Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
2.
Cell Death Differ ; 30(5): 1155-1165, 2023 05.
Article in English | MEDLINE | ID: mdl-36828915

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a continuum that includes epithelial, partial EMT, and mesenchymal states, each of which is associated with cancer progression, invasive capabilities, and ultimately, metastasis. We used a lineage-traced sporadic model of pancreatic cancer to generate a murine organoid biobank from primary and secondary tumors, including sublines that underwent partial EMT and complete EMT. Using an unbiased proteomics approach, we found that organoid morphology predicts the EMT state, and the solid organoids are associated with a partial EMT signature. We also observed that exogenous TGFß1 induces solid organoid morphology that is associated with changes in the S100 family, complete EMT, and the formation of high-grade tumors. S100A4 may be a useful biomarker for predicting EMT state, disease progression, and outcome in patients with pancreatic cancer.


Subject(s)
Pancreatic Neoplasms , S100 Proteins , Humans , Animals , Mice , S100 Proteins/genetics , S100 Proteins/metabolism , Epithelial-Mesenchymal Transition , Pancreatic Neoplasms/pathology , Cell Line, Tumor , Pancreatic Neoplasms
3.
Cancers (Basel) ; 13(19)2021 Oct 04.
Article in English | MEDLINE | ID: mdl-34638463

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid malignancies. While immortalized cancer cell lines and genetically engineered murine models have increased our understanding of PDAC tumorigenesis, they do not recapitulate inter- and intra-patient heterogeneity. PDAC patient derived organoid (PDO) biobanks have overcome this hurdle, and provide an opportunity for the high throughput screening of potential new therapies. This review provides a summary of the PDAC PDO biobanks established to date, and discusses how they have advanced our understanding of PDAC biology. Looking forward, the development of coculturing techniques for specific immune or stromal cell populations will enable a better understanding of the crosstalk that occurs within the tumor microenvironment, and the impact of this crosstalk on treatment response.

4.
Cell Death Differ ; 28(5): 1466-1476, 2021 05.
Article in English | MEDLINE | ID: mdl-33230260

ABSTRACT

Chronic inflammation of the large intestine is associated with an increased risk of developing colorectal cancer (CRC), the second most common cause of cancer-related deaths worldwide. Necroptosis has emerged as a form of lytic programmed cell death that, distinct from apoptosis, triggers an inflammatory response. Dysregulation of necroptosis has been linked to multiple chronic inflammatory diseases, including inflammatory bowel disease and cancer. Here, we used murine models of acute colitis, colitis-associated CRC, sporadic CRC, and spontaneous intestinal tumorigenesis to investigate the role of necroptosis in these gastrointestinal pathologies. In the Dextran Sodium Sulfate-induced acute colitis model, in some experiments, mice lacking the terminal necroptosis effector protein, MLKL, or its activator RIPK3, exhibited greater weight loss compared to wild-type mice, consistent with some earlier reports. However, the magnitude of weight loss and accompanying inflammatory pathology upon Mlkl deletion varied substantially between independent repeats. Such variation provides a possible explanation for conflicting literature reports. Furthermore, contrary to earlier reports, we observed that genetic deletion of MLKL had no impact on colon cancer development using several mouse models. Collectively, these data do not support an obligate role for necroptosis in inflammation or cancer within the gastrointestinal tract.


Subject(s)
Colonic Neoplasms/genetics , Inflammation/genetics , Necroptosis/genetics , Animals , Disease Models, Animal , Mice
5.
J Biol Chem ; 295(24): 8285-8301, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32332100

ABSTRACT

Interleukin (IL) 11 activates multiple intracellular signaling pathways by forming a complex with its cell surface α-receptor, IL-11Rα, and the ß-subunit receptor, gp130. Dysregulated IL-11 signaling has been implicated in several diseases, including some cancers and fibrosis. Mutations in IL-11Rα that reduce signaling are also associated with hereditary cranial malformations. Here we present the first crystal structure of the extracellular domains of human IL-11Rα and a structure of human IL-11 that reveals previously unresolved detail. Disease-associated mutations in IL-11Rα are generally distal to putative ligand-binding sites. Molecular dynamics simulations showed that specific mutations destabilize IL-11Rα and may have indirect effects on the cytokine-binding region. We show that IL-11 and IL-11Rα form a 1:1 complex with nanomolar affinity and present a model of the complex. Our results suggest that the thermodynamic and structural mechanisms of complex formation between IL-11 and IL-11Rα differ substantially from those previously reported for similar cytokines. This work reveals key determinants of the engagement of IL-11 by IL-11Rα that may be exploited in the development of strategies to modulate formation of the IL-11-IL-11Rα complex.


Subject(s)
Interleukin-11 Receptor alpha Subunit/chemistry , Interleukin-11 Receptor alpha Subunit/metabolism , Interleukin-11/metabolism , Area Under Curve , Cell Line, Tumor , Entropy , Humans , Interleukin-11 Receptor alpha Subunit/genetics , Models, Molecular , Mutation/genetics , Protein Binding , Protein Domains , Structure-Activity Relationship , Thermodynamics
6.
Adv Exp Med Biol ; 1240: 59-72, 2020.
Article in English | MEDLINE | ID: mdl-32060888

ABSTRACT

Interleukin (IL)-18, a member of the IL-1 family of cytokines, has emerged as a key regulator of mucosal homeostasis within the gastrointestinal tract. Like other members of this family, IL-18 is secreted as an inactive protein and is processed into its active form by caspase-1, although other contributors to precursor processing are emerging.Numerous studies have evaluated the role of IL-18 within the gastrointestinal tract using genetic or complementary pharmacological tools and have revealed multiple roles in tumorigenesis. Most striking among these are the divergent roles for IL-18 in colon and gastric cancers. Here, we review our current understanding of IL-18 biology and how this applies to colorectal and gastric cancers.


Subject(s)
Colorectal Neoplasms/pathology , Interleukin-18/metabolism , Stomach Neoplasms/pathology , Tumor Microenvironment , Animals , Caspase 1/metabolism , Humans
7.
Cell Death Differ ; 27(2): 742-757, 2020 02.
Article in English | MEDLINE | ID: mdl-31296963

ABSTRACT

Gastrointestinal epithelial cells provide a selective barrier that segregates the host immune system from luminal microorganisms, thereby contributing directly to the regulation of homeostasis. We have shown that from early embryonic development Bcl-G, a Bcl-2 protein family member with unknown function, was highly expressed in gastrointestinal epithelial cells. While Bcl-G was dispensable for normal growth and development in mice, the loss of Bcl-G resulted in accelerated progression of colitis-associated cancer. A label-free quantitative proteomics approach revealed that Bcl-G may contribute to the stability of a mucin network, which when disrupted, is linked to colon tumorigenesis. Consistent with this, we observed a significant reduction in Bcl-G expression in human colorectal tumors. Our study identifies an unappreciated role for Bcl-G in colon cancer.


Subject(s)
Colorectal Neoplasms/metabolism , Inflammation/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Colitis/metabolism , Colitis/pathology , Colorectal Neoplasms/pathology , Humans , Inflammation/pathology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/genetics
8.
Growth Factors ; 37(1-2): 1-11, 2019 04.
Article in English | MEDLINE | ID: mdl-31161823

ABSTRACT

Interleukin (IL)-11 belongs to the IL-6 family of cytokines, discovered over 30 years ago. While early studies focused on the ability of IL-11 to stimulate megakaryocytopoiesis, the importance of this cytokine to inflammatory disease and cancers is only just beginning to be uncovered. This review outlines recent advances in our understanding of IL-11 biology, and highlights the development of novel therapeutics with the potential for clinical targeting of signaling by this cytokine in multiple diseases.


Subject(s)
Cardiovascular Diseases/genetics , Craniofacial Dysostosis/genetics , Immune System Diseases/genetics , Interleukin-11/genetics , Neoplasms/genetics , Animals , Humans , Interleukin-11/metabolism , Mutation
9.
Cytokine ; 118: 8-18, 2019 06.
Article in English | MEDLINE | ID: mdl-29396054

ABSTRACT

Chronic inflammation is recognized as a key tumor-promoting factor in a number of epithelial cancers, including gastric cancer (GC). The production of pro-inflammatory cytokines in the tumor microenvironment by both the innate and the adaptive immune response can activate signaling pathways that are associated with increased cell survival and proliferation of cancer cells. Among the cytokines that have most commonly been linked to inflammation-associated cancers, are the Th17 cell-associated cytokines IL-17A, IL-23, IL-22, and the IL-1 family members IL-1ß and IL-18. However, whether their contribution to inflammation-associated cancers is universal, or specific to individual types of cancers, remains to be elucidated. This review will explore our current understanding of the known roles of these cytokines in gastritis and discuss how their therapeutic inhibition may be useful for GC.


Subject(s)
Antineoplastic Agents/immunology , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Interleukin-18/antagonists & inhibitors , Interleukin-18/immunology , Stomach Neoplasms/immunology , Stomach Neoplasms/therapy , Animals , Humans , Inflammation/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
10.
Mol Immunol ; 110: 48-56, 2019 06.
Article in English | MEDLINE | ID: mdl-29174430

ABSTRACT

Innate lymphoid cells (ILCs) contribute to the regulation of gastrointestinal (GI) homeostasis. Over the past 15 years, there has been a large effort to dissect the mechanisms required for GI homeostasis, with a major focus on different immune cell populations and the cytokines that they produce. In contrast to T-helper (Th) cells, ILCs respond rapidly to cytokines in their microenvironment in the absence of specific antigens; however, once activated both cell populations have similar effector functions. Two effector cytokines produced by both ILC3 and Th17 cell populations, Interleukin (IL)-17 and IL-22, have taken center stage for their ability to signal directly to GI epithelial cells and promote epithelial cell survival. In this review, we outline our current understanding of ILCs in the GI tract, and focus on GI cancers associated with aberrant production of IL-17 and IL-22. We highlight evidence from both mouse and patient-based analyses and discuss how tumor cells may hijack the potential evolutionary redundancy of these two cell populations.


Subject(s)
Gastrointestinal Neoplasms/immunology , Immunity, Innate , Lymphocytes/physiology , T-Lymphocytes/physiology , Animals , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/therapy , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Mice , Tumor Escape/genetics , Tumor Escape/immunology , Tumor Microenvironment/immunology
11.
Cancer Immunol Res ; 6(4): 409-421, 2018 04.
Article in English | MEDLINE | ID: mdl-29463593

ABSTRACT

Interleukin 33 (IL33) is an inflammatory cytokine released during necrotic cell death. The epithelium and stroma of the intestine express large amounts of IL33 and its receptor St2. IL33 is therefore continuously released during homeostatic turnover of the intestinal mucosa. Although IL33 can prevent colon cancer associated with inflammatory colitis, the contribution of IL33 signaling to sporadic colon cancer remains unknown. Here, we utilized a mouse model of sporadic colon cancer to investigate the contribution of IL33 signaling to tumorigenesis in the absence of preexisting inflammation. We demonstrated that genetic ablation of St2 enhanced colon tumor development. Conversely, administration of recombinant IL33 reduced growth of colon cancer cell allografts. In reciprocal bone marrow chimeras, the concurrent loss of IL33 signaling within radioresistant nonhematopoietic, and the radiosensitive hematopoietic, compartments was associated with increased tumor burden. We detected St2 expression within the radioresistant mesenchymal cell compartment of the colon whose stimulation with IL33 induced expression of bona fide NF-κB target genes. Mechanistically, we discovered that St2 deficiency within the nonhematopoietic compartment coincided with increased abundance of regulatory T cells and suppression of an IFNγ gene expression signature, whereas IL33 administration triggered IFNγ expression by tumor allograft-infiltrating T cells. The decrease of this IFNγ gene expression signature was associated with more aggressive disease in human colon cancer patients, suggesting that lack of IL33 signaling impaired the generation of a potent IFNγ-mediated antitumor immune response. Collectively, our data reveal that IL33 functions as a tumor suppressor in sporadic colon cancer. Cancer Immunol Res; 6(4); 409-21. ©2018 AACR.


Subject(s)
Colonic Neoplasms/metabolism , Interferon-gamma/metabolism , Interleukin-33/metabolism , Signal Transduction , Allografts , Animals , Biomarkers , Biopsy , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Gene Expression , Gene Expression Profiling , Interferon-gamma/genetics , Interleukin-33/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , NF-kappa B/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Transcriptome
12.
J Interferon Cytokine Res ; 35(5): 340-50, 2015 May.
Article in English | MEDLINE | ID: mdl-25760898

ABSTRACT

The gastrointestinal tract is lined by a single layer of epithelial cells that secrete mucus toward the lumen, which collectively separates the immune sentinels in the underlying lamina propria from the intestinal microflora to prevent aberrant immune responses. Inflammatory bowel disease (IBD) describes a group of autoimmune diseases that arise from defects in epithelial barrier function and, as a consequence, aberrant production of inflammatory cytokines. Among these, interleukin (IL)-6, IL-11, and IL-22 are elevated in human IBD patients and corresponding mouse models and, through activation of the JAK/STAT3 pathway, can both propagate and ameliorate disease. In particular, cytokine-mediated activation of STAT3 in the epithelial lining cells affords cellular protection, survival, and proliferation, thereby affording therapeutic opportunities for the prevention and treatment of colitis. In this review, we focus on recent insights gained from therapeutic modulation of the activities of IL-6, IL-11, and IL-22 in models of IBD and advocate a cautionary approach with these cytokines to minimize their tumor-promoting activities on neoplastic epithelium.


Subject(s)
Cytokines/metabolism , Inflammatory Bowel Diseases/metabolism , STAT3 Transcription Factor/metabolism , Animals , Homeostasis , Humans , Inflammatory Bowel Diseases/drug therapy , Interleukin-10/metabolism , Interleukin-11/metabolism , Interleukin-6/metabolism , Interleukins/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Interleukin-22
13.
Semin Immunol ; 26(1): 29-37, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24434062

ABSTRACT

A contiguous intestinal epithelial barrier safeguards against aberrant activation of the immune system and therefore requires molecular mechanisms that ensure effective wound-healing responses. During this processes cytokine-producing myeloid cells serve as rheostats that link the degree of wounding and local inflammation to the epithelial repair response. Likewise, intestinal inflammation is an important factor by which the microenvironment promotes tumorigenesis and the progression of established cancers by facilitating neoplastic cell survival and proliferation. Among the cytokines and chemokines orchestrating this process, those comprising the interleukin (IL) IL6, IL10/IL22 and IL17/IL23 families play a prominent role by virtue of converging on the latent Signal Transducer and Activator of Transcription (Stat)-3. Accordingly, aberrant and persistent Stat3 activation is a frequent observation in cancers of the gastrointestinal tract where it promotes "cancer hallmark capabilities" in the malignant epithelium and suppresses the anti-tumor response of innate and adaptive immune cells. Here, we discuss recent insights arising from situations where persistent activation of the gp130/Stat3 signaling cascades result from excessive abundance of IL6 family cytokines. In particular, we highlight novel and unique roles for IL11 in promoting intestinal wound-healing and, in its corrupted form, enabling and facilitating growth of inflammation-associated and sporadic gastrointestinal tumors.


Subject(s)
Cytokine Receptor gp130/metabolism , Intestinal Mucosa/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cytokine Receptor gp130/genetics , Cytokines/metabolism , Homeostasis , Humans , Inflammation/genetics , Inflammation/metabolism , Intestinal Mucosa/pathology , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction
14.
Cancer Cell ; 24(2): 257-71, 2013 Aug 12.
Article in English | MEDLINE | ID: mdl-23948300

ABSTRACT

Among the cytokines linked to inflammation-associated cancer, interleukin (IL)-6 drives many of the cancer "hallmarks" through downstream activation of the gp130/STAT3 signaling pathway. However, we show that the related cytokine IL-11 has a stronger correlation with elevated STAT3 activation in human gastrointestinal cancers. Using genetic mouse models, we reveal that IL-11 has a more prominent role compared to IL-6 during the progression of sporadic and inflammation-associated colon and gastric cancers. Accordingly, in these models and in human tumor cell line xenograft models, pharmacologic inhibition of IL-11 signaling alleviated STAT3 activation, suppressed tumor cell proliferation, and reduced the invasive capacity and growth of tumors. Our results identify IL-11 signaling as a potential therapeutic target for the treatment of gastrointestinal cancers.


Subject(s)
Cell Transformation, Neoplastic/immunology , Gastrointestinal Neoplasms/immunology , Interleukin-11/metabolism , Interleukin-6/metabolism , Animals , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/therapy , Humans , Interleukin-11/genetics , Interleukin-11/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Molecular Targeted Therapy , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL