Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Nat Immunol ; 25(5): 820-833, 2024 May.
Article in English | MEDLINE | ID: mdl-38600356

ABSTRACT

Human bone marrow permanently harbors high numbers of neutrophils, and a tumor-supportive bias of these cells could significantly impact bone marrow-confined malignancies. In individuals with multiple myeloma, the bone marrow is characterized by inflammatory stromal cells with the potential to influence neutrophils. We investigated myeloma-associated alterations in human marrow neutrophils and the impact of stromal inflammation on neutrophil function. Mature neutrophils in myeloma marrow are activated and tumor supportive and transcribe increased levels of IL1B and myeloma cell survival factor TNFSF13B (BAFF). Interactions with inflammatory stromal cells induce neutrophil activation, including BAFF secretion, in a STAT3-dependent manner, and once activated, neutrophils gain the ability to reciprocally induce stromal activation. After first-line myeloid-depleting antimyeloma treatment, human bone marrow retains residual stromal inflammation, and newly formed neutrophils are reactivated. Combined, we identify a neutrophil-stromal cell feed-forward loop driving tumor-supportive inflammation that persists after treatment and warrants novel strategies to target both stromal and immune microenvironments in multiple myeloma.


Subject(s)
B-Cell Activating Factor , Interleukin-1beta , Multiple Myeloma , Neutrophils , Stromal Cells , Tumor Microenvironment , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Humans , Tumor Microenvironment/immunology , Neutrophils/immunology , Neutrophils/metabolism , Stromal Cells/metabolism , Stromal Cells/immunology , B-Cell Activating Factor/metabolism , Interleukin-1beta/metabolism , Neutrophil Activation , STAT3 Transcription Factor/metabolism , Bone Marrow/immunology , Bone Marrow/pathology
2.
Nat Rev Immunol ; 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38491073

ABSTRACT

Tissue inflammation is a hallmark of tumour microenvironments. In the bone marrow, tumour-associated inflammation impacts normal niches for haematopoietic progenitor cells and mature immune cells and supports the outgrowth and survival of malignant cells residing in these niche compartments. This Review provides an overview of our current understanding of inflammatory changes in the bone marrow microenvironment of myeloid and lymphoid malignancies, using acute myeloid leukaemia and multiple myeloma as examples and highlights unique and shared features of inflammation in niches for progenitor cells and plasma cells. Importantly, inflammation exerts profoundly different effects on normal bone marrow niches in these malignancies, and we provide context for possible drivers of these divergent effects. We explore the role of tumour cells in inflammatory changes, as well as the role of cellular constituents of normal bone marrow niches, including myeloid cells and stromal cells. Integrating knowledge of disease-specific dynamics of malignancy-associated bone marrow inflammation will provide a necessary framework for future targeting of these processes to improve patient outcome.

3.
Blood Cancer Discov ; 4(5): 394-417, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37470778

ABSTRACT

Cancer initiation is orchestrated by an interplay between tumor-initiating cells and their stromal/immune environment. Here, by adapted single-cell RNA sequencing, we decipher the predicted signaling between tissue-resident hematopoietic stem/progenitor cells (HSPC) and their neoplastic counterparts with their native niches in the human bone marrow. LEPR+ stromal cells are identified as central regulators of hematopoiesis through predicted interactions with all cells in the marrow. Inflammatory niche remodeling and the resulting deprivation of critical HSPC regulatory factors are predicted to repress high-output hematopoietic stem cell subsets in NPM1-mutated acute myeloid leukemia (AML), with relative resistance of clonal cells. Stromal gene signatures reflective of niche remodeling are associated with reduced relapse rates and favorable outcomes after chemotherapy across all genetic risk categories. Elucidation of the intercellular signaling defining human AML, thus, predicts that inflammatory remodeling of stem cell niches drives tissue repression and clonal selection but may pose a vulnerability for relapse-initiating cells in the context of chemotherapeutic treatment. SIGNIFICANCE: Tumor-promoting inflammation is considered an enabling characteristic of tumorigenesis, but mechanisms remain incompletely understood. By deciphering the predicted signaling between tissue-resident stem cells and their neoplastic counterparts with their environment, we identify inflammatory remodeling of stromal niches as a determinant of normal tissue repression and clinical outcomes in human AML. See related commentary by Lisi-Vega and Méndez-Ferrer, p. 349. This article is featured in Selected Articles from This Issue, p. 337.


Subject(s)
Hematopoietic Stem Cells , Leukemia, Myeloid, Acute , Humans , Bone Marrow , Leukemia, Myeloid, Acute/genetics , Hematopoiesis/genetics , Stromal Cells
4.
Nat Immunol ; 22(6): 769-780, 2021 06.
Article in English | MEDLINE | ID: mdl-34017122

ABSTRACT

Progression and persistence of malignancies are influenced by the local tumor microenvironment, and future eradication of currently incurable tumors will, in part, hinge on our understanding of malignant cell biology in the context of their nourishing surroundings. Here, we generated paired single-cell transcriptomic datasets of tumor cells and the bone marrow immune and stromal microenvironment in multiple myeloma. These analyses identified myeloma-specific inflammatory mesenchymal stromal cells, which spatially colocalized with tumor cells and immune cells and transcribed genes involved in tumor survival and immune modulation. Inflammatory stromal cell signatures were driven by stimulation with proinflammatory cytokines, and analyses of immune cell subsets suggested interferon-responsive effector T cell and CD8+ stem cell memory T cell populations as potential sources of stromal cell-activating cytokines. Tracking stromal inflammation in individuals over time revealed that successful antitumor induction therapy is unable to revert bone marrow inflammation, predicting a role for mesenchymal stromal cells in disease persistence.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mesenchymal Stem Cells/immunology , Multiple Myeloma/immunology , Neoplasm Recurrence, Local/immunology , Tumor Microenvironment/immunology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Marrow/drug effects , Bone Marrow/immunology , Bone Marrow/pathology , Cell Line, Tumor , Disease Progression , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Male , Mesenchymal Stem Cells/pathology , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Primary Cell Culture , Prospective Studies , RNA-Seq , Single-Cell Analysis , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...