Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Genome Med ; 16(1): 1, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38281962

ABSTRACT

BACKGROUND: Despite therapeutic advances, once a cancer has metastasized to the bone, it represents a highly morbid and lethal disease. One third of patients with advanced clear cell renal cell carcinoma (ccRCC) present with bone metastasis at the time of diagnosis. However, the bone metastatic niche in humans, including the immune and stromal microenvironments, has not been well-defined, hindering progress towards identification of therapeutic targets. METHODS: We collected fresh patient samples and performed single-cell transcriptomic profiling of solid metastatic tissue (Bone Met), liquid bone marrow at the vertebral level of spinal cord compression (Involved), and liquid bone marrow from a different vertebral body distant from the tumor site but within the surgical field (Distal), as well as bone marrow from patients undergoing hip replacement surgery (Benign). In addition, we incorporated single-cell data from primary ccRCC tumors (ccRCC Primary) for comparative analysis. RESULTS: The bone marrow of metastatic patients is immune-suppressive, featuring increased, exhausted CD8 + cytotoxic T cells, T regulatory cells, and tumor-associated macrophages (TAM) with distinct transcriptional states in metastatic lesions. Bone marrow stroma from tumor samples demonstrated a tumor-associated mesenchymal stromal cell population (TA-MSC) that appears to be supportive of epithelial-to mesenchymal transition (EMT), bone remodeling, and a cancer-associated fibroblast (CAFs) phenotype. This stromal subset is associated with poor progression-free and overall survival and also markedly upregulates bone remodeling through the dysregulation of RANK/RANKL/OPG signaling activity in bone cells, ultimately leading to bone resorption. CONCLUSIONS: These results provide a comprehensive analysis of the bone marrow niche in the setting of human metastatic cancer and highlight potential therapeutic targets for both cell populations and communication channels.


Subject(s)
Carcinoma, Renal Cell , Humans , Carcinoma, Renal Cell/genetics , Stromal Cells/pathology , Signal Transduction , Gene Expression Profiling , Single-Cell Analysis , Tumor Microenvironment
2.
Blood Adv ; 7(21): 6608-6623, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37450380

ABSTRACT

Myelodysplastic syndromes (MDSs) are a heterogenous group of diseases affecting the hematopoietic stem cell that are curable only by stem cell transplantation. Both hematopoietic cell intrinsic changes and extrinsic signals from the bone marrow (BM) niche seem to ultimately lead to MDS. Animal models of MDS indicate that alterations in specific mesenchymal progenitor subsets in the BM microenvironment can induce or select for abnormal hematopoietic cells. Here, we identify a subset of human BM mesenchymal cells marked by the expression of CD271, CD146, and CD106. This subset of human mesenchymal cells is comparable with mouse mesenchymal cells that, when perturbed, result in an MDS-like syndrome. Its transcriptional analysis identified Osteopontin (SPP1) as the most overexpressed gene. Selective depletion of Spp1 in the microenvironment of the mouse MDS model, Vav-driven Nup98-HoxD13, resulted in an accelerated progression as demonstrated by increased chimerism, higher mutant myeloid cell burden, and a more pronounced anemia when compared with that in wild-type microenvironment controls. These data indicate that molecular perturbations can occur in specific BM mesenchymal subsets of patients with MDS. However, the niche adaptations to dysplastic clones include Spp1 overexpression that can constrain disease fitness and potentially progression. Therefore, niche changes with malignant disease can also serve to protect the host.


Subject(s)
Bone Marrow , Myelodysplastic Syndromes , Humans , Mice , Animals , Bone Marrow/pathology , Myelodysplastic Syndromes/genetics , Hematopoietic Stem Cells/metabolism , Bone Marrow Cells/metabolism , Disease Models, Animal , Disease Progression
3.
bioRxiv ; 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37066307

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) within the bone marrow microenvironment (BMME) support normal hematopoietic stem and progenitor cells (HSPCs). However, the heterogeneity of human MSCs has limited the understanding of their contribution to clonal dynamics and evolution to myelodysplastic syndromes (MDS). We combined three MSC cell surface markers, CD271, VCAM-1 (Vascular Cell Adhesion Molecule-1) and CD146, to isolate distinct subsets of human MSCs from bone marrow aspirates of healthy controls (Control BM). Based on transcriptional and functional analysis, CD271+CD106+CD146+ (NGFR+/VCAM1+/MCAM+/Lin-; NVML) cells display stem cell characteristics, are compatible with murine BM-derived Leptin receptor positive MSCs and provide superior support for normal HSPCs. MSC subsets from 17 patients with MDS demonstrated shared transcriptional changes in spite of mutational heterogeneity in the MDS clones, with loss of preferential support of normal HSPCs by MDS-derived NVML cells. Our data provide a new approach to dissect microenvironment-dependent mechanisms regulating clonal dynamics and progression of MDS.

4.
Nat Commun ; 14(1): 663, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36750562

ABSTRACT

The treatment of low-risk primary prostate cancer entails active surveillance only, while high-risk disease requires multimodal treatment including surgery, radiation therapy, and hormonal therapy. Recurrence and development of metastatic disease remains a clinical problem, without a clear understanding of what drives immune escape and tumor progression. Here, we comprehensively describe the tumor microenvironment of localized prostate cancer in comparison with adjacent normal samples and healthy controls. Single-cell RNA sequencing and high-resolution spatial transcriptomic analyses reveal tumor context dependent changes in gene expression. Our data indicate that an immune suppressive tumor microenvironment associates with suppressive myeloid populations and exhausted T-cells, in addition to high stromal angiogenic activity. We infer cell-to-cell relationships from high throughput ligand-receptor interaction measurements within undissociated tissue sections. Our work thus provides a highly detailed and comprehensive resource of the prostate tumor microenvironment as well as tumor-stromal cell interactions.


Subject(s)
Prostatic Neoplasms , Transcriptome , Male , Humans , Prostate/pathology , Tumor Microenvironment , Gene Expression Profiling , Prostatic Neoplasms/genetics
5.
Nat Commun ; 13(1): 5747, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36180422

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is the most common type of kidney cancer in adults. When ccRCC is localized to the kidney, surgical resection or ablation of the tumor is often curative. However, in the metastatic setting, ccRCC remains a highly lethal disease. Here we use fresh patient samples that include treatment-naive primary tumor tissue, matched adjacent normal kidney tissue, as well as tumor samples collected from patients with bone metastases. Single-cell transcriptomic analysis of tumor cells from the primary tumors reveals a distinct transcriptional signature that is predictive of metastatic potential and patient survival. Analysis of supporting stromal cells within the tumor environment demonstrates vascular remodeling within the endothelial cells. An in silico cell-to-cell interaction analysis highlights the CXCL9/CXCL10-CXCR3 axis and the CD70-CD27 axis as potential therapeutic targets. Our findings provide biological insights into the interplay between tumor cells and the ccRCC microenvironment.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Adult , Carcinoma, Renal Cell/pathology , Endothelial Cells/metabolism , Humans , Kidney/metabolism , Kidney Neoplasms/pathology , Transcriptome , Tumor Microenvironment/genetics
6.
Blood Adv ; 6(17): 5072-5084, 2022 09 13.
Article in English | MEDLINE | ID: mdl-35793392

ABSTRACT

Genome-wide CRISPR screens have been extremely useful in identifying therapeutic targets in diverse cancers by defining genes that are essential for malignant growth. However, most CRISPR screens were performed in vitro and thus cannot identify genes that are essential for interactions with the microenvironment in vivo. Here, we report genome-wide CRISPR screens in 2 in vivo murine models of acute myeloid leukemia (AML) driven by the KMT2A/MLLT3 fusion or by the constitutive coexpression of Hoxa9 and Meis1. Secondary validation using a focused library identified 72 genes specifically essential for leukemic growth in vivo, including components of the major histocompatibility complex class I complex, Cd47, complement receptor Cr1l, and the ß-4-galactosylation pathway. Importantly, several of these in vivo-specific hits have a prognostic effect or are inferred to be master regulators of protein activity in human AML cases. For instance, we identified Fermt3, a master regulator of integrin signaling, as having in vivo-specific dependency with high prognostic relevance. Overall, we show an experimental and computational pipeline for genome-wide functional screens in vivo in AML and provide a genome-wide resource of essential drivers of leukemic growth in vivo.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Leukemia, Myeloid, Acute , Animals , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Signal Transduction , Tumor Microenvironment/genetics
7.
Nat Cardiovasc Res ; 1(1): 28-44, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35747128

ABSTRACT

Abnormal hematopoiesis advances cardiovascular disease by generating excess inflammatory leukocytes that attack the arteries and the heart. The bone marrow niche regulates hematopoietic stem cell proliferation and hence the systemic leukocyte pool, but whether cardiovascular disease affects the hematopoietic organ's microvasculature is unknown. Here we show that hypertension, atherosclerosis and myocardial infarction (MI) instigate endothelial dysfunction, leakage, vascular fibrosis and angiogenesis in the bone marrow, altogether leading to overproduction of inflammatory myeloid cells and systemic leukocytosis. Limiting angiogenesis with endothelial deletion of Vegfr2 (encoding vascular endothelial growth factor (VEGF) receptor 2) curbed emergency hematopoiesis after MI. We noted that bone marrow endothelial cells assumed inflammatory transcriptional phenotypes in all examined stages of cardiovascular disease. Endothelial deletion of Il6 or Vcan (encoding versican), genes shown to be highly expressed in mice with atherosclerosis or MI, reduced hematopoiesis and systemic myeloid cell numbers in these conditions. Our findings establish that cardiovascular disease remodels the vascular bone marrow niche, stimulating hematopoiesis and production of inflammatory leukocytes.

8.
Cancer Cell ; 39(11): 1464-1478.e8, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34719426

ABSTRACT

Bone metastases are devastating complications of cancer. They are particularly common in prostate cancer (PCa), represent incurable disease, and are refractory to immunotherapy. We seek to define distinct features of the bone marrow (BM) microenvironment by analyzing single cells from bone metastatic prostate tumors, involved BM, uninvolved BM, and BM from cancer-free, orthopedic patients, and healthy individuals. Metastatic PCa is associated with multifaceted immune distortion, specifically exhaustion of distinct T cell subsets, appearance of macrophages with states specific to PCa bone metastases. The chemokine CCL20 is notably overexpressed by myeloid cells, as is its cognate CCR6 receptor on T cells. Disruption of the CCL20-CCR6 axis in mice with syngeneic PCa bone metastases restores T cell reactivity and significantly prolongs animal survival. Comparative high-resolution analysis of PCa bone metastases shows a targeted approach for relieving local immunosuppression for therapeutic effect.


Subject(s)
Bone Neoplasms/pathology , Bone Neoplasms/secondary , Chemokine CCL20/genetics , Prostatic Neoplasms/pathology , Receptors, CCR6/genetics , Up-Regulation , Animals , Bone Neoplasms/genetics , Bone Neoplasms/immunology , Case-Control Studies , Cell Line, Tumor , Chemokine CCL20/metabolism , Gene Expression Regulation, Neoplastic , Humans , Macrophages/immunology , Male , Mice , Myeloid Cells/immunology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Receptors, CCR6/metabolism , Single-Cell Analysis , T-Lymphocytes/immunology , Tumor Microenvironment
9.
Cell Stem Cell ; 28(12): 2090-2103.e9, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34551362

ABSTRACT

Extracellular vesicles (EVs) transfer complex biologic material between cells. However, the role of this process in vivo is poorly defined. Here, we demonstrate that osteoblastic cells in the bone marrow (BM) niche elaborate extracellular vesicles that are taken up by hematopoietic progenitor cells in vivo. Genotoxic or infectious stress rapidly increased stromal-derived extracellular vesicle transfer to granulocyte-monocyte progenitors. The extracellular vesicles contained processed tRNAs (tiRNAs) known to modulate protein translation. 5'-ti-Pro-CGG-1 was preferentially abundant in osteoblast-derived extracellular vesicles and, when transferred to granulocyte-monocyte progenitors, increased protein translation, cell proliferation, and myeloid differentiation. Upregulating EV transfer improved hematopoietic recovery from genotoxic injury and survival from fungal sepsis. Therefore, EV-mediated tiRNA transfer provides a stress-modulated signaling axis in the BM niche distinct from conventional cytokine-driven stress responses.


Subject(s)
Extracellular Vesicles , Hematopoietic Stem Cells , Bone Marrow , Bone Marrow Cells , Hematopoiesis
10.
Cell Stem Cell ; 25(4): 570-583.e7, 2019 Oct 03.
Article in English | MEDLINE | ID: mdl-31279774

ABSTRACT

Stromal cell populations that maintain hematopoietic stem and progenitor cells (HSPCs) are generally characterized in steady-state conditions. Here, we report a comprehensive atlas of bone marrow stromal cell subpopulations under homeostatic and stress conditions using mass cytometry (CyTOF)-based single-cell protein analysis. We identified 28 subsets of non-hematopoietic cells during homeostasis, 14 of which expressed hematopoietic regulatory factors. Irradiation-based conditioning for HSPC transplantation led to the loss of most of these populations, including the LeptinR+ and Nestin+ subsets. In contrast, a subset expressing Ecto-5'-nucleotidase (CD73) was retained and a specific CD73+NGFRhigh population expresses high levels of cytokines during homeostasis and stress. Genetic ablation of CD73 compromised HSPC transplantation in an acute setting without long-term changes in bone marrow HSPCs. Thus, this protein-based expression mapping reveals distinct sets of stromal cells in the bone marrow and how they change in clinically relevant stress settings to contribute to early stages of hematopoietic regeneration.


Subject(s)
Bone Marrow Cells/metabolism , Stress, Physiological/physiology , Stromal Cells/metabolism , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Animals , Atlases as Topic , Bone Marrow Cells/pathology , Cells, Cultured , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Homeostasis , Humans , Mass Spectrometry , Mice , Mice, Knockout , Nestin/metabolism , Receptor, Nerve Growth Factor/metabolism , Receptors, Leptin/metabolism , Stem Cell Niche , Stromal Cells/pathology
11.
Cell ; 177(7): 1915-1932.e16, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31130381

ABSTRACT

Stroma is a poorly defined non-parenchymal component of virtually every organ with key roles in organ development, homeostasis, and repair. Studies of the bone marrow stroma have defined individual populations in the stem cell niche regulating hematopoietic regeneration and capable of initiating leukemia. Here, we use single-cell RNA sequencing (scRNA-seq) to define a cellular taxonomy of the mouse bone marrow stroma and its perturbation by malignancy. We identified seventeen stromal subsets expressing distinct hematopoietic regulatory genes spanning new fibroblastic and osteoblastic subpopulations including distinct osteoblast differentiation trajectories. Emerging acute myeloid leukemia impaired mesenchymal osteogenic differentiation and reduced regulatory molecules necessary for normal hematopoiesis. These data suggest that tissue stroma responds to malignant cells by disadvantaging normal parenchymal cells. Our taxonomy of the stromal compartment provides a comprehensive bone marrow cell census and experimental support for cancer cell crosstalk with specific stromal elements to impair normal tissue function and thereby enable emergent cancer.


Subject(s)
Bone Marrow Cells/metabolism , Cell Differentiation , Homeostasis , Leukemia, Myeloid, Acute/metabolism , Osteoblasts/metabolism , Osteogenesis , Tumor Microenvironment , Animals , Bone Marrow Cells/pathology , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Osteoblasts/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
12.
J Clin Invest ; 127(4): 1405-1413, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28319048

ABSTRACT

The mTOR pathway is a critical determinant of cell persistence and growth wherein mTOR complex 1 (mTORC1) mediates a balance between growth factor stimuli and nutrient availability. Amino acids or glucose facilitates mTORC1 activation by inducing RagA GTPase recruitment of mTORC1 to the lysosomal outer surface, enabling activation of mTOR by the Ras homolog Rheb. Thereby, RagA alters mTORC1-driven growth in times of nutrient abundance or scarcity. Here, we have evaluated differential nutrient-sensing dependence through RagA and mTORC1 in hematopoietic progenitors, which dynamically drive mature cell production, and hematopoietic stem cells (HSC), which provide a quiescent cellular reserve. In nutrient-abundant conditions, RagA-deficient HSC were functionally unimpaired and upregulated mTORC1 via nutrient-insensitive mechanisms. RagA was also dispensable for HSC function under nutritional stress conditions. Similarly, hyperactivation of RagA did not affect HSC function. In contrast, RagA deficiency markedly altered progenitor population function and mature cell output. Therefore, RagA is a molecular mechanism that distinguishes the functional attributes of reactive progenitors from a reserve stem cell pool. The indifference of HSC to nutrient sensing through RagA contributes to their molecular resilience to nutritional stress, a characteristic that is relevant to organismal viability in evolution and in modern HSC transplantation approaches.


Subject(s)
Hematopoietic Stem Cells/physiology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptation, Physiological , Amino Acids/physiology , Animals , Cell Differentiation , Cells, Cultured , Homeostasis , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , Mice, Transgenic , Monomeric GTP-Binding Proteins/metabolism , Stress, Physiological
13.
Exp Hematol ; 49: 68-72, 2017 05.
Article in English | MEDLINE | ID: mdl-28043821

ABSTRACT

Sharing reagents is of self-evident value in life science research, however, primary cell populations often do not cryopreserve well or can require extensive preparation by collaborators, making shipping difficult. Here we report an evaluation of different conditions for the storage shipment of mouse bone marrow (BM) cells that would best preserve the number, viability, and frequency of different hematopoietic lineages, as well as functionality of progenitor populations. Bones were either crushed to release BM cells or stored intact in one of three media: Phosphate buffered saline (PBS) supplemented with 2% fetal bovine serum (FBS), Plasmalyte, or RPMI at 4°C. Cell numbers, viability, phenotype, and functionality were assessed 16 hours and 40 hours later and compared to freshly prepared samples. Whereas BM cells stored in suspension for 16 hours and BM cells kept in bone for 40 hours suffered major losses in cell number, hematopoietic lineages that were kept in the bone for 16 hours had only minor differences compared to fresh cells. With no significant differences among the different media used, intact long bones stored in media, Plasmalyte, or PBS 2% FBS for up to 16 hours provided a reasonable means of preserving bone marrow cell populations.


Subject(s)
Bone Marrow , Culture Media , Hematopoietic Stem Cells , Specimen Handling/methods , Tissue Preservation/methods , Animals , Cattle , Cell Survival , Mice
14.
Cell ; 167(1): 171-186.e15, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27641501

ABSTRACT

While acute myeloid leukemia (AML) comprises many disparate genetic subtypes, one shared hallmark is the arrest of leukemic myeloblasts at an immature and self-renewing stage of development. Therapies that overcome differentiation arrest represent a powerful treatment strategy. We leveraged the observation that the majority of AML, despite their genetically heterogeneity, share in the expression of HoxA9, a gene normally downregulated during myeloid differentiation. Using a conditional HoxA9 model system, we performed a high-throughput phenotypic screen and defined compounds that overcame differentiation blockade. Target identification led to the unanticipated discovery that inhibition of the enzyme dihydroorotate dehydrogenase (DHODH) enables myeloid differentiation in human and mouse AML models. In vivo, DHODH inhibitors reduced leukemic cell burden, decreased levels of leukemia-initiating cells, and improved survival. These data demonstrate the role of DHODH as a metabolic regulator of differentiation and point to its inhibition as a strategy for overcoming differentiation blockade in AML.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Molecular Targeted Therapy , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Cell Differentiation , Dihydroorotate Dehydrogenase , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , High-Throughput Screening Assays , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Myeloid Cells/pathology , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pyrimidines/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/isolation & purification , Small Molecule Libraries/therapeutic use , Xenograft Model Antitumor Assays
15.
Cell Stem Cell ; 19(4): 530-543, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27524439

ABSTRACT

Physiological stem cell function is regulated by secreted factors produced by niche cells. In this study, we describe an unbiased approach based on the differential single-cell gene expression analysis of mesenchymal osteolineage cells close to, and further removed from, hematopoietic stem/progenitor cells (HSPCs) to identify candidate niche factors. Mesenchymal cells displayed distinct molecular profiles based on their relative location. We functionally examined, among the genes that were preferentially expressed in proximal cells, three secreted or cell-surface molecules not previously connected to HSPC biology-the secreted RNase angiogenin, the cytokine IL18, and the adhesion molecule Embigin-and discovered that all of these factors are HSPC quiescence regulators. Therefore, our proximity-based differential single-cell approach reveals molecular heterogeneity within niche cells and can be used to identify novel extrinsic stem/progenitor cell regulators. Similar approaches could also be applied to other stem cell/niche pairs to advance the understanding of microenvironmental regulation of stem cell function.


Subject(s)
Hematopoietic Stem Cells/cytology , Single-Cell Analysis/methods , Stem Cell Niche , Animals , Bone Marrow Cells/cytology , Bone and Bones/cytology , Cell Lineage/genetics , Cell Self Renewal/genetics , Cell Separation , Gene Deletion , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Interleukin-18/metabolism , Membrane Glycoproteins/metabolism , Ribonuclease, Pancreatic/metabolism , Time Factors , Transcription, Genetic , Vascular Cell Adhesion Molecule-1/metabolism
16.
Nat Biotechnol ; 34(7): 738-45, 2016 07.
Article in English | MEDLINE | ID: mdl-27272386

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) offers curative therapy for patients with hemoglobinopathies, congenital immunodeficiencies, and other conditions, possibly including AIDS. Autologous HSCT using genetically corrected cells would avoid the risk of graft-versus-host disease (GVHD), but the genotoxicity of conditioning remains a substantial barrier to the development of this approach. Here we report an internalizing immunotoxin targeting the hematopoietic-cell-restricted CD45 receptor that effectively conditions immunocompetent mice. A single dose of the immunotoxin, CD45-saporin (SAP), enabled efficient (>90%) engraftment of donor cells and full correction of a sickle-cell anemia model. In contrast to irradiation, CD45-SAP completely avoided neutropenia and anemia, spared bone marrow and thymic niches, enabling rapid recovery of T and B cells, preserved anti-fungal immunity, and had minimal overall toxicity. This non-genotoxic conditioning method may provide an attractive alternative to current conditioning regimens for HSCT in the treatment of non-malignant blood diseases.


Subject(s)
DNA Damage/immunology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/immunology , Leukocyte Common Antigens/immunology , Ribosome Inactivating Proteins, Type 1/genetics , Ribosome Inactivating Proteins, Type 1/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , DNA Damage/genetics , Female , Genetic Enhancement/methods , Immunogenetic Phenomena/genetics , Immunotoxins , Mice , Mice, Inbred C57BL , Saporins
17.
Annu Rev Pathol ; 11: 555-81, 2016 05 23.
Article in English | MEDLINE | ID: mdl-27193455

ABSTRACT

Regulation of stem cells in adult tissues is a key determinant of how well an organism can respond to the stresses of physiological challenge and disease. This is particularly true of the hematopoietic system, where demands on host defenses can call for an acute increase in cell production. Hematopoietic stem cells receive the regulatory signals for cell production in adult mammals in the bone marrow, a tissue with higher-order architectural and functional organization than previously appreciated. Here, we review the data defining particular structural components and heterologous cells in the bone marrow that participate in hematopoietic stem cell function. Further, we explore the case for stromal-hematopoietic cell interactions contributing to neoplastic myeloid disease. As the hematopoietic regulatory networks in the bone marrow are revealed, it is anticipated that strategies will emerge for how to enhance or inhibit production of specific blood cells. In that way, the control of hematopoiesis will enter the domain of therapies to modulate broad aspects of hematopoiesis, both normal and malignant.


Subject(s)
Adult Stem Cells/cytology , Bone Marrow Cells/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Stem Cell Niche , Animals , Humans
18.
Cell Stem Cell ; 16(3): 239-53, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25748931

ABSTRACT

Mesenchymal stromal cells (MSCs) are heterogeneous and primitive cells discovered first in the bone marrow (BM). They have putative roles in maintaining tissue homeostasis and are increasingly recognized as components of stem cell niches, which are best defined in the blood. The absence of in vivo MSC markers has limited our ability to track their behavior in vivo and draw comparisons with in vitro observations. Here we review the historical background of BM-MSCs, advances made in their prospective isolation, their developmental origin and contribution to maintaining subsets of hematopoietic cells, and how mesenchymal cells contribute to other stem cell niches.


Subject(s)
Antigens, Differentiation/metabolism , Bone Marrow Cells/metabolism , Mesenchymal Stem Cells/metabolism , Stem Cell Niche/physiology , Animals , Bone Marrow Cells/cytology , Humans , Mesenchymal Stem Cells/cytology
19.
Blood ; 125(3): 474-82, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25395419

ABSTRACT

The human T-cell lymphotropic virus type I (HTLV-1) Tax transactivator initiates transformation in adult T-cell leukemia/lymphoma (ATL), a highly aggressive chemotherapy-resistant malignancy. The arsenic/interferon combination, which triggers degradation of the Tax oncoprotein, selectively induces apoptosis of ATL cell lines and has significant clinical activity in Tax-driven murine ATL or human patients. However, the role of Tax loss in ATL response is disputed, and the molecular mechanisms driving degradation remain elusive. Here we demonstrate that ATL-derived or HTLV-1-transformed cells are dependent on continuous Tax expression, suggesting that Tax degradation underlies clinical responses to the arsenic/interferon combination. The latter enforces promyelocytic leukemia protein (PML) nuclear body (NB) formation and partner protein recruitment. In arsenic/interferon-treated HTLV-1 transformed or ATL cells, Tax is recruited onto NBs and undergoes PML-dependent hyper-sumoylation by small ubiquitin-like modifier (SUMO)2/3 but not SUMO1, ubiquitination by RNF4, and proteasome-dependent degradation. Thus, the arsenic/interferon combination clears ATL through degradation of its Tax driver, and this regimen could have broader therapeutic value by promoting degradation of other pathogenic sumoylated proteins.


Subject(s)
Arsenicals/pharmacology , Gene Products, tax/metabolism , Interferons/pharmacology , Leukemia-Lymphoma, Adult T-Cell/virology , Nuclear Proteins/metabolism , Proteolysis/drug effects , SUMO-1 Protein/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Antiviral Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Cell Transformation, Viral/drug effects , Drug Therapy, Combination , Flow Cytometry , Fluorescent Antibody Technique , HeLa Cells , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/pathogenicity , Humans , Immunoprecipitation , Leukemia-Lymphoma, Adult T-Cell/genetics , Promyelocytic Leukemia Protein , Sulfhydryl Reagents/pharmacology , Sumoylation/drug effects , Ubiquitination/drug effects
20.
Stem Cell Reports ; 3(5): 832-40, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25418727

ABSTRACT

Central to cellular proliferative, survival, and metabolic responses is the serine/threonine kinase mTOR, which is activated in many human cancers. mTOR is present in distinct complexes that are either modulated by AKT (mTORC1) or are upstream and regulatory of it (mTORC2). Governance of mTORC2 activity is poorly understood. Here, we report a transmembrane molecule in hematopoietic progenitor cells that physically interacts with and inhibits RICTOR, an essential component of mTORC2. Upstream of mTORC2 (UT2) negatively regulates mTORC2 enzymatic activity, reducing AKT(S473), PKCα, and NDRG1 phosphorylation and increasing FOXO transcriptional activity in an mTORC2-dependent manner. Modulating UT2 levels altered animal survival in a T cell acute lymphoid leukemia (T-ALL) model that is known to be mTORC2 sensitive. These studies identify an inhibitory component upstream of mTORC2 in hematopoietic cells that can reduce mortality from NOTCH-induced T-ALL. A transmembrane inhibitor of mTORC2 may provide an attractive target to affect this critical cell regulatory pathway.


Subject(s)
Carrier Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Membrane Transport Proteins/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HEK293 Cells , Hematopoietic Stem Cells/cytology , Humans , Immunoblotting , Intracellular Signaling Peptides and Proteins/metabolism , Mechanistic Target of Rapamycin Complex 2 , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Multiprotein Complexes/genetics , NIH 3T3 Cells , Phosphorylation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Binding , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Rapamycin-Insensitive Companion of mTOR Protein , Receptors, Notch/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...