Network


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

Hotspot


Dive into the research topics where Nicolas Theys is active.

Publication


Featured researches published by Nicolas Theys.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2009

Maternal low protein diet alters pancreatic islet mitochondrial function in a sex-specific manner in the adult rat.

Nicolas Theys; Thomas Bouckenooghe; Marie-Thérèse Ahn; Claude Remacle; Brigitte Reusens

Mitochondrial dysfunction may be a long-term consequence of a poor nutritional environment during early life. Our aim was to investigate whether a maternal low-protein (LP) diet may program mitochondrial dysfunction in islets of adult progeny before glucose intolerance ensues. To address this, pregnant Wistar rats were fed isocaloric diets containing either 20% protein (control) or 8% protein (LP diet) throughout gestation. From birth, offspring received the control diet. The mitochondrial function was analyzed in islets of 3-mo-old offspring. Related to their basal insulin release, cultured islets from both male and female LP offspring presented a lower response to glucose challenge and a blunted ATP production compared with control offspring. The expression of malate dehydrogenase as well as the subunit 6 of the ATP synthase encoded by mitochondrial genome (mtDNA) was lower in these islets, reducing the capacity of ATP production through the Krebs cycle and oxidative phosphorylation. However, mtDNA content was unchanged in LP islets compared with control. Several consequences of protein restriction during fetal life were more marked in male offspring. Only LP males showed an increased reactive oxygen species production associated with a higher expression of mitochondrial subunits of the electron transport chain NADH-ubiquinone oxireductase subunit 4L, an overexpression of peroxisome proliferator-activated receptor-gamma and uncoupling protein-2, and a strongly reduced beta-cell mass. In conclusion, mitochondrial function is clearly altered in islets from LP adult offspring in a sex-specific manner. That may provide a cellular explanation for the earlier development of glucose intolerance in male than in female offspring of dams fed an LP diet.


Diabetes, Obesity and Metabolism | 2007

Intrauterine programming of the endocrine pancreas.

Claude Remacle; O Dumortier; Vanesa Bol; K Goosse; Pierre Romanus; Nicolas Theys; T. Bouckenooghe; Brigitte Reusens

Epidemiological studies have revealed strong relationships between poor foetal growth and subsequent development of the metabolic syndrome. Persisting effects of early malnutrition become translated into pathology, thereby determine chronic risk for developing glucose intolerance and diabetes. These epidemiological observations identify the phenomena of foetal programming without explaining the underlying mechanisms that establish the causal link. Animal models have been established and studies have demonstrated that reduction in the availability of nutrients during foetal development programs the endocrine pancreas and insulin‐sensitive tissues. Whatever the type of foetal malnutrition, whether there are not enough calories or protein in food or after placental deficiency, malnourished pups are born with a defect in their β‐cell population that will never completely recover, and insulin‐sensitive tissues will be definitively altered. Despite the similar endpoint, different cellular and physiological mechanisms are proposed. Hormones operative during foetal life like insulin itself, insulin‐like growth factors and glucocorticoids, as well as specific molecules like taurine, or islet vascularization were implicated as possible factors amplifying the defect. The molecular mechanisms responsible for intrauterine programming of the β cells are still elusive, but two hypotheses recently emerged: the first one implies programming of mitochondria and the second, epigenetic regulation.


Journal of Nutritional Biochemistry | 2011

Maternal malnutrition programs pancreatic islet mitochondrial dysfunction in the adult offspring

Nicolas Theys; Marie-Thérèse Ahn; Thomas Bouckenooghe; Brigitte Reusens; Claude Remacle

Accumulating evidence has shown that maternal malnutrition increases the risk of metabolic disease in the progeny. We previously reported that prenatal exposure to a low-protein diet (LP) leads to mitochondrial dysfunction in pancreatic islets from adult rodent offspring that could relate physiological and cellular alterations due to early diet. We aim to determine whether mitochondrial dysfunction could be a common consequence of prenatal nutritional unbalances. Pregnant Wistar rats received either a global food restriction (GFR), consisting in the reduction by 50% of the normal daily food intake, or a high-fat diet (HF) throughout gestation. GFR or HF diet during pregnancy leads to a lack of increase in insulin release and ATP content in response to glucose stimulation in islets from 3-month-old male and female offspring. These similar consequences originated from impairment in either glucose sensing or glucose metabolism, depending on the type of early malnutrition and on the sex of the progeny. Indeed, the glucose transport across β-cell membrane seemed compromised in female HF offspring, since GLUT-2 gene was markedly underexpressed. Additionally, for each progeny, consequences downstream the entry of glucose were also apparent. Expression of genes involved in glycolysis, TCA cycle and oxidative phosphorylations was altered in GFR and HF rats in a sex- and diet-dependent manner. Moreover, prenatal malnutrition affected the regulators of mitochondrial biogenesis, namely, PPAR coactivator 1 alpha (PGC-1α), since its expression was higher in islets from GFR rats. In conclusion, programming of mitochondrial dysfunction is a consequence of maternal malnutrition, which may predispose to glucose intolerance in the adult offspring.


PLOS ONE | 2009

Early low protein diet aggravates unbalance between antioxidant enzymes leading to islet dysfunction.

Nicolas Theys; André Clippe; Thomas Bouckenooghe; Brigitte Reusens; Claude Remacle

Background Islets from adult rat possess weak antioxidant defense leading to unbalance between superoxide dismutase (SOD) and hydrogen peroxide-inactivating enzymatic activities, catalase (CAT) and glutathione peroxidase (GPX) rending them susceptible to oxidative stress. We have shown that this vulnerability is influenced by maternal diet during gestation and lactation. Methodology/Principal Findings The present study investigated if low antioxidant activity in islets is already observed at birth and if maternal protein restriction influences the development of islet antioxidant defenses. Rats were fed a control diet (C group) or a low protein diet during gestation (LP) or until weaning (LPT), after which offspring received the control diet. We found that antioxidant enzymatic activities varied with age. At birth and after weaning, normal islets possessed an efficient GPX activity. However, the antioxidant capacity decreased thereafter increasing the potential vulnerability to oxidative stress. Maternal protein malnutrition changed the antioxidant enzymatic activities in islets of the progeny. At 3 months, SOD activity was increased in LP and LPT islets with no concomitant activation of CAT and GPX. This unbalance could lead to higher hydrogen peroxide production, which may concur to oxidative stress causing defective insulin gene expression due to modification of critical factors that modulate the insulin promoter. We found indeed that insulin mRNA level was reduced in both groups of malnourished offspring compared to controls. Analyzing the expression of such critical factors, we found that c-Myc expression was strongly increased in islets from both protein-restricted groups compared to controls. Conclusion and Significance Modification in antioxidant activity by maternal low protein diet could predispose to pancreatic islet dysfunction later in life and provide new insights to define a molecular mechanism responsible for intrauterine programming of endocrine pancreas.


The American Journal of Clinical Nutrition | 2011

Maternal malnutrition programs the endocrine pancreas in progeny

Brigitte Reusens; Nicolas Theys; Olivier Dumortier; Kevin Goosse; Claude Remacle

Type 2 diabetes arises when the endocrine pancreas fails to secrete sufficient insulin to cope with metabolic demands resulting from β cell secretory dysfunction, decreased β cell mass, or both. Epidemiologic studies have shown strong relations between poor fetal and early postnatal nutrition and susceptibility to diabetes later in life. Animal models have been established, and studies have shown that a reduction in the availability of nutrients during fetal development programs the endocrine pancreas and insulin-sensitive tissues. We investigated several modes of early malnutrition in rats. Regardless of the type of diet investigated, whether there was a deficit in calories or protein in food or even in the presence of a high-fat diet, malnourished pups were born with a defect in their β cell population, with fewer β cells that did not secrete enough insulin and that were more vulnerable to oxidative stress; such populations of β cells will never completely recover. Despite the similar endpoint, the cellular and physiologic mechanisms that contribute to alterations in β cell mass differ depending on the nature of the nutritional insult. Hormones that are operative during fetal life, such as insulin, insulin-like growth factors, and glucocorticoids; specific molecules, such as taurine; and islet vascularization have been implicated as possible factors in amplifying this defect. The molecular mechanisms responsible for intrauterine programming of β cells are still elusive, but among them the programming of mitochondria may be a strong central candidate.


American Journal of Physiology-endocrinology and Metabolism | 2013

Maternal calorie restriction modulates placental mitochondrial biogenesis and bioenergetic efficiency: putative involvement in fetoplacental growth defects in rats

Sylvain Mayeur; Steve Lancel; Nicolas Theys; Marie-Amélie Lukaszewski; Sophie Duban-Deweer; Bruno Bastide; Johan Hachani; Roméo Cecchelli; Christophe Breton; Anne Gabory; Laurent Storme; Brigitte Reusens; Claudine Junien; Didier Vieau; Jean Lesage

Low birth weight is associated with an increased risk for developing type 2 diabetes and metabolic diseases. The placental capacity to supply nutrients and oxygen to the fetus represents the main determiner of fetal growth. However, few studies have investigated the effects of maternal diet on the placenta. We explored placental adaptive proteomic processes implicated in response to maternal undernutrition. Rat term placentas from 70% food-restricted (FR30) mothers were used for a proteomic screen. Placental mitochondrial functions were evaluated using molecular and functional approaches, and ATP production was measured. FR30 drastically reduced placental and fetal weights. FR30 placentas displayed 14 proteins that were differentially expressed, including several mitochondrial proteins. FR30 induced a marked increase in placental mtDNA content and changes in mitochondrial functions, including modulation of the expression of genes implicated in biogenesis and bioenergetic pathways. FR30 mitochondria showed higher oxygen consumption but failed to maintain their ATP production. Maternal undernutrition induces placental mitochondrial abnormalities. Although an increase in biogenesis and bioenergetic efficiency was noted, placental ATP level was reduced. Our data suggest that placental mitochondrial defects may be implicated in fetoplacental pathologies.


PLOS ONE | 2011

Impairment of rat fetal beta-cell development by maternal exposure to dexamethasone during different time-windows.

Olivier Dumortier; Nicolas Theys; Marie-Thérèse Ahn; Claude Remacle; Brigitte Reusens

Aim Glucocorticoids (GCs) take part in the direct control of cell lineage during the late phase of pancreas development when endocrine and exocrine cell differentiation occurs. However, other tissues such as the vasculature exert a critical role before that phase. This study aims to investigate the consequences of overexposure to exogenous glucocorticoids during different time-windows of gestation for the development of the fetal endocrine pancreas. Methods Pregnant Wistar rats received dexamethasone acetate in their drinking water (1 µg/ml) during the last week or throughout gestation. Fetuses and their pancreases were analyzed at day 15 and 21 of gestation. Morphometrical analysis was performed on pancreatic sections after immunohistochemistry techniques and insulin secretion was evaluated on fetal islets collected in vitro. Results Dexamethasone given the last week or throughout gestation reduced the beta-cell mass in 21-day-old fetuses by respectively 18% or 62%. This was accompanied by a defect in insulin secretion. The alpha-cell mass was reduced similarly. Neither islet vascularization nor beta-cell proliferation was affected when dexamethasone was administered during the last week, which was however the case when given throughout gestation. When given from the beginning of gestation, dexamethasone reduced the number of cells expressing the early marker of endocrine lineage neurogenin-3 when analyzed at 15 days of fetal age. Conclusions GCs reduce the beta- and alpha-cell mass by different mechanisms according to the stage of development during which the treatment was applied. In fetuses exposed to glucocorticoids the last week of gestation only, beta-cell mass is reduced due to impairment of beta-cell commitment, whereas in fetuses exposed throughout gestation, islet vascularization and lower beta-cell proliferation are involved as well, amplifying the reduction of the endocrine mass.


Brain Structure & Function | 2016

Systematic expression analysis of Hox genes at adulthood reveals novel patterns in the central nervous system

Bertrand Hutlet; Nicolas Theys; Cécile Coste; Marie-Thérèse Ahn; Konstantin Doshishti-Agolli; Benoit Lizen; Françoise Gofflot

Hox proteins are key regulators of animal development, providing positional identity and patterning information to cells along the rostrocaudal axis of the embryo. Although their embryonic expression and function are well characterized, their presence and biological importance in adulthood remains poorly investigated. We provide here the first detailed quantitative and neuroanatomical characterization of the expression of the 39 Hox genes in the adult mouse brain. Using RT-qPCR we determined the expression of 24 Hox genes mainly in the brainstem of the adult brain, with low expression of a few genes in the cerebellum and the forebrain. Using in situ hybridization (ISH) we have demonstrated that expression of Hox genes is maintained in territories derived from the early segmental Hox expression domains in the hindbrain. Indeed, we show that expression of genes belonging to paralogy groups PG2-8 is maintained in the hindbrain derivatives at adulthood. The spatial colinearity, which characterizes the early embryonic expression of Hox genes, is still observed in sequential antero-posterior boundaries of expression. Moreover, the main mossy and climbing fibres precerebellar nuclei express PG2-8 Hox genes according to their migration origins. Second, ISH confirms the presence of Hox gene transcripts in territories where they are not detected during development, suggesting neo-expression in these territories in adulthood. Within the forebrain, we have mapped Hoxb1, Hoxb3, Hoxb4, Hoxd3 and Hoxa5 expression in restricted areas of the sensory cerebral cortices as well as in specific thalamic relay nuclei. Our data thus suggest a requirement of Hox genes beyond their role of patterning genes, providing a new dimension to their functional relevance in the central nervous system.


World Journal of Diabetes | 2011

Alteration of mitochondrial function in adult rat offspring of malnourished dams

Brigitte Reusens; Nicolas Theys; Claude Remacle

Under-nutrition as well as over-nutrition during pregnancy has been associated with the development of adult diseases such as diabetes and obesity. Both epigenetic modifications and programming of the mitochondrial function have been recently proposed to explain how altered intrauterine metabolic environment may produce such a phenotype. This review aims to report data reported in several animal models of fetal malnutrition due to maternal low protein or low calorie diet, high fat diet as well as reduction in placental blood flow. We focus our overview on the β cell. We highlight that, notwithstanding early nutritional events, mitochondrial dysfunctions resulting from different alteration by diet or gender are programmed. This may explain the higher propensity to develop obesity and diabetes in later life.


International Journal of Developmental Neuroscience | 2010

Quantitative and anatomical mapping of the expression of Hox genes belonging to paralogs groups 1 to 5 in the adult central nervous system: towards a functional analysis of Hox genes in the adult brain.

Nicolas Theys; A. Van Ecke; Konstantin Doshishti-Agolli; Marie-Thérèse Ahn; Françoise Gofflot

We have shown that Sox5 is expressed in neural progenitors in the spinal cord, and in a subpopulation of dorsal dI3 interneurons. Through gainand loss-of-function analyses, we found that Sox5 controls the timing of cell cycle exit in neural progenitors at the G1-S transition by counteracting the mitotic effect of the Wnt/ catenin pathway. Mechanistically, by increasing the transcriptional levels of the negative regulator Axin2, Sox5 would control the feedback repressor pathway regulating Wnt signalling. Furthermore, we have found that Sox5 downregulation in postmitotic cells is necessary for the progression of the differentiation program and that Sox5 is essential for the survival of dI3 interneurons. Hence, these data situate Sox5 as an important brake of Wnt/ -catenin mitogenic activity during the progression of neurogenesis.

Collaboration


Dive into the Nicolas Theys's collaboration.

Top Co-Authors

Avatar

Brigitte Reusens

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Claude Remacle

Lawson Health Research Institute

View shared research outputs
Top Co-Authors

Avatar

Marie-Thérèse Ahn

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Thomas Bouckenooghe

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Françoise Gofflot

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Konstantin Doshishti-Agolli

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Olivier Dumortier

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

A. Van Ecke

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

André Clippe

Université catholique de Louvain

View shared research outputs
Researchain Logo
Decentralizing Knowledge