Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Lett ; 587: 216678, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38360143

ABSTRACT

Endoplasmic reticulum (ER) stress and the adaptive response that follows, termed the unfolded protein response (UPR), are crucial molecular mechanisms to maintain cellular integrity by safeguarding proper protein synthesis. Next to being important in protein homeostasis, the UPR is intricate in cell fate decisions such as proliferation, differentiation, and stemness. In the intestine, stem cells are critical in governing epithelial homeostasis and they are the cell of origin of gastrointestinal malignancies. In this review, we will discuss the role of ER stress and the UPR in the gastrointestinal tract, focusing on stem cells and carcinogenesis. Insights in mechanisms that connect ER stress and UPR with stemness and carcinogenesis may broaden our understanding in the development of cancer throughout the gastrointestinal tract and how we can exploit these mechanisms to target these malignancies.


Subject(s)
Neoplasms , Unfolded Protein Response , Humans , Endoplasmic Reticulum Stress/physiology , Carcinogenesis , Neoplasms/pathology , Stem Cells/pathology , Gastrointestinal Tract
2.
Cell Death Dis ; 10(7): 490, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31227689

ABSTRACT

The unfolded protein response (UPR) acts through its downstream branches, PERK-eIF2α signaling, IRE1α-XBP1 signaling and ATF6 signaling. In the intestine, activation of the UPR through the kinase PERK results in differentiation of intestinal epithelial stem cells and colon cancer stem cells, whereas deletion of XBP1 results in increased stemness and adenomagenesis. How downstream activation of XBP1 and ATF6 influences intestinal stemness and proliferation remains largely unknown. We generated colorectal cancer cells (LS174T) that harbor doxycycline inducible expression of the active forms of either XBP1(s) or ATF61-373. Activation of either XBP1 or ATF6 resulted in reduced cellular proliferation and reduced expression of markers of intestinal epithelial stemness. Moreover, XBP1 and ATF6 activation reduced global protein synthesis and lowered the threshold for UPR activation. XBP1-mediated loss of stemness and proliferation resulted from crossactivation of PERK-eIF2α signaling and could be rescued by constitutive expression of eIF2α phosphatase GADD34. We thus find that enforced activation of XBP1 and ATF6 results in reduction of stemness and proliferation. We expose a novel interaction between XBP1 and PERK-eIF2α signaling.


Subject(s)
Activating Transcription Factor 6/metabolism , Colonic Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response/physiology , X-Box Binding Protein 1/metabolism , Activating Transcription Factor 6/genetics , Blotting, Western , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Colonic Neoplasms/genetics , Humans , Neoplastic Stem Cells/cytology , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Unfolded Protein Response/genetics , X-Box Binding Protein 1/genetics
3.
Cancer Res ; 78(21): 6098-6106, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30232220

ABSTRACT

Deletion of endoplasmic reticulum resident chaperone Grp78 results in activation of the unfolded protein response and causes rapid depletion of the entire intestinal epithelium. Whether modest reduction of Grp78 may affect stem cell fate without compromising intestinal integrity remains unknown. Here, we employ a model of epithelial-specific, heterozygous Grp78 deletion by use of VillinCreERT2-Rosa26ZsGreen/LacZ-Grp78+/fl mice and organoids. We examine models of irradiation and tumorigenesis, both in vitro and in vivo Although we observed no phenotypic changes in Grp78 heterozygous mice, Grp78 heterozygous organoid growth was markedly reduced. Irradiation of Grp78 heterozygous mice resulted in less frequent regeneration of crypts compared with nonrecombined (wild-type) mice, exposing reduced capacity for self-renewal upon genotoxic insult. We crossed mice to Apc-mutant animals for adenoma studies and found that adenomagenesis in Apc heterozygous-Grp78 heterozygous mice was reduced compared with Apc heterozygous controls (1.43 vs. 3.33; P < 0.01). In conclusion, epithelium-specific Grp78 heterozygosity compromises epithelial fitness under conditions requiring expansive growth such as adenomagenesis or regeneration after γ-irradiation. These results suggest that Grp78 may be a therapeutic target in prevention of intestinal neoplasms without affecting normal tissue.Significance: Heterozygous disruption of chaperone protein Grp78 reduces tissue regeneration and expansive growth and protects from tumor formation without affecting intestinal homeostasis. Cancer Res; 78(21); 6098-106. ©2018 AACR.


Subject(s)
Adenoma/metabolism , Heat-Shock Proteins/metabolism , Intestinal Neoplasms/metabolism , Intestines/cytology , Stem Cells/cytology , Adenoma/genetics , Alleles , Animals , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Endoplasmic Reticulum Chaperone BiP , Female , Gene Deletion , Genotype , Heat-Shock Proteins/genetics , Heterozygote , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Neoplasms/genetics , Male , Mice , Molecular Chaperones , Organoids , Phenotype , Regeneration , Unfolded Protein Response
4.
Cell Rep ; 13(3): 489-494, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26456824

ABSTRACT

Colon cancer stem cells (colon-CSCs) are more resistant to conventional chemotherapy than differentiated cancer cells. This subset of therapy refractory cells is therefore believed to play an important role in post-therapeutic tumor relapse. In order to improve the rate of sustained response to conventional chemotherapy, development of approaches is warranted that specifically sensitize colon-CSCs to treatment. Here, we report that ER-stress-induced activation of the unfolded protein response (UPR) forces colon-CSCs to differentiate, resulting in their enhanced sensitivity to chemotherapy in vitro and in vivo. Our data suggest that agents that induce activation of the UPR may be used to specifically increase sensitivity of colon-CSCs to the effects of conventional chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Differentiation , Colonic Neoplasms/metabolism , Endoplasmic Reticulum Stress , Neoplastic Stem Cells/metabolism , Organoplatinum Compounds/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Humans , Mice , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Spheroids, Cellular , Unfolded Protein Response
5.
J Vis Exp ; (98)2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25938265

ABSTRACT

Intestinal crypt-villus structures termed organoids, can be kept in sustained culture three dimensionally when supplemented with the appropriate growth factors. Since organoids are highly similar to the original tissue in terms of homeostatic stem cell differentiation, cell polarity and presence of all terminally differentiated cell types known to the adult intestinal epithelium, they serve as an essential resource in experimental research on the epithelium. The possibility to express transgenes or interfering RNA using lentiviral or retroviral vectors in organoids has increased opportunities for functional analysis of the intestinal epithelium and intestinal stem cells, surpassing traditional mouse transgenics in speed and cost. In the current video protocol we show how to utilize transduction of small intestinal organoids with lentiviral vectors illustrated by use of doxycylin inducible transgenes, or IPTG inducible short hairpin RNA for overexpression or gene knockdown. Furthermore, considering organoid culture yields minute cell counts that may even be reduced by experimental treatment, we explain how to process organoids for downstream analysis aimed at quantitative RT-PCR, RNA-microarray and immunohistochemistry. Techniques that enable transgene expression and gene knock down in intestinal organoids contribute to the research potential that these intestinal epithelial structures hold, establishing organoid culture as a new standard in cell culture.


Subject(s)
Intestinal Mucosa/physiology , Intestine, Small/physiology , Lentivirus/genetics , Transduction, Genetic/methods , Animals , Cell Differentiation/physiology , Gene Knockdown Techniques/methods , Genetic Vectors/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/virology , Intestine, Small/cytology , Intestine, Small/virology , Mice , Mice, Transgenic , Organ Culture Techniques , Organoids/physiology , Organoids/virology , RNA Interference , Stem Cells/cytology , Stem Cells/virology , Transgenes
6.
World J Gastroenterol ; 20(44): 16683-9, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25469037

ABSTRACT

AIM: To investigate if azathioprine could reduce adenoma formation in Apc(Min/+) , a mouse model of sporadic intestinal tumorigenesis. METHODS: Azathioprine was administered via drinking water (estimated 6-20 mg/kg body weight per day) to Apc(Min/+) and wildtype mice. Control animals received vehicle only (DMSO) dissolved in drinking water. At 15 wk of age all mice were sacrificed and intestines of Apc(Min/+) were harvested for evaluation of polyp number. Azathioprine induced toxicity was investigated by immunohistochemical analysis on spleens. RESULTS: All azathioprine treated mice showed signs of drug-associated toxicity such as weight loss and development of splenic T-cell lymphomas. Although this suggests that the thiopurine concentration was clearly in the therapeutic range, it did not reduce tumor formation (48 ± 3.1 adenomas vs 59 ± 5.7 adenomas, P = 0.148). CONCLUSION: We conclude that in the absence of inflammation, azathioprine does not affect intestinal tumorigenesis.


Subject(s)
Adenomatous Polyposis Coli/prevention & control , Anticarcinogenic Agents/pharmacology , Azathioprine/pharmacology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Animals , Anticarcinogenic Agents/toxicity , Azathioprine/toxicity , Female , Genes, APC , Lymphoma, T-Cell/chemically induced , Lymphoma, T-Cell/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Splenic Neoplasms/chemically induced , Splenic Neoplasms/pathology , Time Factors
7.
Gut ; 63(2): 310-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23408349

ABSTRACT

BACKGROUND: Hormone replacement therapy increases the risk of developing ulcerative colitis in postmenopausal women. Chronic intestinal inflammation predisposes to colon cancer development, but effects of female hormones on colitis-associated cancer development have not been examined. AIM: To investigate the role of female hormones in the dextran sodium sulfate (DSS)-azoxymethane (AOM) mouse model for colitis-associated cancer. DESIGN: We performed ovariectomies, or sham operations, on mice, and supplemented these animals with indicated hormones. Additionally, we used oestrogen receptor α or ß (Erα or Erß) mutant mice. To study colitis or colitis-associated cancer, we used DSS only, or DSS and AOM, respectively. RESULTS: Ovariectomy protects female mice against colitis-associated tumour development. Hormone replacement in ovariectomised mice with either oestradiol (E2), medroxyprogesterone acetate or a combination of both suggests that oestrogens are the ovary-derived factor that promotes tumour development in the context of inflammatory damage. E2-treated animals showed increased clinical symptoms and Il-6 production upon DSS-induced colitis and enhanced epithelial proliferation. Treatment with E2 markedly increased the numbers of polyps in ovariectomised mice and also strongly promoted tumour progression with all E2-treated animals developing at least one invasive adenocarcinoma, whereas, placebo-treated animals developed adenomas only. Using Er mutant mice, we find that the protumorigenic effect of oestrogen depends on both Erα and Erß. CONCLUSIONS: Our results suggest that oestrogens promote inflammation-associated cancer development by impairing the mucosal response to inflammatory damage.


Subject(s)
Carcinogenesis/chemically induced , Colitis/chemically induced , Colonic Neoplasms/chemically induced , Disease Models, Animal , Estradiol/adverse effects , Estrogens/adverse effects , Medroxyprogesterone/adverse effects , Animals , Azoxymethane/toxicity , Cytokines/metabolism , Dextran Sulfate/toxicity , Female , Hormone Replacement Therapy/adverse effects , Immunohistochemistry , Mice , Ovariectomy
8.
Cell Rep ; 3(4): 1128-39, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23545496

ABSTRACT

Stem cells generate rapidly dividing transit-amplifying cells that have lost the capacity for self-renewal but cycle for a number of times until they exit the cell cycle and undergo terminal differentiation. We know very little of the type of signals that trigger the earliest steps of stem cell differentiation and mediate a stem cell to transit-amplifying cell transition. We show that in normal intestinal epithelium, endoplasmic reticulum (ER) stress and activity of the unfolded protein response (UPR) are induced at the transition from stem cell to transit-amplifying cell. Induction of ER stress causes loss of stemness in a Perk-eIF2α-dependent manner. Inhibition of Perk-eIF2α signaling results in stem cell accumulation in organoid culture of primary intestinal epithelium. Our findings show that the UPR plays an important role in the regulation of intestinal epithelial stem cell differentiation.


Subject(s)
Endoplasmic Reticulum Stress , Intestinal Mucosa/metabolism , Stem Cells/cytology , Unfolded Protein Response , Animals , Cell Differentiation , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Intestinal Mucosa/cytology , Mice , Mutation , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Stem Cells/metabolism , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...