Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 13(1): 6021, 2022 10 12.
Article in English | MEDLINE | ID: mdl-36224199

ABSTRACT

Drug-induced cytopenias are a prevalent and significant issue that worsens clinical outcomes and hinders the effective treatment of cancer. While reductions in blood cell numbers are classically associated with traditional cytotoxic chemotherapies, they also occur with newer targeted small molecules and the factors that determine the hematotoxicity profiles of oncologic drugs are not fully understood. Here, we explore why some Aurora kinase inhibitors cause preferential neutropenia. By studying drug responses of healthy human hematopoietic cells in vitro and analyzing existing gene expression datasets, we provide evidence that the enhanced vulnerability of neutrophil-lineage cells to Aurora kinase inhibition is caused by early developmental changes in ATP-binding cassette (ABC) transporter expression. These data show that hematopoietic cell-intrinsic expression of ABC transporters may be an important factor that determines how some Aurora kinase inhibitors affect the bone marrow.


Subject(s)
ATP-Binding Cassette Transporters , Neutrophils , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate , Aurora Kinases/metabolism , Hematopoiesis/genetics , Humans , Neoplasm Proteins/metabolism , Neutrophils/metabolism , Protein Kinase Inhibitors/pharmacology
3.
Nat Biomed Eng ; 4(4): 394-406, 2020 04.
Article in English | MEDLINE | ID: mdl-31988457

ABSTRACT

The inaccessibility of living bone marrow (BM) hampers the study of its pathophysiology under myelotoxic stress induced by drugs, radiation or genetic mutations. Here, we show that a vascularized human BM-on-a-chip (BM chip) supports the differentiation and maturation of multiple blood cell lineages over 4 weeks while improving CD34+ cell maintenance, and that it recapitulates aspects of BM injury, including myeloerythroid toxicity after clinically relevant exposures to chemotherapeutic drugs and ionizing radiation, as well as BM recovery after drug-induced myelosuppression. The chip comprises a fluidic channel filled with a fibrin gel in which CD34+ cells and BM-derived stromal cells are co-cultured, a parallel channel lined by human vascular endothelium and perfused with culture medium, and a porous membrane separating the two channels. We also show that BM chips containing cells from patients with the rare genetic disorder Shwachman-Diamond syndrome reproduced key haematopoietic defects and led to the discovery of a neutrophil maturation abnormality. As an in vitro model of haematopoietic dysfunction, the BM chip may serve as a human-specific alternative to animal testing for the study of BM pathophysiology.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow/pathology , Hematopoiesis , Microfluidics/methods , Animals , Antigens, CD34 , Bone Marrow/drug effects , Bone Marrow/radiation effects , Bone Marrow Transplantation , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Lab-On-A-Chip Devices , Mesenchymal Stem Cells , Microfluidics/instrumentation
4.
Cell Mol Gastroenterol Hepatol ; 9(3): 507-526, 2020.
Article in English | MEDLINE | ID: mdl-31778828

ABSTRACT

BACKGROUND & AIMS: The mucus layer in the human colon protects against commensal bacteria and pathogens, and defects in its unique bilayered structure contribute to intestinal disorders, such as ulcerative colitis. However, our understanding of colon physiology is limited by the lack of in vitro models that replicate human colonic mucus layer structure and function. Here, we investigated if combining organ-on-a-chip and organoid technologies can be leveraged to develop a human-relevant in vitro model of colon mucus physiology. METHODS: A human colon-on-a-chip (Colon Chip) microfluidic device lined by primary patient-derived colonic epithelial cells was used to recapitulate mucus bilayer formation, and to visualize mucus accumulation in living cultures noninvasively. RESULTS: The Colon Chip supports spontaneous goblet cell differentiation and accumulation of a mucus bilayer with impenetrable and penetrable layers, and a thickness similar to that observed in the human colon, while maintaining a subpopulation of proliferative epithelial cells. Live imaging of the mucus layer formation on-chip showed that stimulation of the colonic epithelium with prostaglandin E2, which is increased during inflammation, causes rapid mucus volume expansion via an Na-K-Cl cotransporter 1 ion channel-dependent increase in its hydration state, but no increase in de novo mucus secretion. CONCLUSIONS: This study shows the production of colonic mucus with a physiologically relevant bilayer structure in vitro, which can be analyzed in real time noninvasively. The Colon Chip may offer a new preclinical tool to analyze the role of mucus in human intestinal homeostasis as well as diseases, such as ulcerative colitis and cancer.


Subject(s)
Colon/metabolism , Intestinal Mucosa/metabolism , Lab-On-A-Chip Devices , Mucus/metabolism , Cells, Cultured , Dinoprostone/metabolism , Goblet Cells/physiology , Humans , Organoids , Primary Cell Culture/methods , Solute Carrier Family 12, Member 1/metabolism
5.
Nat Commun ; 10(1): 5580, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31811125

ABSTRACT

Mucosal healing plays a critical role in combatting the effects of inflammatory bowel disease, fistulae and ulcers. While most treatments for such diseases focus on systemically delivered anti-inflammatory drugs, often leading to detrimental side effects, mucosal healing agents that target the gut epithelium are underexplored. We genetically engineer Escherichia coli Nissle 1917 (EcN) to create fibrous matrices that promote gut epithelial integrity in situ. These matrices consist of curli nanofibers displaying trefoil factors (TFFs), known to promote intestinal barrier function and epithelial restitution. We confirm that engineered EcN can secrete the curli-fused TFFs in vitro and in vivo, and is non-pathogenic. We observe enhanced protective effects of engineered EcN against dextran sodium sulfate-induced colitis in mice, associated with mucosal healing and immunomodulation. This work lays a foundation for the development of a platform in which the in situ production of therapeutic protein matrices from beneficial bacteria can be exploited.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drug Delivery Systems/methods , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Engineering/methods , Probiotics/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Bacterial Proteins/genetics , Caco-2 Cells , Colitis/chemically induced , Colitis/drug therapy , Colitis/immunology , Colitis/pathology , Dextran Sulfate/adverse effects , Disease Models, Animal , Epithelium , Female , Humans , Immunomodulation , Inflammatory Bowel Diseases/drug therapy , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Probiotics/pharmacology , Trefoil Factors/genetics
6.
Microbiome ; 7(1): 43, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30890187

ABSTRACT

BACKGROUND: Species-specific differences in tolerance to infection are exemplified by the high susceptibility of humans to enterohemorrhagic Escherichia coli (EHEC) infection, whereas mice are relatively resistant to this pathogen. This intrinsic species-specific difference in EHEC infection limits the translation of murine research to human. Furthermore, studying the mechanisms underlying this differential susceptibility is a difficult problem due to complex in vivo interactions between the host, pathogen, and disparate commensal microbial communities. RESULTS: We utilize organ-on-a-chip (Organ Chip) microfluidic culture technology to model damage of the human colonic epithelium induced by EHEC infection, and show that epithelial injury is greater when exposed to metabolites derived from the human gut microbiome compared to mouse. Using a multi-omics approach, we discovered four human microbiome metabolites-4-methyl benzoic acid, 3,4-dimethylbenzoic acid, hexanoic acid, and heptanoic acid-that are sufficient to mediate this effect. The active human microbiome metabolites preferentially induce expression of flagellin, a bacterial protein associated with motility of EHEC and increased epithelial injury. Thus, the decreased tolerance to infection observed in humans versus other species may be due in part to the presence of compounds produced by the human intestinal microbiome that actively promote bacterial pathogenicity. CONCLUSION: Organ-on-chip technology allowed the identification of specific human microbiome metabolites modulating EHEC pathogenesis. These identified metabolites are sufficient to increase susceptibility to EHEC in our human Colon Chip model and they contribute to species-specific tolerance. This work suggests that higher concentrations of these metabolites could be the reason for higher susceptibility to EHEC infection in certain human populations, such as children. Furthermore, this research lays the foundation for therapeutic-modulation of microbe products in order to prevent and treat human bacterial infection.


Subject(s)
Bacteria/metabolism , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/pathology , Intestines/cytology , Organ Culture Techniques/methods , Animals , Benzoates/pharmacology , Caproates/pharmacology , Cells, Cultured , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Female , Gastrointestinal Microbiome , Heptanoic Acids/pharmacology , Humans , Intestines/microbiology , Male , Mice , Microchip Analytical Procedures , Species Specificity
7.
J Leukoc Biol ; 92(1): 123-31, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22493080

ABSTRACT

Inflammation alters hematopoiesis, often by decreasing erythropoiesis and enhancing myeloid output. The mechanisms behind these changes and how the BM stroma contributes to this process are active areas of research. In this study, we examine these questions in the setting of murine Toxoplasma gondii infection. Our data reveal that infection alters early myeloerythroid differentiation, blocking erythroid development beyond the Pre MegE stage, while expanding the GMP population. IL-6 was found to be a critical mediator of these differences, independent of hepcidin-induced iron restriction. Comparing the BM with the spleen showed that the hematopoietic response was driven by the local microenvironment, and BM chimeras demonstrated that radioresistant cells were the relevant source of IL-6 in vivo. Finally, direct ex vivo sorting revealed that VCAM(+)CD146(lo) BM stromal fibroblasts significantly increase IL-6 secretion after infection. These data suggest that BMSCs regulate the hematopoietic changes during inflammation via IL-6.


Subject(s)
Erythroid Precursor Cells/drug effects , Interleukin-6/pharmacology , Myeloid Progenitor Cells/drug effects , Stromal Cells/drug effects , Toxoplasma/drug effects , Toxoplasmosis/drug therapy , Animals , Bone Marrow/drug effects , Bone Marrow/parasitology , Bone Marrow/pathology , Cell Differentiation/drug effects , Cells, Cultured , Colony-Forming Units Assay , Erythroid Precursor Cells/parasitology , Erythroid Precursor Cells/pathology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/parasitology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/parasitology , Hematopoietic Stem Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Progenitor Cells/parasitology , Myeloid Progenitor Cells/pathology , Stromal Cells/parasitology , Stromal Cells/pathology , Toxoplasma/pathogenicity , Toxoplasmosis/parasitology , Toxoplasmosis/pathology
8.
Immunity ; 34(3): 435-47, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21419664

ABSTRACT

Vitamin A and its metabolite, retinoic acid (RA) are implicated in the regulation of immune homeostasis via the peripheral induction of regulatory T cells. Here we showed RA was also required to elicit proinflammatory CD4(+) helper T cell responses to infection and mucosal vaccination. Retinoic acid receptor alpha (RARα) was the critical mediator of these effects. Antagonism of RAR signaling and deficiency in RARα (Rara(-/-)) resulted in a cell-autonomous CD4(+) T cell activation defect, which impaired intermediate signaling events, including calcium mobilization. Altogether, these findings reveal a fundamental role for the RA-RARα axis in the development of both regulatory and inflammatory arms of adaptive immunity and establish nutritional status as a broad regulator of adaptive T cell responses.


Subject(s)
Adaptive Immunity/immunology , CD4-Positive T-Lymphocytes/immunology , Receptors, Retinoic Acid/immunology , Tretinoin/immunology , Animals , Female , Homeostasis/immunology , Male , Mice , Mice, Inbred C57BL , Retinoic Acid Receptor alpha , Signal Transduction , Toxoplasmosis/immunology
9.
Clin Cancer Res ; 16(23): 5824-34, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20947516

ABSTRACT

PURPOSE: Esophageal cancer is one of the most aggressive and deadly forms of cancer; highlighting the need to identify biomarkers for early detection and prognostic classification. Our recent studies have identified inflammatory gene and microRNA signatures derived from tumor and nontumor tissues as prognostic biomarkers of hepatocellular, lung, and colorectal adenocarcinoma. Here, we examine the relationship between expression of these inflammatory genes and micro RNA (miRNA) expression in esophageal adenocarcinoma and patient survival. EXPERIMENTAL DESIGN: We measured the expression of 23 inflammation-associated genes in tumors and adjacent normal tissues from 93 patients (58 Barrett's and 35 Sporadic adenocarcinomas) by quantitative reverse transcription-polymerase chain reaction. These data were used to build an inflammatory risk model, based on multivariate Cox regression, to predict survival in a training cohort (n = 47). We then determined whether this model could predict survival in a cohort of 46 patients. Expression data for miRNA-375 were available for these patients and was combined with inflammatory gene expression. RESULTS: IFN-γ, IL-1α, IL-8, IL-21, IL-23, and proteoglycan expression in tumor and nontumor samples were each associated with poor prognosis based on Cox regression [(Z-score)>1.5] and therefore were used to generate an inflammatory risk score (IRS). Patients with a high IRS had poor prognosis compared with those with a low IRS in the training (P = 0.002) and test (P = 0.012) cohorts. This association was stronger in the group with Barrett's history. When combining with miRNA-375, the combined IRS/miR signature was an improved prognostic classifier than either one alone. CONCLUSION: Transcriptional profiling of inflammation-associated genes and miRNA expression in resected esophageal Barrett's-associated adenocarcinoma tissues may have clinical utility as predictors of prognosis.


Subject(s)
Adenocarcinoma/diagnosis , Barrett Esophagus/diagnosis , Esophageal Neoplasms/diagnosis , Inflammation/genetics , MicroRNAs/genetics , Adenocarcinoma/classification , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Algorithms , Barrett Esophagus/classification , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Esophageal Neoplasms/classification , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/physiology , Humans , Inflammation/pathology , Male , Middle Aged , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...