Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Am J Transplant ; 23(2): 291-293, 2023 02.
Article in English | MEDLINE | ID: mdl-36804136

ABSTRACT

AL amyloidosis is a rare condition characterized by the overproduction of an unstable free light chain, protein misfolding and aggregation, and extracellular deposition that can progress to multiorgan involvement and failure. To our knowledge, this is the first worldwide report to describe triple organ transplantation for AL amyloidosis and triple organ transplantation using thoracoabdominal normothermic regional perfusion recovery with a donation from a circulatory death (DCD) donor. The recipient was a 40-year-old man with multiorgan AL amyloidosis with a terminal prognosis without multiorgan transplantation. An appropriate DCD donor was selected for sequential heart, liver, and kidney transplants via our center's thoracoabdominal normothermic regional perfusion pathway. The liver was additionally placed on an ex vivo normothermic machine perfusion, and the kidney was maintained on hypothermic machine perfusion while awaiting implantation. The heart transplant was completed first (cold ischemic time [CIT]: 131 minutes), followed by the liver transplant (CIT: 87 minutes, normothermic machine perfusion: 301 minutes). Kidney transplantation was performed the following day (CIT: 1833 minutes). He is 8 months posttransplant without evidence of heart, liver, or kidney graft dysfunction or rejection. This case highlights the feasibility of normothermic recovery and storage modalities for DCD donors, which can expand transplant opportunities for allografts previously not considered for multiorgan transplantations.


Subject(s)
Immunoglobulin Light-chain Amyloidosis , Kidney Transplantation , Tissue and Organ Procurement , Male , Humans , Adult , Organ Preservation , Tissue Donors , Perfusion , Liver , Death
2.
Biochem Soc Trans ; 51(6): 2017-2028, 2023 12 20.
Article in English | MEDLINE | ID: mdl-38031753

ABSTRACT

Advances in immunotherapy in the last decade have revolutionized treatment paradigms across multiple cancer diagnoses. However, only a minority of patients derive durable benefit and progress with traditional approaches, such as cancer vaccines, remains unsatisfactory. A key to overcoming these barriers resides with a deeper understanding of tumor antigen presentation and the complex and dynamic heterogeneity of tumor-infiltrating antigen-presenting cells (APCs). Reminiscent of the 'second touch' hypothesis proposed by Klaus Ley for CD4+ T cell differentiation, the acquisition of full effector potential by lymph node- primed CD8+ T cells requires a second round of co-stimulation at the site where the antigen originated, i.e. the tumor bed. The tumor stroma holds a prime role in this process by hosting specialized APC niches, apparently distinct from tertiary lymphoid structures, that support second antigenic touch encounters and CD8+ T cell effector proliferation and differentiation. We propose that APC within second-touch niches become licensed for co-stimulation through stromal-derived instructive signals emulating embryonic or wound-healing provisional matrix remodeling. These immunostimulatory roles of stroma contrast with its widely accepted view as a physical and functional 'immune barrier'. Stromal control of antigen presentation makes evolutionary sense as the host stroma-tumor interface constitutes the prime line of homeostatic 'defense' against the emerging tumor. In this review, we outline how stroma-derived signals and cells regulate tumor antigen presentation and T-cell effector differentiation in the tumor bed. The re-definition of tumor stroma as immune rheostat rather than as inflexible immune barrier harbors significant untapped therapeutic opportunity.


Subject(s)
Antigen Presentation , Neoplasms , Humans , Antigen-Presenting Cells , CD4-Positive T-Lymphocytes , Lymphocyte Activation , Antigens, Neoplasm , CD8-Positive T-Lymphocytes , Dendritic Cells
3.
Blood ; 137(1): 61-74, 2021 01 07.
Article in English | MEDLINE | ID: mdl-32640012

ABSTRACT

NRAS Q61 mutations are prevalent in advanced/relapsed multiple myeloma (MM) and correlate with poor patient outcomes. Thus, we generated a novel MM model by conditionally activating expression of endogenous NrasQ61R and an MYC transgene in germinal center (GC) B cells (VQ mice). VQ mice developed a highly malignant MM characterized by a high proliferation index, hyperactivation of extracellular signal-regulated kinase and AKT signaling, impaired hematopoiesis, widespread extramedullary disease, bone lesions, kidney abnormalities, preserved programmed cell death protein 1 and T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibition motif domain immune-checkpoint pathways, and expression of human high-risk MM gene signatures. VQ MM mice recapitulate most of the biological and clinical features of human advanced/high-risk MM. These MM phenotypes are serially transplantable in syngeneic recipients. Two MM cell lines were also derived to facilitate future genetic manipulations. Combination therapies based on MEK inhibition significantly prolonged the survival of VQ mice with advanced-stage MM. Our study provides a strong rationale to develop MEK inhibition-based therapies for treating advanced/relapsed MM.


Subject(s)
B-Lymphocytes/pathology , Disease Models, Animal , Monomeric GTP-Binding Proteins/genetics , Multiple Myeloma/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , Germinal Center/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Myeloma/pathology , Transgenes
4.
Am J Physiol Cell Physiol ; 323(3): C678-C693, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35876288

ABSTRACT

Cancer immunoediting progresses through elimination, equilibrium, and escape. Each of these phases is characterized by breaching, remodeling, and rebuilding tissue planes and structural barriers that engage extracellular matrix (ECM) components, in particular matrix proteoglycans. Some of the signals emanating from matrix proteoglycan remodeling are readily co-opted by the growing tumor to sustain an environment of tumor-promoting and immune-suppressive inflammation. Yet other matrix-derived cues can be viewed as part of a homeostatic response by the host, aiming to eliminate the tumor and restore tissue integrity. These latter signals may be harnessed for therapeutic purposes to tip the polarity of the tumor immune milieu toward anticancer immunity. In this review, we attempt to showcase the importance and complexity of matrix proteoglycan signaling in both cancer-restraining and cancer-promoting inflammation. We propose that the era of matrix diagnostics and therapeutics for cancer is fast approaching the clinic.


Subject(s)
Neoplasms , Proteoglycans , Extracellular Matrix/pathology , Humans , Inflammation , Neoplasms/pathology , Signal Transduction
5.
J Transl Med ; 20(1): 527, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36371231

ABSTRACT

BACKGROUND: The role of the inflammatory milieu in prostate cancer progression is not well understood. Differences in inflammatory signaling between localized and metastatic disease may point to opportunities for early intervention. METHODS: We modeled PCa disease progression by analyzing RNA-seq of localized vs. metastatic patient samples, followed by CIBERSORTx to assess their immune cell populations. The VHA CDW registry of PCa patients was analyzed for anti-TNF clinical outcomes. RESULTS: We observed statistically significant opposing patterns of IL-6 and TNFα expression between localized and metastatic disease. IL-6 was robustly expressed in localized disease and downregulated in metastatic disease. The reverse was observed with TNFα expression. Metastatic disease was also characterized by downregulation of adhesion molecule E-selectin, matrix metalloproteinase ADAMTS-4 and a shift to M2 macrophages whereas localized disease demonstrated a preponderance of M1 macrophages. Treatment with anti-TNF agents was associated with earlier stage disease at diagnosis. CONCLUSIONS: Our data points to clearly different inflammatory contexts between localized and metastatic prostate cancer. Primary localized disease demonstrates local inflammation and adaptive immunity, whereas metastases are characterized by immune cold microenvironments and a shift towards resolution of inflammation and tissue repair. Therapies that interfere with these inflammatory networks may offer opportunities for early intervention in monotherapy or in combination with immunotherapies and anti-angiogenic approaches.


Subject(s)
Immune Evasion , Prostatic Neoplasms , Male , Humans , Interleukin-6 , Tumor Necrosis Factor-alpha , Tumor Necrosis Factor Inhibitors , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Inflammation , Tumor Microenvironment
6.
Adv Exp Med Biol ; 1272: 55-72, 2020.
Article in English | MEDLINE | ID: mdl-32845502

ABSTRACT

Versican is an extracellular matrix proteoglycan with nonredundant roles in diverse biological and cellular processes, ranging from embryonic development to adult inflammation and cancer. Versican is essential for cardiovascular morphogenesis, neural crest migration, and skeletal development during embryogenesis. In the adult, versican acts as an inflammation "amplifier" and regulator of immune cell activation and cytokine production. Increased versican expression has been observed in a wide range of malignant tumors and has been associated with poor patient outcomes. The main sources of versican production in the tumor microenvironment include accessory cells (myeloid cells and stromal components) and, in some contexts, the tumor cells themselves. Versican has been implicated in several classical hallmarks of cancer such as proliferative signaling, evasion of growth suppressor signaling, resistance to cell death, angiogenesis, and tissue invasion and metastasis. More recently, versican has been implicated in escape from tumor immune surveillance, e.g., through dendritic cell dysfunction. Versican's multiple contributions to benign and malignant biological processes are further diversified through the generation of versican-derived bioactive proteolytic fragments (matrikines), with versikine being the most studied to date. Versican and versican-derived matrikines hold promise as targets in the management of inflammatory and malignant conditions as well as in the development of novel predictive and prognostic biomarkers.


Subject(s)
Neoplasms , Tumor Microenvironment , Versicans , Extracellular Matrix , Humans , Neoplasms/metabolism , Neoplasms/pathology , Versicans/metabolism
7.
J Biol Chem ; 293(7): 2452-2465, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29279332

ABSTRACT

Nuclear factor-κB (NF-κB) is a family of transcription factors that play a key role in cell survival and proliferation in many hematological malignancies, including multiple myeloma (MM). Bortezomib, a proteasome inhibitor used in the management of MM, can inhibit both canonical and noncanonical activation of NF-κB in MM cells. However, we previously reported that a significant fraction of freshly isolated MM cells harbor bortezomib-resistant NF-κB activity. Here, we report that hyaluronan and proteoglycan link protein 1 (HAPLN1) is produced in bone marrow stromal cells from MM patients, is detected in patients' bone marrow plasma, and can activate an atypical bortezomib-resistant NF-κB pathway in MM cells. We found that this pathway involves bortezomib-resistant degradation of the inhibitor of NF-κB (IκBα), despite efficient bortezomib-mediated inhibition of proteasome activity. Moreover, HAPLN1 can also confer bortezomib-resistant survival of MM cells. We propose that HAPLN1 is a novel pathogenic factor in MM that induces an atypical NF-κB activation and thereby promotes bortezomib resistance in MM cells.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Extracellular Matrix Proteins/metabolism , Multiple Myeloma/metabolism , NF-kappa B/metabolism , Proteoglycans/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Drug Resistance, Neoplasm , Extracellular Matrix Proteins/genetics , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , NF-kappa B/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteoglycans/genetics , Proteolysis
9.
J Immunol ; 199(5): 1933-1941, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28754680

ABSTRACT

Colorectal cancer originates within immunologically complex microenvironments. To date, the benefits of immunotherapy have been modest, except in neoantigen-laden mismatch repair-deficient tumors. Approaches to enhance tumor-infiltrating lymphocytes in the tumor bed may substantially augment clinical immunotherapy responses. In this article, we report that proteolysis of the tolerogenic matrix proteoglycan versican (VCAN) strongly correlated with CD8+ T cell infiltration in colorectal cancer, regardless of mismatch repair status. Tumors displaying active VCAN proteolysis and low total VCAN were associated with robust (10-fold) CD8+ T cell infiltration. Tumor-intrinsic WNT pathway activation was associated with CD8+ T cell exclusion and VCAN accumulation. In addition to regulating VCAN levels at the tumor site, VCAN proteolysis results in the generation of bioactive fragments with novel functions (VCAN-derived matrikines). Versikine, a VCAN-derived matrikine, enhanced the generation of CD103+CD11chiMHCIIhi conventional dendritic cells (cDCs) from Flt3L-mobilized primary bone marrow-derived progenitors, suggesting that VCAN proteolysis may promote differentiation of tumor-seeding DC precursors toward IRF8- and BATF3-expressing cDCs. Intratumoral BATF3-dependent DCs are critical determinants for T cell antitumor immunity, effector T cell trafficking to the tumor site, and response to immunotherapies. Our findings provide a rationale for testing VCAN proteolysis as a predictive and/or prognostic immune biomarker and VCAN-derived matrikines as novel immunotherapy agents.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Dendritic Cells/immunology , Extracellular Matrix/immunology , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Versicans/immunology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation , Cell Movement , Cells, Cultured , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Proteolysis , Repressor Proteins/metabolism , Tumor Microenvironment
10.
Blood ; 128(5): 680-5, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27259980

ABSTRACT

Myeloma immunosurveillance remains incompletely understood. We have demonstrated proteolytic processing of the matrix proteoglycan, versican (VCAN), in myeloma tumors. Whereas intact VCAN exerts tolerogenic activities through Toll-like receptor 2 (TLR2) binding, the immunoregulatory consequences of VCAN proteolysis remain unknown. Here we show that human myeloma tumors displaying CD8(+) infiltration/aggregates underwent VCAN proteolysis at a site predicted to generate a glycosaminoglycan-bereft N-terminal fragment, versikine Myeloma-associated macrophages (MAMs), rather than tumor cells, chiefly produced V1-VCAN, the precursor to versikine, whereas stromal cell-derived ADAMTS1 was the most robustly expressed VCAN-degrading protease. Purified versikine induced early expression of inflammatory cytokines interleukin 1ß (IL-1ß) and IL-6 by human myeloma marrow-derived MAMs. We show that versikine signals through pathways both dependent and independent of Tpl2 kinase, a key regulator of nuclear factor κB1-mediated MAPK activation in macrophages. Unlike intact VCAN, versikine-induced Il-6 production was partially independent of Tlr2. In a model of macrophage-myeloma cell crosstalk, versikine induced components of "T-cell inflammation," including IRF8-dependent type I interferon transcriptional signatures and T-cell chemoattractant CCL2. Thus the interplay between stromal cells and myeloid cells in the myeloma microenvironment generates versikine, a novel bioactive damage-associated molecular pattern that may facilitate immune sensing of myeloma tumors and modulate the tolerogenic consequences of intact VCAN accumulation. Therapeutic versikine administration may potentiate T-cell-activating immunotherapies.


Subject(s)
Immunomodulation , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Proteolysis , Tumor Microenvironment , Versicans/metabolism , Alarmins/metabolism , Animals , Humans , Interferon Regulatory Factors/metabolism , Transcription, Genetic
11.
Handb Exp Pharmacol ; 249: 251-288, 2018.
Article in English | MEDLINE | ID: mdl-28315070

ABSTRACT

Multiple myeloma (MM) is an incurable hematopoietic cancer that is characterized by malignant plasma cell infiltration of the bone marrow and/or extramedullary sites. Multi-modality approaches including "novel agents," traditional chemotherapy, and/or stem cell transplantation are used in MM therapy. Drug resistance, however, ultimately develops and the disease remains incurable for the vast majority of patients. In this chapter, we review both tumor cell-autonomous and non-autonomous (microenvironment-dependent) mechanisms of drug resistance. MM provides an attractive paradigm highlighting a number of current concepts and challenges in oncology. Firstly, identification of MM cancer stem cells and their unique drug resistance attributes may provide rational avenues towards MM eradication and cure. Secondly, the oligoclonal evolution of MM and alternation of "clonal tides" upon therapy challenge our current understanding of treatment responses. Thirdly, the success of MM "novel agents" provides exemplary evidence for the impact of therapies that target the immune and non-immune microenvironment. Fourthly, the rapid pace of drug approvals for MM creates an impetus for development of precision medicine strategies and biomarkers that promote efficacy and mitigate toxicity and cost. While routine cure of the disease remains the ultimate and yet unattainable prize, MM advances in the last 10-15 years have provided an astounding paradigm for the treatment of blood cancers in the modern era and have radically transformed patient outcomes.


Subject(s)
Drug Resistance, Neoplasm , Multiple Myeloma/drug therapy , Tumor Microenvironment , Bone Marrow/pathology , Humans , Precision Medicine , Stem Cell Transplantation
12.
Proc Natl Acad Sci U S A ; 112(25): 7689-94, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26056298

ABSTRACT

Multiple myeloma (MM), a malignancy of plasma cells, is characterized by widespread genomic heterogeneity and, consequently, differences in disease progression and drug response. Although recent large-scale sequencing studies have greatly improved our understanding of MM genomes, our knowledge about genomic structural variation in MM is attenuated due to the limitations of commonly used sequencing approaches. In this study, we present the application of optical mapping, a single-molecule, whole-genome analysis system, to discover new structural variants in a primary MM genome. Through our analysis, we have identified and characterized widespread structural variation in this tumor genome. Additionally, we describe our efforts toward comprehensive characterization of genome structure and variation by integrating our findings from optical mapping with those from DNA sequencing-based genomic analysis. Finally, by studying this MM genome at two time points during tumor progression, we have demonstrated an increase in mutational burden with tumor progression at all length scales of variation.


Subject(s)
DNA Copy Number Variations , Multiple Myeloma/genetics , DNA/genetics , Humans , Loss of Heterozygosity , Multiple Myeloma/pathology , Polymorphism, Single Nucleotide
13.
J Biol Chem ; 291(7): 3268-79, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26631723

ABSTRACT

B cell malignancies comprise a diverse group of cancers that proliferate in lymph nodes, bone marrow, and peripheral blood. SIRT3 (sirtuin 3) is the major deacetylase within the mitochondrial matrix that promotes aerobic metabolism and controls reactive oxygen species (ROS) by deacetylating and activating isocitrate dehydrogenase 2 (IDH2) and superoxide dismutase 2 (SOD2). There is controversy as to whether SIRT3 acts as an oncogene or a tumor suppressor, and here we investigated its role in B cell malignancies. In mantle cell lymphoma patient samples, we found that lower SIRT3 protein expression was associated with worse overall survival. Further, SIRT3 protein expression was reduced in chronic lymphocytic leukemia primary samples and malignant B cell lines compared to primary B cells from healthy donors. This lower level of expression correlated with hyperacetylation of IDH2 and SOD2 mitochondrial proteins, lowered enzymatic activities, and higher ROS levels. Overexpression of SIRT3 decreased proliferation and diminished the Warburg-like phenotype in SIRT3-deficient cell lines, and this effect is largely dependent on deacetylation of IDH2 and SOD2. Lastly, depletion of SIRT3 from malignant B cell lines resulted in greater susceptibility to treatment with an ROS scavenger but did not result in greater sensitivity to inhibition of the hypoxia-inducible factor-1α pathway, suggesting that loss of SIRT3 increases proliferation via ROS-dependent but hypoxia-inducible factor-1α-independent mechanisms. Our study suggests that SIRT3 acts as a tumor suppressor in B cell malignancies, and activating the SIRT3 pathway might represent a novel therapeutic approach for treating B cell malignancies.


Subject(s)
Burkitt Lymphoma/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphoma, Follicular/metabolism , Lymphoma, Mantle-Cell/metabolism , Neoplasm Proteins/metabolism , Reactive Oxygen Species/metabolism , Sirtuin 3/metabolism , Acetylation , Aged , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Cell Line, Tumor , Cell Proliferation , Enzyme Activation , Gene Expression Regulation, Neoplastic , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Staging , Protein Processing, Post-Translational , RNA Interference , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sirtuin 3/antagonists & inhibitors , Sirtuin 3/genetics , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Survival Analysis , Tumor Cells, Cultured
14.
Blood ; 123(21): 3305-15, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24723682

ABSTRACT

Targeted modulation of microenvironmental regulatory pathways may be essential to control myeloma and other genetically/clonally heterogeneous cancers. Here we report that human myeloma-associated monocytes/macrophages (MAM), but not myeloma plasma cells, constitute the predominant source of interleukin-1ß (IL-1ß), IL-10, and tumor necrosis factor-α at diagnosis, whereas IL-6 originates from stromal cells and macrophages. To dissect MAM activation/cytokine pathways, we analyzed Toll-like receptor (TLR) expression in human myeloma CD14(+) cells. We observed coregulation of TLR2 and TLR6 expression correlating with local processing of versican, a proteoglycan TLR2/6 agonist linked to carcinoma progression. Versican has not been mechanistically implicated in myeloma pathogenesis. We hypothesized that the most readily accessible target in the versican-TLR2/6 pathway would be the mitogen-activated protein 3 (MAP3) kinase, TPL2 (Cot/MAP3K8). Ablation of Tpl2 in the genetically engineered in vivo myeloma model, Vκ*MYC, led to prolonged disease latency associated with plasma cell growth defect. Tpl2 loss abrogated the "inflammatory switch" in MAM within nascent myeloma lesions and licensed macrophage repolarization in established tumors. MYC activation/expression in plasma cells was independent of Tpl2 activity. Pharmacologic TPL2 inhibition in human monocytes led to dose-dependent attenuation of IL-1ß induction/secretion in response to TLR2 stimulation. Our results highlight a TLR2/6-dependent TPL2 pathway as novel therapeutic target acting nonautonomously through macrophages to control myeloma progression.


Subject(s)
MAP Kinase Kinase Kinases/immunology , Macrophages/pathology , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Proto-Oncogene Proteins/immunology , Animals , Cytokines/analysis , Cytokines/immunology , Drug Discovery , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Interleukin-1beta/analysis , Interleukin-1beta/immunology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/genetics , Macrophages/immunology , Mice , Mice, Inbred C57BL , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Toll-Like Receptor 2/agonists , Toll-Like Receptor 6/genetics , Toll-Like Receptor 6/immunology , Tumor Microenvironment
15.
Blood ; 132(16): 1629-1630, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30337318
16.
STAR Protoc ; 5(3): 103195, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39096495

ABSTRACT

Conventional type 1 dendritic cells (cDC1s) are critical for innate sensing of cancer, yet they are scarce in the tumor microenvironment (TME). Here, we present a protocol to identify and isolate cDC1 subsets from murine implantable tumors for subsequent transcriptomic profiling using a flow sorting-based strategy. We describe steps for cell culture of mouse tumors, tumoral growth, dissociation and isolation of tumoral cells, extracellular staining, and cell sorting. We then detail procedures for RNA isolation, mRNA library preparation, and sequencing. For complete details on the use and execution of this protocol, please refer to Papadas et al.1.


Subject(s)
Dendritic Cells , Gene Expression Profiling , Animals , Dendritic Cells/cytology , Dendritic Cells/metabolism , Mice , Gene Expression Profiling/methods , Flow Cytometry/methods , Tumor Microenvironment/genetics , Transcriptome/genetics , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Cell Separation/methods , Mice, Inbred C57BL
17.
Br J Haematol ; 160(6): 779-84, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23252623

ABSTRACT

Benefit from cytotoxic therapy in myeloma may be limited by the persistence of residual tumour cells within protective niches. We have previously shown that monocytes/macrophages acquire a proinflammatory transcriptional profile in the myeloma microenvironment. Here we report constitutive activation of MAP3K8 kinase-dependent pathways that regulate the magnitude and extent of inflammatory activity of monocytes/macrophages within myeloma niches. In myeloma tumour cells, MAP3K8 acts as mitogen-induced MAP3K in mitosis and is required for TNFα-mediated ERK activation. Pharmacological MAP3K8 inhibition results in dose-dependent, tumour cell-autonomous apoptosis despite contact with primary stroma. MAP3K8 blockade may disrupt crucial macrophage-tumour cell interactions within myeloma niches.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Macrophages/enzymology , Macrophages/pathology , Monocytes/enzymology , Monocytes/pathology , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Proto-Oncogene Proteins/metabolism , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Multiple Myeloma/drug therapy , Neoplasm, Residual , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Tumor Cells, Cultured
18.
Leuk Lymphoma ; 64(1): 87-97, 2023 01.
Article in English | MEDLINE | ID: mdl-36218226

ABSTRACT

The gut microbiome is an important feature of host immunity with associations to hematologic malignancies and cellular therapy. We evaluated the gut microbiome and dietary intake in patients with multiple myeloma undergoing autologous stem cell transplantation. Thirty patients were enrolled, and samples were collected at four timepoints: pre-transplant, engraftment, day +100 (D + 100), and 9-12 months post-transplant. Microbiome analysis demonstrated a loss of alpha diversity at the engraftment timepoint driven by decreases in Blautia, Ruminococcus, and Faecalibacterium genera and related to intravenous antibiotic exposure. Higher fiber intake was associated with increased relative abundance of Blautia at the pre-transplant timepoint. Lower alpha diversity at engraftment was associated with a partial response to therapy compared with complete response (CR) or very good partial response (VGPR) (CR/VGPR vs. PR, p < 0.05). We conclude that loss of bacterial diversity at engraftment may be associated with impaired response to stem cell transplantation in multiple myeloma.


Subject(s)
Gastrointestinal Microbiome , Hematopoietic Stem Cell Transplantation , Multiple Myeloma , Humans , Multiple Myeloma/therapy , Multiple Myeloma/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Treatment Outcome , Transplantation, Autologous , Disease-Free Survival , Antineoplastic Combined Chemotherapy Protocols , Stem Cell Transplantation/adverse effects
19.
Oncogene ; 42(21): 1751-1762, 2023 05.
Article in English | MEDLINE | ID: mdl-37031341

ABSTRACT

Multiple myeloma (MM) is a cancer of malignant plasma cells in the bone marrow and extramedullary sites. We previously characterized a VQ model for human high-risk MM. The various VQ lines display different disease phenotypes and survival rates, suggesting significant intra-model variation. Here, we use whole-exome sequencing and copy number variation (CNV) analysis coupled with RNA-Seq to stratify the VQ lines into corresponding clusters: Group A cells had monosomy chromosome (chr) 5 and overexpressed genes and pathways associated with sensitivity to bortezomib (Btz) treatment in human MM patients. By contrast, Group B VQ cells carried recurrent amplification (Amp) of chr3 and displayed high-risk MM features, including downregulation of Fam46c, upregulation of cancer growth pathways associated with functional high-risk MM, and expression of Amp1q and high-risk UAMS-70 and EMC-92 gene signatures. Consistently, in sharp contrast to Group A VQ cells that showed short-term response to Btz, Group B VQ cells were de novo resistant to Btz in vivo. Our study highlights Group B VQ lines as highly representative of the human MM subset with ultrahigh risk.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/pathology , DNA Copy Number Variations/genetics , Bortezomib/pharmacology , Bone Marrow/pathology , Down-Regulation , Drug Resistance, Neoplasm/genetics
20.
Br J Haematol ; 158(3): 336-46, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22583117

ABSTRACT

Multiple myeloma (MM) is characterized by almost exclusive tropism of malignant cells for the bone marrow (BM) milieu. The survival and proliferation of malignant plasma cells have been shown to rely on interactions with nonmalignant stromal cells, in particular mesenchymal stromal cells (MSCs), in the BM microenvironment. However, the BM microenvironment is composed of a diverse array of cell types. This study examined the role of macrophages, an abundant component of BM stroma, as a potential niche component that supports malignant plasma cells. We investigated the proliferation of MM tumour cell lines when cultured alone or together with MSCs, macrophages, or a combination of MSCs and macrophages, using the carboxyfluorescein succinimidyl ester assay. Consistently, we observed increased proliferation of MM cell lines in the presence of either MSCs or macrophages compared to cell line-only control. Furthermore, the combined co-culture of MSCs plus macrophages induced the greatest degree of proliferation of myeloma cells. In addition to increased proliferation, MSCs and macrophages decreased the rate of apoptosis of myeloma cells. Our in vitro studies provide evidence that highlights the role of macrophages as a key component of the BM microenvironment facilitating the growth of malignant plasma cells in MM.


Subject(s)
Cell Communication/physiology , Macrophages/pathology , Mesenchymal Stem Cells/pathology , Multiple Myeloma/pathology , Apoptosis/physiology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Boronic Acids/pharmacology , Bortezomib , Cell Differentiation/physiology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Coculture Techniques , Humans , Interleukin-6/metabolism , Lipopolysaccharide Receptors/metabolism , Macrophages/metabolism , Multiple Myeloma/metabolism , Pyrazines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL