associated protein in the proteomic screen. DNMT1 is required to maintain HbF silencing in primary human adult erythroid cells. DNMT1 haploinsufficiency combined with BCL11A deficiency further enhances γ-globin expression in adult animals. Our findings provide important insights into the mechanistic roles of BCL11A in HbF silencing and clues for therapeutic targeting of BCL11A in β-hemoglobinopathies.
- Published
- 2013
- Full Text
- View/download PDF
32. TiF1-gamma plays an essential role in murine hematopoiesis and regulates transcriptional elongation of erythroid genes.
- Author
-
Bai X, Trowbridge JJ, Riley E, Lee JA, DiBiase A, Kaartinen VM, Orkin SH, and Zon LI
- Subjects
- Animals, B-Lymphocytes cytology, B-Lymphocytes metabolism, Bone Marrow Cells cytology, Bone Marrow Cells metabolism, Cell Differentiation genetics, Cell Lineage genetics, Erythroid Cells cytology, Gene Deletion, Granulocyte-Macrophage Progenitor Cells cytology, Granulocyte-Macrophage Progenitor Cells metabolism, Granulocytes cytology, Granulocytes metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Myelopoiesis genetics, Spleen metabolism, Transcription Factors deficiency, Erythroid Cells metabolism, Gene Expression Regulation, Developmental, Hematopoiesis genetics, Transcription Elongation, Genetic, Transcription Factors metabolism
- Abstract
Transcriptional regulators play critical roles in the regulation of cell fate during hematopoiesis. Previous studies in zebrafish have identified an essential role for the transcriptional intermediary factor TIF1γ in erythropoiesis by regulating the transcription elongation of erythroid genes. To study if TIF1γ plays a similar role in murine erythropoiesis and to assess its function in other blood lineages, we generated mouse models with hematopoietic deletion of TIF1γ. Our results showed a block in erythroid maturation in the bone marrow following tif1γ deletion that was compensated with enhanced spleen erythropoiesis. Further analyses revealed a defect in transcription elongation of erythroid genes in the bone marrow. In addition, loss of TIF1γ resulted in defects in other blood compartments, including a profound loss of B cells, a dramatic expansion of granulocytes and decreased HSC function. TIF1γ exerts its functions in a cell-autonomous manner as revealed by competitive transplantation experiments. Our study therefore demonstrates that TIF1γ plays essential roles in multiple murine blood lineages and that its function in transcription elongation is evolutionally conserved., (Published by Elsevier Inc.)
- Published
- 2013
- Full Text
- View/download PDF
33. Haploinsufficiency of Dnmt1 impairs leukemia stem cell function through derepression of bivalent chromatin domains.
- Author
-
Trowbridge JJ, Sinha AU, Zhu N, Li M, Armstrong SA, and Orkin SH
- Subjects
- Animals, DNA Methylation, Gene Knockout Techniques, Kaplan-Meier Estimate, Leukemia pathology, Mice, Neoplastic Stem Cells cytology, Tumor Suppressor Proteins genetics, Chromatin metabolism, DNA (Cytosine-5-)-Methyltransferases genetics, DNA (Cytosine-5-)-Methyltransferases metabolism, Gene Expression Regulation, Neoplastic, Haploinsufficiency, Leukemia enzymology, Neoplastic Stem Cells enzymology
- Abstract
Epigenetic mechanisms regulating leukemia stem cells (LSCs) are an attractive target for therapy of blood cancers. Here, we report that conditional knockout of the DNA methyltransferase Dnmt1 blocked development of leukemia, and haploinsufficiency of Dnmt1 was sufficient to delay progression of leukemogenesis and impair LSC self-renewal without altering normal hematopoiesis. Haploinsufficiency of Dnmt1 resulted in tumor suppressor gene derepression associated with reduced DNA methylation and bivalent chromatin marks. These results suggest that LSCs depend on not only active expression of leukemogenic programs, but also DNA methylation-mediated silencing of bivalent domains to enforce transcriptional repression.
- Published
- 2012
- Full Text
- View/download PDF
34. Dnmt3a silences hematopoietic stem cell self-renewal.
- Author
-
Trowbridge JJ and Orkin SH
- Subjects
- Cell Differentiation genetics, DNA Methylation, DNA Methyltransferase 3A, Cell Division genetics, DNA (Cytosine-5-)-Methyltransferases genetics, Hematopoietic Stem Cells physiology, RNA Interference
- Abstract
DNA methylation is an epigenetic mark stably directing gene expression throughout development. A new study uncovers a role for the DNA methyltransferase Dnmt3a in silencing self-renewal genes in hematopoietic stem cells (HSCs) to permit efficient hematopoietic differentiation.
- Published
- 2011
- Full Text
- View/download PDF
35. Context-dependent function of "GATA switch" sites in vivo.
- Author
-
Snow JW, Trowbridge JJ, Johnson KD, Fujiwara T, Emambokus NE, Grass JA, Orkin SH, and Bresnick EH
- Subjects
- Amino Acid Motifs, Animals, Erythropoiesis genetics, Erythropoiesis physiology, GATA2 Transcription Factor chemistry, Gene Expression, Gene Knock-In Techniques, Genes, Switch, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Mice, Mice, Transgenic, Models, Biological, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Transcription Initiation Site, GATA2 Transcription Factor genetics, GATA2 Transcription Factor metabolism, Hematopoiesis genetics, Hematopoiesis physiology
- Abstract
Master transcriptional regulators of development often function through dispersed cis elements at endogenous target genes. While cis-elements are routinely studied in transfection and transgenic reporter assays, it is challenging to ascertain how they function in vivo. To address this problem in the context of the locus encoding the critical hematopoietic transcription factor Gata2, we engineered mice lacking a cluster of GATA motifs 2.8 kb upstream of the Gata2 transcriptional start site. We demonstrate that the -2.8 kb site confers maximal Gata2 expression in hematopoietic stem cells and specific hematopoietic progenitors. By contrast to our previous demonstration that a palindromic GATA motif at the neighboring -1.8 kb site maintains Gata2 repression in terminally differentiating erythroid cells, the -2.8 kb site was not required to initiate or maintain repression. These analyses reveal qualitatively distinct functions of 2 GATA motif-containing regions in vivo.
- Published
- 2011
- Full Text
- View/download PDF
36. A single cis element maintains repression of the key developmental regulator Gata2.
- Author
-
Snow JW, Trowbridge JJ, Fujiwara T, Emambokus NE, Grass JA, Orkin SH, and Bresnick EH
- Subjects
- Animals, Base Pairing genetics, CpG Islands genetics, DNA Methylation genetics, Erythroid Cells metabolism, Erythropoiesis genetics, GATA2 Transcription Factor metabolism, Gene Deletion, Gene Targeting, Genetic Loci genetics, Histones metabolism, Mice, Mice, Mutant Strains, Nucleoproteins metabolism, Protein Binding, RNA Polymerase II metabolism, Stress, Physiological genetics, GATA2 Transcription Factor genetics, Gene Expression Regulation, Developmental, Promoter Regions, Genetic, Repressor Proteins metabolism
- Abstract
In development, lineage-restricted transcription factors simultaneously promote differentiation while repressing alternative fates. Molecular dissection of this process has been challenging as transcription factor loci are regulated by many trans-acting factors functioning through dispersed cis elements. It is not understood whether these elements function collectively to confer transcriptional regulation, or individually to control specific aspects of activation or repression, such as initiation versus maintenance. Here, we have analyzed cis element regulation of the critical hematopoietic factor Gata2, which is expressed in early precursors and repressed as GATA-1 levels rise during terminal differentiation. We engineered mice lacking a single cis element -1.8 kb upstream of the Gata2 transcriptional start site. Although Gata2 is normally repressed in late-stage erythroblasts, the -1.8 kb mutation unexpectedly resulted in reactivated Gata2 transcription, blocked differentiation, and an aberrant lineage-specific gene expression pattern. Our findings demonstrate that the -1.8 kb site selectively maintains repression, confers a specific histone modification pattern and expels RNA Polymerase II from the locus. These studies reveal how an individual cis element establishes a normal developmental program via regulating specific steps in the mechanism by which a critical transcription factor is repressed., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2010
- Full Text
- View/download PDF
37. Wnt3a activates dormant c-Kit(-) bone marrow-derived cells with short-term multilineage hematopoietic reconstitution capacity.
- Author
-
Trowbridge JJ, Guezguez B, Moon RT, and Bhatia M
- Subjects
- Animals, Cell Cycle genetics, Cell Cycle physiology, Cell Differentiation genetics, Cell Differentiation physiology, Cells, Cultured, Female, Hematopoietic Stem Cell Transplantation, Liver cytology, Liver embryology, Liver metabolism, Mice, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, SCID, Polymerase Chain Reaction, Pregnancy, Signal Transduction genetics, Signal Transduction physiology, Wnt Proteins genetics, Wnt3 Protein, Wnt3A Protein, Bone Marrow Cells cytology, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Proto-Oncogene Proteins c-kit genetics, Wnt Proteins metabolism
- Abstract
Quiescent cells lacking expression of mature lineage makers and the c-Kit receptor reside in adult bone marrow. Despite their phenotypic similarity to hematopoietic stem cells, these Lin(-)Sca-1(+)c-Kit(-) cells lack myeloid and erythroid potential and long-term hematopoietic repopulating capacity, whereas, recent studies have functionally demonstrated that the Lin(-)Sca-1(+)c-Kit(-) population contains early lymphoid-committed progenitors. Examining the role of Wnt signaling in regulation of this population, we found that c-Kit(-) cells express diverse Wnt receptors and proliferate upon Wnt pathway activation in vitro and in vivo. Stimulation with Wnt3a, but not Wnt5a or Wnt11, promoted c-Kit(-) cells to give rise to myeloid and erythroid progenitors with robust self-renewal capacity measured by clonal replating. In addition, Wnt3a-stimulated c-Kit(-) cells gave rise to all hematopoietic lineages (lymphoid, myeloid, and erythroid) upon transplant into the liver of newborn recipient mice. Our study reveals that Wnt3a activates unique cell fate decisions of dormant c-Kit(-) that promotes short-term multilineage reconstitution capacity in vivo, thereby revealing a unique role for Wnt activation in hematopoiesis. Overall, our results highlight the potential of utilizing signaling molecules known to have instructive roles in regeneration to discover cell subsets residing in adult organisms with unexploited regenerative capacity.
- Published
- 2010
- Full Text
- View/download PDF
38. DNA methylation in adult stem cells: new insights into self-renewal.
- Author
-
Trowbridge JJ and Orkin SH
- Subjects
- Adult Stem Cells cytology, Animals, DNA Modification Methylases metabolism, Humans, Mice, Adult Stem Cells metabolism, DNA Methylation genetics
- Abstract
Methylation of cytosine residues in the context of CpG dinucleotides within mammalian DNA is an epigenetic modification with profound effects on transcriptional regulation. A group of enzymes, the DNA methyltransferases (DNMTs) tightly regulate both the initiation and maintenance of these methyl marks. Loss of critical components of this enzymatic machinery results in growth, viability, and differentiation defects in both mice and humans, supporting the notion that this epigenetic modification is essential for proper development. Beyond this, DNA methylation also provides a potent epigenetic mechanism for cellular memory needed to silence repetitive elements and preserve lineage specificity over repeated cell divisions throughout adulthood. Recent work highlighting the specialized roles of DNA methylation and methyltransferases in maintaining adult somatic stem cell function suggests that further dissection of these mechanisms will shed new light on the complex nature of self-renewal.
- Published
- 2010
- Full Text
- View/download PDF
39. DNA methyltransferase 1 is essential for and uniquely regulates hematopoietic stem and progenitor cells.
- Author
-
Trowbridge JJ, Snow JW, Kim J, and Orkin SH
- Subjects
- Animals, Blotting, Western, Bone Marrow Transplantation, DNA (Cytosine-5-)-Methyltransferase 1, DNA (Cytosine-5-)-Methyltransferases genetics, Female, Flow Cytometry, Mice, Polymerase Chain Reaction, DNA (Cytosine-5-)-Methyltransferases physiology, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism
- Abstract
DNA methylation is essential for development and in diverse biological processes. The DNA methyltransferase Dnmt1 maintains parental cell methylation patterns on daughter DNA strands in mitotic cells; however, the precise role of Dnmt1 in regulation of quiescent adult stem cells is not known. To examine the role of Dnmt1 in adult hematopoietic stem cells (HSCs), we conditionally disrupted Dnmt1 in the hematopoietic system. Defects were observed in Dnmt1-deficient HSC self-renewal, niche retention, and in the ability of Dnmt1-deficient HSCs to give rise to multilineage hematopoiesis. Loss of Dnmt1 also had specific impact on myeloid progenitor cells, causing enhanced cell cycling and inappropriate expression of mature lineage genes. Dnmt1 regulates distinct patterns of methylation and expression of discrete gene families in long-term HSCs and multipotent and lineage-restricted progenitors, suggesting that Dnmt1 differentially controls these populations. These findings establish a unique and critical role for Dnmt1 in the primitive hematopoietic compartment.
- Published
- 2009
- Full Text
- View/download PDF
40. Hematopoietic stem cell biology: too much of a Wnt thing.
- Author
-
Trowbridge JJ, Moon RT, and Bhatia M
- Subjects
- Animals, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Signal Transduction, Wnt Proteins genetics, beta Catenin genetics, Hematopoietic Stem Cells physiology, Wnt Proteins metabolism, beta Catenin metabolism
- Published
- 2006
- Full Text
- View/download PDF
41. Hedgehog modulates cell cycle regulators in stem cells to control hematopoietic regeneration.
- Author
-
Trowbridge JJ, Scott MP, and Bhatia M
- Subjects
- Animals, Antigens, Ly genetics, Antigens, Ly metabolism, Bone Marrow Cells cytology, Bone Marrow Cells physiology, Cell Cycle Proteins genetics, Cell Differentiation physiology, Cell Lineage, Gene Expression Profiling, Hedgehog Proteins genetics, Hematopoietic Stem Cells cytology, Humans, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Mice, Inbred Strains, Patched Receptors, Proto-Oncogene Proteins c-kit genetics, Proto-Oncogene Proteins c-kit metabolism, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism, Regeneration, Spleen cytology, Transcription Factors genetics, Transcription Factors metabolism, Transcription, Genetic, Veratrum Alkaloids metabolism, Cell Cycle physiology, Cell Cycle Proteins metabolism, Hedgehog Proteins metabolism, Hematopoiesis, Hematopoietic Stem Cells physiology, Signal Transduction physiology
- Abstract
The signals that control the regenerative ability of hematopoietic stem cells (HSCs) in response to damage are unknown. Here, we demonstrate that downstream activation of the Hedgehog (Hh) signaling pathway induces cycling and expansion of primitive bone marrow hematopoietic cells under homeostatic conditions and during acute regeneration. However, this effect is at the expense of HSC function, because continued Hh activation during regeneration represses expression of specific cell cycle regulators, leading to HSC exhaustion. In vivo treatment with an inhibitor of the Hh pathway rescues these transcriptional and functional defects in HSCs. Our study establishes Hh signaling as a regulator of the HSC cell cycle machinery that balances hematopoietic homeostasis and regeneration in vivo.
- Published
- 2006
- Full Text
- View/download PDF
42. Glycogen synthase kinase-3 is an in vivo regulator of hematopoietic stem cell repopulation.
- Author
-
Trowbridge JJ, Xenocostas A, Moon RT, and Bhatia M
- Subjects
- Animals, Blood Glucose metabolism, Cell Proliferation, Cell Transplantation, Cells, Cultured, Culture Media, Serum-Free pharmacology, Enzyme Inhibitors pharmacology, Fetal Blood cytology, Flow Cytometry, Green Fluorescent Proteins metabolism, Hedgehog Proteins, Humans, Leukocytes, Mononuclear metabolism, Megakaryocytes cytology, Mice, Mice, Inbred C57BL, Mice, SCID, Mice, Transgenic, Models, Biological, Neutrophils cytology, Receptors, Notch metabolism, Reverse Transcriptase Polymerase Chain Reaction, Stem Cells, Time Factors, Trans-Activators metabolism, Wnt Proteins metabolism, Gene Expression Regulation, Enzymologic, Glycogen Synthase Kinase 3 physiology, Hematopoietic Stem Cells cytology, Stem Cell Transplantation methods
- Abstract
The in vivo regulation of hematopoietic stem cell (HSC) function is poorly understood. Here, we show that hematopoietic repopulation can be augmented by administration of a glycogen synthase kinase-3 (GSK-3) inhibitor to recipient mice transplanted with mouse or human HSCs. GSK-3 inhibitor treatment improved neutrophil and megakaryocyte recovery, recipient survival and resulted in enhanced sustained long-term repopulation. The output of primitive Lin(-)c-Kit(+)Sca-1(+) cells and progenitors from HSCs increased upon GSK-3 inhibitor treatment without altering secondary repopulating ability, suggesting that the HSC pool is maintained while overall hematopoietic reconstitution is increased. GSK-3 inhibitors were found to modulate gene targets of Wnt, Hedgehog and Notch pathways in cells comprising the primitive hematopoietic compartment without affecting mature cells. Our study establishes GSK-3 as a specific in vivo modulator of HSC activity, and suggests that administration of GSK-3 inhibitors may provide a clinical means to directly enhance the repopulating capacity of transplanted HSCs.
- 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.