Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mireille Andriamihaja is active.

Publication


Featured researches published by Mireille Andriamihaja.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2010

Colon luminal content and epithelial cell morphology are markedly modified in rats fed with a high-protein diet

Mireille Andriamihaja; Anne-Marie Davila; Mamy Eklou-Lawson; Nathalie Petit; Serge Delpal; Fadhila Allek; Anne Blais; Corine Delteil; Daniel Tomé; François Blachier

Hyperproteic diets are used in human nutrition to obtain body weight reduction. Although increased protein ingestion results in an increased transfer of proteins from the small to the large intestine, there is little information on the consequences of the use of such diets on the composition of large intestine content and on epithelial cell morphology and metabolism. Rats were fed for 15 days with either a normoproteic (NP, 14% protein) or a hyperproteic isocaloric diet (HP, 53% protein), and absorptive colonocytes were observed by electron microscopy or isolated for enzyme activity studies. The colonic luminal content was recovered for biochemical analysis. Absorbing colonocytes were characterized by a 1.7-fold reduction in the height of the brush-border membranes (P = 0.0001) after HP diet consumption when compared with NP. This coincided in the whole colon content of HP animals with a 1.8-fold higher mass content (P = 0.0020), a 2.2-fold higher water content (P = 0.0240), a 5.2-fold higher protease activity (P = 0.0104), a 5.5-fold higher ammonia content (P = 0.0008), and a more than twofold higher propionate, valerate, isobutyrate, and isovalerate content (P < 0.05). The basal oxygen consumption of colonocytes was similar in the NP and HP groups, but ammonia was found to provoke a dose-dependent decrease of oxygen consumption in the isolated absorbing colonocytes. The activity of glutamine synthetase (which condenses ammonia and glutamate) was found to be much higher in colonocytes than in small intestine enterocytes and was 1.6-fold higher (P = 0.0304) in colonocytes isolated from HP animals than NP. Glutaminase activity remained unchanged. Thus hyperproteic diet ingestion causes marked changes both in the luminal environment of colonocytes and in the characteristics of these cells, demonstrating that hyperproteic diet interferes with colonocyte metabolism and morphology. Possible causal relationships between energy metabolism, reduced height of colonocyte brush-border membranes, and reduced water absorption are discussed.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2014

High protein diet modifies colonic microbiota and luminal environment but not colonocyte metabolism in the rat model: The increased luminal bulk connection

Xinxin Liu; Jean-Marc Blouin; Arlette Santacruz; Annaïg Lan; Mireille Andriamihaja; Sabina Wilkanowicz; Pierre-Henri Benetti; Daniel Tomé; Yolanda Sanz; François Blachier; Anne-Marie Davila

High-protein diets are used for body weight reduction, but consequences on the large intestine ecosystem are poorly known. Here, rats were fed for 15 days with either a normoproteic diet (NP, 14% protein) or a hyperproteic-hypoglucidic isocaloric diet (HP, 53% protein). Cecum and colon were recovered for analysis. Short- and branched-chain fatty acids, as well as lactate, succinate, formate, and ethanol contents, were markedly increased in the colonic luminal contents of HP rats (P < 0.05 or less) but to a lower extent in the cecal luminal content. This was associated with reduced concentrations of the Clostridium coccoides and C. leptum groups and Faecalibacterium prausnitzii in both the cecum and colon (P < 0.05 or less). In addition, the microbiota diversity was found to be higher in the cecum of HP rats but was lower in the colon compared with NP rats. In HP rats, the colonic and cecal luminal content weights were markedly higher than in NP rats (P < 0.001), resulting in similar butyrate, acetate, and propionate concentrations. Accordingly, the expression of monocarboxylate transporter 1 and sodium monocarboxylate transporter 1 (which is increased by higher butyrate concentration) as well as the colonocyte capacity for butyrate oxidation were not modified by the HP diet, whereas the amount of butyrate in feces was increased (P < 0.01). It is concluded that an increased bulk in the large intestine content following HP diet consumption allows maintenance in the luminal butyrate concentration and thus its metabolism in colonocytes despite modified microbiota composition and increased substrate availability.


Journal of Cellular Physiology | 2009

Butyrate metabolism in human colon carcinoma cells: Implications concerning its growth‐inhibitory effect

Mireille Andriamihaja; Catherine Chaumontet; Daniel Tomé; François Blachier

Butyrate and acetate are bacterial metabolites present in the large intestine lumen. Although butyrate is well known to inhibit the in vitro proliferation of human colon carcinoma cells in a process involving the hyperacetylation of specific nuclear histones, little is known about the possible link between butyrate metabolism and its growth‐inhibitory effect. In a previous study (Leschelle et al., 2000, Eur J Biochem 267: 6435–6442), we showed that butyrate accumulates and is metabolized in HT‐29 Glc−/+ cells without increasing oxygen consumption. In the present study, using the same cell line incubated with 14C‐labeled butyrate, we determined that a minor part of 14C from butyrate was recovered in nuclear histones. Unlike butyrate, acetate exerted no effect on cell growth but was a precursor for overall net histone acetylation. Although butyrate was able to increase the cellular AMP/ADP ratio, it did not affect the ATP cell content or the adenylate charge or the oxidation of endogenous L‐glutamine. Butyrate oxidation was found to be markedly sensitive to the presence of other substrates with D‐glucose decreasing this oxidation and L‐malate stimulating it. Furthermore, in the presence of L‐malate, the growth‐inhibitory effect of butyrate was significantly weaker than in its absence. From these data, we conclude that the metabolism of butyrate downstream acetyl‐CoA synthesis is not involved in the butyrate antiproliferative effect. The suggestion that butyrate metabolism in mitochondria is not used in these cells as a fuel but acts as a regulator of butyrate free concentrations (thus limiting its action upon cellular targets), is discussed. J. Cell. Physiol. 218: 58–65, 2009.


Journal of Nutritional Biochemistry | 2015

High-protein diet differently modifies intestinal goblet cell characteristics and mucosal cytokine expression in ileum and colon

Annaïg Lan; Mireille Andriamihaja; Jean-Marc Blouin; Xinxin Liu; Véronique Descatoire; Caroline Desclée de Maredsous; Anne-Marie Davila; Francine Walker; Daniel Tomé; François Blachier

We have previously shown that high-protein (HP) diet ingestion causes marked changes in the luminal environment of the colonic epithelium. This study aimed to evaluate the impact of such modifications on small intestinal and colonic mucosa, two segments with different transit time and physiological functions. Rats were fed with either normal protein (NP; 14% protein) or HP (53% protein) isocaloric diet for 2 weeks, and parameters related to intestinal mucous-secreting cells and to several innate/adaptive immune characteristics (myeloperoxidase activity, cytokine and epithelial TLR expression, proportion of immune cells in gut-associated lymphoid tissues) were measured in the ileum and colon. In ileum from HP animals, we observed hyperplasia of mucus-producing cells concomitant with an increased expression of Muc2 at both gene and protein levels, reduction of mucosal myeloperoxidase activity, down-regulation of Tlr4 gene expression in enterocytes and down-regulation of mucosal Th cytokines associated with CD4+ lymphocyte reduction in mesenteric lymph nodes. These changes coincided with an increased amount of acetate in the ileal luminal content. In colon, HP diet ingestion resulted in a lower number of goblet cells at the epithelial surface but increased goblet cell number in colonic crypts together with an increased Muc3 and a slight reduction of Il-6 gene expression. Our data suggest that HP diet modifies the goblet cell distribution in colon and, in ileum, increases goblet cell activity and decreases parameters related to basal gut inflammatory status. The impact of HP diet on intestinal mucosa in terms of beneficial or deleterious effects is discussed.


Inflammatory Bowel Diseases | 2013

Beneficial effects of an amino acid mixture on colonic mucosal healing in rats.

Xinxin Liu; Martin Beaumont; Francine Walker; Catherine Chaumontet; Mireille Andriamihaja; Hideki Matsumoto; Nadezda Khodorova; Annaïg Lan; Claire Gaudichon; Robert Benamouzig; Daniel Tomé; Anne-Marie Davila; Jean-Claude Marie; François Blachier

Background:Mucosal healing (MH) decreases the relapse risk in patients with inflammatory bowel disease, but the role of dietary supplementation in this process has been poorly investigated. Here, we investigated the effect of an amino acid mixture supplement on rat MH. Methods:Colitis was induced using 5% of dextran sodium sulfate for 6 days. Then, rats received a mixture of threonine (0.50 g/d), methionine (0.31 g/d), and monosodium glutamate (0.57 g/d) or an isonitrogenous amount of alanine (control group). Colons were recovered after colitis induction and after dietary supplementation for measuring colon characteristics, myeloperoxidase, cytokine gene expression, glutathione content, protein synthesis rate, and for histological analysis. Short-chain fatty acids were measured in the colonic content. Results:Colitis induction resulted in anorexia, thickening and shortening of the colon, and ulceration. Colonic cytokine expression and neutrophil infiltration were increased. An increased amount of water and a decreased amount of butyrate, propionate, and acetate were measured in the colonic content. Supplementation with the amino acid mixture coincided with a reduced protein synthesis rate in the colon compatible with the observed increased colonic MH. Mucosal regeneration/re-epithelialization was visible within 3 days after colitis induction at a time when mucosal inflammation was severe. Histological analysis revealed an increased regeneration/re-epithelialization after 10-day supplementation. In contrast, the spontaneous resolution of inflammation was not affected by the supplementation. Conclusions:Amino acid supplementation ameliorates colonic MH but not mucosal inflammatory status. Our data sustain the use of adjuvant dietary intervention on initiated intestinal MH.


Journal of Agricultural and Food Chemistry | 2016

Deleterious Effect of p-Cresol on Human Colonic Epithelial Cells Prevented by Proanthocyanidin-Containing Polyphenol Extracts from Fruits and Proanthocyanidin Bacterial Metabolites

Ximena Wong; Catalina Carrasco-Pozo; Elizabeth Escobar; Paola Navarrete; Franςois Blachier; Mireille Andriamihaja; Annaïg Lan; Daniel Tomé; Maria Jose Cires; Edgar Pastene; Martin Gotteland

The protective effect of proanthocyanidin-containing polyphenol extracts from apples, avocados, cranberries, grapes, or proanthocyanidin microbial metabolites was evaluated in colonic epithelial cells exposed to p-cresol, a deleterious compound produced by the colonic microbiota from l-tyrosine. In HT29 Glc(-/+) cells, p-cresol significantly increased LDH leakage and decreased ATP contents, whereas in Caco-2 cell monolayers, it significantly decreased the transepithelial electrical resistance and increased the paracellular transport of FITC-dextran. The alterations induced by p-cresol in HT29 Glc(-/+) cells were prevented by the extracts from cranberries and avocados, whereas they became worse by extracts from apples and grapes. The proanthocyanidin bacterial metabolites decreased LDH leakage, ameliorating cell viability without improving intracellular ATP. All of the polyphenol extracts and proanthocyanidin bacterial metabolites prevented the p-cresol-induced alterations of barrier function. These results suggest that proanthocyanidin-containing polyphenol extracts and proanthocyanidin metabolites likely contribute to the protection of the colonic mucosa against the deleterious effects of p-cresol.


Amino Acids | 2013

Comparative efficiency of microbial enzyme preparations versus pancreatin for in vitro alimentary protein digestion

Mireille Andriamihaja; Alain Guillot; Allan Svendsen; Joerg Hagedorn; Sahondra Rakotondratohanina; Daniel Tomé; François Blachier

Utilisation of microbial enzymes may represent an alternative strategy to the use of conventional pancreatin obtained from pig pancreas for the treatment of severe pancreatic insufficiency. In this study, we focused on the capacity of two microbial preparations for their capacity to digest alimentary proteins (caseins and soya proteins) in comparison with pancreatin. These microbial enzymatic preparations were found to be able to generate small, medium-size and larger polypeptides from caseins and soya proteins but were inactivated at pH 3.0. As determined by Liquid Chromatography–Mass Spectrometry analysis, microbial enzymes generated very different peptides from caseins when compared with peptides generated through pancreatin action. These microbial preparations were characterised by relatively low trypsin- and low carboxypeptidase-like activities but high chymotrypsin-like activities and strong capacity for cleavage of caseins at the methionine sites. Although the efficiency of these microbial preparations to increase the rate of absorption of nitrogen-containing compounds in severe pancreatic insufficiency remains to be tested in vivo, our in vitro data indicate proteolytic capacities of such preparations for alimentary protein digestion.


Nature Communications | 2016

Sulfheme formation during homocysteine S-oxygenation by catalase in cancers and neurodegenerative diseases

Dominique Padovani; Assia Hessani; Francine T. Castillo; Géraldine Liot; Mireille Andriamihaja; Annaïg Lan; Camilla Pilati; François Blachier; Suvajit Sen; Erwan Galardon; Isabelle Artaud

Accumulating evidence suggests that abnormal levels of homocysteine are associated with vascular dysfunctions, cancer cell proliferation and various neurodegenerative diseases. With respect to the latter, a perturbation of transition metal homeostasis and an inhibition of catalase bioactivity have been reported. Herein, we report on some of the molecular bases for the cellular toxicity of homocysteine and demonstrate that it induces the formation of sulfcatalase, an irreversible inactive state of the enzyme, without the intervention of hydrogen sulfide. Initially, homocysteine reacts with native catalase and/or redox-active transition metal ions to generate thiyl radicals that mediate compound II formation, a temporarily inactive state of the enzyme. Then, the ferryl centre of compound II intervenes into the unprecedented S-oxygenation of homocysteine to engender the corresponding sulfenic acid species that further participates into the prosthetic heme modification through the formation of an unusual Fe(II) sulfonium. In addition, our ex cellulo studies performed on cancer cells, models of neurodegenerative diseases and ulcerative colitis suggest the likelihood of this scenario in a subset of cancer cells, as well as in a cellular model of Parkinsons disease. Our findings expand the repertoire of heme modifications promoted by biological compounds and point out another deleterious trait of disturbed homocysteine levels that could participate in the aetiology of these diseases.


Biochimica et Biophysica Acta | 2005

Adaptative metabolic response of human colonic epithelial cells to the adverse effects of the luminal compound sulfide

Xavier Leschelle; Marc Goubern; Mireille Andriamihaja; Hervé M. Blottière; Elodie Couplan; Maria-del-Mar Gonzalez-Barroso; Caroline Petit; Anthony Pagniez; Catherine Chaumontet; Bernard Mignotte; Frédéric Bouillaud; François Blachier


Archive | 2017

10 Glutamine and Intestinal Physiology and Pathology

François Blachier; Moïse Coeffier; Weiyun Zhu; Chunlong Mu; Yuxiang Yang; Guoyao Wu; Xiangfeng Kong; Antonio Herbert Lancha; Mireille Andriamihaja; Daniel Tomé; Yulong Yin

Collaboration


Dive into the Mireille Andriamihaja's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xinxin Liu

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alain Guillot

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Anthony Pagniez

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Camilla Pilati

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Caroline Desclée de Maredsous

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge