Network


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

Hotspot


Dive into the research topics where Kadiombo Bantubungi is active.

Publication


Featured researches published by Kadiombo Bantubungi.


Journal of Neurochemistry | 2004

Dopamine D3 receptor stimulation promotes the proliferation of cells derived from the post-natal subventricular zone

Valérie Coronas; Kadiombo Bantubungi; J. Fombonne; S. Krantic; Serge N. Schiffmann; Michel Roger

In the adult mammalian brain, neural stem cells persist in the subventricular zone (SVZ) where dopamine D3 receptors are expressed. Here, we demonstrate that addition of 1 µm apomorphine increases cell numbers in post‐natal SVZ cell cultures. This effect was prevented by a co‐treatment with haloperidol, sulpiride or U‐99194A, a D3‐preferring antagonist, and mimicked by the dopamine D3 receptor selective agonist 7‐hydroxy‐dipropylaminotetralin (7‐OH‐DPAT). EC50 values were 4.04 ± 1.54 nm for apomorphine and 0.63 ± 0.13 nm for 7‐OH‐DPAT, which fits the pharmacological profile of the D3 receptor. D3 receptors were detected in SVZ cells by RT‐PCR and immunocytochemistry. D3 receptors were expressed in numerous β‐III tubulin immunopositive cells. The fraction of apoptotic nuclei remained unchanged following apomorphine treatment, thus ruling out any possible effect on cell survival. In contrast, proliferation was increased as both the proportion of nuclei incorporating bromo‐deoxyuridine and the expression of the cell division marker cyclin D1 were enhanced. These findings provide support for a regulatory role of dopamine over cellular dynamics in post‐natal SVZ.


Molecular and Cellular Neuroscience | 2008

Stem cell factor and mesenchymal and neural stem cell transplantation in a rat model of Huntington's disease.

Kadiombo Bantubungi; David Blum; Laetitia Cuvelier; Sabine Wislet-Gendebien; Bernard Rogister; Emmanuel Brouillet; Serge N. Schiffmann

Neural and mesenchymal stem cells have been proposed as alternative sources of cells for transplantation into the brain in neurodegenerative disorders. However, the endogenous factors controlling their engraftment within the injured parenchyma remain ill-defined. Here, we demonstrate significant engraftment of undifferentiated exogenous mesenchymal or neural stem cells throughout the lesioned area in a rat model for Huntingtons disease, as late as 8 weeks post-transplantation. We show that stem cell factor (SCF), strongly up-regulated within host cells in the damaged striatum, is able to activate the SCF receptor c-kit and its signaling pathway and to promote the migration and proliferation of mesenchymal and neural stem cells in vitro. Furthermore, c-kit receptor blockade alters neural stem cell distribution within the lesioned striatum. Host SCF expression is observed in atypical cells expressing glial fibrillary acidic protein and doublecortin in the lesioned striatum and in migrating doublecortin-positive progenitors. Taken together, these data demonstrate that SCF produced in situ in the lesioned striatum is an important factor in promoting the engraftment of stem cells within the lesioned brain.


Neurobiology of Disease | 2004

Death of cortical and striatal neurons induced by mitochondrial defect involves differential molecular mechanisms.

Marie-Christine Galas; Nicolas Bizat; Laetitia Cuvelier; Kadiombo Bantubungi; Emmanuel Brouillet; Serge N. Schiffmann; David Blum

An important aspect of Huntingtons disease (HD) pathogenesis which may have important therapeutic implications is that the cellular events leading to cell death may be different in cortical and striatal neurons. In the present study, we characterized cellular changes in cortical and striatal neurons treated with the mitochondrial toxin 3-nitropropionic acid (3NP) in culture. Degeneration induced by 3NP was similar in both striatal and cortical neurons as observed using markers of cell viability and DNA fragmentation. However, in striatal neurons, 3NP produced a marked delocalization of Bad, Bax, cytochrome c and Smac while this was not observed in cortical neurons. Death of striatal neurons was preceded by activation of calpain and was blocked by calpain inhibitor I. In cortical neurons, calpain was not activated and calpain inhibitor I was without effect. In both cell types, caspase-9 and -3 were not activated by 3NP and the caspase inhibitor zVAD-fmk did not provide neuroprotective effect. Interestingly, treatment with staurosporine (STS) triggered caspase-9 and -3 in cortical and striatal cells, suggesting that the molecular machinery related to caspase-dependent apoptosis was functional in both cell types even though this machinery was not involved in 3NP toxicity. The present results clearly demonstrate that under mitochondrial inhibition, striatal and cortical neurons die through different pathways. This suggests that mitochondrial defects in HD may trigger the death of cortical and striatal neurons through different molecular events.


Blood | 2011

p16INK4a deficiency promotes IL-4-induced polarization and inhibits proinflammatory signaling in macrophages.

Céline Cudejko; Kristiaan Wouters; Lucía Fuentes; Sarah Anissa Hannou; Charlotte Paquet; Kadiombo Bantubungi; Emmanuel Bouchaert; Jonathan Vanhoutte; Sébastien Fleury; Patrick Rémy; Anne Tailleux; Giulia Chinetti; David Dombrowicz; Bart Staels; Réjane Paumelle

The CDKN2A locus, which contains the tumor suppressor gene p16(INK4a), is associated with an increased risk of age-related inflammatory diseases, such as cardiovascular disease and type 2 diabetes, in which macrophages play a crucial role. Monocytes can polarize toward classically (CAMϕ) or alternatively (AAMϕ) activated macrophages. However, the molecular mechanisms underlying the acquisition of these phenotypes are not well defined. Here, we show that p16(INK4a) deficiency (p16(-/-)) modulates the macrophage phenotype. Transcriptome analysis revealed that p16(-/-) BM-derived macrophages (BMDMs) exhibit a phenotype resembling IL-4-induced macrophage polarization. In line with this observation, p16(-/-) BMDMs displayed a decreased response to classically polarizing IFNγ and LPS and an increased sensitivity to alternative polarization by IL-4. Furthermore, mice transplanted with p16(-/-) BM displayed higher hepatic AAMϕ marker expression levels on Schistosoma mansoni infection, an in vivo model of AAMϕ phenotype skewing. Surprisingly, p16(-/-) BMDMs did not display increased IL-4-induced STAT6 signaling, but decreased IFNγ-induced STAT1 and lipopolysaccharide (LPS)-induced IKKα,β phosphorylation. This decrease correlated with decreased JAK2 phosphorylation and with higher levels of inhibitory acetylation of STAT1 and IKKα,β. These findings identify p16(INK4a) as a modulator of macrophage activation and polarization via the JAK2-STAT1 pathway with possible roles in inflammatory diseases.


Journal of Experimental Medicine | 2017

Tau deletion promotes brain insulin resistance

Elodie Marciniak; Antoine Leboucher; Emilie Caron; Tariq Ahmed; Anne Tailleux; Julie Dumont; Tarik Issad; Ellen Gerhardt; Patrick Pagesy; Margaux Vileno; Clément Bournonville; Malika Hamdane; Kadiombo Bantubungi; Steve Lancel; Dominique Demeyer; Sabiha Eddarkaoui; Emmanuelle Vallez; Didier Vieau; Sandrine Humez; Emilie Faivre; Benjamin Grenier-Boley; Tiago F. Outeiro; Bart Staels; Philippe Amouyel; Detlef Balschun; Luc Buée; David Blum

The molecular pathways underlying tau pathology–induced synaptic/cognitive deficits and neurodegeneration are poorly understood. One prevalent hypothesis is that hyperphosphorylation, misfolding, and fibrillization of tau impair synaptic plasticity and cause degeneration. However, tau pathology may also result in the loss of specific physiological tau functions, which are largely unknown but could contribute to neuronal dysfunction. In the present study, we uncovered a novel function of tau in its ability to regulate brain insulin signaling. We found that tau deletion leads to an impaired hippocampal response to insulin, caused by altered IRS-1 and PTEN (phosphatase and tensin homologue on chromosome 10) activities. Our data also demonstrate that tau knockout mice exhibit an impaired hypothalamic anorexigenic effect of insulin that is associated with energy metabolism alterations. Consistently, we found that tau haplotypes are associated with glycemic traits in humans. The present data have far-reaching clinical implications and raise the hypothesis that pathophysiological tau loss-of-function favors brain insulin resistance, which is instrumental for cognitive and metabolic impairments in Alzheimer’s disease patients.


Atherosclerosis | 2011

PPARα activation differently affects microparticle content in atherosclerotic lesions and liver of a mouse model of atherosclerosis and NASH

Morgane Baron; Aurélie S. Leroyer; Zouher Majd; Fanny Lalloyer; Emmanuelle Vallez; Kadiombo Bantubungi; Giulia Chinetti-Gbaguidi; Chantal M. Boulanger; Bart Staels; Anne Tailleux

BACKGROUND Atherosclerosis and non-alcoholic fatty liver disease (NAFLD) are complex pathologies characterized by lipid accumulation, chronic inflammation and extensive tissue remodelling. Microparticles (MPs), small membrane vesicles produced by activated and apoptotic cells, might not only be biomarkers, but also functional actors in these pathologies. The apoE2-KI mouse is a model of atherosclerosis and NAFLD. Activation of the nuclear receptor PPARα decreases atherosclerosis and components of non-alcoholic steatohepatitis (NASH) in the apoE2-KI mouse. OBJECTIVES (1) To determine whether MPs are present in atherosclerotic lesions, liver and plasma during atherosclerosis and NASH progression in apoE2-KI mice, and (2) to study whether PPARα activation modulates MP concentrations. METHODS ApoE2-KI mice were fed a Western diet to induce atherosclerosis and NASH. MPs were isolated from atherosclerotic lesions, liver and blood and quantified by flow cytometry. RESULTS An increase of MPs was observed in the atherosclerotic lesions and in the liver of apoE2-KI mice upon Western diet feeding. PPARα activation with fenofibrate decreased MP levels in the atherosclerotic lesions in a PPARα-dependent manner, but did not influence MP concentrations in the liver. CONCLUSION Here we report that MPs are present in atherosclerotic lesions and in the liver of apoE2-KI mice. Their concentration increased during atherosclerosis and NASH development. PPARα activation differentially modulates MP levels in a tissue-specific manner.


Diabetes | 2014

Cdkn2a/p16Ink4a Regulates Fasting-Induced Hepatic Gluconeogenesis Through the PKA-CREB-PGC1α Pathway

Kadiombo Bantubungi; Sarah-Anissa Hannou; Sandrine Caron-Houde; Emmanuelle Vallez; Morgane Baron; Anthony Lucas; Emmanuel Bouchaert; Réjane Paumelle; Anne Tailleux; Bart Staels

Type 2 diabetes (T2D) is hallmarked by insulin resistance, impaired insulin secretion, and increased hepatic glucose production. The worldwide increasing prevalence of T2D calls for efforts to understand its pathogenesis in order to improve disease prevention and management. Recent genome-wide association studies have revealed strong associations between the CDKN2A/B locus and T2D risk. The CDKN2A/B locus contains genes encoding cell cycle inhibitors, including p16Ink4a, which have not yet been implicated in the control of hepatic glucose homeostasis. Here, we show that p16Ink4a deficiency enhances fasting-induced hepatic glucose production in vivo by increasing the expression of key gluconeogenic genes. p16Ink4a downregulation leads to an activation of PKA-CREB-PGC1α signaling through increased phosphorylation of PKA regulatory subunits. Taken together, these results provide evidence that p16Ink4a controls fasting glucose homeostasis and could as such be involved in T2D development.


Neuroscience Letters | 2006

Unilateral induction of progenitors in the spinal cord of hSOD1(G93A) transgenic rats correlates with an asymmetrical hind limb paralysis.

Isabelle de Hemptinne; Cédric Boucherie; Roland Pochet; Kadiombo Bantubungi; Serge N. Schiffmann; Jean-Marie Maloteaux; Emmanuel Hermans

Transgenic rats expressing a mutated form of the human Cu/Zn superoxide dismutase (hSOD1(G93A)) develop an amyotrophic lateral sclerosis (ALS)-like phenotype, including motor neurone degeneration and reactive gliosis in the spinal cord. This study aimed at examining the presence of endogenous neural progenitors in the lumbar spinal cord of these rats at the end-stage of the disease. Immunohistochemical data clearly demonstrated the induced expression of the stem cell factor reported as a chemoattractant and survival factor for neural stem cells as well as nestin (neuro-epithelial stem cell intermediate filament) in the spinal cord sections. While the stem cell factor immunolabelling appeared diffuse throughout the gray matter, nestin labelling was restricted to clusters within the ventral horn. Interestingly, as paralysis regularly develops asymmetrically, induction of nestin was only detected on the ipsilateral side of the predominant symptoms. Finally, immunohistochemical detection of the stem cell factor receptor (c-Kit) revealed its specific induction which coincided with nestin immunolabelling. Together, these results are indicative of endogenous recruitment of neural progenitors within lesioned tissues and could support the development of treatments involving endogenous or exogenous stem cells.


PLOS ONE | 2012

Bone Marrow p16INK4a-Deficiency Does Not Modulate Obesity, Glucose Homeostasis or Atherosclerosis Development

Kristiaan Wouters; Céline Cudejko; Marion J. J. Gijbels; Lucía Fuentes; Kadiombo Bantubungi; Jonathan Vanhoutte; Rébecca Dièvart; Charlotte Paquet; Emmanuel Bouchaert; Sarah Anissa Hannou; Florence Gizard; Anne Tailleux; Menno P.J. de Winther; Bart Staels; Réjane Paumelle

Objective A genomic region near the CDKN2A locus, encoding p16INK4a, has been associated to type 2 diabetes and atherosclerotic vascular disease, conditions in which inflammation plays an important role. Recently, we found that deficiency of p16INK4a results in decreased inflammatory signaling in murine macrophages and that p16INK4a influences the phenotype of human adipose tissue macrophages. Therefore, we investigated the influence of immune cell p16INK4a on glucose tolerance and atherosclerosis in mice. Methods and Results Bone marrow p16INK4a-deficiency in C57Bl6 mice did not influence high fat diet-induced obesity nor plasma glucose and lipid levels. Glucose tolerance tests showed no alterations in high fat diet-induced glucose intolerance. While bone marrow p16INK4a-deficiency did not affect the gene expression profile of adipose tissue, hepatic expression of the alternative markers Chi3l3, Mgl2 and IL10 was increased and the induction of pro-inflammatory Nos2 was restrained on the high fat diet. Bone marrow p16INK4a-deficiency in low density lipoprotein receptor-deficient mice did not affect western diet-induced atherosclerotic plaque size or morphology. In line, plasma lipid levels remained unaffected and p16INK4a-deficient macrophages displayed equal cholesterol uptake and efflux compared to wild type macrophages. Conclusion Bone marrow p16INK4a-deficiency does not affect plasma lipids, obesity, glucose tolerance or atherosclerosis in mice.


The Journal of Neuroscience | 2003

A Dual Role of Adenosine A2A Receptors in 3-Nitropropionic Acid-Induced Striatal Lesions: Implications for the Neuroprotective Potential of A2A Antagonists

David Blum; Marie-Christine Galas; Annita Pintor; Emmanuel Brouillet; Catherine Ledent; Christa E. Müller; Kadiombo Bantubungi; Mariangela Galluzzo; David Gall; Laetitia Cuvelier; Anne-Sophie Rolland; Patrizia Popoli; Serge N. Schiffmann

Collaboration


Dive into the Kadiombo Bantubungi's collaboration.

Top Co-Authors

Avatar

Serge N. Schiffmann

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emmanuel Brouillet

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Laetitia Cuvelier

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Annita Pintor

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Patrizia Popoli

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

David Gall

Université libre de Bruxelles

View shared research outputs
Researchain Logo
Decentralizing Knowledge