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.
Sci Rep ; 13(1): 16920, 2023 10 07.
Article in English | MEDLINE | ID: mdl-37805544

ABSTRACT

M3 muscarinic receptors (M3R) modulate ß-catenin signaling and colon neoplasia. CDC42/RAC guanine nucleotide exchange factor, ßPix, binds to ß-catenin in colon cancer cells, augmenting ß-catenin transcriptional activity. Using in silico, in vitro, and in vivo approaches, we explored whether these actions are regulated by M3R. At the invasive fronts of murine and human colon cancers, we detected co-localized nuclear expression of ßPix and ß-catenin in stem cells overexpressing M3R. Using immunohistochemistry, immunoprecipitation, proximity ligand, and fluorescent cell sorting assays in human tissues and established and primary human colon cancer cell cultures, we detected time-dependent M3R agonist-induced cytoplasmic and nuclear association of ßPix with ß-catenin. ßPix knockdown attenuated M3R agonist-induced human colon cancer cell proliferation, migration, invasion, and expression of PTGS2, the gene encoding cyclooxygenase-2, a key player in colon neoplasia. Overexpressing ßPix dose-dependently augmented ß-catenin binding to the transcription factor TCF4. In a murine model of sporadic colon cancer, advanced neoplasia was attenuated in conditional knockout mice with intestinal epithelial cell deficiency of ßPix. Expression levels of ß-catenin target genes and proteins relevant to colon neoplasia, including c-Myc and Ptgs2, were reduced in colon tumors from ßPix-deficient conditional knockout mice. Targeting the M3R/ßPix/ß-catenin axis may have therapeutic potential.


Subject(s)
Colonic Neoplasms , beta Catenin , Mice , Humans , Animals , beta Catenin/metabolism , Cyclooxygenase 2/metabolism , Colonic Neoplasms/pathology , Rho Guanine Nucleotide Exchange Factors/metabolism , Receptors, Muscarinic/metabolism , Mice, Knockout , Gene Expression Regulation, Neoplastic
2.
AIMS Public Health ; 8(1): 124-136, 2021.
Article in English | MEDLINE | ID: mdl-33575412

ABSTRACT

OBJECTIVES: The COVID-19 pandemic (caused by SARS-CoV-2) has introduced significant challenges for accurate prediction of population morbidity and mortality by traditional variable-based methods of estimation. Challenges to modelling include inadequate viral physiology comprehension and fluctuating definitions of positivity between national-to-international data. This paper proposes that accurate forecasting of COVID-19 caseload may be best preformed non-parametrically, by vector autoregression (VAR) of verifiable data regionally. METHODS: A non-linear VAR model across 7 major demographically representative New York City (NYC) metropolitan region counties was constructed using verifiable daily COVID-19 caseload data March 12-July 23, 2020. Through association of observed case trends with a series of (county-specific) data-driven dynamic interdependencies (lagged values), a systematically non-assumptive approximation of VAR representation for COVID-19 patterns to-date and prospective upcoming trends was produced. RESULTS: Modified VAR regression of NYC area COVID-19 caseload trends proves highly significant modelling capacity of observed patterns in longitudinal disease incidence (county R2 range: 0.9221-0.9751, all p < 0.001). Predictively, VAR regression of daily caseload results at a county-wide level demonstrates considerable short-term forecasting fidelity (p < 0.001 at one-step ahead) with concurrent capacity for longer-term (tested 11-week period) inferences of consistent, reasonable upcoming patterns from latest (model data update) disease epidemiology. CONCLUSIONS: In contrast to macroscopic variable-assumption projections, regionally-founded VAR modelling may substantially improve projection of short-term community disease burden, reduce potential for biostatistical error, as well as better model epidemiological effects resultant from intervention. Predictive VAR extrapolation of existing public health data at an interdependent regional scale may improve accuracy of current pandemic burden prognoses.

3.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G627-G643, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33566751

ABSTRACT

Rho guanine nucleotide exchange factors (RhoGEFs) regulate Rho GTPase activity and cytoskeletal and cell adhesion dynamics. ßPix, a CDC42/RAC family RhoGEF encoded by ARHGEF7, is reported to modulate human colon cancer cell proliferation and postwounding restitution of rat intestinal epithelial monolayers. We hypothesized that ßPix plays a role in maintaining intestinal epithelial homeostasis. To test this hypothesis, we examined ßPix distribution in the human and murine intestine and created mice with intestinal epithelial-selective ßPix deletion [ßPixflox/flox/Tg(villin-Cre); Arhgef7 CKO mice]. Using Arhgef7 conditional knockout (CKO) and control mice, we investigated the consequences of ßPix deficiency in vivo on intestinal epithelial and enteroid development, dextran sodium sulfate-induced mucosal injury, and gut permeability. In normal human and murine intestines, we observed diffuse cytoplasmic and moderate nuclear ßPix immunostaining in enterocytes. Arhgef7 CKO mice were viable and fertile, with normal gross intestinal architecture but reduced small intestinal villus height, villus-to-crypt ratio, and goblet cells; small intestinal crypt cells had reduced Ki67 staining, compatible with impaired cell proliferation. Enteroids derived from control mouse small intestine were viable for more than 20 passages, but those from Arhgef7 CKO mice did not survive beyond 24 h despite addition of Wnt proteins or conditioned media from normal enteroids. Adding a Rho kinase (ROCK) inhibitor partially rescued CKO enteroid development. Compared with littermate control mice, dextran sodium sulfate-treated ßPix-deficient mice lost more weight and had greater impairment of intestinal barrier function, and more severe colonic mucosal injury. These findings reveal ßPix expression is important for enterocyte development, intestinal homeostasis, and resistance to toxic injury.NEW & NOTEWORTHY To explore the role of ßPix, a guanine nucleotide exchange factor encoded by ARHGEF7, in intestinal development and physiology, we created mice with intestinal epithelial cell Arhgef7/ßPix deficiency. We found ßPix essential for normal small intestinal epithelial cell proliferation, villus development, and mucosal resistance to injury. Moreover, Rho kinase signaling mediated developmental arrest observed in enteroids derived from ßPix-deficient small intestinal crypts. Our studies provide insights into the role Arhgef7/ßPix plays in intestinal epithelial homeostasis.


Subject(s)
Cell Proliferation , Colitis/metabolism , Colon/metabolism , Enterocytes/metabolism , Intestinal Mucosa/metabolism , Microvilli/metabolism , Rho Guanine Nucleotide Exchange Factors/deficiency , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Enterocytes/pathology , Female , Gene Deletion , Humans , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Microvilli/pathology , Organoids , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Tissue Culture Techniques , rho-Associated Kinases/metabolism
4.
AME Case Rep ; 4: 21, 2020.
Article in English | MEDLINE | ID: mdl-33178993

ABSTRACT

In this case, a 78-year-old female with no previous medical history of crystalline arthropathy presented with pain, effusion, and erythema about a total knee arthroplasty (TKA) performed 13 years prior. Implementation of a novel synovial fluid alpha-defensin assay ruled out periprosthetic joint infection (PJI) despite a positive 2018 Musculoskeletal Infection Society (MSIS) minor criteria score of 8 points, a significant diagnostic differentiation which prevented secondary invasive debridement or joint irrigation intervention. Confirmatory histologic study was positive for calcium pyrophosphate crystals, indicative of acute pseudogout inflammation rather than PJI or septic arthritis manifestation. The patient was then conservatively managed medically for a pseudogout flare and had no evidence of infection with normal physical exam and laboratory study at one- and two-years post treatment, respectively. Given the predominantly clinical nature of current PJI assessment in-clinic coupled with notable risks associated with aggressive re-intervention in the setting of suspected infection, critical need exists for the maturation of sensitive, reliable empiric measures which may assist in guiding orthopaedic surgeon evaluation of patients presenting with inflammatory symptomology around a previous surgical site. In this case, we conclude that patients with a negative alpha-defensin assay alongside crystalline arthropathy on histology may be cautiously yet successfully treated non-operatively despite clinical MSIS criteria concerning for PJI.

5.
Oncotarget ; 9(39): 25572-25585, 2018 May 22.
Article in English | MEDLINE | ID: mdl-29876009

ABSTRACT

Fibroblast growth factor-19 (human FGF19; murine FGF15) suppresses bile acid synthesis. In FGF19 deficiency, diarrhea resulting from bile acid spillage into the colon mimics irritable bowel syndrome. To seek other consequences of FGF19/15 deficiency, we used Fgf15-/- and wild-type (WT) mice to assess gallbladder filling, the bile acid pool, fecal bile acid levels, and colon neoplasia. We fasted mice for six hours before assessing gallbladder size by magnetic resonance imaging (MRI). We measured bile acid levels in different compartments by enzymatic assay, and induced colon neoplasia with azoxymethane (AOM)/dextran sodium sulfate (DSS) and quantified epithelial Ki67 immunostaining and colon tumors 20 weeks later. In vivo MRI confirmed the gross finding of tubular gallbladders in FGF15-deficient compared to WT mice, but fasting gallbladder volumes overlapped. After gavage with a bile acid analogue, ex vivo MRI revealed diminished gallbladder filling in FGF15-deficient mice (P = 0.0399). In FGF15-deficient mice, the total bile acid pool was expanded 45% (P <0.05) and fecal bile acid levels were increased 2.26-fold (P <0.001). After AOM/DSS treatment, colons from FGF15-deficient mice had more epithelial cell Ki67 staining and tumors (7.33 ± 1.32 vs. 4.57 ± 0.72 tumors/mouse; P = 0.003 compared to WT mice); carcinomas were more common in FGF15-deficient mice (P = 0.01). These findings confirm FGF15, the murine homolog of FGF19, plays a key role in modulating gallbladder filling and bile acid homeostasis. In a well-characterized animal model of colon cancer, increased fecal bile acid levels in FGF15-deficient mice promoted epithelial proliferation and advanced neoplasia.

6.
Article in English | MEDLINE | ID: mdl-31742233

ABSTRACT

AIM: Strong evidence reveals important differences between cancers in the proximal vs. distal colon. Animal models of metastatic colon cancer are available but with varying degrees of reproducibility and several important limitations. We explored whether there were regional differences in the location of murine colon cancers and assessed the utility of murine models to explore the biological basis for such differences. METHODS: We re-analyzed data from our previous studies to assess the regional distribution of murine colon cancer. In survival surgery experiments, we injected HT-29 human colon cancer cells into the wall of the cecum or distal colon of Nu(NCr)-Foxn1nu or NOD.Cg-PrkdcscidIl2rgTim1Wji/SzJ mice and compared the development of primary tumors and metastases. RESULTS: Within 7-17 weeks after intramural cecal injection of HT-29 cells, eight mice failed to develop solid primary tumors or metastases. In contrast, within four weeks after cell injection into the distal colon, 13 mice developed metastases - 12 mice developed subcutaneous metastases; of these, four developed liver metastases and one developed both liver and lung metastases. One mouse developed liver metastases only. Histological examination confirmed these lesions were adenocarcinomas. CONCLUSION: Our findings reveal the preferential growth of murine colon neoplasia and invasive human orthotopic xenografts in the distal mouse colon. The new approach of injecting cells into the distal colon wall results in a pattern of colon cancer development that closely mimics the progression of metastatic colon cancer in humans. This novel model of colon neoplasia has great potential for exploring anatomical differences in colon cancer and testing novel therapeutics.

7.
Oncotarget ; 8(13): 21106-21114, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28416748

ABSTRACT

M3 muscarinic receptor (M3R) activation promotes colon cancer cell proliferation, migration, and invasion in vitro. Although over-expression of CHRM3, the gene encoding M3R, is reported in primary colon cancers, expression of M3R itself has not been studied in colon neoplasia. We compared M3R expression in normal colon to colon adenomas, and primary and metastatic colon cancers. Compared to adjacent normal colon, CHRM3 expression was increased up to 128-fold in 10 of 18 consecutive surgical cancer specimens (56%) and associated with metastatic spread (P < 0.05). To analyze M3R protein expression we interrogated 29 consecutive paraffin-embedded colon adenocarcinomas and adjacent normal colon using a specific anti-M3R antibody and immunoperoxidase staining. This revealed weak M3R expression in normal colonocytes, primarily on basolateral surfaces. In contrast, in 25 of 29 cancer tissues (86%) we observed both cytoplasmic and plasma membrane over-expression of M3R; compared to normal epithelium, mean M3R staining intensity was increased more than two-fold in colon cancer (P < 0.001). M3R staining was also increased in 22 colon adenomas compared to adjacent normal colon (P < 0.001). In contrast, M3R staining intensity was not increased in lymph node or liver metastases. These findings suggest M3R expression plays an important role in early progression and invasion of colon neoplasia but is less important once tumors have spread.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Receptors, Muscarinic/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Adenoma/metabolism , Adenoma/pathology , Cell Membrane/metabolism , Cell Proliferation , Colon/cytology , Colon/metabolism , Humans , Immunohistochemistry , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Lymphatic Metastasis , Matrix Metalloproteinase 1/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Muscarinic M3 , Receptors, Muscarinic/genetics
8.
J Vis Exp ; (117)2016 11 27.
Article in English | MEDLINE | ID: mdl-27929465

ABSTRACT

Along with their traditional role as detergents that facilitate fat absorption, emerging literature indicates that bile acids are potent signaling molecules that affect multiple organs; they modulate gut motility and hormone production, and alter vascular tone, glucose metabolism, lipid metabolism, and energy utilization. Changes in fecal bile acids may alter the gut microbiome and promote colon pathology including cholerrheic diarrhea and colon cancer. Key regulators of fecal bile acid composition are the small intestinal Apical Sodium-dependent Bile Acid Transporter (ASBT) and fibroblast growth factor-19 (FGF19). Reduced expression and function of ASBT decreases intestinal bile acid up-take. Moreover, in vitro data suggest that some FDA-approved drugs inhibit ASBT function. Deficient FGF19 release increases hepatic bile acid synthesis and release into the intestines to levels that overwhelm ASBT. Either ASBT dysfunction or FGF19 deficiency increases fecal bile acids and may cause chronic diarrhea and promote colon neoplasia. Regrettably, tools to measure bile acid malabsorption and the actions of drugs on bile acid transport in vivo are limited. To understand the complex actions of bile acids, techniques are required that permit simultaneous monitoring of bile acids in the gut and metabolic tissues. This led us to conceive an innovative method to measure bile acid transport in live animals using a combination of proton (1H) and fluorine (19F) magnetic resonance imaging (MRI). Novel tracers for fluorine (19F)-based live animal MRI were created and tested, both in vitro and in vivo. Strengths of this approach include the lack of exposure to ionizing radiation and translational potential for clinical research and practice.


Subject(s)
Bile Acids and Salts , Biological Transport , Magnetic Resonance Imaging , Animals , Bile , Fluorine Compounds , Humans , Intestines
9.
Carcinogenesis ; 36(10): 1193-200, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26210740

ABSTRACT

Although epidemiological evidence in humans and bile acid feeding studies in rodents implicate bile acids as tumor promoters, the role of endogenous bile acids in colon carcinogenesis remains unclear. In this study, we exploited mice deficient in the ileal apical sodium-dependent bile acid transporter (ASBT, encoded by SLC10A2) in whom fecal bile acid excretion is augmented more than 10-fold. Wild-type and Asbt-deficient (Slc10a2 (-/-) ) male mice were treated with azoxymethane (AOM) alone to examine the development of aberrant crypt foci, the earliest histological marker of colon neoplasia and a combination of AOM and dextran sulfate sodium to induce colon tumor formation. Asbt-deficient mice exhibited a 54% increase in aberrant crypt foci, and 70 and 59% increases in colon tumor number and size, respectively. Compared to littermate controls, Asbt-deficient mice had a striking, 2-fold increase in the number of colon adenocarcinomas. Consistent with previous studies demonstrating a role for muscarinic and epidermal growth factor receptor signaling in bile acid-induced colon neoplasia, increasing bile acid malabsorption was associated with M3 muscarinic and epidermal growth factor receptor expression, and activation of extracellular signal-related kinase, a key post-receptor signaling molecule.


Subject(s)
Bile Acids and Salts/toxicity , Colonic Neoplasms/metabolism , Ileum/metabolism , Organic Anion Transporters, Sodium-Dependent/genetics , Symporters/genetics , Animals , Bile Acids and Salts/genetics , Bile Acids and Salts/metabolism , Colonic Neoplasms/etiology , Colonic Neoplasms/pathology , Disease Models, Animal , Feces , Humans , Ileum/pathology , Mice , Mice, Knockout , Organic Anion Transporters, Sodium-Dependent/metabolism , Signal Transduction/drug effects , Symporters/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...