1
|
Goodlad RA. Quantification of epithelial cell proliferation, cell dynamics, and cell kinetics in vivo. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2017; 6. [PMID: 28474479 DOI: 10.1002/wdev.274] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 02/21/2017] [Accepted: 02/22/2017] [Indexed: 12/12/2022]
Abstract
The measurement of cell proliferation in vivo is usually carried out by the examination of static measures. These comprise the mitotic index or labeling indices using incorporation of DNA synthesis markers such as bromodeoxyuridine or tritiated thymidine, or intrinsic markers, such as Ki67 and proliferative cell nuclear antigen (PCNA). But static measures only provide a 'snapshot' of cell proliferation. Rate measures, including double labeling methods and the metaphase arrest method, can actually measure cell production rates but they are far less utilized at present. Transit times and migration rates can also be measured using pulse and chase labeling or by following the transit of labeled cells through the tissue. Simple indices of cell division can easily be confounded by concomitant changes in the compartment size and many alleged markers of proliferation have serious shortcomings, as the markers may be involved in multiple aspects of cell regulation. The complexities of studying proliferation in vivo are illustrated here with a focus on the gastrointestinal tract. Some of these methods can help elucidate the role of the stem cells and their relationship to label retaining cells. WIREs Dev Biol 2017, 6:e274. doi: 10.1002/wdev.274 For further resources related to this article, please visit the WIREs website.
Collapse
|
2
|
The Na+-D-glucose cotransporters SGLT1 and SGLT2 are targets for the treatment of diabetes and cancer. Pharmacol Ther 2017; 170:148-165. [DOI: 10.1016/j.pharmthera.2016.10.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
3
|
Kitchen PA, Goodlad RA, FitzGerald AJ, Mandir N, Ghatei MA, Bloom SR, Berlanga-Acosta J, Playford RJ, Forbes A, Walters JRF. Intestinal Growth in Parenterally-Fed Rats Induced by the Combined Effects of Glucagon-like Peptide 2 and Epidermal Growth Factor. JPEN J Parenter Enteral Nutr 2017; 29:248-54. [PMID: 15961680 DOI: 10.1177/0148607105029004248] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Parenteral nutrition and the absence of luminal feeding result in impaired intestinal growth and differentiation of enterocytes. Glucagon-like peptide 2 (GLP-2) and epidermal growth factor (EGF) have each been shown to have trophic effects on the intestine, and thus have the potential to benefit patients fed parenterally, such as those with intestinal failure from short bowel syndrome. We report studies aimed to determine whether there may be synergistic effects of these 2 peptides. METHODS Rats were established on parenteral nutrition (PN) and infused for 6 days with GLP-2 (20 microg/d), EGF (20 microg/d), or GLP-2 + EGF (20 microg/d of each). These groups were compared with untreated PN-fed and orally-fed controls. Tissue was obtained from small intestine and colon to determine growth, proliferation, and representative gene expression. RESULTS Small intestinal weight was increased by 75%, 43%, and 116% in the GLP-2, EGF, and GLP-2 + EGF groups, respectively, compared with PN controls (all p < .001). Cell proliferation increased with GLP-2, EGF, and GLP-2 + EGF in proximal small intestine by factors of 2.3, 1.7, and 3.4 respectively (p < .001). A synergistic effect on villous and crypt area was observed in the proximal small intestine when GLP-2 and EGF were combined (p < .05). GLP-2 had no effect in the colon, unlike EGF. Further studies showed GLP-2 + EGF significantly increased expression in distal small intestine of transcripts for the bile acid transport protein IBABP (p < .05) and showed a significant correlation between the expression of IBABP and the transcription factor HNF-4. CONCLUSIONS Both GLP-2 and EGF upregulate growth of the small intestine, and this is augmented when GLP-2 and EGF are combined. These findings may lead to improved treatment of patients receiving PN.
Collapse
Affiliation(s)
- Paul A Kitchen
- St Mark's Hospital, Imperial College London, Harrow, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Geng Y, Li J, Wang F, Li Q, Wang X, Sun L, Li W. Epidermal growth factor promotes proliferation and improves restoration after intestinal ischemia-reperfusion injury in rats. Inflammation 2014; 36:670-9. [PMID: 23397076 DOI: 10.1007/s10753-012-9591-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Epidermal growth factor (EGF) is an attractive and promising therapeutic application for intestinal disorders. The current study examined its influence on proliferation and restoration after ischemia-reperfusion (I/R) injury in rat small intestine. Six groups were performed: sham operation (Con); ischemia for 30 min with subsequent reperfusion for 30 min (I/R); I/R injured with 500 μg/kg EGF injected 5 min before ischemia (Pre-l); I/R injured with 50 μg/kg EGF injected 5 min before ischemia (Pre-s); I/R injured with 500 μg/kg EGF injected 5 min after reperfusion (Post-l); and I/R injured with 50 μg/kg EGF injected 5 min after reperfusion (Post-s). Intestinal histological damage, crypt cell proliferation degree, mucosal permeability, tight junction proteins expression, and levels of inflammation factors were studied for each group. Compared with the I/R group, administration of EGF in the Pre-l, Pre-s, and Post-l groups all presented a significant proliferation effect. The levels of FD4, IL-6, and TNF-α were dramatically decreased in all EGF-treated groups. Histological destruction was improved and TJs recovery was notably accelerated in all EGF-treated groups except the Post-s group. D-lactate concentration was only diminished in the Pre-l group. These results suggest that mucosally applied EGF can promote intestinal proliferation and improve restoration after I/R injury. EGF intraluminal administration is an effective treatment against intestinal I/R injury.
Collapse
Affiliation(s)
- Yanxia Geng
- Department of Intensive Care Unit, Jiangsu Province Chinese Medicine Hospital, 155 Han Zhong Road, Nanjing 210029, China
| | | | | | | | | | | | | |
Collapse
|
5
|
VINTER-JENSEN LARS. Pharmacological effects of epidermal growth factor (EGF) with focus on the urinary and gastrointestinal tracts. APMIS 2011. [DOI: 10.1111/j.1600-0463.1999.tb05690.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
6
|
Gusterson BA, Hunter KD. Should we be surprised at the paucity of response to EGFR inhibitors? Lancet Oncol 2009; 10:522-7. [PMID: 19410197 DOI: 10.1016/s1470-2045(09)70034-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Data suggest that neither our current understanding of the function and signalling of epidermal growth factor receptor (EGFR), nor measurements of receptor expression are reliably predictive of therapeutic responses to EGFR inhibitors. The time has now come to consider whether such poor correlation between receptor expression and clinical response is caused by poor assays or by more fundamental issues relating to the in-vivo function of EGFR. Revisiting some of the early findings of the biology of EGFR function and understanding the limitations of immunohistochemistry as a quantitative technique might provide some clues. However, we still have a lot to learn about this receptor, its many ligands, and its binding partners in normal physiology and disease.
Collapse
Affiliation(s)
- Barry A Gusterson
- Department of Pathology, Division of Cancer Sciences and Molecular Pathology, Faculty of Medicine, University of Glasgow, Western Infirmary, Glasgow, UK.
| | | |
Collapse
|
7
|
Transenteric signalling: mammary messages or white noise? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009. [PMID: 19227536 DOI: 10.1007/978-1-4020-8749-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
8
|
Resistant carbohydrates stimulate cell proliferation and crypt fission in wild-type mice and in the Apc(Min/+) mouse model of intestinal cancer, association with enhanced polyp development. Br J Nutr 2008; 100:711-21. [PMID: 18279550 DOI: 10.1017/s0007114508901276] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Fermentation of carbohydrates in the colon can stimulate cell proliferation and could thus be a cancer risk. The effects of resistant carbohydrates, i.e. those not digested and absorbed in the small intestine, on cell proliferation, crypt fission and polyp development were investigated in wild-type and adenomatous polyposis coli multiple intestinal neoplasia (Apc(Min/+)) mice. Fifteen 4-week-old female wild-type and fifteen Apc(Min/+) mice were used for each group and fed a chow diet, a semi-synthetic diet or the semi-synthetic supplemented with wheat bran or an apple pomace preparation, both high in resistant carbohydrates, for 8 weeks. Tissue from all mice was used to measure cell proliferation and crypt fission and tissue from the Apc(Min/+) mice was scored for polyp number and tumour burden. There were slight reductions in intestinal mass in the mice fed the semi-synthetic diets and this was increased by the inclusion of resistant carbohydrates. The Apc(Min/+) mice had elevated cell proliferation and crypt fission in the distal small intestine and colon and these were increased by the resistant carbohydrates. Bran or apple pomace significantly increased polyp number in the proximal third of the small intestine. Apple pulp more than doubled polyp number throughout the small bowel (99.2 (SEM 11.1) v. 40.0 (SEM 8.2), P<0.004). Bran and apple pomace increased polyp diameter and hence burden in the colon by 243 and 150 %, respectively (P<0.05). In conclusion, both types of resistant carbohydrates increased polyp number and tumour burden and this was associated with elevated epithelial cell proliferation and crypt fission.
Collapse
|
9
|
Alferez D, Goodlad RA. To best measure cell proliferation in samples from the intestine. Cell Prolif 2007; 40:231-40. [PMID: 17472729 PMCID: PMC6496137 DOI: 10.1111/j.1365-2184.2007.00427.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Accepted: 11/23/2006] [Indexed: 01/28/2023] Open
Abstract
Arrangement of the intestinal cell lining, as it is, into distinct anatomically defined zones where proliferation is confined to the crypts, makes it an ideal tissue to study growth control mechanisms. While many methods have been used to quantify cell proliferation in the gut, several of them have severe limitations and others (although potentially better) have been misused and misinterpreted. Here, correct use and interpretation of labelling studies will be described as will a well established alternative method that provides equivalent results for one-sixth of the effort.
Collapse
Affiliation(s)
- D Alferez
- Department of Histopathology, Division of Investigative Science, Imperial College, Hammersmith Hospital, London, UK
| | | |
Collapse
|
10
|
YANG HUA, TEITELBAUM DANIELH. Novel agents in the treatment of intestinal failure: humoral factors. Gastroenterology 2006; 130:S117-21. [PMID: 16473058 PMCID: PMC1522079 DOI: 10.1053/j.gastro.2005.08.059] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2004] [Accepted: 08/24/2005] [Indexed: 12/14/2022]
Affiliation(s)
| | - DANIEL H. TEITELBAUM
- Address requests for reprints to: Daniel H. Teitelbaum, MD, University of Michigan Medical School and the C.S. Mott Children’s Hospital, Ann Arbor, Michigan 48109. e-mail
| |
Collapse
|
11
|
FitzGerald AJ, Mandir N, Goodlad RA. Leptin, cell proliferation and crypt fission in the gastrointestinal tract of intravenously fed rats. Cell Prolif 2005; 38:25-33. [PMID: 15679864 PMCID: PMC6496203 DOI: 10.1111/j.1365-2184.2005.00327.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2004] [Accepted: 11/11/2004] [Indexed: 12/25/2022] Open
Abstract
Many peptides, hormones and growth factors have been implicated in the control of cell renewal in the gastrointestinal epithelium. Leptin is present in the stomach and salivary glands and leptin receptors are seen throughout the gut. Leptin can stimulate mitogen-activated protein kinase activity in vitro and short-term infusion has been reported to have a proliferative action on the colon in vivo, suggesting a biological link between obesity, physical activity and colon cancer. Food intake is one of the most important determinants of intestinal mucosal cell renewal, thus any direct effects of leptin on the gut may be hidden. This problem has been avoided experimentally by maintaining animals on total parenteral nutrition (TPN). Male Wistar rats were anaesthetized and cannulae were inserted into the jugular vein to deliver the TPN diet to which had been added 0, 0.5, 2.5, or 10 mg/kg of recombinant murine leptin. Orally fed rats were also studied. After 6 days of treatment, all animals were injected with vincristine and killed 2 h later. Tissue weight was recorded and crypt cell proliferation (arrested metaphases) and crypt fission were scored in 'microdissected' crypts. Leptin infusion led to a small decrease in body weight and in the weight of the caecum. Intestinal cell proliferation was significantly reduced by TPN when compared to the orally fed rats, but the addition of leptin had no effect on the small intestine or colon. Crypt fission was also significantly lowered in the TPN group. Fission was slightly but significantly increased in the proximal and mid-colon of the leptin-treated rats, but was decreased in the distal colon. Although leptin did not significantly alter cell proliferation, it had significant effects on the process of crypt fission in the colon, which varied according to the exact locality.
Collapse
Affiliation(s)
- A J FitzGerald
- Histopathology Imperial College School of Medicine, Hammersmith Hospital, London, UK
| | | | | |
Collapse
|
12
|
Cellini C, Xu J, Arriaga A, Buchmiller-Crair TL. Effect of epidermal growth factor infusion on fetal rabbit intrauterine growth retardation and small intestinal development. J Pediatr Surg 2004; 39:891-7; discussion 891-7. [PMID: 15185220 DOI: 10.1016/j.jpedsurg.2004.02.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND/PURPOSE Intrauterine growth retardation (IUGR) infants have impaired gastrointestinal function with resultant feeding difficulties and predisposition to necrotizing enterocolitis. Supplemented amniotic fluid swallowed by the developing fetus is a potential prenatal treatment for IUGR. Rabbits have naturally occurring IUGR fetuses based on uterine position. To determine intestinal response to epidermal growth factor (EGF) infusion, this rabbit model of IUGR was studied. METHODS Eight pregnant rabbits underwent placement of intraamniotic catheters into 2 normal and 2 IUGR fetuses per mother on gestational day 24 of a 31-day gestation. Miniosmotic pumps infused either EGF (about 300 microg/kg/d) or control solution forming 4 study groups (EGF-Favored [Fav] v. Cont-Fav; EGF-IUGR v. Cont-IUGR). On gestational day 31, the fetal gastrointestinal tracts were harvested for analysis. Intestinal epithelial cell proliferation was studied by 5-bromo-2-deoxyuridine (BrdU) incorporation, villus heights were measured, and EGF mRNA was measured by reverse transcriptase polymerase chain reaction (RT-PCR). Statistical analysis was performed using Students' t test. RESULTS Fetal survival rate was 87%. EGF-IUGR fetal weights were increased compared with Cont-IUGR fetuses. EGF infusion significantly increased IUGR fetal small intestinal villus height and BrdU-positive small intestinal (SI) crypt cells, all approaching Cont-Fav levels. EGF mRNA was expressed throughout the gastrointestinal tract. CONCLUSIONS Supplemental amniotic EGF normalizes fetal weight and intestinal proliferation in the IUGR fetal rabbit. The inclusion of EGF in supplemental amniotic feeding solutions is supported.
Collapse
Affiliation(s)
- Christina Cellini
- Division of Pediatric Surgery, Children's Hospital of New York Presbyterian Hospital, Weill Medical College of Cornell University, New York, NY, USA
| | | | | | | |
Collapse
|
13
|
Sasaki M, Fitzgerald AJ, Grant G, Ghatei MA, Wright NA, Goodlad RA. Lectins can reverse the distal intestinal atrophy associated with elemental diets in mice. Aliment Pharmacol Ther 2002; 16:633-42. [PMID: 11876719 DOI: 10.1046/j.1365-2036.2002.01160.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Elemental diets cause intestinal atrophy and reduced intestinal integrity, which can lead to significant increases in intestinal permeability and bacterial translocation. Recently, several lectins have been shown to have trophic effects on the intestine. AIMS We examined the effects of concanavalin-A and phytohaemagglutinin on cell proliferation and crypt fission throughout the intestine of mice fed on elemental diets. METHODS Mice were randomized to chow fed, elemental diet, elemental diet plus concanavalin-A and elemental diet plus phytohaemagglutinin groups. Cell proliferation and crypt fission were estimated in microdissected crypts. Plasma gastrin and enteroglucagon levels were measured by radioimmunoassay. RESULTS Elemental diet feeding significantly decreased cell proliferation and crypt fission of the middle and distal small intestine and throughout the colon. Phytohaemagglutinin significantly increased the weight of the intestine, but concanavalin-A had little effect. Cell proliferation in the small intestine was significantly increased by both lectins. However, in the stomach and colon, only phytohaemagglutinin increased proliferation. Crypt fission in the colon was dramatically increased by phytohaemagglutinin. Phytohaemagglutinin increased the plasma gastrin level, but not the enteroglucagon level. CONCLUSIONS Lectins have significant trophic effects on the small intestine and colon of mice fed elemental diets, and these actions vary between different sites in the gastrointestinal tract.
Collapse
Affiliation(s)
- M Sasaki
- Department of Histopathology, Imperial College School of Medicine, Hammersmith Hospital, London, UK
| | | | | | | | | | | |
Collapse
|
14
|
Aly A, Shulkes A, Baldwin GS. Short term infusion of glycine-extended gastrin(17) stimulates both proliferation and formation of aberrant crypt foci in rat colonic mucosa. Int J Cancer 2001; 94:307-13. [PMID: 11745407 DOI: 10.1002/ijc.1483] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Evidence is accumulating that gastrin precursors may act as growth factors for the colonic mucosa in vivo and for colorectal carcinoma cell lines in vitro. The effect of short term administration of synthetic gastrins on the colonic mucosa in vivo, however, has not been reported. The aim of our study was to determine whether continuous systemic infusion of glycine-extended gastrin(17) stimulated proliferation and accelerated carcinogenesis in the colorectal mucosa. A significant increase in colonic mucosal proliferation as assessed by metaphase index was seen in the caecum (23%, p < 0.02) and distal colon (27%, p < 0.001), but not the rectum, after treatment of intact rats with glycine-extended gastrin(17) for 1 week using implanted miniosmotic pumps. Defunctioning of the rectum reduced both the proliferative index and crypt height of the rectal mucosa of untreated rats. Treatment of rectally defunctioned animals with glycine-extended gastrin(17) for either 1 or 4 weeks resulted in a significant increase in both the proliferative index (40% and 93%, respectively) and crypt height (11% and 19%, respectively) of the rectal mucosa. The total number of aberrant crypt foci in intact rats treated with the procarcinogen azoxymethane plus glycine-extended gastrin(17) was increased by 48% compared to the value in controls treated with azoxymethane only (p = 0.01). We conclude that short term administration of glycine-extended gastrin(17) to mature rats not only has a proliferative effect upon colonic mucosa, but also increases the number of aberrant crypt foci formed in the colorectal mucosa after treatment with azoxymethane. Glycine-extended gastrin(17) could thus potentially act as a promoter of carcinogenesis.
Collapse
Affiliation(s)
- A Aly
- University of Melbourne Department of Surgery, Austin Campus, A&RMC, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
15
|
Jeffrey SC, Murray MJ, Eichorn ES. Distribution of epidermal growth factor receptor (EGFr) in normal and acute peptic-injured equine gastric squamous epithelium. Equine Vet J 2001; 33:562-9. [PMID: 11720027 DOI: 10.2746/042516401776563481] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Growth factors are important in healing and restoration of injured gastrointestinal tissues and, therefore, we characterised temporally the distribution and density of epidermal growth factor receptor (EGFr) in normal and peptic-injured gastric squamous epithelium of horses. Lesions were induced in the equine gastric squamous epithelium using a feed deprivation protocol that results in prolonged increased gastric acidity. Fifteen mature horses, 9 geldings and 6 mares, age 3 to 20 years, were used and divided into 3 groups: Group 1 (n = 5) were subjected to euthanasia for problems unrelated to the gastrointestinal tract and had normal-appearing gastric squamous mucosal epithelium; Groups 2 (n = 5) and 3 (n = 5) had lesions induced in the gastric squamous epithelium by alternating 24 h periods of feed deprivation and ad libitum access to hay, for a total of 48 h and 96 h, respectively. Following lethal injection of a barbiturate, stomachs were removed and fixed by filling with 4- 6 l 10% buffered formalin. Sections were made from normal stomachs and lesions in the gastric squamous epithelium adjacent to the margo plicatus along the right side of the stomach/greater curvature and the lesser curvature. A modified avidin-biotin immunoperoxidase technique was used to stain the formalin-fixed tissue specimens for EGFr. A computerised image analysis system was used to measure area occupied by EGFr (EGFr area) and mean EGFr density in 4 zones within the epithelium extending from the basal cell layers toward the lumen. Measurements were made of epithelium in an erosion bed, at the margin of an ulcer or erosion, and 10-15 mm distant from the lesion margin. Additionally, EGFr area and density were measured in epithelial cells adjacent to capillaries in the epithelium. Intermittent feed deprivation resulted in erosion and ulceration of the gastric squamous epithelium of each horse. Mean EGFr area and density were greatest (P<0.05) in the basal layer of epithelia from all horses, and EGFr staining diminished progressively toward the lumen. Tissues from Group 3 had significantly greater EGFr area in the lesion margin than epithelia from Group 2. EGFr density was less in the epithelia of erosion beds from Groups 2 and 3 compared to normal epithelium, and EGFr area in Group 2 erosion bed epithelia was significantly less than in normal epithelium and epithelia of Group 3. EGFr area in cells adjacent to epithelial capillaries of Group 3 was significantly greater than that of Group 1. Mitotic cell activity was significantly greater in epithelia associated with ulcers and erosions in Groups 2 and 3 compared to normal tissues from Group 1 horses. Staining for EGFr in the glandular mucosa adjacent to squamous epithelium at the margo plicatus was inconsistent and typically faint when present. EGFr distribution in equine gastric squamous epithelium was greatest in regions of greatest cell proliferation, and these areas were in the basal layers of epithelium and immediately adjacent to capillaries. There was evidence that EGFr is induced in peptic-injured equine gastric squamous epithelium. A receptor ligand, EGF or transforming growth factoralpha, may be a factor in healing of gastric squamous mucosal ulcers in horses. Further research should be directed at identifying this ligand and determining its origin in equine gastric mucosa.
Collapse
Affiliation(s)
- S C Jeffrey
- Marion duPont Scott Equine Medical Center, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Leesburg 20177, USA
| | | | | |
Collapse
|
16
|
Sasaki M, Fitzgerald AJ, Mandir N, Sasaki K, Wright NA, Goodlad RA. Glicentin, an active enteroglucagon, has a significant trophic role on the small intestine but not on the colon in the rat. Aliment Pharmacol Ther 2001; 15:1681-6. [PMID: 11564010 DOI: 10.1046/j.1365-2036.2001.01082.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
BACKGROUND Many experiments have indicated that the gut glucagons (enteroglucagons) are associated with cell proliferation in the small intestine. However, recent studies have failed to show trophic effects of glicentin (enteroglucagon) on the intestine. AIMS To examine the effects of glicentin on intestinal proliferation in vivo in the rat. METHODS Rats were established on total parenteral nutrition for 6 days. Four experimental groups were given daily doses of 1, 4, 20 and 80 microg/rat of glicentin via the jugular vein. Rats fed by total parenteral nutrition and rats fed chow ad libitum were used as controls. Tissues taken from the duodenum, jejunum, ileum and colon were fixed in Carnoy's fluid and microdissected to determine the metaphase arrest scores and crypt fission ratios. RESULTS The mean metaphase arrest scores per crypt of the small intestine were significantly increased in the rats given 4, 20 and 80 microg of glicentin. These responses were dose-dependent, and were most prominent in the ileum. Crypt fission of the ileum was significantly decreased in the 20 and 80 microg glicentin groups. Glicentin had no effects on proliferation or fission in the colon. CONCLUSIONS Glicentin is trophic to the rat small intestine, but not the colon.
Collapse
Affiliation(s)
- M Sasaki
- Department of Histopathology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Road, London, UK
| | | | | | | | | | | |
Collapse
|
17
|
Fitzgerald AJ, Jordinson M, Rhodes JM, Singh R, Calam J, Goodlad RA. Comparison of the effects of concanavalin-A and epidermal growth factor on epithelial cell proliferation in the rat intestine. Aliment Pharmacol Ther 2001; 15:1077-84. [PMID: 11421885 DOI: 10.1046/j.1365-2036.2001.01025.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Concanavalin-A, the lectin present in Jack beans, binds to mannose- and glucose-containing residues and can interact with the epidermal growth factor receptor and moderate cell proliferation in vitro. AIM To compare the actions of concanavalin-A and epidermal growth factor on the gastrointestinal tract in vivo. METHODS Rats maintained on total parenteral nutrition were given intragastric concanavalin-A, intravenous epidermal growth factor or concanavalin-A and epidermal growth factor. Cell proliferation and crypt fission were assayed in 'micro-dissected' crypts. RESULTS Concanavalin-A and epidermal growth factor both significantly elevated proliferation in the small intestine and colon. No significant interaction between the effects of these two agents was seen, except in the mid small intestine where there was a synergistic interaction. Concanavalin-A had no effect on crypt branching. Epidermal growth factor significantly reduced branching in the distal small intestine and mid colon. CONCLUSION The effects of the two agents appeared to be separate, except in the mid small intestine where they were additive. This is in marked contrast with the actions reported in vitro, where concanavalin-A is a powerful inhibitor of epidermal growth factor-induced cell proliferation. Concanavalin-A thus has potential for enhancing the functions of the small intestine.
Collapse
Affiliation(s)
- A J Fitzgerald
- Department of Histopathology, Division of Investigative Science, Imperial College School of Medicine, Hammersmith Hospital, London, UK
| | | | | | | | | | | |
Collapse
|
18
|
Ghatei MA, Goodlad RA, Taheri S, Mandir N, Brynes AE, Jordinson M, Bloom SR. Proglucagon-derived peptides in intestinal epithelial proliferation: glucagon-like peptide-2 is a major mediator of intestinal epithelial proliferation in rats. Dig Dis Sci 2001; 46:1255-63. [PMID: 11414302 DOI: 10.1023/a:1010615429639] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Proglucagon-derived peptides have been implicated in the control of intestinal mucosal cell division. To investigate the actions of these peptides on intestinal cell proliferation, different doses of enteroglucagon, oxyntomodulin, glucagon-like peptide-1 (GLP-1) and glucagon-like peptide-2 (GLP-2) were tested in male Wistar rats maintained on total parenteral nutrition. Crypt cell proliferation was assessed by the analysis of arrested metaphases in microdissected crypts. Enteroglucagon and oxyntomodulin had no effect on intestinal weight or cell proliferation. GLP-1 had a slight effect on stomach and small intestinal weights and on epithelial cell proliferation in the small and large intestines. GLP-2 infusion dose-dependently increased the weights of the stomach, small intestine, colon, and cecum and increased crypt cell proliferation in the small and large intestines of parenterally fed rats. In orally fed animals, GLP-2 increased intestinal weight but had little effect on proliferation. Therefore, of the proglucagon-derived peptides, GLP-2 appears to be a major mediator of intestinal epithelial proliferation.
Collapse
Affiliation(s)
- M A Ghatei
- Department of Endocrinology and Metabolism, Imperial College School of Medicine, The Hammersmith Hospital, London, UK
| | | | | | | | | | | | | |
Collapse
|
19
|
Berlanga-Acosta J, Playford RJ, Mandir N, Goodlad RA. Gastrointestinal cell proliferation and crypt fission are separate but complementary means of increasing tissue mass following infusion of epidermal growth factor in rats. Gut 2001; 48:803-7. [PMID: 11358899 PMCID: PMC1728321 DOI: 10.1136/gut.48.6.803] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Epidermal growth factor (EGF) is a potent mitogen for the gastrointestinal tract and also influences the number of new crypts formed by crypt fission. The time course of these events and possible linkage between these two complementary mechanisms is however poorly understood. We therefore examined the temporal relationship of proliferation and fission in rats treated with EGF. METHODS Osmotic minipumps were implanted subcutaneously into male Wistar rats to infuse EGF continuously (60 microg/rat/day) for periods of 1-14 days. Proliferation and crypt branching were quantified following vincristine induced metaphase arrest and morphometric assessment of microdissected tissue. RESULTS In the small intestine, EGF significantly increased epithelial cell proliferation and crypt and villus area after 24 hours of EGF, although maximal effects were only reached following six days of infusion. EGF also resulted in an approximate 30% reduction in crypt fission in the small bowel. In the colon, EGF caused a twofold increase in epithelial cell proliferation one day after infusion, from 15.3 (2.3) to 29.6 (3.5) metaphases per crypt (p<0.01). Maximal effects were seen in rats receiving EGF for seven days. For all time points, colonic crypt size increased in response to EGF. The amount of branching increased following one day of infusion with EGF (from 15.3 (1.9) to 32.4 (5.5)%; p<0.001) but was significantly lower (approximately 25% of control values) following longer periods of infusion. Crypt fission did not correlate with crypt area. CONCLUSION EGF has profound effects on cell proliferation and also altered crypt fission, with its actions on crypt fission most pronounced in the colon where it first increased and then decreased fission. EGF can thus be a potent stimulus for crypt fission during short term infusion and may reduce the number of branched crypts present in a resting or quiescent stage. Growth factors can alter cell mass by two separate but linked mechanisms, namely altered cell production and crypt fission.
Collapse
Affiliation(s)
- J Berlanga-Acosta
- Center for Genetic Engineering and Biotechnology, PO Box 6162, Havana, Cuba.
| | | | | | | |
Collapse
|
20
|
Westphal V, Murch S, Kim S, Srikrishna G, Winchester B, Day R, Freeze HH. Reduced heparan sulfate accumulation in enterocytes contributes to protein-losing enteropathy in a congenital disorder of glycosylation. THE AMERICAN JOURNAL OF PATHOLOGY 2000; 157:1917-25. [PMID: 11106564 PMCID: PMC1885788 DOI: 10.1016/s0002-9440(10)64830-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Intestinal biopsy in a boy with gastroenteritis-induced protein-losing enteropathy (PLE) showed loss of heparan sulfate (HS) and syndecan-1 core protein from the basolateral surface of the enterocytes, which improved after PLE subsided. Isoelectric focusing analysis of serum transferrin indicated a congenital disorder of glycosylation (CDG) and subsequent analysis showed three point mutations in the ALG6 gene encoding an alpha1,3-glucosyltransferase needed for the addition of the first glucose to the dolichol-linked oligosaccharide. The maternal mutation, C998T, causing an A333V substitution, has been shown to cause CDG-Ic, whereas the two paternal mutations, T391C (Y131H) and C924A (S308R) have not previously been reported. The mutations were tested for their ability to rescue faulty N:-linked glycosylation of carboxypeptidase Y in an ALG6-deficient Saccharomyces cerevisiae strain. Normal human ALG6 rescues glycosylation and A333V partially rescues, whereas the combined paternal mutations (Y131H and S308R) are ineffective. Underglycosylation resulting from each of these mutations is much more severe in rapidly dividing yeast. Similarly, incomplete protein glycosylation in the patient is most severe in rapidly dividing enterocytes during gastroenteritis-induced stress. Incomplete N:-linked glycosylation of an HS core protein and/or other biosynthetic enzymes may explain the selective localized loss of HS and PLE.
Collapse
Affiliation(s)
- V Westphal
- The Burnham Institute, La Jolla, California. Royal Free and University College School of Medicine, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
21
|
Wright NA. Epithelial stem cell repertoire in the gut: clues to the origin of cell lineages, proliferative units and cancer. Int J Exp Pathol 2000; 81:117-43. [PMID: 10762441 PMCID: PMC2517719 DOI: 10.1046/j.1365-2613.2000.00146.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/1999] [Accepted: 01/11/2000] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal stem cells are shown to be pluripotential and to give rise to all cell lineages in the epithelium. After damage, gut stem cells produce reparative cell lineages that produce a wide range of peptides with important actions on cell proliferation and migration, and promote regeneration and healing. Increase in stem cell number is considered to induce crypt fission, and lead to increases in the number of crypts, even in the adult; it is also the mode of spread of mutated clones in the colorectal mucosa. Stem cell repertoire is defined by both intrinsic programming of the stem cell itself, but signalling from the mesenchyme is also vitally important for defining both stem cell progeny and proliferation. Carcinogenesis in the colon occurs through sequential mutations, possibly occurring in a single cell. A case is made for this being the stem cell, but recent studies indicate that several stem cells may need to be so involved, since early lesions appear to be polyclonal in derivation.
Collapse
Affiliation(s)
- N A Wright
- ICRF Histopathology Unit, Lincoln's Inn Fields and Department of Histopathology, Imperial College School of Medicine, Hammersmith Hospital, London, UK
| |
Collapse
|
22
|
Goodlad RA, Mandir N, Meeran K, Ghatei MA, Bloom SR, Playford RJ. Does the response of the intestinal epithelium to keratinocyte growth factor vary according to the method of administration? REGULATORY PEPTIDES 2000; 87:83-90. [PMID: 10710291 DOI: 10.1016/s0167-0115(99)00113-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND Keratinocyte growth factor (KGF) is a potent mitogen and may be of value for the treatment of conditions such as short bowel syndrome and chemotherapy-induced mucositis. However the most efficacious route and method of administration is unclear. METHODS Rats maintained by total parenteral nutrition (TPN) were given KGF (1 mg/kg/rat/day, i.v.) infused continuously or as a once-daily injection. The same dose was also given s.c. to chow-fed rats. Changes in gut growth were assessed by measurement of wet weight, proliferation (vincristine induced metaphase arrest) and crypt branching index. Changes in gut hormone profile were also determined to examine if any trophic effects were mediated via this mechanism. RESULTS KGF caused a 70-100% increase in wet weight of the stomach, small and large intestine of TPN-fed rats (P < 0.01) with no significant differences seen between the two methods of administration. The increase in metaphase counts was greatest in the stomach (about seven-fold P < 0.01), but was less pronounced in the distal small intestine and colon (about 50% increase). The trophic effect of KGF was much less prominent in orally-fed rats. Crypt branching index was significantly reduced by KGF in the proximal small intestine of TPN, but not orally-fed rats. Plasma gastrin, PYY, total glucagon, enteroglucagon and GLP-1 all increased by two-three-fold (all P < 0.01) in response to KGF whereas insulin levels fell by about 25% in the TPN group. CONCLUSIONS The mitogenic action of KGF occurred predominantly in the stomach and proximal small intestine. Its efficacy was less pronounced in orally-fed animals, suggesting KGF may be of greatest benefit in conditions associated with lowered intestinal proliferation. Clinical trials of KGF can probably use single daily i.v. injections without reduction in efficacy.
Collapse
Affiliation(s)
- R A Goodlad
- Histopathology Unit, Imperial Cancer Research Fund, London, UK.
| | | | | | | | | | | |
Collapse
|
23
|
Pillai SB, Hinman CE, Luquette MH, Nowicki PT, Besner GE. Heparin-binding epidermal growth factor-like growth factor protects rat intestine from ischemia/reperfusion injury. J Surg Res 1999; 87:225-31. [PMID: 10600353 DOI: 10.1006/jsre.1999.5764] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND We have shown previously that heparin-binding epidermal growth factor (EGF)-like growth factor (HB-EGF) is cytoprotective for intestinal epithelial cells exposed to hypoxia in vitro. We now examine the effects of HB-EGF on the recovery of small intestine from ischemic injury in vivo. METHODS Segmental intestinal ischemia of 60-min duration was produced in adult rats by occlusion of a first-order branch of the superior mesenteric artery. Recombinant HB-EGF (100 microg) was injected intraluminally into the proximal small bowel after 45 min of ischemia in experimental animals, and buffered saline was injected in control animals. Animals were sacrificed after 48 h, and the affected bowel was resected, processed, and examined microscopically, with histologic grading of the ischemic injury. Additional animals were allowed to recover for up to 1 month to evaluate mortality differences. RESULTS Intraluminal administration of HB-EGF resulted in significantly decreased extent and severity of ischemia/reperfusion injury, with significantly decreased grade of injury in the HB-EGF-treated compared with nontreated animals (average injury grade 0.66 compared with 2.44, respectively). Moreover, the mortality rate was significantly lower in the HB-EGF-treated animals compared with nontreated animals (0% vs 25%, respectively). HB-EGF-treated animals had increased weight gain in the postischemia recovery period. CONCLUSIONS We conclude that HB-EGF, given intraluminally, reduces both the amount and the severity of ischemia/reperfusion injury in the small bowel, reduces the mortality associated with intestinal ischemia, and may enhance intestinal recovery. The in vitro and in vivo cytoprotective effects of this growth factor suggest that it may, in the future, be clinically useful in treating patients with intestinal ischemia.
Collapse
Affiliation(s)
- S B Pillai
- Department of Surgery, The Ohio State University and Children's Hospital, Columbus, Ohio 43205, USA
| | | | | | | | | |
Collapse
|
24
|
Wong WM, Wright NA. Epidermal growth factor, epidermal growth factor receptors, intestinal growth, and adaptation. JPEN J Parenter Enteral Nutr 1999; 23:S83-8. [PMID: 10483903 DOI: 10.1177/014860719902300521] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Epidermal growth factor (EGF) is an important constituent of several gastrointestinal secretions. Many studies in both animals and humans have shown EGF to have multiple effects upon gut epithelial cells. These include cytoprotection, stimulatory effects on cell proliferation and migration, induction of gene expression such as mucosal enzymes and trefoil proteins, and inhibitory effects on gastric acid secretion. The main conundrum associated with EGF action is the disparity between experimental studies on its luminal and systemic actions. Opinion is sharply divided as to whether or not EGF has any action when given luminally and on the site of the EGF receptors on gut epithelial cells. Most studies agree that the EGF receptors are located on the basolateral surface, in which case EGF should only be active if surface ulceration has occurred--unless there is translocation across the epithelium. There are several clinical situations in which EGF might be useful in cytoprotection and in stimulating repair and regeneration in the gut. These include necrotizing enterocolitis and mucositis, and it is to be hoped that the solid basis of experimental studies on EGF might stimulate work on this topic.
Collapse
Affiliation(s)
- W M Wong
- Department of Histopathology, Imperial College of Science, Technology and Medicine, London, England, UK
| | | |
Collapse
|
25
|
Abstract
BACKGROUND Several papers have indicated that glutamine is a preferred fuel for the enterocyte and that it can increase intestinal epithelial cell proliferation. AIMS To investigate the effects of glutamine on intestinal epithelial cell proliferation in the parenterally fed rat. METHODS Five groups of six rats were fed parenterally; a group of orally fed rats was also studied. Crypt cell proliferation was studied after six days using native mitoses in microdissected crypts and bromodeoxyuridine labelling. RESULTS No effect of treatment was seen on intestinal weight; however, the weights of the small intestine, caecum, and colon were all significantly heavier in the orally fed group than in the total parenteral nutrition groups (p<0.001). There was no effect of any of the glutamine treatments on mitotic activity in the small intestine. In the colon there was a small increase in native mitoses with glutamine (p=0.03). There was also an indication of increased proliferative activity in the first fifth of the small intestine and colon with glutamine. Little effect of glutamine on bromodeoxyuridine labelling in either site was observed, but there was a small but significant reduction in growth fraction of the colon of the glutamine treated group. The labelling distribution curve from sections and the mitotic distribution curve obtained from crypt squashes showed a good correlation. CONCLUSION Glutamine has a small, but significant effect on mitotic activity but only in the colon. Modest effects on the distribution of labelled cells were also seen.
Collapse
Affiliation(s)
- N Mandir
- Histopathology Unit, Imperial Cancer Research Fund, 44 Lincoln's Inn Fields, London WC2A 3PX, UK
| | | |
Collapse
|
26
|
Abstract
Gastrointestinal cell proliferation plays an important role in the maintenance of the integrity of the gastrointestinal system. The study of gastrointestinal proliferation kinetics allows a better understanding of the complexity of the system, and also has important implications for the study of gastrointestinal carcinogenesis. Gastrointestinal stem cells are shown to be pluripotential and to give rise to all cell lineages in the epithelium. Carcinogenesis in the colon occurs through sequential mutations, possibly occurring in a single cell--the stem cell.
Collapse
Affiliation(s)
- W M Wong
- Department of Histopathology, Imperial College of Science, Technology and Medicine, London, UK
| | | |
Collapse
|
27
|
Jordinson M, Goodlad RA, Brynes A, Bliss P, Ghatei MA, Bloom SR, Fitzgerald A, Grant G, Bardocz S, Pusztai A, Pignatelli M, Calam J. Gastrointestinal responses to a panel of lectins in rats maintained on total parenteral nutrition. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:G1235-42. [PMID: 10330015 DOI: 10.1152/ajpgi.1999.276.5.g1235] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Total parenteral nutrition (TPN) causes atrophy of gastrointestinal epithelia, so we asked whether lectins that stimulate epithelial proliferation can reverse this effect of TPN. Two lectins stimulate pancreatic proliferation by releasing CCK, so we asked whether lectins that stimulate gastrointestinal proliferation also release hormones that might mediate their effects. Six rats per group received continuous infusion of TPN and a once daily bolus dose of purified lectin (25 mg. rat-1. day-1) or vehicle alone (control group) for 4 days via an intragastric cannula. Proliferation rates were estimated by metaphase arrest, and hormones were measured by RIAs. Phytohemagglutinin (PHA) increased proliferation by 90% in the gastric fundus (P < 0.05), doubled proliferation in the small intestine (P < 0.001), and had a small effect in the midcolon (P < 0.05). Peanut agglutinin (PNA) had a minor trophic effect in the proximal small intestine (P < 0.05) and increased proliferation by 166% in the proximal colon (P < 0.001) and by 40% in the midcolon (P < 0.001). PNA elevated circulating gastrin and CCK by 97 (P < 0.05) and 81% (P < 0.01), respectively, and PHA elevated plasma enteroglucagon by 69% and CCK by 60% (both P < 0.05). Only wheat germ agglutinin increased the release of glucagon-like peptide-1 by 100% (P < 0.05). PHA and PNA consistently reverse the fall in gastrointestinal and pancreatic growth associated with TPN in rats. Both lectins stimulated the release of specific hormones that may have been responsible for the trophic effects. It is suggested that lectins could be used to prevent gastrointestinal atrophy during TPN. Their hormone-releasing effects might be involved.
Collapse
Affiliation(s)
- M Jordinson
- Imperial College School of Medicine, Division of Investigative Sciences, Hammersmith Hospital, London W12 0NN, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Helmrath MA, Shin CE, Fox JW, Erwin CR, Warner BW. Adaptation after small bowel resection is attenuated by sialoadenectomy: the role for endogenous epidermal growth factor. Surgery 1998. [PMID: 9823398 DOI: 10.1016/s0039-6060(98)70008-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Epidermal growth factor (EGF) is likely involved during adaptation after small bowel resection (SBR) because some studies have shown enhanced adaptation by EGF administration. Because the major source of endogenous EGF in mice is the submandibular glands, we sought to determine the effect of submandibular gland excision (SAL) and luminal or systemic EGF replacement on adaptation after SBR. METHODS A 50% proximal SBR or Sham-SBR (bowel transection and reanastomosis) was performed on male C57BL/6 mice after either SAL or gland mobilization only. Additional mice underwent both SBR and SAL and then received daily EGF or saline solution by intraperitoneal or orogastric administration. At 1 week, adaptation was characterized in the ileum as changes in villus height, DNA, and protein content. RESULTS SAL significantly attenuated the increase in ileal villus height, total protein, and DNA content after SBR. Both systemic and oral EGF reversed these findings equally and significantly augmented all parameters of intestinal adaptation after SAL. CONCLUSIONS Submandibular EGF is important for the adaptive response to massive SBR. As both luminal and systemic EGF equally reversed the findings following SAL and SBR, the specific site of action for endogenous EGF during adaptation is either the luminal or basolateral surface of the enterocyte.
Collapse
Affiliation(s)
- M A Helmrath
- Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Ohio 45229-3039, USA
| | | | | | | | | |
Collapse
|
29
|
Abstract
Growth factors play a key role in maintaining gut integrity and in stimulating repair. By understanding their physiological function we can use this information to apply to the treatment of conditions in which mucosal homeostasis has broken down.
Collapse
Affiliation(s)
- N J Seare
- Research Office, Leicester General Hospital, UK.
| | | |
Collapse
|
30
|
Acra SA, Bulus N, Bogatcheva G, Coffey RJ, Barnard JA. Increased intestinal epithelial proliferation in metallothioneine-transforming growth factor alpha transgenic mice. REGULATORY PEPTIDES 1998; 74:105-12. [PMID: 9712170 DOI: 10.1016/s0167-0115(98)00029-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIM The epidermal growth factor (EGF) peptide family includes six closely-related proteins, all of which bind to the EGF receptor. In the intestinal epithelium, transforming growth factor alpha (TGFalpha) appears to be the most physiological ligand for the EGF receptor. The present studies were designed to examine the effect of TGFalpha overexpression on duodenal epithelial proliferation using a metallothioneine-inducible promoter/enhancer transgenic mouse (MT-TGFalpha). The MT-TGFalpha mouse model was further studied to determine the in vivo effect of unregulated TGFalpha production on the physiological proliferative responses to fasting and refeeding. METHODS MT-TGFalpha mice were given 25 mM oral ZnSO4 to induce transgene expression and were studied 1 to 2 months later. Duodenal histology was analyzed morphometrically in well-oriented transverse sections. The vincristine metaphase-arrest technique was used to assess proliferation in duodenal crypts. Immunohistochemical staining and in situ hybridization were used to assess transgenic TGFalpha protein and mRNA expression, respectively. RESULTS Normally fed MT-TGFalpha mice had deeper crypts (0.12 vs. 0.08 mm), longer villi (0.66 vs. 0.54 mm), and greater luminal diameters (2.65 vs. 2.19 mm) compared to controls (P<0.05 for all three dimensions). The crypt cell mitotic index in normally fed transgenic mice was 1.5 fold greater than the index in normally fed controls (20+/-2 vs. 35+/-4 mitoses per crypt; P <0.05). Fasting and refeeding MT-TGFalpha mice resulted in no significant change in their high baseline rate of crypt proliferation, while proliferation in control mice rose from a lower baseline during fasting to a level with refeeding that approximated rates in MT-TGFalpha mice. Transgenic TGFalpha protein and mRNA were localized to the villus epithelial compartment with little or no evidence of mRNA or protein expression in the crypt epithelium. CONCLUSION Overproduction of TGFalpha in the mouse duodenal epithelium results in a pronounced increase in crypt epithelial cell proliferation and a resulting increase in the dimension of the crypt/villus unit. This appears to be mediated through a paracrine and/or juxtacrine effect on crypt cells by TGFalpha produced in the villus epithelium. Fasting and refeeding experiments suggest that TGFalpha may also play a role in the proliferative response to refeeding or that the full potential for proliferation is realized by TGFalpha overexpression alone. Collectively, these studies suggest that TGFalpha is a physiological autocrine and paracrine regulator of small intestinal epithelial proliferation.
Collapse
Affiliation(s)
- S A Acra
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232 2576, USA
| | | | | | | | | |
Collapse
|
31
|
Wright NA. Aspects of the biology of regeneration and repair in the human gastrointestinal tract. Philos Trans R Soc Lond B Biol Sci 1998; 353:925-33. [PMID: 9684290 PMCID: PMC1692279 DOI: 10.1098/rstb.1998.0257] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The main pathways of epithelial differentiation in the intestine, Paneth, mucous, endocrine and columnar cell lineages are well recognized. However, in abnormal circumstances, for example in mucosal ulceration, a cell lineage with features distinct from these emerges, which has often been dismissed in the past as 'pyloric' metaplasia, because of its morphological resemblance to the pyloric mucosa in the stomach. However, we can conclude that this cell lineage has a defined phenotype unique in gastrointestinal epithelia, has a histogenesis that resembles that of Brunner's glands, but acquires a proliferative organization similar to that of the gastric gland. It expresses several peptides of particular interest, including epidermal growth factor, the trefoil peptides TFF1, TFF2, TFF3, lysozyme and PSTI. The presence of this lineage also appears to cause altered gene expression in adjacent indigenous cell lineages. We propose that this cell lineage is induced in gastrointestinal stem cells as a result of chronic mucosal ulceration, and plays an important part in ulcer healing; it should therefore be added to the repertoire of gastrointestinal stem cells.
Collapse
Affiliation(s)
- N A Wright
- Histopathology Unit, ICRF Laboratories, London, UK
| |
Collapse
|
32
|
|
33
|
Park HS, Goodlad RA, Ahnen DJ, Winnett A, Sasieni P, Lee CY, Wright NA. Effects of epidermal growth factor and dimethylhydrazine on crypt size, cell proliferation, and crypt fission in the rat colon. Cell proliferation and crypt fission are controlled independently. THE AMERICAN JOURNAL OF PATHOLOGY 1997; 151:843-52. [PMID: 9284833 PMCID: PMC1857833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Crypt fission is now established as an important mechanism of intestinal growth and regeneration. It has been proposed that increased crypt size is the stimulus for crypt fission, because crypts preparing for fission are generally larger. Consequently, we investigated the effects of epidermal growth factor (EGF) and dimethylhydrazine, which are both known to stimulate crypt cell proliferation, on crypt fission in the rat intestine. We also examined whether the effects of EGF on both proliferation and crypt fission are modified by the pretreatment with dimethylhydrazine for 16 weeks, dimethylhydrazine was then discontinued for 8 weeks, followed by intravenous infusion of EGF for 1 week. There were four groups: vehicle alone, EGF alone, dimethylhydrazine alone, and dimethylhydrazine followed by EGF infusion. The rats were killed at 25 weeks and rates of intestinal crypt cell production, crypt size, and crypt fission were determined. Intravenously infused EGF significantly increased crypt cell production rate, but the magnitude of the effect decreased from the proximal to the distal colon. EGF caused an increase in crypt area, possibly reflecting an increase in crypt size. Importantly dimethylhydrazine had no significant effect on crypt cell production rate nor on crypt area in the distal colon, but it did cause an increase in crypt area in the mid-colon. The crypt fission index was significantly decreased by EGF and increased by dimethylhydrazine. There was no qualitative interaction between EGF and dimethylhydrazine. These results demonstrate the marked proliferative effect of intravenously infused EGF in the colon of orally fed rats, with significant site effects (P = 0.0007); the effect was greatest in the proximal colon and disappeared in the distal colon. The observation that EGF reduced crypt fission indicates that increased cell proliferation, per se, is not a stimulus for crypt fission. This is further supported by the observation that dimethylhydrazine increases crypt fission in crypts of normal size in the distal colon without significantly increasing cell proliferation. These results suggest that increasing crypt cellularity by proliferation is not sufficient to induce crypt fission, and factors other than increased crypt size by proliferation can control crypt fission. It is also probable that cell proliferation and crypt fission are independently regulated. Crypt fission appears to play a considerable role in the intestinal response to carcinogens.
Collapse
Affiliation(s)
- H S Park
- Department of Histopathology, Royal Postgraduate Medical School, Hammersmith Hospital, London, UK
| | | | | | | | | | | | | |
Collapse
|
34
|
Warner BW, Vander Kolk WE, Can G, Helmrath MA, Shin CE, Erwin CR. Epidermal growth factor receptor expression following small bowel resection. J Surg Res 1997; 70:171-7. [PMID: 9245568 DOI: 10.1006/jsre.1997.5125] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Adaptation following massive small bowel resection (SBR) is an important compensatory response. While epidermal growth factor (EGF) has been shown to augment this response, the mechanism and role of EGF and its intestinal receptor (EGF-R) during adaptation are not known. The purpose of this study was to determine the effect of massive SBR and adaptation on intestinal expression of EGF-R. MATERIALS AND METHODS Male Sprague-Dawley rats underwent either a 75% mid-SBR with primary reanastomosis or sham operation (bowel transection with reanastomosis). Ileal mucosa was harvested from animals of each group at 6 and 12 hr, 1, 3, and 5 days, and 1, 2, and 4 weeks after operation. Expression of EGF-R protein was studied by Western blotting. Expression of EGF-R mRNA was determined by quantitative reverse-transcriptase polymerase chain reactions normalized to beta-actin. RESULTS When compared with sham, SBR resulted in a modest (32%) increase in the expression of EGF-R mRNA at 1 week (P < 0.003). A twofold greater expression of EGF-R protein corresponded to this time point. While not statistically significant, expression of EGF-R mRNA was slightly greater after SBR at every other time point measured. CONCLUSION Following massive SBR, expression of both EGF-R mRNA and protein is slightly increased in the ileum with a more pronounced increase in protein. These findings, which have not previously been reported, suggest that changes in EGF-R signaling may not play a major role during the initiation and or progression of intestinal adaptation following massive SBR.
Collapse
Affiliation(s)
- B W Warner
- Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Ohio 45229, USA
| | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Lynch DA, Mapstone NP, Lewis F, Pentith J, Axon AT, Dixon MF, Quirke P. Serum and gastric luminal epidermal growth factor in Helicobacter pylori-associated gastritis and peptic ulcer disease. Helicobacter 1996; 1:219-26. [PMID: 9398872 DOI: 10.1111/j.1523-5378.1996.tb00042.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Helicobacter pylori is the cause of chronic (type B) gastritis, duodenal ulceration (DU), and gastric ulceration (GU). Smoking is associated with delayed ulcer healing. Epidermal growth factor (EGF) is produced in the salivary and Brunner's glands of the upper gastrointestinal tract, inhibits gastric acid secretion, and is a powerful mitogen. MATERIALS AND METHODS We sought to determine gastric luminal EGF (GL-EGF) in smokers and patients with Hp-associated DU and the effects of Hp eradication. Our aim was to determine GL-EGF in patients with GU and the effect of ulcer healing and to measure serum EGF in patients with Hp gastritis with or without DU disease. RESULTS GL-EGF was reduced in smokers compared to controls (p = .008). Subjects with HP gastritis had reduced GL-EGF compared to controls (p = .0002). There was no difference in GL-EGF between Hp-positive subjects who had DU and those with chronic gastritis alone. Eradication of Hp from those patients with DU had no effect on the low levels of GL-EGF. There was no difference between GL-EGF in Hp gastritis alone and in Hp-associated active GU. GL-EGF fell after ulcer healing (p = .04), a difference confirmed by analysis of paired samples from patients before and after ulcer healing (p = .03). There was no difference in serum EGF between controls and subjects with Hp infection. There was no difference in serum EGF in subjects with DU associated and non-ulcer-associated gastritis. CONCLUSIONS Subjects with Hp gastritis, or those who smoke, had low concentrations of GL-EGF regardless of whether DU was present. Eradication of Hp did not return the concentrations of GL-EGF to normal in DU subjects. Individuals and Hp gastritis and inactive GU had low levels of GL-EGF compared to non-ulcer Hp infection. The relative increase in GL-EGF that occurred with ulceration of the gastric mucosa may have resulted from the development of an ulcer-associated cell lineage. Serum EGF did not play a role in the pathogenesis of Hp gastritis or of associated DU ulcer disease.
Collapse
Affiliation(s)
- D A Lynch
- Centre for Digestive Diseases, General Infirmary at Leeds, England
| | | | | | | | | | | | | |
Collapse
|
37
|
Wong ME, Hollinger JO, Pinero GJ. Integrated processes responsible for soft tissue healing. ORAL SURGERY, ORAL MEDICINE, ORAL PATHOLOGY, ORAL RADIOLOGY, AND ENDODONTICS 1996; 82:475-92. [PMID: 8936509 DOI: 10.1016/s1079-2104(96)80190-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Wounded soft tissues undergo repair through a complex series of interrelated events that involve both physical and chemical activities. These processes are currently undergoing extensive investigation as efforts are directed toward achieving augmented and accelerated healing. Early wound-healing research focused on expanding traditional histologic descriptions of tissue healing by attempting to characterize the environment and biologic mediators responsible for healing. These initial studies successfully identified a number of agents and physiochemical factors present in healing wounds, but their precise roles and importance remain largely unknown. This review article summarizes the current literature on soft tissue healing. An effort has been made to correlate the activities of the major growth factors and cytokines with the individual reparative processes including the inflammatory response, hemostasis, fibroplasia, angiogenesis, and remodeling. Explanations and characteristics of growth factor function as well as brief descriptions of several major factors and their spectrum of activity are also provided.
Collapse
Affiliation(s)
- M E Wong
- Division of Oral and Maxillofacial Surgery, LBJ General Hospital, University of Texas Health Science Center at Houston, USA
| | | | | |
Collapse
|
38
|
Reindel JF, Pilcher GD, Gough AW, Haskins JR, de la Iglesia FA. Recombinant human epidermal growth factor1-48-induced structural changes in the digestive tract of cynomolgus monkeys (Macaca fascicularis). Toxicol Pathol 1996; 24:669-80. [PMID: 9082544 DOI: 10.1177/019262339602400601] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To determine the cellular effects and potential toxicity of exogenously administered recombinant human epidermal growth factor1-48 (EGF1-48) in primates, intravenous bolus injections were given to 2 cynomolgus monkeys per sex at 0 (vehicle control). 10, 100, 500 (females only), and 1,000 micrograms/kg/day (males only) for up to 2 wk. Males given the suprapharmacologic dose of 1,000 micrograms/kg did not tolerate treatment and were necropsied after 5 days of dosing. All other monkeys completed the 2-wk study. Necropsy findings included enlarged, discolored, pale tan livers at 500 micrograms/kg and greater, firm, thickened pancreata in 500-micrograms/kg females, and enlarged salivary glands at all doses. Relative liver weights were increased at 500 and 1,000 micrograms/kg: mean salivary gland weights in all dose groups were greater than in controls. Histopathologic changes were primarily those of diffuse epithelial cell hypertrophy and hyperplasia in liver (hepatocytes and biliary tract), pancreas, salivary glands, tongue, esophagus, stomach, small and large intestine, and gallbladder. Alterations were dose-related in intensity and occurred in at last some tissues at the lowest dose. In gastric glands, colon crypts, pancreatic ducts, biliary tract, and salivary glands, differentiated epithelial cells were replaced by cells of less differentiated phenotype. These morphologic alterations were consistent with exuberant proliferation induced by this epithelial mitogen. The extent of the proliferative response in tissues of the digestive tract attests to the potency of this fragment of human EGF1-53 in primates. Furthermore, the epithelial proliferation was significantly greater than that reported previously in EGF-treated rodents.
Collapse
Affiliation(s)
- J F Reindel
- Department of Pathology and Experimental Toxicology, Parke-Davis Pharmaceutical Research Division of Warner-Lambert Company, Ann Arbor, Michigan 48105, USA
| | | | | | | | | |
Collapse
|
39
|
Baskin LS, Hayward SW, Sutherland RA, DiSandro MJ, Thomson AA, Goodman J, Cunha GR. Mesenchymal-epithelial interactions in the bladder. World J Urol 1996; 14:301-9. [PMID: 8912470 DOI: 10.1007/bf00184602] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
During bladder development, undifferentiated mesenchymal and epithelial cells undergo an orderly sequence of differentiation defined by the expression of smooth-muscle (alpha-actin, myosin, vinculin, desmin, vimentin, and laminin) and epithelial (cytokeratins 5, 7, 8, 14, 18 and 19) protein markers. This process requires mesenchymal-epithelial interactions with bladder epithelium (urothelium) necessary for the differentiation of bladder smooth muscle. Peptide growth factors such as keratinocyte growth factor (KGF) and transforming growth factors (TGF) alpha and beta are likely candidates as mediators of these mesenchymal-epithelial interactions. Transcripts for KGF, TGF alpha, and TGF beta are regulated during bladder development and during smooth-muscle hypertrophy secondary to bladder-outlet obstruction. Finally, two experimental bladder models--(1) partial outlet obstruction and (2) regeneration of bladder smooth muscle into an acellular tissue matrix--are described in the context of mesenchymal-epithelial interactions in the bladder.
Collapse
Affiliation(s)
- L S Baskin
- Department of Urology, University of California, San Francisco 94143, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Playford RJ, Hanby AM, Gschmeissner S, Peiffer LP, Wright NA, McGarrity T. The epidermal growth factor receptor (EGF-R) is present on the basolateral, but not the apical, surface of enterocytes in the human gastrointestinal tract. Gut 1996; 39:262-6. [PMID: 8977341 PMCID: PMC1383309 DOI: 10.1136/gut.39.2.262] [Citation(s) in RCA: 113] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND While it is clear that luminal epidermal growth factor (EGF) stimulates repair of the damaged bowel, its significance in maintaining normal gut growth remains uncertain. If EGF is important in maintaining normal gut growth, the EGF receptor (EGF-R) should be present on the apical (luminal) surface in addition to the basolateral surface. AIMS/SUBJECTS/METHODS: This study examined the distribution of the EGF-R in the epithelium throughout the human gastro-intestinal tract using immunohistochemistry, electron microscopy, and western blotting of brush border preparations. RESULTS Immunostaining of the oesophagus showed circumferential EGF-R positivity in the cells of the basal portions of the stratified squamous epithelium but surface cells were EGF-R negative. In the normal stomach, small intestine, and colon, immunostaining localised the receptor to the basolateral surface with the apical membranes being consistently negative. EGF-R positivity within the small intestine appeared to be almost entirely restricted to the proliferative (crypt) region. Western blotting demonstrated a 170 kDa protein in whole tissue homogenates but not in the brush border vesicle preparations. CONCLUSIONS As the EGF-R is located only on the basolateral surfaces in the normal adult gastrointestinal tract, the major role of luminal EGF is probably to stimulate repair rather than to maintain normal gut growth.
Collapse
Affiliation(s)
- R J Playford
- Department of Medicine and Therapeutics, University of Leicester
| | | | | | | | | | | |
Collapse
|
41
|
Sizemore N, Dudeck RC, Barksdale CM, Nordblom GD, Mueller WT, McConnell P, Wright DS, Guglietta A, Kuo BS. Development and validation of two solid-phase enzyme immunoassays (ELISA) for quantitation of human epidermal growth factors (hEGFs). Pharm Res 1996; 13:1088-94. [PMID: 8842051 DOI: 10.1023/a:1016075128086] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE The purpose of the present investigation was to develop and validate two separate enzyme-linked immunosorbent assays (ELISA) for quantitation of exogenous human epidermal growth factor (hEGF1-53) and its truncated fragment (hEGF1-48) in rat plasma. METHODS The present assay systems were based on the sandwiching of the antigen between a monoclonal mouse anti-hEGF1-53 antibody, pre-coated on a 96-well polystyrene plate, and a polyclonal rabbit anti-hEGF1-48 antibody, which is then detected with a peroxidase-labeled goat anti-rabbit antibody. RESULTS The calibration curves for hEGF1-48 and hEGF1-53 in plasma were validated over a concentration range of 7.8-250 and 62.5-1000 pg/ml, respectively. Determined from replicate assays of hEGF1-48 quality control samples, the intra-assay precision and accuracy were < or = 8.8% RSD and within +/- 9.8%; and the inter-assay precision and accuracy were < or = 14.8% RSD and within +/- 9.7% RE, respectively. Determined from replicate assays of hEGF1-53 quality control samples, the intra-assay precision and accuracy were < or = 10.0% RSD and within +/- 8.5%; and the inter-assay precision and accuracy were < or = 10.0% RSD and within +/- 5.7% RE, respectively. The limit of quantitation of the hEGF1-48 and hEGF1-53 assay using 200 microliters plasma per well is 7.8 and 62.5 pg/ml, respectively. These two ELISA methods are specific to hEGFs and do not cross-react with mouse EGF or other growth factors (TGF alpha, TGF beta, PDGF, and FGF) or lymphokines (IL1 beta and TNF alpha). These validated methods have been routinely applied to assay of plasma samples from various pharmacokinetic studies in rats receiving intravenous hEGFs. Both assay methods were also adapted to assay endogenous hEGFs in biological fluids of different animal species. CONCLUSIONS Two sensitive ELISA methods have been validated for quantitation of hEGF1-53 and hEGF1-48 in rat plasma. Their utility has been demonstrated in the application of assaying immunoreactive concentration of exogenous and endogenous epidermal growth factors.
Collapse
Affiliation(s)
- N Sizemore
- Department of Oncology, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kissmeyer-Nielsen P, Vinter-Jensen L, Smerup M. Effects of longterm epidermal growth factor treatment on the normal rat colon. Gut 1996; 38:582-6. [PMID: 8707092 PMCID: PMC1383119 DOI: 10.1136/gut.38.4.582] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Epidermal growth factor (EGF) exerts trophic effects on the mucosa of damaged and defunctioned colon, but the effects on the normal large bowel wall are not known. AIMS To investigate the effect of systemic EGF treatment on growth and morphology of normal rat colon. METHODS Rats were treated with subcutaneous biosynthetic EGF injections of 150 micrograms/kg/day for 28 days. The weight of the histological colonic wall layers and the luminal surface area were measured using quantitative morphometric analysis (stereology). The colon was subdivided into proximal and distal parts. RESULTS EGF treatment increased the total colon wet weight by 23% compared with controls (p < 0.005). The weight increase occurred in the mucosal (33%) and the submucosal layers of the bowel wall (36%) and there was a 69% increase of the total luminal surface area (p = 0.001). In the proximal part of colon of EGF rats there was a 68% increase in mucosal weight (p < 0.005) accompanied by a 79% increase in the mucosal surface area compared with controls (p < 0.005), whereas submucosal and muscularis propria weights were identical. In distal colon, the mucosal weight increased 28% in the EGF group (p < 0.005), the mucosal surface area increased by 72% after treatment (p < 0.01). Furthermore there was a 34% increase in the weight of submucosa (p < 0.001) in the distal colon among EGF rats. CONCLUSIONS Treatment of rats with EGF has a stimulating role on the mucosa and luminal surface area of the entire functioning colon and a trophic effect on the submucosa of the distal colon.
Collapse
Affiliation(s)
- P Kissmeyer-Nielsen
- Department of Surgery L, Amtssygehuset, University Hospital of Aarhus, Denmark
| | | | | |
Collapse
|
43
|
Abstract
Despite the wealth of information concerning EGF and its related peptides, its precise role in the control of gastrointestinal functions is still not fully resolved. However, there is no doubt that it can have some very potent effects on the gastrointestinal tract. These may be related to the control of growth and development and to the regular control of cell renewal. Nevertheless, in the adult, EGF may only be active in response to luminal damage and repair, and furthermore this may also only occur if the luminal EGF is protected from proteolytic degradation. Notwithstanding this, 'EGF'-like responses may be evoked in the gut by intestinal TGF-alpha. The possible therapeutic use of EGF and members of its family in ulcer therapy will be discussed in later Chapters of this volume, other potential uses are in the control of necrotising enteritis and in the alleviation of the mucositis associated with cancer treatment.
Collapse
Affiliation(s)
- R A Goodlad
- Histopathology Unit, Imperial Cancer Research Fund, London, UK
| | | |
Collapse
|
44
|
Abstract
The jury is still out on the role of luminal EGF in the normal gastrointestinal tract. Recent evidence, however, suggests that its major function is to act as a 'luminal surveillance' peptide, which is available to stimulate repair and that it is not of major importance in maintaining normal gut growth. Recombinant EGF administered via the gut lumen could still prove a valuable tool for the treatment of gastrointestinal ulceration. Perhaps the old term for EGF, 'epidermal healing factor' or EHF, was more appropriate after all!
Collapse
Affiliation(s)
- R J Playford
- Department of Gastroenterology, Leicester General Hospital
| | | |
Collapse
|
45
|
Vinter-Jensen L, Duch BU, Petersen JA, Ryslev A, Gregersen H. Systemic treatment with epidermal growth factor in the rat. Biomechanical properties of the growing small intestine. REGULATORY PEPTIDES 1996; 61:135-42. [PMID: 8852816 DOI: 10.1016/0167-0115(95)00158-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Prolonged treatment with epidermal growth factor (EGF) in the rat provides an experimental model to growth of the gastrointestinal tract. We treated female Wistar rats for 0 (n = 15), 1 (n = 8), 2 (n = 8), and 4 (n = 8) weeks with subcutaneous EGF (i50 micrograms.kg-1.day-1). Segments were taken from locations at 10, 50 and 90% along the length of the small intestine, weighed, the wall thickness was measured and the luminal cross-sectional area and passive biomechanical properties were assessed using impedance planimetry. In addition, the wall composition was evaluated on histological sections. The weight of the total small intestine and of the three segments (measured in mg.cm-1) increased with the duration of the EGF treatment due to mucosal and muscular growth. After 1 week of treatment the wall thickness increased. After 2 weeks of treatment the cross-sectional area began to increase. The circumferential stress-strain distributions revealed translation of the curves to the right in the graphs implying reduced wall stiffness during EGF treatment. In conclusion EGF treatment for 1 to 4 weeks caused a time-dependent increase in intestinal weight. The growth was characterized by increased wall thickness, increased cross-sectional area and reduced wall stiffness.
Collapse
Affiliation(s)
- L Vinter-Jensen
- Department of Clinical Biochemistry, KH Aarhus University Hospital, Denmark
| | | | | | | | | |
Collapse
|
46
|
Vinter-Jensen L, Smerup M, Jørgensen PE, Juhl CO, Orntoft T, Poulsen SS, Nexø E. Chronic treatment with epidermal growth factor stimulates growth of the urinary tract in the rat. UROLOGICAL RESEARCH 1996; 24:15-21. [PMID: 8966836 DOI: 10.1007/bf00296727] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Twenty-four male Wistar rats, 8 weeks old, were allocated into three groups and treated with human recombinant epidermal growth factor (EGF) administered subcutaneously in doses of 0, 30, and 150 micrograms/kg per day for 4 weeks. Blood sampling was done every 2nd week and urine sampling was done for 2 consecutive days every week. The most striking finding was that the ureters were dose dependently enlarged, due to growth of all layers of the ureteric wall. The urothelium of the bladder showed considerable hyperplasticity with a widening of the basal proliferative compartment and a normal differentiation pattern as observed by the expression of carbohydrate epitopes, characterized with lectinohistochemistry. Blood examination revealed a decrease in blood haemoglobin concentration and a slight increase in serum creatinine concentration in the high-dose group. There were no effects of EGF on the urinary excretion of electrolytes, proteins, and endogenous EGF.
Collapse
Affiliation(s)
- L Vinter-Jensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Denmark
| | | | | | | | | | | | | |
Collapse
|
47
|
Otto WR, Rao J, Cox HM, Kotzian E, Lee CY, Goodlad RA, Lane A, Gorman M, Freemont PA, Hansen HF, Pappin D, Wright NA. Effects of pancreatic spasmolytic Polypeptide (PSP) on epithelial cell function. EUROPEAN JOURNAL OF BIOCHEMISTRY 1996; 235:64-72. [PMID: 8631368 DOI: 10.1111/j.1432-1033.1996.00064.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Trefoil peptides are expressed near endodermal ulcerations and may modulate epithelial repair. The trefoil pancreatic spasmolytic polypeptide (PSP) was tested for growth activity in vitro on epithelial cells and in vivo following intragastric or intravenous infusion in parenterally fed intact rats. Ion transport was assessed as changes in short-circuit current in rat intestine and adenocarcinoma cells in Ussing chambers. PSP stimulated growth of MCF-7 and Colo-357 cells, but only in the presence of extracellular glutathione (GSH). The effect was attenuated by GSH depletion with buthionine sulphoximine, even in GSH-containing media. When GSH-reduced PSP was carboxymethylated with iodoacetic acid, it still depended on extracellular GSH for its growth effect. Intestinal epithelial proliferation in rats was not affected by either intravenous or intraluminal infusion. PSP had no effect on basal or stimulated ion flux in rat jejunum or epithelial monolayers. The peptide did not compete with 125I-labeled epidermal growth factor for its receptor. [14C]Iodoacetamide treatment of PSP, followed by prolonged tryptic digestion yielded predominantly a 14C-labeled tetrapeptide fragment containing Cys1O4, with a lesser quantity of a 14C-labeled 15-amino-acid peptide containing Cys95 (molar ratio 15:1). GSH may predominantly reduce the Cys6-Cys1O4 terminal disulphide bond in PSP. We conclude that some epithelia may exhibit a growth response to PSP if extracellular GSH is present. Reduction of PSP by GSH is not necessary for this response, suggesting that the trefoil receptor or its signal transduction is GSH sensitive. PSP could assist wound healing by interactions with epithelial cells exposed concurrently to a local high GSH concentration.
Collapse
Affiliation(s)
- W R Otto
- Histopathology Unit, Imperial Cancer Research Fund, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Foltzer-Jourdainne C, Raul F. Facteurs de croissance intestinaux. NUTR CLIN METAB 1996. [DOI: 10.1016/s0985-0562(96)80010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
49
|
|
50
|
Sizemore N, Wright DS, Mueller WT, Kuo BS. Impact of receptor downregulation on clearance of two human EGFs with different receptor binding activity. Peptides 1996; 17:1229-36. [PMID: 8959761 DOI: 10.1016/s0196-9781(96)00191-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Human epidermal growth factor [hEGF(1-53)] has been thought to be cleared mainly via an EGF receptor (EGFR) endocytosis pathway. Pretreatment of rats with hEGF(1-53) has been shown previously to cause a dramatic reduction in clearance of the peptide contributable to EGFR downregulation. The impact of receptor downregulation has raised concerns for rational design of dosage regimen for this potential wound-healing therapeutic peptide. However, following a similar protocol, we could not reproduce the dramatic reduction in clearance reported previously mediated by an i.v. bolus acute dose. As EGFR downregulation may be sensitive to the length of exposure and to the activation of the receptor tyrosine kinase activity, two other pretreatment protocols were also evaluated: a 4-h i.v. infusion (prolonged exposure) of the peptide and an i.v. bolus of a potent synthetic kinase inhibitor pretreatment were evaluated for effects on clearance. However, neither pretreatment affected the peptide's clearance profile. Further, no effects on clearance and other kinetic parameters were observed for any pretreatment paradigms with a truncated analogue hEGF (1-48), whose EGF receptor binding activity is much weaker but plasma clearance is much higher than hEGF (1-53). In addition, a study in a second rat strain showed no difference in clearance profile of hEGF-(1-53) following pretreatment. Results of the present investigation suggest that receptor binding does not have a direct relationship with plasma clearance, and that the EGF clearance mechanisms is highly refractory with EGF receptors possibly recovering rapidly from downregulation through the recycling process.
Collapse
Affiliation(s)
- N Sizemore
- Department of Pharmacokinetics, Parke-Davis Pharmaceutical Research, Division of Warner-Lambert Company, Ann Arbor, MI 48105, USA
| | | | | | | |
Collapse
|