Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Scand J Immunol ; : e13391, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773691

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are innate-like T cells that can be activated by microbial antigens and cytokines and are abundant in mucosal tissues including the colon. MAIT cells have cytotoxic and pro-inflammatory functions and have potentials for use as adoptive cell therapy. However, studies into their anti-cancer activity, including their role in colon cancer, are limited. Using an animal model of colon cancer, we showed that peritumoral injection of in vivo-expanded MAIT cells into RAG1-/- mice with MC38-derived tumours inhibits tumour growth compared to control. Multiplex cytokine analyses showed that tumours from the MAIT cell-treated group have higher expression of markers for eosinophil-activating cytokines, suggesting a potential association between eosinophil recruitment and tumour inhibition. In a human peripheral leukocyte co-culture model, we showed that leukocytes stimulated with MAIT ligand showed an increase in eotaxin-1 production and activation of eosinophils, associated with increased cancer cell killing. In conclusion, we showed that MAIT cells have a protective role in a murine colon cancer model, associated with modulation of the immune response to cancer, potentially involving eosinophil-associated mechanisms. Our results highlight the potential of MAIT cells for non-donor restricted colon cancer immunotherapy.

2.
Biomedicines ; 12(2)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38397942

ABSTRACT

RNA binding proteins (RBPs) post-transcriptionally regulate gene expression by associating with regulatory sequences in the untranslated regions of mRNAs. Cold-inducible RBP (CIRP) is a stress-induced RBP that was recently shown to modulate inflammation in response to cellular stress, where it increases or decreases pro-tumorigenic (proinflammatory) cytokines in different contexts. CIRP expression is altered in several cancers, including breast cancer, but the effects of CIRP on inflammation in breast cancer is not known. Here, we investigate if CIRP alters growth and the inflammatory profile of breast tumors. Transgenic mice overexpressing CIRP in the mammary epithelium were crossed with the PyMT mouse model of breast cancer, and the effects on both early and late tumorigenesis and inflammation were assessed. The effects of CIRP knockdown were also assessed in Py2T cell grafts. Overexpression of CIRP led to decreased tumorigenesis in the PyMT mouse model. Conversely, the knockdown of CIRP in Py2T cell grafts led to increased tumor growth. Luminex cytokine assays assessed the effects on the inflammatory environment. CIRP/PyMT mammary glands/mammary tumors and serum had decreased cytokines that promote inflammation, angiogenesis, and metastasis compared to PyMT mammary glands and serum, documenting a shift towards an environment less supportive of tumorigenesis. CIRP overexpression also decreased CD4+ helper T cells and increased CD8+ cytotoxic T cells in mammary tumors. Overall, these data support a role for CIRP as a potent antitumor molecule that suppresses both local and systemic pro-tumorigenic inflammation.

3.
J Crohns Colitis ; 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38224550

ABSTRACT

BACKGROUND: Mesenchymal stromal cells are suggested to play a critical role in the Crohn's Disease (CD) associated fibrosis. MAPKAPK2 (MK2) has emerged as a potential therapeutic target to reduce inflammation in CD. However, cell-specific pattern of pMK2 activation and its role in the CD associated fibrosis are unknown. The objectives of this study were to evaluate cell-specific changes in MK2 activity between predominantly inflammatory CD versus CD with fibrotic complication and define the role of stromal cell-specific MK2 activation in CD-associated fibrosis. METHODS: CD tissue, CD tissue derived mesenchymal stromal cells known as myo-/fibroblasts (CD-MFs), fibroblast specific MK2 conditional KO mice were used. RESULTS: We observed that in the inflamed area of predominantly inflammatory CD, high MK2 activity was equally distributed between mesenchymal and hematopoietic cells. By contrast, in CD with fibrotic complications, high MK2 activity was mostly associated with mesenchymal stromal cells. Using ex vivo CD tissue explants and IL-10KO murine colitis model, we demonstrated that pro-fibrotic responses are significantly reduced by treatment with the MK2 inhibitor PF-3644022. Inhibition of MK2 activity in primary cultures of CD-MFs significantly reduced basal and TGF-ß1-induced profibrotic responses. Using fibroblast-specific MK2 knockout mice in chronic DSS colitis, we demonstrated that fibroblast intrinsic MK2 signaling is among the key processes involved in the chronic inflammation induced profibrotic responses. CONCLUSIONS: Our data suggest that activation of MK2 within fibroblasts contributes to the chronic inflammation induced fibrosis in CD and that targeting MK2 has potential for the development of novel therapeutic approaches for fibrosis in CD.

4.
bioRxiv ; 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38293128

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are innate-like T cells that can be activated by microbial antigens and cytokines and are abundant in mucosal tissues including the colon. MAIT cells have cytotoxic and pro-inflammatory functions and have potentials for use as adoptive cell therapy. However, studies into their anti-cancer activity, including their role in colon cancer, are limited. Using an animal model of colon cancer, we show that peritumoral injection of in vivo-expanded MAIT cells into RAG1-/- mice with MC38-derived tumors inhibits tumor growth compared to control. Multiplex cytokine analyses show that tumors from the MAIT cell-treated group have higher expression of markers for eosinophil-activating cytokines, suggesting an association between eosinophil recruitment and tumor inhibition. In a human peripheral leukocyte co-culture model, we show that leukocytes stimulated with MAIT ligand show an increase in eotaxin-1 production and activation of eosinophils, associated with increased cancer cell killing. In conclusion, we show that MAIT cells have a protective role in a murine colon cancer model, associated with modulation of the immune response to cancer, potentially involving eosinophil-associated mechanisms. Our results highlight the potential of MAIT cells for non-donor restricted colon cancer immunotherapy.

5.
Front Immunol ; 14: 1212100, 2023.
Article in English | MEDLINE | ID: mdl-37415974

ABSTRACT

Background: Immune cell composition is a critical and dynamic component of the tumor microenvironment, which has an impact on immunosuppression and progression of cancer. T cells, especially CD8+ T cells, are one of the major immune cell types responsible for tumor cell killing employing receptor-ligand mediated apoptosis and/or releasing lytic granules among others. Accumulating evidence highlighted that adoptive transfer of activated and/or modified immune cells can enhance anti-tumorigenic immune responses and serve as promising therapy approach for patients with cancers. The mitogen-activated protein kinase-activated protein kinase 2 (MK2) is a serine/threonine protein kinase, which controls production and secretion of numerous pro-inflammatory cytokines and chemokines involved in tumorigenesis. However, limited efforts have been made to learn how MK2 may affects CD8+ T cell action and function in the tumor microenvironment especially in gastrointestinal cancers. Methods: To explore the therapeutic potential of MK2 in the immune response mediated by CD8+ T cells, RAG1 knockout mice with PK5L1940 and BRAF cells-derived allograft tumors were treated with WT or MK2 knockout CD8+ T cells. The phenotype of CD8+ T cells with MK2 depletion were evaluated in vitro. Immunofluorescence staining, real-time PCR and multiplex analysis were utilized to estimate the expression of apoptotic and lytic factors. Results: Here, we show that CD8+ T cells with MK2 depletion prevent gastrointestinal cancer growth, which is accompanied by enhanced expression and secretion of factors related to apoptosis. Moreover, using in vitro and in vivo approaches, we found that depletion of MK2 lead to hyperactivation of CD8+ T cells and enhanced anti-tumor immunity. Conclusion: Overall, we documented that MK2 drives the progression of gastrointestinal cancers and prevents immune response generated by CD8+ T cells suggesting potential implications of MK2 in the immunotherapy of gastrointestinal cancers.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Animals , Mice , CD8-Positive T-Lymphocytes/metabolism , Colonic Neoplasms/therapy , Immunotherapy , Intracellular Signaling Peptides and Proteins , Pancreas/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Microenvironment
6.
Matrix Biol ; 121: 149-166, 2023 08.
Article in English | MEDLINE | ID: mdl-37391162

ABSTRACT

In response to tissue injury, changes in the extracellular matrix (ECM) can directly affect the inflammatory response and contribute to disease progression or resolution. During inflammation, the glycosaminoglycan hyaluronan (HA) becomes modified by tumor necrosis factor stimulated gene-6 (TSG6). TSG6 covalently transfers heavy chain (HC) proteins from inter-α-trypsin inhibitor (IαI) to HA in a transesterification reaction and is to date is the only known HC-transferase. By modifying the HA matrix, TSG6 generates HC:HA complexes that are implicated in mediating both protective and pathological responses. Inflammatory bowel disease (IBD) is a lifelong chronic disorder with well-described remodeling of the ECM and increased mononuclear leukocyte influx into the intestinal mucosa. Deposition of HC:HA matrices is an early event in inflamed gut tissue that precedes and promotes leukocyte infiltration. However, the mechanisms by which TSG6 contributes to intestinal inflammation are not well understood. The aim of our study was to understand how the TSG6 and its enzymatic activity contributes to the inflammatory response in colitis. Our findings indicate that inflamed tissues of IBD patients show an elevated level of TSG6 and increased HC deposition and that levels of HA strongly associate with TSG6 levels in patient colon tissue specimens. Additionally, we observed that mice lacking TSG6 are more vulnerable to acute colitis and exhibit an aggravated macrophage-associated mucosal immune response characterized by elevated pro-inflammatory cytokines and chemokines and diminished anti-inflammatory mediators including IL-10. Surprisingly, along with significantly increased levels of inflammation in the absence of TSG6, tissue HA levels in mice were found to be significantly reduced and disorganized, absent of typical "HA-cable" structures. Inhibition of TSG6 HC-transferase activity leads to a loss of cell surface HA and leukocyte adhesion, indicating that the enzymatic functions of TSG6 are a major contributor to stability of the HA ECM during inflammation. Finally, using biochemically generated HC:HA matrices derived by TSG6, we show that HC:HA complexes can attenuate the inflammatory response of activated monocytes. In conclusion, our data suggests that TSG6 exerts a tissue-protective, anti-inflammatory effect via the generation of HC:HA complexes that become dysregulated in IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Mice , Cell Adhesion , Colitis/chemically induced , Colitis/genetics , Hyaluronic Acid/metabolism , Inflammation/genetics , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism
7.
Biochim Biophys Acta Mol Basis Dis ; 1869(5): 166683, 2023 06.
Article in English | MEDLINE | ID: mdl-36878303

ABSTRACT

Among the numerous adhesion G protein-coupled receptors (GPCRs), adhesion G protein-coupled estrogen receptor F5 (ADGRF5) contains unique domains in the long N-terminal tail which can determine cell-cell and cell-matrix interaction as well as cell adhesion. Nevertheless, the biology of ADGRF5 is complex and still poorly explored. Accumulating evidence suggests that the ADGRF5 activity is fundamental in health and disease. For instance, ADGRF5 is essential in the proper function of lungs and kidney as well as the endocrine system, and its signification in vascularization and tumorigenesis has been demonstrated. The most recent studies have provided findings about the diagnostic potential of ADGRF5 in osteoporosis and cancers, and ongoing studies suggest other diseases as well. Here, we elaborate on the current state of knowledge about the ADGRF5 in the physiology and pathophysiology of human diseases and highlight its high potential as a novel target in various therapeutic areas.


Subject(s)
Clinical Relevance , Neoplasms , Humans , Cell Adhesion , Lung/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
8.
Br J Cancer ; 128(4): 537-548, 2023 02.
Article in English | MEDLINE | ID: mdl-36482184

ABSTRACT

BACKGROUND: Increases in IL-6 by cancer-associated fibroblasts (CAFs) contribute to colon cancer progression, but the mechanisms involved in the increase of this tumor-promoting cytokine are unknown. The aim of this study was to identify novel targets involved in the dysregulation of IL-6 expression by CAFs in colon cancer. METHODS: Colonic normal (N), hyperplastic, tubular adenoma, adenocarcinoma tissues, and tissue-derived myo-/fibroblasts (MFs) were used in these studies. RESULTS: Transcriptomic analysis demonstrated a striking decrease in alcohol dehydrogenase 1B (ADH1B) expression, a gene potentially involved in IL-6 dysregulation in CAFs. ADH1B expression was downregulated in approximately 50% of studied tubular adenomas and all T1-4 colon tumors, but not in hyperplastic polyps. ADH1B metabolizes alcohols, including retinol (RO), and is involved in the generation of all-trans retinoic acid (atRA). LPS-induced IL-6 production was inhibited by either RO or its byproduct atRA in N-MFs, but only atRA was effective in CAFs. Silencing ADH1B in N-MFs significantly upregulated LPS-induced IL-6 similar to those observed in CAFs and lead to the loss of RO inhibitory effect on inducible IL-6 expression. CONCLUSION: Our data identify ADH1B as a novel potential mesenchymal tumor suppressor, which plays a critical role in ADH1B/retinoid-mediated regulation of tumor-promoting IL-6.


Subject(s)
Cancer-Associated Fibroblasts , Colonic Neoplasms , Interleukin-6 , Humans , Alcohol Dehydrogenase , Cancer-Associated Fibroblasts/metabolism , Colonic Neoplasms/pathology , Fibroblasts/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Tretinoin , Vitamin A/metabolism
9.
Br J Cancer ; 128(2): 387-397, 2023 01.
Article in English | MEDLINE | ID: mdl-36376448

ABSTRACT

BACKGROUND: Immunotherapy of gastrointestinal cancers is challenging; however, several lines of evidence suggest that adoptive transfer of stimulated or modified immune cells support not only protective role of immune cells in tumor microenvironment, but actively participate in the elimination of cancer cells. METHODS: In vivo studies employing cancer cell-derived allograft murine models of gastrointestinal cancers were performed. The effects of T helper (Th) 2 cells on gastrointestinal cancers growth and tumor microenvironment composition using adoptive transfer of Th2 cells, interleukin (IL)-5 treatment, and immunofluorescence, multiplex and real-time PCR were explored. RESULTS: Here, we show that Th2 cells play an essential role in the inhibition of colon and pancreas cancers progression. In murine models of gastrointestinal tumors using adoptive transfer of Th2 cells, we identify that Th2 cells are responsible for generation of apoptotic factors and affect macrophage as well as eosinophil recruitment into tumors where they produce cytotoxic factors. Moreover, we found that Th2 cells lead to IL-5 hypersecretion, which links the anti-tumorigenic function of Th2 cells and eosinophils. Importantly, we noted that recombinant IL-5 administration is also related with inhibition of gastrointestinal tumor growth. Finally, using an in vitro approach, we documented that both Th2 cells and eosinophils are directly responsible for gastrointestinal cancer cell killing. CONCLUSIONS: These data demonstrate the significance of Th2 cells, eosinophils and IL-5 in the inhibition of gastrointestinal tumor growth, and pointed toward tumor microenvironment reprogramming as a Th2 cell-mediated anti-tumorigenic mechanism of action.


Subject(s)
Pancreatic Neoplasms , Th2 Cells , Humans , Mice , Animals , Eosinophils , Interleukin-5/pharmacology , Colon , Macrophages , Th1 Cells , Cytokines/pharmacology , Tumor Microenvironment
10.
Pharmaceuticals (Basel) ; 15(12)2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36558958

ABSTRACT

Crohn's Disease (CD) and Ulcerative Colitis (UC) are the two major forms of inflammatory bowel disease (IBD), which are incurable chronic immune-mediated diseases of the gastrointestinal tract. Both diseases present with chronic inflammation that leads to epithelial barrier dysfunction accompanied by loss of immune tolerance and inflammatory damage to the mucosa of the GI tract. Despite extensive research in the field, some of the mechanisms associated with the pathology in IBD remain elusive. Here, we identified a mechanism by which the MAPK-activated protein kinase 2 (MK2) pathway contributes to disease pathology in CD by regulating the expression of matrix metalloproteinases (MMPs), which cleave checkpoint molecules on immune cells and enhance T cell activity. By utilizing pharmaceuticals targeting MMPs and MK2, we show that the cleavage of checkpoint molecules and enhanced T cell responses may be reduced. The data presented here suggest the potential for MK2 inhibitors as a therapeutic approach for the treatment of CD.

11.
Int J Mol Sci ; 23(21)2022 Nov 05.
Article in English | MEDLINE | ID: mdl-36362348

ABSTRACT

Cases of pancreatic neuroendocrine tumors (PNETs) are growing in number, and new treatment options are needed in order to improve patient outcomes. The mitogen-activated protein kinase-activated protein kinase 2 (MK2) is a crucial regulator of cytokine/chemokine production. The significance of MK2 expression and signaling pathway mediated by MK2 in PNETs has not been investigated. To characterize the impact of MK2 on PNET growth, we used the RipTag2 transgenic murine model of PNETs, and we developed a primary PNET cell line for both in vitro and in vivo studies. In the transgenic murine model of PNETs, we found that MK2 inhibition improves survival of mice and prevents PNET progression. MK2 blockade abolished cytokine/chemokine production, which was related to macrophage function. A role for MK2 in the regulation of metabolic factor secretion in PNETs was identified, making this the first study to identify a potential role for the MK2 pathway in regulation of tumor metabolism. Moreover, using an in vitro approach and allograft model of PNETs, we were able to show that macrophages with MK2 depletion exhibit increased cytotoxicity against PNET cells and substantially decreased production of pro-inflammatory cytokines and chemokines, as well as metabolic factors. Taken together, our work identifies MK2 as a potent driver of immune response and metabolic effectors in PNETs, suggesting it is a potential therapeutic target for patients with PNETs.


Subject(s)
Neuroectodermal Tumors, Primitive , Neuroendocrine Tumors , Pancreatic Neoplasms , Animals , Mice , Neuroendocrine Tumors/metabolism , Disease Models, Animal , Macrophages/metabolism , Cytokines/metabolism , Chemokines/metabolism , Pancreatic Neoplasms/metabolism , Neuroectodermal Tumors, Primitive/metabolism
12.
Biochim Biophys Acta Rev Cancer ; 1876(2): 188640, 2021 12.
Article in English | MEDLINE | ID: mdl-34695532

ABSTRACT

Pancreatic neuroendocrine tumors are rare types of pancreatic cancer formed from islet cells of pancreas. Clinical presentation of pancreatic neuroendocrine tumors depends on both tumor progression and hormone secretion status, which generate several complications in both diagnosis and treatment. Despite numerous strategies, treatment of patients with pancreatic neuroendocrine tumors still needs improvement. It is suggested that immune response modulation may be essential in the regulation of pancreatic neuroendocrine tumor progression and patient's symptomology. Accumulating evidence indicates that immunotherapy seems to be a promising treatment option for patients with pancreatic neuroendocrine tumors. Nevertheless, several challenges in pre-clinical and clinical studies are present. This review provides knowledge about microenvironment of pancreatic neuroendocrine tumors including significance of cytokine and chemokine as well as specific immune cell types. Additionally, in vitro and in vivo models of pancreatic neuroendocrine tumors and translational challenges are highlighted.


Subject(s)
Immunotherapy/methods , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Cell Line, Tumor , Humans , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Tumor Microenvironment
13.
Analyst ; 146(20): 6124-6131, 2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34543367

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is produced at high levels in several cancers and is directly linked with metastasis in gastrointestinal (GI) cancers. In order to further understand the alteration of molecular compositions and biochemical features triggered by G-CSF treatment at molecular and cell levels, we sought to investigate the long term treatment of G-CSF on colon and breast cancer cells measured by label-free, non-invasive single-cell Raman microspectroscopy. Raman spectrum captures the molecule-specific spectral signatures ("fingerprints") of different biomolecules presented on cells. In this work, mouse breast cancer line 4T1 and mouse colon cancer line CT26 were treated with G-CSF for 7 weeks and subsequently analyzed by machine learning based Raman spectroscopy and gene/cytokine expression. The principal component analysis (PCA) identified the Raman bands that most significantly changed between the control and G-CSF treated cells. Notably, here we proposed the concept of aggressiveness score, which can be derived from the posterior probability of linear discriminant analysis (LDA), for quantitative spectral analysis of tumorigenic cells. The aggressiveness score was effectively applied to analyze and differentiate the overall cell biochemical changes of G-CSF-treated two model cancer cells. All these tumorigenic progressions suggested by Raman analysis were confirmed by pro-tumorigenic cytokine and gene analysis. A high correlation between gene expression data and Raman spectra highlights that the machine learning based non-invasive Raman spectroscopy offers emerging and powerful tools to better understand the regulation mechanism of cytokines in the tumor microenvironment that could lead to the discovery of new targets for cancer therapy.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Colon , Female , Granulocyte Colony-Stimulating Factor , Humans , Machine Learning , Mice , Phenotype , Spectrum Analysis, Raman , Tumor Microenvironment
14.
JCI Insight ; 6(17)2021 09 08.
Article in English | MEDLINE | ID: mdl-34314391

ABSTRACT

Vascular injury has emerged as a complication contributing to morbidity in coronavirus disease 2019 (COVID-19). The glycosaminoglycan hyaluronan (HA) is a major component of the glycocalyx, a protective layer of glycoconjugates that lines the vascular lumen and regulates key endothelial cell functions. During critical illness, as in the case of sepsis, enzymes degrade the glycocalyx, releasing fragments with pathologic activities into circulation and thereby exacerbating disease. Here, we analyzed levels of circulating glycosaminoglycans in 46 patients with COVID-19 ranging from moderate to severe clinical severity and measured activities of corresponding degradative enzymes. This report provides evidence that the glycocalyx becomes significantly damaged in patients with COVID-19 and corresponds with severity of disease. Circulating HA fragments and hyaluronidase, 2 signatures of glycocalyx injury, strongly associate with sequential organ failure assessment scores and with increased inflammatory cytokine levels in patients with COVID-19. Pulmonary microvascular endothelial cells exposed to COVID-19 milieu show dysregulated HA biosynthesis and degradation, leading to production of pathological HA fragments that are released into circulation. Finally, we show that HA fragments present at high levels in COVID-19 patient plasma can directly induce endothelial barrier dysfunction in a ROCK- and CD44-dependent manner, indicating a role for HA in the vascular pathology of COVID-19.


Subject(s)
COVID-19/metabolism , Endothelium, Vascular/metabolism , Hyaluronic Acid/metabolism , Aged , COVID-19/blood , COVID-19/pathology , Cytokines/blood , Endothelium, Vascular/pathology , Female , Glycocalyx/metabolism , Glycocalyx/pathology , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/blood , Hyaluronoglucosaminidase/blood , Hyaluronoglucosaminidase/metabolism , Male , Middle Aged , rho-Associated Kinases/metabolism
15.
Cytokine ; 142: 155479, 2021 06.
Article in English | MEDLINE | ID: mdl-33677228

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is a cytokine most well-known for maturation and mobilization of bone marrow neutrophils. Although it is used therapeutically to treat chemotherapy induced neutropenia, it is also highly expressed in some tumors. Case reports suggest that tumors expressing high levels of G-CSF are aggressive, more difficult to treat, and present with poor prognosis and high mortality rates. Research on this topic suggests that G-CSF has tumor-promoting effects on both tumor cells and the tumor microenvironment. G-CSF has a direct effect on tumor cells to promote tumor stem cell longevity and overall tumor cell proliferation and migration. Additionally, it may promote pro-tumorigenic immune cell phenotypes such as M2 macrophages, myeloid-derived suppressor cells, and regulatory T cells. Overall, the literature suggests a plethora of pro-tumorigenic activity that should be balanced with the therapeutic use. In this review, we present an overview of the multiple complex roles of G-CSF and G-CSFR in tumors and their microenvironment and discuss how clinical advances and strategies may open new therapeutic avenues.


Subject(s)
Granulocyte Colony-Stimulating Factor/metabolism , Leukocytes/metabolism , Neoplasms/immunology , Neoplasms/pathology , Tumor Microenvironment/immunology , Animals , Humans , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
16.
J Crohns Colitis ; 15(8): 1362-1375, 2021 Aug 02.
Article in English | MEDLINE | ID: mdl-33506258

ABSTRACT

BACKGROUND AND AIMS: Little is known about the presence and function of tissue-resident mesenchymal stem cells [MtSCs] within the gastrointestinal mucosa in health and inflammatory bowel disease [IBD]. The contribution of MtSCs to the generation of inflammatory fibroblasts during IBD is also poorly understood. We hypothesized that IBD-MtSCs are impaired and contribute to the generation of the pathological myofibroblasts in IBD. METHODS: In a cohort of clinically and endoscopically active IBD patients and normal controls, we used quantitative RT-PCR and stem cell differentiation assays, as well as confocal microscopy, to characterize MtSCs. RESULTS: Expression of two stem cell markers, Oct4 and ALDH1A, was increased in the inflamed IBD colonic mucosa and correlated with an increase of the mesenchymal lineage marker Grem1 in ulcerative colitis [UC], but not Crohn's disease [CD]. Increased proliferation and aberrant differentiation of Oct4+Grem1+ MtSC-like cells was observed in UC, but not in CD colonic mucosa. In contrast to normal and UC-derived MtSCs, CD-MtSCs lose their clonogenic and most of their differentiation capacities. Our data also suggest that severe damage to these cells in CD may account for the pathological PD-L1low phenotype of CD myofibroblasts. In contrast, aberrant differentiation of MtSCs appears to be involved in the appearance of pathological partially differentiated PD-L1high myofibroblasts within the inflammed colonic mucosa in UC. CONCLUSION: Our data show, for the first time, that the progenitor functions of MtSCs are differentially impaired in CD vs UC, providing a scientific rationale for the use of allogeneic MSC therapy in IBD, and particularly in CD.


Subject(s)
Colitis, Ulcerative/pathology , Crohn Disease/pathology , Mesenchymal Stem Cells/pathology , Adolescent , Adult , Aldehyde Dehydrogenase 1 Family/metabolism , Case-Control Studies , Cell Differentiation , Cohort Studies , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Mucosa/pathology , Male , Microscopy, Confocal , Myofibroblasts/pathology , Octamer Transcription Factor-3/metabolism , Real-Time Polymerase Chain Reaction , Retinal Dehydrogenase/metabolism , Young Adult
17.
J Leukoc Biol ; 109(1): 55-66, 2021 01.
Article in English | MEDLINE | ID: mdl-32930456

ABSTRACT

COVID-19 rapidly emerged as a crippling public health crisis in the last few months, which has presented a series health risk. Understanding of the immune response and biomarker analysis is needed to progress toward understanding disease pathology and developing improved treatment options. The goal of this study is to identify pathogenic factors that are linked to disease severity and patient characteristics. Patients with COVID-19 who were hospitalized from March 17 to June 5, 2020 were analyzed for clinical features of disease and soluble plasma cytokines in association with disease severity and sex. Data from COVID-19 patients with acute illness were examined along with an age- and gender-matched control cohort. We identified a group of 16 soluble factors that were found to be increased in COVID-19 patients compared to controls, whereas 2 factors were decreased. In addition to inflammatory cytokines, we found significant increases in factors known to mediate vasculitis and vascular remodeling (PDGF-AA, PDGF-AB-BB, soluble CD40L (sCD40L), FGF, and IP10). Four factors such as platelet-derived growth factors, fibroblast growth factor-2, and IFN-γ-inducible protein 10 were strongly associated with severe disease and ICU admission. Th2-related factors (IL-4 and IL-13) were increased with IL-4 and sCD40L present at increased levels in males compared with females. Our analysis revealed networking clusters of cytokines and growth factors, including previously unknown roles of vascular and stromal remodeling, activation of the innate immunity, as well activation of type 2 immune responses in the immunopathogenesis of COVID-19. These data highlight biomarker associations with disease severity and sex in COVID-19 patients.


Subject(s)
Blood Platelets , COVID-19 , Cytokine Release Syndrome , Cytokines , Immunity, Innate , SARS-CoV-2 , Sex Characteristics , Adult , Aged , Biomarkers/blood , Blood Platelets/immunology , Blood Platelets/metabolism , COVID-19/blood , COVID-19/complications , COVID-19/immunology , Cytokine Release Syndrome/blood , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Cytokines/blood , Cytokines/immunology , Female , Humans , Male , Middle Aged , Prospective Studies , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Severity of Illness Index , Th2 Cells/immunology
18.
Int J Mol Sci ; 21(23)2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33271941

ABSTRACT

The gastrointestinal (GI) mucosa is among the most complex systems in the body. It has a diverse commensal microbiome challenged continuously by food and microbial components while delivering essential nutrients and defending against pathogens. For these reasons, regulatory cells and receptors are likely to play a central role in maintaining the gut mucosal homeostasis. Recent lessons from cancer immunotherapy point out the critical role of the B7 negative co-stimulator PD-L1 in mucosal homeostasis. In this review, we summarize the current knowledge supporting the critical role of PD-L1 in gastrointestinal mucosal tolerance and how abnormalities in its expression and signaling contribute to gut inflammation and cancers. Abnormal expression of PD-L1 and/or the PD-1/PD-L1 signaling pathways have been observed in the pathology of the GI tract. We also discuss the current gap in our knowledge with regards to PD-L1 signaling in the GI tract under homeostasis and pathology. Finally, we summarize the current understanding of how this pathway is currently targeted to develop novel therapeutic approaches.


Subject(s)
B7-H1 Antigen/metabolism , Immune Tolerance , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Biomarkers , Disease Progression , Disease Susceptibility , Fibrosis , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology , Homeostasis , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immune Tolerance/genetics , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/therapy , Molecular Targeted Therapy
19.
Cancers (Basel) ; 12(10)2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33036138

ABSTRACT

Tumor-associated macrophages (TAMs) in the gastrointestinal tumor microenvironment (TME) are known to polarize into populations exhibiting pro- or anti-tumoral activity in response to stimuli such as growth factors and cytokines. Our previous work has recognized granulocyte colony-stimulating factor (G-CSF) as a cytokine capable of influencing immune cells of the TME exhibiting pro-tumoral activity. Here, we aimed to focus on how G-CSF regulates TAM phenotype and function and the effects on gastrointestinal (GI) tumor progression. Thus, wildtype (WT) and G-CSFR-/- macrophages were examined for cytokine production, gene expression, and transcription factor activity. Adoptive transfer of WT or G-CSFR-/- macrophages into tumor-bearing mice was performed to study their influence in the progression of colon (MC38) and pancreatic (PK5L1940) tumor mouse models. Finally, the difference in cytotoxic potential between WT and G-CSFR-/- macrophages was examined both in vitro and in vivo. Our results indicate that G-CSF promotes increased IL-10 production and decreased IL-12 production, which was reversed in G-CSFR-/- macrophages for a pro-inflammatory phenotype. Furthermore, G-CSFR-/- macrophages were characterized by higher levels of NOS2 expression and NO production, which led to greater tumor related cytotoxicity both in vitro and in vivo. Our results suggest that in the absence of G-CSFR, macrophage-related tumor cytotoxicity was amplified. These findings, along with our previous reports, pinpoint G-CSF /G-CSFR as a prominent target for possible clinical applications that aim to control the TME and the GI tumor progression.

20.
Front Immunol ; 11: 1885, 2020.
Article in English | MEDLINE | ID: mdl-33042110

ABSTRACT

Cytokines are known to shape the tumor microenvironment and although progress has been made in understanding their role in carcinogenesis, much remains to learn regarding their role in tumor growth and progression. We have identified granulocyte colony-stimulating factor (G-CSF) as one such cytokine, showing that G-CSF is linked with metastasis in human gastrointestinal tumors and neutralizing G-CSF in a mouse model of colitis-associated cancer is protective. Here, we set out to identify the role of G-CSF and its receptor, G-CSFR, in CD4+ and CD8+ T cell responses in the tumor microenvironment. MC38 colon cancer cells were injected into WT, G-CSFR-/- mice, or Rag2-/- mice. Flow cytometry, Real Time PCR and Multiplex cytokine array analysis were used for in vitro T cell phenotype analysis. Adoptive transfer of WT or G-CSFR-/- CD4+ of CD8+ T cells were performed. Mouse tumor size, cytokine expression, T cell phenotype, and cytotoxic activity were analyzed. We established that in G-CSFR-/- mice, tumor growth of MC38 colon cancer cells is significantly decreased. T cell phenotype and cytokine production were also altered, as both in vitro and in vivo approaches revealed that the G-CSF/G-CSFR stimulate IL-10-producing, FoxP3-expressing CD4+ and CD8+ T cells, whereas G-CSFR-/- T cells exhibit increased IFNγ and IL-17A production, leading to increased cytotoxic activity in the tumor microenvironment. Furthermore, peritumoral injection of recombinant IFNγ or IL-17A inhibited colon and pancreas tumor growth compared to controls. Taken together, our data reveal an unknown mechanism by which G-CSF, through its receptor G-CSFR, promotes an inhibitory Treg phenotype that limits tumor immune responses and furthermore suggest that targeting this cytokine/receptor axis could represent a novel therapeutic approach for gastrointestinal, and likely other tumors with high expression of these factors.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Colonic Neoplasms/metabolism , Cytotoxicity, Immunologic , Granulocyte Colony-Stimulating Factor/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Receptors, Colony-Stimulating Factor/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Coculture Techniques , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-17/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Colony-Stimulating Factor/deficiency , Receptors, Colony-Stimulating Factor/genetics , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Tumor Burden , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...