Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Immunol ; 212(4): 505-512, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38315950

ABSTRACT

As COVID-19 continues, an increasing number of patients develop long COVID symptoms varying in severity that last for weeks, months, or longer. Symptoms commonly include lingering loss of smell and taste, hearing loss, extreme fatigue, and "brain fog." Still, persistent cardiovascular and respiratory problems, muscle weakness, and neurologic issues have also been documented. A major problem is the lack of clear guidelines for diagnosing long COVID. Although some studies suggest that long COVID is due to prolonged inflammation after SARS-CoV-2 infection, the underlying mechanisms remain unclear. The broad range of COVID-19's bodily effects and responses after initial viral infection are also poorly understood. This workshop brought together multidisciplinary experts to showcase and discuss the latest research on long COVID and chronic inflammation that might be associated with the persistent sequelae following COVID-19 infection.


Subject(s)
COVID-19 , Post-Acute COVID-19 Syndrome , Humans , SARS-CoV-2 , Inflammation , Disease Progression
2.
Cancer Metastasis Rev ; 41(2): 383-404, 2022 06.
Article in English | MEDLINE | ID: mdl-35366154

ABSTRACT

Cancer, especially when it has metastasized to different locations in the body, is notoriously difficult to treat. Metastatic cancer accounts for most cancer deaths and thus remains an enormous challenge. During the metastasis process, cancer cells negotiate a series of steps termed the "metastatic cascadeË® that offer potential for developing anti-metastatic therapy strategies. Currently available conventional treatment and diagnostic methods addressing metastasis come with their own pitfalls and roadblocks. In this contribution, we comprehensively discuss the potential improvements that nanotechnology-aided approaches are able to bring, either alone or in combination with the existing conventional techniques, to the identification and treatment of metastatic disease. We tie specific nanotechnology-aided strategies to the complex biology of the different steps of the metastatic cascade in order to open up new avenues for fine-tuned targeting and development of anti-metastatic agents designed specifically to prevent or mitigate the metastatic outgrowth of cancer. We also present a viewpoint on the progress of translation of nanotechnology into cancer metastasis patient care.


Subject(s)
Nanotechnology , Neoplasm Metastasis , Humans , Nanotechnology/methods , Neoplasm Metastasis/prevention & control , Neoplasms/pathology
3.
Cancer Metastasis Rev ; 39(4): 1223-1243, 2020 12.
Article in English | MEDLINE | ID: mdl-32780245

ABSTRACT

Pancreatic cancer is the third leading cause of cancer death in the USA, and pancreatic ductal adenocarcinoma (PDA) constitutes 85% of pancreatic cancer diagnoses. PDA frequently metastasizes to the peritoneum, but effective treatment of peritoneal metastasis remains a clinical challenge. Despite this unmet need, understanding of the biological mechanisms that contribute to development and progression of PDA peritoneal metastasis is sparse. By contrast, a vast number of studies have investigated mechanisms of peritoneal metastasis in ovarian and gastric cancers. Here, we contrast similarities and differences between peritoneal metastasis in PDA as compared with those in gastric and ovarian cancer by outlining molecular mediators involved in each step of the peritoneal metastasis cascade. This review aims to provide mechanistic insights that could be translated into effective targeted therapies for patients with peritoneal metastasis from PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Peritoneal Neoplasms/secondary , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/therapy , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/therapy
4.
Mol Cancer Res ; 18(2): 229-239, 2020 02.
Article in English | MEDLINE | ID: mdl-31676721

ABSTRACT

Over 90% of pancreatic ductal adenocarcinomas (PDAC) express mesothelin (MSLN). Overexpression or knockdown of MSLN has been implicated in PDAC aggressiveness. This activity has been ascribed to MSLN-induced activation of MAPK or NF-κB signaling pathways and to interaction of MSLN with its only known binding partner, MUC16. Here, we used CRISPR/Cas9 gene editing to delete MSLN from PDAC, then restored expression of wild-type (WT) or Y318A mutant MSLN by viral transduction. We found that MSLN KO cells grew in culture and as subcutaneous tumors in mouse xenografts at the same rate as WT cells but formed intraperitoneal metastases poorly. Complementation with WT MSLN restored intraperitoneal growth, whereas complementation with Y318A mutant MSLN, which does not bind MUC16, was ineffective at enhancing growth in both MUC16(+) and MUC16(-) models. Restoration of WT MSLN did enhance growth but did not affect cell-to-cell binding, cell viability in suspension or signaling pathways previously identified as contributing to the protumorigenic effect of MSLN. RNA deep sequencing of tumor cells identified no changes in transcriptional profile that could explain the observed phenotype. Furthermore, no histologic changes in tumor cell proliferation or morphology were observed in mature tumors. Examination of nascent MSLN KO tumors revealed decreased microvascular density as intraperitoneal tumors were forming, followed by decreased proliferation, which resolved by 2 weeks postimplantation. These data support a model whereby MSLN expression by tumor cells contributes to metastatic colonization. IMPLICATIONS: MSLN confers a growth advantage to tumor cells during colonization of peritoneal metastasis. Therapeutic blockade of MSLN might limit peritoneal spread.


Subject(s)
Antigens, Neoplasm/therapeutic use , Carcinoma, Pancreatic Ductal/complications , GPI-Linked Proteins/therapeutic use , Peritoneal Neoplasms/secondary , Animals , Antigens, Neoplasm/pharmacology , Cell Line, Tumor , Female , GPI-Linked Proteins/pharmacology , Humans , Mesothelin , Mice , Mice, Nude , Neoplasm Metastasis
5.
Cell Mol Gastroenterol Hepatol ; 5(4): 591-609, 2018.
Article in English | MEDLINE | ID: mdl-29930980

ABSTRACT

BACKGROUND & AIMS: Human enteroids present a novel tool to study human intestinal ion transport physiology and pathophysiology. The present study describes the contributions of Cl- and HCO3- secretion to total cyclic adenosine monophosphate (cAMP)-stimulated electrogenic anion secretion in human duodenal enteroid monolayers and the relevant changes after differentiation. METHODS: Human duodenal enteroids derived from 4 donors were grown as monolayers and differentiated by a protocol that includes the removal of Wnt3A, R-spondin1, and SB202190 for 5 days. The messenger RNA level and protein expression of selected ion transporters and carbonic anhydrase isoforms were determined by quantitative real-time polymerase chain reaction and immunoblotting, respectively. Undifferentiated and differentiated enteroid monolayers were mounted in the Ussing chamber/voltage-current clamp apparatus, using solutions that contained as well as lacked Cl- and HCO3-/CO2, to determine the magnitude of forskolin-induced short-circuit current change and its sensitivity to specific inhibitors that target selected ion transporters and carbonic anhydrase(s). RESULTS: Differentiation resulted in a significant reduction in the messenger RNA level and protein expression of cystic fibrosis transmembrane conductance regulator, (CFTR) Na+/K+/2Cl- co-transporter 1 (NKCC1), and potassium channel, voltage gated, subfamily E, regulatory subunit 3 (KCNE3); and, conversely, increase of down-regulated-in-adenoma (DRA), electrogenic Na+/HCO3- co-transporter 1 (NBCe1), carbonic anhydrase 2 (CA2), and carbonic anhydrase 4 (CA4). Both undifferentiated and differentiated enteroids showed active cAMP-stimulated anion secretion that included both Cl- and HCO3- secretion as the magnitude of total active anion secretion was reduced after the removal of extracellular Cl- or HCO3-/CO2. The magnitude of total anion secretion in differentiated enteroids was approximately 33% of that in undifferentiated enteroids, primarily owing to the reduction in Cl- secretion with no significant change in HCO3- secretion. Anion secretion was consistently lower but detectable in differentiated enteroids compared with undifferentiated enteroids in the absence of extracellular Cl- or HCO3-/CO2. Inhibiting CFTR, NKCC1, carbonic anhydrase(s), cAMP-activated K+ channel(s), and Na+/K+-adenosine triphosphatase reduced cAMP-stimulated anion secretion in both undifferentiated and differentiated enteroids. CONCLUSIONS: Human enteroids recapitulate anion secretion physiology of small intestinal epithelium. Enteroid differentiation is associated with significant alterations in the expression of several ion transporters and carbonic anhydrase isoforms, leading to a reduced but preserved anion secretory phenotype owing to markedly reduced Cl- secretion but no significant change in HCO3- secretion.

6.
Am J Physiol Gastrointest Liver Physiol ; 314(1): G81-G90, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28882822

ABSTRACT

The intestinal epithelial brush border Na+/H+ exchanger NHE3 accounts for a large component of intestinal Na absorption. NHE3 is regulated during digestion by signaling complexes on its COOH terminus that include the four multi-PDZ domain-containing NHERF family proteins. All bind to NHE3 and take part in different aspects of NHE3 regulation. Because the roles of each NHERF appear to vary on the basis of the cell model or intestinal segment studied and because of our recent finding that a NHERF3-NHERF2 heterodimer appears important for NHE3 regulation in Caco-2 cells, we examined the role of NHERF3 and NHERF2 in C57BL/6 mouse jejunum using homozygous NHERF2 and NHERF3 knockout mice. NHE3 activity was determined with two-photon microscopy and the dual-emission pH-sensitive dye SNARF-4F. The jejunal apical membrane of NHERF3-null mice appeared similar to wild-type (WT) mice in surface area, microvillus number, and height, which is similar to results previously reported for jejunum of NHERF2-null mice. NHE3 basal activity was not different from WT in either NHERF2- or NHERF3-null jejunum, while d-glucose-stimulated NHE3 activity was reduced in NHERF2, but similar to WT in NHERF3 KO. LPA stimulation and UTP (elevated Ca2+) and cGMP inhibition of NHE3 were markedly reduced in both NHERF2- and NHERF3-null jejunum. Forskolin inhibited NHE3 in NHERF3-null jejunum, but the extent of inhibition was reduced compared with WT. The forskolin inhibition of NHE3 in NHERF2-null mice was too inconsistent to determine whether there was an effect and whether it was altered compared with the WT response. These results demonstrate similar requirement for NHERF2 and NHERF3 in mouse jejunal NHE3 regulation by LPA, Ca2+, and cGMP. The explanation for the similarity is not known but is consistent with involvement of a brush-border NHERF3-NHERF2 heterodimer or sequential NHERF-dependent effects in these aspects of NHE3 regulation. NEW & NOTEWORTHY NHERF2 and NHERF3 are apical membrane multi-PDZ domain-containing proteins that are involved in regulation of intestinal NHE3. This study demonstrates that NHERF2 and NHERF3 have overlapping roles in NHE3 stimulation by LPA and inhibition by elevated Ca2+ and cGMP. These results are consistent with their role being as a NHERF3-NHERF2 heterodimer or via sequential NHERF-dependent signaling steps, and they begin to clarify a role for multiple NHERF proteins in NHE3 regulation.


Subject(s)
Calcium/metabolism , Cyclic GMP/analogs & derivatives , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Jejunum/drug effects , Jejunum/metabolism , Lysophospholipids/pharmacology , Phosphoproteins/metabolism , Sodium-Hydrogen Exchanger 3/metabolism , Sodium-Hydrogen Exchangers/metabolism , Thionucleotides/pharmacology , Animals , Calcium Signaling , Cyclic GMP/pharmacology , Female , Genotype , Glucose/pharmacology , Intestinal Mucosa/ultrastructure , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Jejunum/ultrastructure , Male , Membrane Proteins , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microvilli/drug effects , Microvilli/metabolism , Phenotype , Phosphoproteins/deficiency , Phosphoproteins/genetics , Sodium-Hydrogen Exchanger 3/genetics , Sodium-Hydrogen Exchangers/deficiency , Sodium-Hydrogen Exchangers/genetics , Uridine Triphosphate/pharmacology
7.
Mol Biol Cell ; 26(11): 2030-43, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25851603

ABSTRACT

Sorting nexin 27 (SNX27) contains a PDZ domain that is phylogenetically related to the PDZ domains of the NHERF proteins. Studies on nonepithelial cells have shown that this protein is located in endosomes, where it regulates trafficking of cargo proteins in a PDZ domain-dependent manner. However, the role of SNX27 in trafficking of cargo proteins in epithelial cells has not been adequately explored. Here we show that SNX27 directly interacts with NHE3 (C-terminus) primarily through the SNX27 PDZ domain. A combination of knockdown and reconstitution experiments with wild type and a PDZ domain mutant (GYGF → GAGA) of SNX27 demonstrate that the PDZ domain of SNX27 is required to maintain basal NHE3 activity and surface expression of NHE3 in polarized epithelial cells. Biotinylation-based recycling and degradation studies in intestinal epithelial cells show that SNX27 is required for the exocytosis (not endocytosis) of NHE3 from early endosome to plasma membrane. SNX27 is also required to regulate the retention of NHE3 on the plasma membrane. The findings of the present study extend our understanding of PDZ-mediated recycling of cargo proteins from endosome to plasma membrane in epithelial cells.


Subject(s)
Epithelial Cells/metabolism , Microvilli/metabolism , Nerve Tissue Proteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Exocytosis , Humans , PDZ Domains , Protein Transport , Sodium-Hydrogen Exchanger 3
8.
Cell Tissue Res ; 359(2): 453-463, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25358399

ABSTRACT

Corticotropin-releasing factor (CRF) and urocortins (UCNs) are important ligands in the CRF signaling pathways, which are most known for their role in the hypothalamic-pituitary-adrenal stress axis. However, peripheral CRF signaling also has profound effects on gastrointestinal functions. Although the murine animal model is highly relevant for the exploration of this complexly balanced pathway via genetic manipulation, little is known about the expression of CRF and UCNs in the mouse intestine. This study aims to investigate the cellular localization of CRF and UCNs in the ileum and to explore whether and how this cellular expression is altered in conditions of intestinal Schistosoma mansoni-induced inflammation. The results show a distinct expression pattern for the different CRF receptor ligands in the ileum. CRF was located in nerve fibers and stromal cells. All UCNs were expressed in polymorphonuclear leukocytes. Furthermore, UCN2 and UCN3 were found in the musculature. During acute schistosomiasis, UCN1 showed an increased immunoreactivity in blood vessels and UCN3 was de novo expressed mainly in submucous neurons. Typical features of S. mansoni-inflamed ileum, such as nerve fiber sprouting, muscle layer thickening and granuloma formation thus all have an impact on the CRF signaling pathways. In conclusion, we outline for the first time the expression of CRF signaling ligands in the mouse ileum; our results point to important changes of this signaling system in S. mansoni-induced intestinal inflammation, which warrants further functional investigation with specific focus on CRF2, given the exclusive binding of UCN2 and UCN3 to this receptor.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Ileum/parasitology , Schistosoma mansoni/physiology , Schistosomiasis mansoni/metabolism , Schistosomiasis mansoni/parasitology , Urocortins/metabolism , Animals , Corticotropin-Releasing Hormone/genetics , Ileum/metabolism , Ileum/pathology , Immunohistochemistry , Inflammation/pathology , Leukocytes/metabolism , Ligands , Male , Mice, Inbred C57BL , Muscles/metabolism , Nerve Fibers/metabolism , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/pathology , Stromal Cells/metabolism , Up-Regulation , Urocortins/genetics
9.
Histochem Cell Biol ; 139(5): 639-58, 2013 May.
Article in English | MEDLINE | ID: mdl-23504129

ABSTRACT

Mas-related gene (Mrg) receptors constitute a subfamily of G protein-coupled receptors that are implicated in nociception, and are as such considered potential targets for pain therapies. Furthermore, some Mrgs have been suggested to play roles in the regulation of inflammatory responses to non-immunological activation of mast cells and in mast cell-neuron communication. Except for MrgD, E and F, whose changed expression has been revealed during inflammation in the mouse intestine in our earlier studies, information concerning the remaining cloned mouse Mrg subtypes in the gastrointestinal tract during (patho) physiological conditions is lacking. Therefore, the present study aimed at identifying the presence and putative function of these remaining cloned Mrg subtypes (n = 19) in the (inflamed) mouse intestine. Using reverse transcriptase-PCR, quantitative-PCR and multiple immunofluorescence staining with commercial and newly custom-developed antibodies, we compared the ileum and the related dorsal root ganglia (DRG) of non-inflamed mice with those of two models of intestinal inflammation, i.e., intestinal schistosomiasis and 2,4,6-trinitrobenzene sulfonic acid-induced ileitis. In the non-inflamed ileum and DRG, the majority of the Mrg subtypes examined were sparsely expressed, showing a neuron-specific expression pattern. However, significant changes in the expression patterns of multiple Mrg subtypes were observed in the inflamed ileum; for instance, MrgA4, MrgB2and MrgB8 were expressed in a clearly increased number of enteric sensory neurons and in nerve fibers in the lamina propria, while de novo expression of MrgB10 was observed in enteric sensory neurons and in newly recruited mucosal mast cells (MMCs). The MrgB10 expressing MMCs were found to be in close contact with nerve fibers in the lamina propria. This is the first report on the expression of all cloned Mrg receptor subtypes in the (inflamed) mouse intestine. The observed changes in the expression and cellular localization of the Mrg subtypes suggest that these receptors are involved in the mediation of primary afferent responses, mast cell responses, and in neuroimmune communication during intestinal inflammation.


Subject(s)
Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestines/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Animals , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/analysis , Reverse Transcriptase Polymerase Chain Reaction
10.
BMC Genomics ; 13: 377, 2012 Aug 06.
Article in English | MEDLINE | ID: mdl-22866923

ABSTRACT

BACKGROUND: Although a number of intestinal inflammatory conditions pertain to the ileum, whole-genome gene expression analyses in animal models of ileal inflammation are lacking to date. Therefore, we aimed to identify and characterize alterations in gene expression in the acutely inflamed ileum of two murine models of intestinal inflammation, namely intestinal schistosomiasis and TNBS-induced ileitis, compared to healthy controls. To this end, we used whole-genome microarrays, followed by bioinformatics analyses to detect over-represented Kyoto Encyclopedia of Genes and Genomes pathways and Gene Ontology categories. RESULTS: Following screening of almost all known mouse genes and transcripts represented on the array, intestinal schistosomiasis and TNBS-induced ileitis yielded 207 and 1417 differentially expressed genes, respectively, with only 30 overlapping concordantly changed genes. Functional category groups consisting of complement and coagulation cascades, extracellular matrix (ECM)-receptor interaction, Fc epsilon receptor I signaling pathways and protein activation cascade, cell adhesion categories were over-represented in the differential gene list of intestinal schistosomiasis. Antigen processing and presentation, cell adhesion molecules, ABC transporters, Toll-like receptor signaling pathways and response to chemical stimulus categories were over-represented in the differential gene list of TNBS-induced ileitis. Although cytokine-cytokine receptor interaction, intestinal immune network for IgA production, focal adhesion pathways and immune, inflammatory and defense response categories were over-represented in the differential gene lists of both inflammation models, the vast majority of the associated genes and changes were unique to each model. CONCLUSIONS: This study characterized two models of ileal inflammation at a whole-genome level and outlined distinct gene expression profiles and patterns in the two models. The results indicate that intestinal schistosomiasis involves Th2 responses, complement activation, protein activation and enhanced ECM turnover, while TNBS-induced ileitis involves Th17 responses, defective antigen processing and presentation and altered Toll-like receptor-mediated responses. Signs of an impaired epithelial barrier are apparent in both inflammation models. Furthermore, the comprehensive differential gene list and functional groups provided by this study constitute an interesting starting point to explore new targets and extended functional networks dealing with small bowel inflammation.


Subject(s)
Ileitis , Ilium , Inflammation/genetics , Transcriptome/genetics , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Female , Gene Expression , Gene Expression Profiling , Genome , Ileitis/chemically induced , Ileitis/genetics , Ileitis/immunology , Ileitis/parasitology , Ilium/drug effects , Ilium/immunology , Ilium/parasitology , Inflammation/chemically induced , Inflammation/parasitology , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Receptors, IgE/metabolism , Schistosoma mansoni , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Trinitrobenzenesulfonic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...