Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Breast Cancer Res Treat ; 166(3): 657-668, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28803352

ABSTRACT

PURPOSE: Breast cancer is the most common malignancy in women in terms of incidence and mortality. Age is undoubtedly the biggest breast cancer risk factor. In this study we examined clinical, histological, and biological characteristics and mortality of breast cancer in elderly women along with their changes with advancing age. METHODS: We reviewed 63 original articles published between 2006 and 2016 concerning women over 70 years with breast cancer. RESULTS: Compared to patients 70-79 years, patients aged 80 and over had larger tumor size with fewer T1 (42.9% vs 57.7%, p < 0.01) and more T2 lesions (43.5% vs 33.0%, p < 0.01). Lymph nodes and distant metastases were more frequent, with more N + (49.5% vs 44.0%, p < 0.01) and more M1 (8.0% vs 5.9%, p < 0.01). Infiltrating mucinous carcinomas were more frequent (4.3% vs 3.7%, p < 0.01). Tumors had lower grades, with more grade 1 (23.2% vs 19.8%, p = 0.01) and fewer grade 3 (21.5% vs 25.5%, p < 0.01), and were more hormone-sensitive: PR was more often expressed (72.6% vs 67.3%, p < 0.01). Lympho-vascular invasion was less frequent in the 80 years and over (22.9% vs 29.7%, p = 0.01). Breast cancer-specific mortality was higher both at 5 years (25.8% vs 17.2%, p < 0.01) and 10 years (32.7% vs 26.6%, p < 0.01). CONCLUSION: Clinico-pathological characteristics, increased incidence, and mortality associated with aging can be explained on one hand by biological changes of the breast such as increased estrogen sensitivity, epithelial cell alterations, immune senescence, and tumor microenvironment modifications. However, sociologic factors such as increased life expectancy, under-treatment, late diagnosis, and insufficient individual screening, are also involved.


Subject(s)
Age Factors , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Aged , Aged, 80 and over , Breast Neoplasms/metabolism , Estrogens/metabolism , Female , Humans , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Tumor Microenvironment
2.
Gynecol Obstet Fertil ; 44(4): 211-7, 2016 Apr.
Article in French | MEDLINE | ID: mdl-27032759

ABSTRACT

INTRODUCTION: Trisomy 21 (T21) is the most common chromosomal abnormality and one of the main causes of intellectual disability. The tumor profile of T21 patients is characterized by the low frequency of solid tumors including breast cancer. METHODS: The objective of this work was to analyze the literature to find possible clues for the low frequency of breast cancer in T21 persons with a focus on one hand to the various risks and protective factors against breast cancer for women T21, and on the other hand to changes in the expression of different genes located on chromosome 21. RESULTS: T21 women have hormonal and societal risk factors for breast cancer: frequent nulliparity, lack of breastfeeding, physical inactivity and high body mass index. The age of menopause, earlier in T21 women, has a modest protective effect against breast cancer. The low rate of breast tumors in T21 women is probably mainly linked to the reduced life expectancy compared to the general population (risk of death before the age of onset of the majority of breast cancers) and the presence of a third chromosome 21, characterizing the disease. It might lead to the increased expression of a number of genes contributing directly or undirectly to tumor suppression, decreased tumor angiogenesis and increased cell apoptosis. Moreover, changes in the mammary stroma of persons T21 could have an inhibitory role on the development of breast tumors. CONCLUSION: The low frequency of breast cancers for T21 patients may not only be explained by hormonal and societal factors, but also by genetic mechanisms which could constitute an interesting axis of research in breast cancer.


Subject(s)
Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Down Syndrome/genetics , Adolescent , Adult , Aged , Child , Chromosomes, Human, Pair 21 , Female , Humans , Middle Aged
3.
Oncogene ; 33(31): 4050-9, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-24141782

ABSTRACT

MMP-11 is a bad prognosis paracrine factor in invasive breast cancers. However, its mammary physiological function remains largely unknown. In the present study we have investigated MMP-11 function during postnatal mammary gland development and function using MMP-11-deficient (MMP-11-/-) mice. Histological and immunohistochemical analyses as well as whole-mount mammary gland staining show alteration of the mammary gland in the absence of MMP-11, where ductal tree, alveolar structures and milk production are reduced. Moreover, a series of transplantation experiments allowed us to demonstrate that MMP-11 exerts an essential local paracrine function that favors mammary gland branching and epithelial cell outgrowth and invasion through adjacent connective tissues. Indeed, MMP-11-/- cleared fat pads are not permissive for wild-type epithelium development, whereas MMP-11-/- epithelium transplants grow normally when implanted in wild-type cleared fat pads. In addition, using primary mammary epithelial organoids, we show in vitro that this MMP-11 pro-branching effect is not direct, suggesting that MMP-11 acts via production/release of stroma-associated soluble factor(s). Finally, the lack of MMP-11 leads to decreased periductal collagen content, suggesting that MMP-11 has a role in collagen homeostasis. Thus, local stromal MMP-11 might also regulate mammary epithelial cell behavior mechanically by promoting extracellular matrix stiffness. Collectively, the present data indicate that MMP-11 is a paracrine factor involved during postnatal mammary gland morphogenesis, and support the concept that the stroma strongly impact epithelial cell behavior. Interestingly, stromal MMP-11 has previously been reported to favor malignant epithelial cell survival and promote cancer aggressiveness. Thus, MMP-11 has a paracrine function during mammary gland development that might be harnessed to promote tumor progression, exposing a new link between development and malignancy.


Subject(s)
Extracellular Matrix/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Matrix Metalloproteinase 11/metabolism , Paracrine Communication , Stromal Cells/cytology , Animals , Animals, Newborn , Cell Differentiation/physiology , Cells, Cultured , Epithelial Cells/cytology , Female , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Knockout , NIH 3T3 Cells
4.
Oncogene ; 30(29): 3261-73, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21358676

ABSTRACT

Although trefoil factor 1 (TFF1; previously named pS2) is abnormally expressed in about 50% of human breast tumors, its physiopathological role in this disease has been poorly studied. Moreover, controversial data have been reported. TFF1 function in the mammary gland therefore needs to be clarified. In this study, using retroviral vectors, we performed TFF1 gain- or loss-of-function experiments in four human mammary epithelial cell lines: normal immortalized TFF1-negative MCF10A, malignant TFF1-negative MDA-MB-231 and malignant TFF1-positive MCF7 and ZR75.1. The expression of TFF1 stimulated the migration and invasion in the four cell lines. Forced TFF1 expression in MCF10A, MDA-MB-231 and MCF7 cells did not modify anchorage-dependent or -independent cell proliferation. By contrast, TFF1 knockdown in MCF7 enhanced soft-agar colony formation. This increased oncogenic potential of MCF7 cells in the absence of TFF1 was confirmed in vivo in nude mice. Moreover, chemically induced tumorigenesis in TFF1-deficient (TFF1-KO) mice led to higher tumor incidence in the mammary gland and larger tumor size compared with wild-type mice. Similarly, tumor development was increased in the TFF1-KO ovary and lung. Collectively, our results clearly show that TFF1 does not exhibit oncogenic properties, but rather reduces tumor development. This beneficial function of TFF1 is in agreement with many clinical studies reporting a better outcome for patients with TFF1-positive breast primary tumors.


Subject(s)
Breast Neoplasms/pathology , Mammary Neoplasms, Experimental/pathology , Peptides/physiology , Tumor Suppressor Proteins/physiology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Base Sequence , Blotting, Western , Breast Neoplasms/genetics , Carcinogens/toxicity , Cell Line, Tumor , Cell Proliferation , DNA , Female , Gene Knockdown Techniques , Humans , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Knockout , Mice, Nude , Molecular Sequence Data , Peptides/genetics , Transplantation, Heterologous , Trefoil Factor-1 , Tumor Suppressor Proteins/genetics
5.
Cell Prolif ; 41(6): 923-935, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19040570

ABSTRACT

OBJECTIVE: It is not known whether or not epithelial progenitors of the pyloric antrum are involved in gastric carcinogenesis. Normally, these progenitors give rise to two main cell lineages: pit and gland mucous cells. This study was designed to examine the changes that occur in pyloric antral mucous cell lineages and their progenitors during development of gastric adenoma and carcinoma in trefoil factor 1 (TFF1) knockout mice. MATERIALS AND METHODS: Pyloric antral mucosal tissues of TFF1 knockout mice at ages from 3 days to 17 months were processed for histochemical analysis using Ulex europaeus and Grifforia simplifolica lectins as markers for pit and gland mucous cells, respectively. The dividing epithelial progenitors were identified by using immunohistochemical and electron microscopy techniques. RESULTS: TFF1 loss was associated with amplification of both mucus-secreting pit and gland cells. Both lectins examined bound not only to mature mucous cells, but also to most of epithelial progenitors which gradually amplified with age and frequently were seen in mitosis. Analysis of 12- to 17-month-old TFF1-deficient stomachs revealed occasional groups of poorly differentiated mucosal cells with features similar to those of epithelial progenitors (or stem cells), in the basal portion of the antral mucosa. These cells eventually invaded the muscularis mucosa while maintaining some capacity to differentiate. CONCLUSION: This study shows that the progenitors of pit and gland mucous cells contribute to gastric carcinogenesis in the pyloric antrum of TFF1 knockout mice, strongly supporting the concept of stem cell origin of cancer.


Subject(s)
Epithelial Cells/pathology , Peptides/deficiency , Stem Cells/pathology , Stomach Neoplasms/pathology , Aging/pathology , Animals , Cell Differentiation , Cell Division , Cell Lineage , Cell Proliferation , Epithelial Cells/ultrastructure , Gastric Mucosa/pathology , Gastric Mucosa/ultrastructure , Lectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , Peptides/metabolism , Pyloric Antrum/pathology , Pyloric Antrum/ultrastructure , Pylorus/pathology , Pylorus/ultrastructure , Stem Cells/ultrastructure , Stomach Neoplasms/ultrastructure , Trefoil Factor-1
6.
Oncogene ; 27(49): 6347-55, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18622425

ABSTRACT

The substrate of matrix metalloproteinase 11 (MMP11) remains unknown. We have recently shown that MMP11 is a negative regulator of adipogenesis, able to reduce and even to revert mature adipocyte differentiation. Here, we have used mouse 3T3L1 cells and human U87MG and SaOS cells to show that MMP11 cleaves the native alpha3 chain of collagen VI, which is an adipocyte-related extracellular matrix component. It is known that extracellular proteolytic processing of this chain is required for correct collagen VI folding. Interestingly, MMP11-deficient fat tissue is less cohesive and exhibits collagen VI alteration, dramatic adipocyte plasma and basement membrane abnormalities and lipid leakage. MMP11 is thus required for correct collagen VI folding and therefore for fat tissue cohesion and adipocyte function. Both MMP11 and collagen VI favor tumor progression. Similar spatio-temporal overexpression at the adipocyte-cancer cell interface has been reported for the two proteins. MMP11-dependent collagen VI processing might therefore be expected to occur during malignancy. Accordingly, collagen VI no longer delineates adipocytes located at the invasive front of breast carcinomas. In conclusion, the native alpha3 chain of collagen VI constitutes a specific MMP11 substrate. This MMP11 collagenolytic activity is functional in fat tissue ontogenesis as well as during cancer invasive steps.


Subject(s)
Collagen Type VI/physiology , Collagen/physiology , Extracellular Matrix Proteins/physiology , Extracellular Matrix/physiology , Matrix Metalloproteinase 11/physiology , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes/pathology , Adipocytes/physiology , Adipocytes/ultrastructure , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cells, Cultured , Collagen/metabolism , Collagen Type VI/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix/ultrastructure , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/physiology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Matrix Metalloproteinase 11/genetics , Matrix Metalloproteinase 11/metabolism , Mice , Osteosarcoma/metabolism , Osteosarcoma/pathology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Silver Staining
7.
Histol Histopathol ; 22(7): 743-51, 2007 07.
Article in English | MEDLINE | ID: mdl-17455148

ABSTRACT

The pathogenesis of colon cancer is not well understood. This common type of cancer is generally believed to occur in a multistep process which involves alterations of various tumor suppressor genes and oncogenes during the progression through benign lesions towards carcinoma. TFF3 is a product of the colonic epithelium and has been implicated in colonic mucosal protection and also in the aggressiveness of colon cancer cells. The aim of this study was to analyze the expression of TFF3 during propagation towards cancer development in the human colon. Colonic tissues representing colitis, adenomatous polyposis, tubulovillous adenoma, and mucoid/adeno-carcinomas were processed for immunohistochemistry using an antibody specific for human TFF3. The results were correlated with those of PCNA-labeling, quantified, and compared with those of control tissues obtained from the safe margin of macroscopically normal colonic mucosa of patients with colon cancer. The data showed marked down-regulation of TFF3 expression in adenomatous polyposis, then TFF3 expression returns to about control level during adenoma and remains high during mucoid- and adeno-carcinomas. Colonic tissues with highly invasive cancer cells were characterized by statistically significant down-regulation of TFF3 expression. The changes observed in expression of TFF3 showed an inverse correlation with cell proliferation and suggest that it might play a protective role against colon carcinogenesis.


Subject(s)
Adenocarcinoma, Mucinous/chemistry , Adenoma, Villous/chemistry , Adenomatous Polyposis Coli/chemistry , Colitis/metabolism , Colonic Neoplasms/chemistry , Peptides/analysis , Adenocarcinoma, Mucinous/pathology , Adenoma, Villous/pathology , Adenomatous Polyposis Coli/pathology , Adult , Cell Proliferation , Cell Transformation, Neoplastic/chemistry , Colitis/pathology , Colon/chemistry , Colonic Neoplasms/pathology , Disease Progression , Humans , Immunohistochemistry , Middle Aged , Neoplasm Invasiveness , Proliferating Cell Nuclear Antigen/analysis , Trefoil Factor-3
8.
Gynecol Obstet Fertil ; 34(12): 1161-9, 2006 Dec.
Article in French | MEDLINE | ID: mdl-17123853

ABSTRACT

Breast cancer is the first cause of death between 35 and 55 years. Genetic alterations and modifications in gene expression are found during different steps of tumor progression. These changes are translated at the protein level where quantitative and qualitative modifications are found in tumor compared to normal samples. Similarly to studies aimed at deciphering transcriptional changes important in cancer, proteomic approaches allow the global and comparative study of proteins in normal and pathological samples. The objective of this article is to present common proteomic methods and to review the first published results concerning proteomics studies applied to breast cancer with an emphasis on reports obtained using the SELDI-TOF MS (Surface Enhanced Laser Desorption Ionization Time-Of-Flight Mass Spectrometry). In breast cancer, it is possible to explore the tumoral proteome and/or the blood derived proteome. The first studies are aimed at globally understanding the disease while the latter are aimed at discovering serum proteins or biomarkers useful for the early detection, diagnosis, prognosis and management of cancer. Promising results are obtained using these emerging methods and these novel biomarkers should be validated in the future and will have an important impact for the management of breast cancer patients.


Subject(s)
Biomarkers, Tumor/chemistry , Breast Neoplasms/metabolism , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Breast Neoplasms/diagnosis , Disease Progression , Female , Humans , Protein Array Analysis/methods
9.
Biochem Soc Trans ; 34(Pt 3): 343-5, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16709157

ABSTRACT

MLN64 (metastatic lymph node 64) and MENTHO (MLN64 N-terminal homologue) are two late-endosomal proteins that share a conserved region of four transmembrane helices with three short intervening loops called the MENTAL domain (MLN64 N-terminal domain). This domain mediates MLN64 and MENTHO homo- and hetero-interactions, targets both proteins to late endosomes and binds cholesterol in vivo. In addition to the MENTAL domain, MLN64 contains a cholesterol-specific START domain [StAR (steroidogenic acute regulatory protein)-related lipid transfer domain]. The START domain is a protein module of approx. 210 residues that binds lipids, including sterols, and is present in 15 distinct proteins in mammals. Thus MLN64 and MENTHO define discrete cholesterol-containing subdomains within the membrane of late endosomes where they may function in cholesterol transport. The MENTAL domain might serve to maintain cholesterol at the membrane of late endosomes prior to its shuttle to cytoplasmic acceptor(s) through the START domain.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/physiology , Cholesterol/chemistry , Endosomes/chemistry , Membrane Proteins/chemistry , Membrane Proteins/physiology , Animals , Biological Transport, Active , Carrier Proteins/metabolism , Cholesterol/metabolism , Cholesterol/physiology , Endosomes/metabolism , Endosomes/physiology , Humans , Membrane Proteins/metabolism
10.
Neuroscience ; 139(3): 1031-8, 2006.
Article in English | MEDLINE | ID: mdl-16549269

ABSTRACT

Intracellular management of cholesterol is a critical process in the brain. Deficits with cholesterol transport and storage are linked to neurodegenerative disorders such as Neimann-Pick disease type C and Alzheimer's disease. One protein putatively involved in cholesterol transport is metastatic lymph node 64 (MLN64). MLN64 localizes to late endosomes which are part of the cholesterol internalization pathway. However, a detailed pattern of MLN64 expression in the brain is unclear. Using immunocytochemical and immunoblot analyses, we demonstrated the presence of MLN64 in several tissue types and various regions within the brain. MLN64 immunostaining in the CNS was heterogeneous, indicating selective expression in discrete specific cell populations and regions. MLN64 immunoreactivity was detected in glia and neurons, which displayed intracellular labeling consistent with an endosomal localization. Although previous studies suggested that MLN64 may promote steroid production in the brain, MLN64 immunoreactivity did not colocalize with steroidogenic cells in the CNS. These results demonstrate that MLN64 is produced in the mouse and human CNS in a restricted pattern of expression, suggesting that MLN64 serves a cell-specific function in cholesterol transport.


Subject(s)
Brain/metabolism , Carrier Proteins/biosynthesis , Cholesterol/metabolism , Membrane Proteins/biosynthesis , Animals , Blotting, Western , Female , HeLa Cells , Humans , Immunohistochemistry , Male , Mice , Neuroglia/metabolism , Neurons/metabolism , Protein Transport/physiology
11.
Peptides ; 27(7): 1603-6, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16426704

ABSTRACT

Ghrelin has been shown to accelerate gastric emptying in animals where its effect appeared mediated through the vagus nerve. We aimed to verify the gastrokinetic capacity of ghrelin in human. Patients with gastroparesis attributed to a neural dysregulation by diabetes (n = 5) or surgical vagotomy (n = 1) were evaluated. The emptying of a test meal (420 kcal) was determined by the C13 octanoic acid breath test. Saline or synthetic ghrelin 1-4 microg/kg were given in 1 min bolus at the end of the meal. T-lag and T-1/2 were shorter during ghrelin than during saline administration [33 +/- 5 min versus 65 +/- 14 min (p < 0.01) and 119 +/- 6 min versus 173 +/- 38 min (p < 0.001)]. Ghrelin injection therefore accelerated gastric emptying of a meal in humans even in presence of a deficient gastric innervation.


Subject(s)
Gastroparesis/drug therapy , Gastroparesis/metabolism , Peptide Hormones/administration & dosage , Peptide Hormones/physiology , Adult , Female , Gastric Emptying/drug effects , Gastric Mucosa/metabolism , Ghrelin , Humans , Middle Aged , Peptide Hormones/metabolism , Peptides/chemistry , Time Factors
12.
Cell Mol Life Sci ; 62(24): 2916-20, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16374579

ABSTRACT

Trefoil Factor 1 (TFF1), the first member of the trefoil factor family, is normally expressed in the stomach mucosa. Ectopic expression is also observed in various human pathological conditions, notably in numerous carcinomas and gastrointestinal acute inflammatory disorders. In vivo experimental data using TFF1-deficient mice highlight the pleiotropic functions of TFF1: (i) it is a gastric tumor suppressor gene involved in gastric ontogenesis and homeostasis; (ii) it protects gut mucosa from aggression; (iii) it participates in folding secreted proteins inside the endoplasmic reticulum. At the cellular level, it limits cell proliferation and apoptosis, and favors cell differentiation. Collectively, these data suggest that TFF1 may provide an alternative pharmacological tool for the prevention and treatment of human gastrointestinal diseases.


Subject(s)
Gastric Mucosa/metabolism , Peptides/deficiency , Stomach Neoplasms/metabolism , Tumor Suppressor Proteins/deficiency , Animals , Cell Differentiation , Cell Lineage , Humans , Inflammatory Bowel Diseases/metabolism , Mice , Peptides/genetics , Peptides/pharmacology , Protein Folding , Stomach Neoplasms/genetics , Trefoil Factor-1 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/pharmacology
13.
Gut ; 53(10): 1408-15, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15361486

ABSTRACT

BACKGROUND: Trefoil factor 1 (TFF1/pS2) is a major secretory product of the stomach and TFF1 knockout mice constantly develop adenomas and occasional carcinomas in the pyloric antrum. AIM: To analyse the role of TFF1 in the differentiation of gastric epithelial cell lineages using oxyntic mucosae from normal and TFF1 knockout mice. METHODS: The various cell lineages were labelled using specific markers of pit, neck, parietal, and enteroendocrine cells. Patterns of TFF1, TFF2, and TFF3 expressions were defined using western blotting, immunohistochemistry, and/or immunogold electron microscopy. RESULTS: In normal mice, starting from postnatal day 1 (P1), TFF1 and TFF2 were produced by mucus secreting cells of the developing epithelium. At P7, TFF3 expression occurred in pit and parietal cells. When oxyntic glands were compartmentalised, at P21 and in older mice, TFF1 and TFF2 were expressed in pit and neck cells, respectively, and TFF3 was no longer in parietal cells but became a feature of zymogenic cells. In TFF1 deficient mice, alteration of oxyntic epithelial differentiation became obvious at P21, showing significant amplification of pit cells at the expense of parietal cells. At the molecular level, lack of TFF1 induced dramatic inhibition of TFF2 expression and more precocious TFF3 expression. CONCLUSION: In the oxyntic mucosa, all three TFFs are produced in a lineage specific manner and TFF1 is essential in maintaining the normal commitment programme of epithelial progenitors.


Subject(s)
Parietal Cells, Gastric/cytology , Peptides/physiology , Animals , Cell Differentiation/physiology , Cell Lineage/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Mucins/metabolism , Muscle Proteins/metabolism , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/ultrastructure , Peptides/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Trefoil Factor-1 , Trefoil Factor-2 , Trefoil Factor-3
14.
Peptides ; 24(4): 531-4, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12860196

ABSTRACT

Peptides can influence gastrointestinal motility, and from data obtained earlier in rats, we hypothesized that MTL-RP/Ghrelin, as well as CGRP receptor antagonist 8-37, could improve gastric post-operative ileus in dog. Dogs submitted to laparotomy were perfused with or saline or CGRP 8-37 or MTL-RP/Ghrelin on days 1-4 post-operatively while gastric emptying was estimated by measuring the postprandial increase in plasma acetaminophen ingested with a meal. As expected, in saline-treated animals the gastric emptying function was impaired post-operatively. The total amount of acetaminophen emptied (AUC over 150 min) on post-operative days 1-4 reached respectively 31+/-5%, 65+/-8%, 60+/-8% and 62+/-8% of the normal emptying capacity. CGRP antagonist increased the total emptying of acetaminophen to 52+/-5% on day 1, 95+/-2% on day 2 and 103+/-3% (P<0.05) on day 3. The delayed emptying of acetaminophen seen post-operatively in saline-treated animals could be completely reversed by MTL-RP/Ghrelin (P<0.01) whether it was given at 100 microg/kg on day 2 (102+/-7% of the normal emptying capacity), 4 microg/kg on day 3 (106+/-7%) or 20 microg/kg on day 4 (132+/-8%). As found earlier in rodents, CGRP receptor antagonist 8-37 as well as MTL-RP/Ghrelin are potent prokinetics to improve post-operative gastric ileus in dog.


Subject(s)
Ileum/pathology , Peptides/pharmacology , Postoperative Complications , Stomach/pathology , Acetaminophen/chemistry , Acetaminophen/pharmacokinetics , Acetaminophen/pharmacology , Animals , Area Under Curve , Calcitonin Gene-Related Peptide/pharmacology , Dogs , Female , Gastric Emptying , Ghrelin , Ileus/metabolism , Kinetics , Peptide Fragments/pharmacology , Peptide Hormones/pharmacology , Peptides/chemistry , Postprandial Period , Time Factors
15.
Am J Physiol Gastrointest Liver Physiol ; 282(6): G948-52, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12016119

ABSTRACT

A novel peptide called ghrelin or motilin-related-peptide (MTLRP) was found in the stomach of various mammals. We studied its effect on the motor function of the rat gastrointestinal tract. In normal, conscious unoperated animals, ghrelin/MTLRP (5 or 20 microg/kg iv) significantly accelerated the gastric emptying of a methylcellulose liquid solution (gastric residue after 15 min: 57 +/- 7, 42 +/- 11, 17 +/- 4, and 9 +/- 3% of the ingested meal with doses of 0, 1, 5, and 20 microg/kg iv, respectively) Transit of the methylcellulose liquid solution was also accelerated by ghrelin/MTLRP in the small intestine but not in the colon. Des-[Gln(14)]ghrelin, also found in the mammalian stomach, was as potent as ghrelin in emptying the stomach (gastric residue after 15 min: 12 +/- 3% at a dose of 20 microg/kg iv). In rats in which postoperative gastrointestinal ileus had been experimentally induced, ghrelin/MTLRP (20 microg/kg iv) reversed the delayed gastric evacuation (gastric residue after 15 min: 28 +/- 7% of the ingested meal vs. 82 +/- 9% with saline). In comparison, the gastric ileus was not modified by high doses of motilin (77 +/- 7%) or erythromycin (82 +/- 6%) and was only partially improved by calcitonin gene-related peptide (CGRP) 8-37 antagonist (59 +/- 7%). Ghrelin/MTLRP, therefore, accelerates the gastric emptying and small intestinal transit of a liquid meal and is a strong prokinetic agent capable of reversing the postoperative gastric ileus in rat.


Subject(s)
Intestinal Obstruction/drug therapy , Motilin/pharmacology , Peptide Hormones , Peptides/pharmacology , Postoperative Complications/drug therapy , Animals , Colon/drug effects , Colon/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Gastric Emptying/drug effects , Gastrointestinal Motility/drug effects , Ghrelin , Intestine, Small/drug effects , Intestine, Small/physiology , Male , Rats , Rats, Sprague-Dawley
16.
Am J Hum Genet ; 68(6): 1373-85, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11333381

ABSTRACT

To obtain more information of the functional domains of the NPC1 protein, the mutational spectrum and the level of immunoreactive protein were investigated in skin fibroblasts from 30 unrelated patients with Niemann-Pick C1 disease. Nine of them were characterized by mild alterations of cellular cholesterol transport (the "variant" biochemical phenotype). The mutations showed a wide distribution to nearly all NPC1 domains, with a cluster (11/32) in a conserved NPC1 cysteine-rich luminal loop. Homozygous mutations in 14 patients and a phenotypically defined allele, combined with a new mutation, in a further 10 patients allowed genotype/phenotype correlations. Premature-termination-codon mutations, the three missense mutations in the sterol-sensing domain (SSD), and A1054T in the cysteine-rich luminal loop all occurred in patients with infantile neurological onset and "classic" (severe) cholesterol-trafficking alterations. By western blot, NPC1 protein was undetectable in the SSD missense mutations studied (L724P and Q775P) and essentially was absent in the A1054T missense allele. Our results thus enhance the functional significance of the SSD and demonstrate a correlation between the absence of NPC1 protein and the most severe neurological form. In the remaining missense mutations studied, corresponding to other disease presentations (including two adults with nonneurological disease), NPC1 protein was present in significant amounts of normal size, without clear-cut correlation with either the clinical phenotype or the "classic"/"variant" biochemical phenotype. Missense mutations in the cysteine-rich luminal loop resulted in a wide array of clinical and biochemical phenotypes. Remarkably, all five mutant alleles (I943M, V950M, G986S, G992R, and the recurrent P1007A) definitively correlated with the "variant" phenotype clustered within this loop, providing new insight on the functional complexity of the latter domain.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cysteine/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Mutation/genetics , Niemann-Pick Diseases/genetics , Niemann-Pick Diseases/metabolism , Sterols/metabolism , Adolescent , Adult , Biological Transport , Blotting, Western , Carrier Proteins/genetics , Child , Child, Preschool , Cholesterol/metabolism , Consanguinity , Conserved Sequence/genetics , Cysteine/genetics , DNA Mutational Analysis , Exons/genetics , Female , Genetic Variation/genetics , Genotype , Humans , Infant , Infant, Newborn , Intracellular Signaling Peptides and Proteins , Male , Membrane Glycoproteins/genetics , Molecular Sequence Data , Niemann-Pick C1 Protein , Niemann-Pick Diseases/physiopathology , Phenotype , Polymorphism, Single-Stranded Conformational , Protein Conformation , Structure-Activity Relationship
17.
Gene ; 266(1-2): 67-75, 2001 Mar 21.
Article in English | MEDLINE | ID: mdl-11290420

ABSTRACT

The mammalian Trefoil Factors (TFFs), TFF1/pS2, TFF2/SP and TFF3/ITF, are expressed and secreted throughout the gastrointestinal tract with a specific and complementary pattern. These proteins exhibit common functions in the protection and repair process of the gastrointestinal epithelial barrier. Here, we report the clustered organization of the three mouse TFF genes in a 40 kb DNA segment, in a head to tail orientation in the following order: TFF1, TFF2, and TFF3. Computer comparison of the mouse TFF promoter sequences to their human counterparts revealed conserved boxes in both mouse and human genes. Promoter methylation analyses showed that, in tissues where these genes are normally expressed, the proximal promoters of TFF1 and TFF2 are specifically not methylated and that of TFF3 is partially demethylated. In contrast, in organs that do not express TFFs, the promoters of the three genes are methylated. These findings strongly argue for the involvement of epigenetic mechanisms in the regulation of TFF expression in normal and pathological conditions.


Subject(s)
Genes/genetics , Growth Substances/genetics , Mucins , Muscle Proteins , Neuropeptides , Peptides/genetics , Animals , Base Sequence , DNA/chemistry , DNA/genetics , DNA/metabolism , Gene Expression , Mice , Molecular Sequence Data , Promoter Regions, Genetic , RNA/genetics , RNA/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Tissue Distribution , Trefoil Factor-2 , Trefoil Factor-3
18.
J Biol Chem ; 276(25): 22954-64, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11304540

ABSTRACT

An aberrant truncated hHb1 hair keratin transcript, named hHb1-DeltaN, was previously identified in breast carcinomas. No normal tissue tested so far, including hairy skin, expressed hHb1-DeltaN, indicating that hHb1-DeltaN is related to carcinogenesis. In the present study, we investigated the mechanism by which such truncated transcript was generated in breast cancer cell lines. We found that hHb1-DeltaN transcription is initiated at an unusual cryptic promoter within the fourth intron of the hHb1 gene and is dependent on two proximal Sp1 binding sites for its baseline activity. Moreover, hHb1-DeltaN transcription is increased in response to DNA demethylation by the 5-aza-2'-deoxycytidine drug. This induction is dependent on protein neosynthesis, indicating that an additional factor is required. In addition, we showed that the hHb1-DeltaN transcript is translated in vivo as a truncated hHb1 protein that is missing the 270 amino-terminal residues. The hHb1-DeltaN protein exhibits a filament pattern throughout the cytoplasm and partially co-localizes with cytokeratin filaments, indicating its participation in the cytoskeleton network. hHb1-DeltaN might alter the adhesive properties of cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Keratins/genetics , Subcellular Fractions/metabolism , Transcription, Genetic , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA Footprinting , DNA Methylation , DNA Primers , Humans , Introns , Keratins/metabolism , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Tumor Cells, Cultured
19.
Neuroendocrinology ; 73(1): 54-61, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11174017

ABSTRACT

Ghrelin (Ghr), a 28 amino acid gastric peptide with an n-octanoylation on Ser 3, has recently been identified as an endogenous ligand of the growth hormone secretagogue (GHS) receptor. A cDNA was also isolated from a mouse stomach library encoding a protein named prepromotilin-related peptide (ppMTLRP) which shares sequence similarities with prepromotilin. Mouse and rat ppMTLRP sequences (rGhr) are identical and show 89% identity with human ghrelin (hGhr). By analogy with promotilin, cleavage of proMTLRP into an 18 amino acid endogenous processed peptide can be assumed on the basis of a conserved dibasic motif in position 9-10 of its sequence. In the present work, we compared the GH-releasing activity of rGhr28/MTLRP and of hGhr28/MTRLP with that of a shorter form of the peptide, hGhr18. A short peptide devoid of Ser-3 n-octanoylation hGhr18[-] was also tested. Addition of rGhr28, hGhr28 and hGhr18 stimulated GH release to the same extent from superfused pituitaries. The effect was dose dependent in a 10(-8) to 10(-6) M concentration range. In contrast, hGhr 18[-] was inactive. In freely moving animals, both rGhr28 and hGhr28 (10 microg, i.v.) stimulated GH release, whereas the same dose of hGhr18 or of hGhr18[-] was ineffective. After rGhr28, GH plasma levels increased as early as 5 min after injection and returned to basal values within 40-60 min. Expressed as percent stimulation, administration of rGhr28 was equally effective when injected during troughs or peaks of GH. Plasma concentrations of prolactin, adrenocorticotropin and leptin were not modified. Spontaneous GH secretory episodes were no longer observed within 3 h of rGhr28 treatment, but repeated administration of the secretagogue at 3- to 4-hour intervals resulted in a similar GH response. Activation of somatostatin (SRIH) release by ether stress did not blunt the GH response to rGhr28. This suggests that the secretagogue acts in part by inhibiting endogenous SRIH, as further substantiated by the ability of rGhr28 (10(-6) M), to decrease the amplitude of 25 mM K+-induced SRIH release from perifused hypothalami. In conclusion, (1) n-octanoylation of Ghrs and the shorter form hGhr18 is essential for the direct pituitary GH-releasing effect of this new family of endogenous GHSs; (2) only the longer forms are active in vivo and (3) inhibition of SRIH release appears involved in the mechanism of Ghr action.


Subject(s)
Growth Hormone/metabolism , Motilin/pharmacology , Peptide Hormones , Peptides/pharmacology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Adrenocorticotropic Hormone/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Ghrelin , In Vitro Techniques , Leptin/metabolism , Male , Molecular Sequence Data , Peptide Fragments/pharmacology , Peptides/chemistry , Pituitary Gland/cytology , Prolactin/metabolism , Rats , Rats, Sprague-Dawley , Somatostatin/metabolism
20.
Protein Expr Purif ; 21(1): 92-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11162392

ABSTRACT

The recombinant protein human trefoil factor 1 (hTFF1), formerly called hpS2, has been produced for the first time in a yeast-based expression in Pichia pastoris. hTFF1 was secreted in large amounts in the extracellular medium of P. pastoris under the control of the glyceraldehyde-3-phosphate dehydrogenase promoter. The fermentation broth containing hTFF1 was concentrated by tangential flow filtration prior to purification by anion- and cation-exchange chromatography, followed by preparative high-performance liquid chromatography. The resulting hTFF1 was found to be intact by Western blot analysis. Further analysis revealed mainly the presence of the monomeric form of the hTFF1 peptide. Finally, in vitro, the recombinant hTFF1 was shown to decrease proliferation of the HCT116 cancer cells.


Subject(s)
Proteins/genetics , Proteins/isolation & purification , Amino Acid Sequence , Cell Division/drug effects , Cell Line , Chromatography, High Pressure Liquid/methods , Chromatography, Ion Exchange/methods , Cloning, Molecular/methods , Escherichia coli , Fermentation , Humans , Mass Spectrometry , Molecular Sequence Data , Peptide Fragments/chemistry , Pichia/growth & development , Polymerase Chain Reaction , Proteins/chemistry , Proteins/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Trefoil Factor-1 , Tumor Cells, Cultured , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...