Network


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

Hotspot


Dive into the research topics where Silvia Pinto is active.

Publication


Featured researches published by Silvia Pinto.


Movement Disorders | 2013

Bladder dysfunction in a transgenic mouse model of multiple system atrophy.

Mathieu Boudes; Pieter Uvin; Silvia Pinto; Thomas Voets; Clare J. Fowler; Gregor K. Wenning; Dirk De Ridder; Nadia Stefanova

Multiple system atrophy (MSA) is an adult‐onset neurodegenerative disorder presenting with motor impairment and autonomic dysfunction. Urological function is altered in the majority of MSA patients, and urological symptoms often precede the motor syndrome. To date, bladder function and structure have never been investigated in MSA models. We aimed to test bladder function in a transgenic MSA mouse featuring oligodendroglial α‐synucleinopathy and define its applicability as a preclinical model to study urological failure in MSA. Experiments were performed in proteolipid protein (PLP)–human α‐synuclein (hαSyn) transgenic and control wild‐type mice. Diuresis, urodynamics, and detrusor strip contractility were assessed to characterize the urological phenotype. Bladder morphology and neuropathology of the lumbosacral intermediolateral column and the pontine micturition center (PMC) were analyzed in young and aged mice. Urodynamic analysis revealed a less efficient and unstable bladder in MSA mice with increased voiding contraction amplitude, higher frequency of nonvoiding contractions, and increased postvoid residual volume. MSA mice bladder walls showed early detrusor hypertrophy and age‐related urothelium hypertrophy. Transgenic hαSyn expression was detected in Schwann cells ensheathing the local nerve fibers in the lamina propria and muscularis of MSA bladders. Early loss of parasympathetic outflow neurons and delayed degeneration of the PMC accompanied the urological deficits in MSA mice. PLP‐hαSyn mice recapitulate major urological symptoms of human MSA that may be linked to αSyn‐related central and peripheral neuropathology and can be further used as a preclinical model to decipher pathomechanisms of MSA.


European Urology | 2015

Essential Role of Transient Receptor Potential M8 (TRPM8) in a Model of Acute Cold-induced Urinary Urgency

Pieter Uvin; Jan Franken; Silvia Pinto; Roma Rietjens; Luc Grammet; Yves Deruyver; Yeranddy A. Alpizar; Karel Talavera; Rudi Vennekens; Wouter Everaerts; Dirk De Ridder; Thomas Voets

BACKGROUND Acute exposure of part of the skin to cold stimuli can evoke urinary urgency, a phenomenon termed acute cold-induced urgency (ACIU). Despite its high prevalence, particularly in patients with overactive bladder, little is known about the mechanisms that induce ACIU. OBJECTIVE To develop an animal model of ACIU and test the involvement of cold-activated ion channels transient receptor potential (TRP) M8 and TRPA1. DESIGN, SETTING, AND PARTICIPANTS Intravesical pressure and micturition were monitored in female mice (wild-type C57BL/6J, Trpa1(-/-), Trpm8(+/+), and Trpm8(-/-)) and Sprague Dawley rats. INTERVENTIONS An intravesical catheter was implanted. Localized cooling of the skin was achieved using a stream of air or topical acetone. The TRPM8 antagonist (N-(3-aminopropyl)-2-{[(3-methylphenyl) methyl]oxy}-N-(2-thienylmethyl)benzamide (AMTB) or vehicle was injected intraperitoneally. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Frequencies of bladder contractions and voids in response to sensory stimuli were compared using the Mann-Whitney or Kruskal-Wallis test. RESULTS AND LIMITATIONS Brief, innocuously cold stimuli applied to different parts of the skin evoked rapid bladder contractions and voids in anesthetized mice and rats. These responses were strongly attenuated in Trpm8(-/-) mice and in rats treated with AMTB. As rodent bladder physiology differs from that of humans, it is difficult to directly extrapolate our findings to human patients. CONCLUSIONS Our findings indicate that ACIU is an evolutionarily conserved reflex rather than subconscious conditioning, and provide a useful in vivo model for further investigation of the underlying mechanisms. Pharmacological inhibition of TRPM8 may be useful for treating ACIU symptoms in patients. PATIENT SUMMARY Brief cold stimuli applied to the skin can evoke a sudden desire to urinate, which can be highly bothersome in patients with overactive bladder. We developed an animal model to study this phenomenon, and found that it depends on a specific molecular cold sensor, transient receptor potential M8 (TRPM8). Pharmacological inhibition of TRPM8 may alleviate acute cold-induced urinary urgency in humans.


Nature Communications | 2017

Steviol glycosides enhance pancreatic beta-cell function and taste sensation by potentiation of TRPM5 channel activity

Koenraad Philippaert; Andy Pironet; Margot Mesuere; William Sones; Laura Vermeiren; Sara Kerselaers; Silvia Pinto; Andrei Segal; Nancy Antoine; Conny Gysemans; Jos Laureys; Katleen Lemaire; Patrick Gilon; Eva Cuypers; Jan Tytgat; Chantal Mathieu; Frans Schuit; Patrik Rorsman; Karel Talavera; Thomas Voets; Rudi Vennekens

Steviol glycosides (SGs), such as stevioside and rebaudioside A, are natural, non-caloric sweet-tasting organic molecules, present in extracts of the scrub plant Stevia rebaudiana, which are widely used as sweeteners in consumer foods and beverages. TRPM5 is a Ca2+-activated cation channel expressed in type II taste receptor cells and pancreatic β-cells. Here we show that stevioside, rebaudioside A and their aglycon steviol potentiate the activity of TRPM5. We find that SGs potentiate perception of bitter, sweet and umami taste, and enhance glucose-induced insulin secretion in a Trpm5-dependent manner. Daily consumption of stevioside prevents development of high-fat-diet-induced diabetic hyperglycaemia in wild-type mice, but not in Trpm5−/− mice. These results elucidate a molecular mechanism of action of SGs and identify TRPM5 as a potential target to prevent and treat type 2 diabetes.


Nature Communications | 2016

VAMP7 regulates constitutive membrane incorporation of the cold-activated channel TRPM8

Debapriya Ghosh; Silvia Pinto; Lydia Danglot; Ine Vandewauw; Andrei Segal; Nele Van Ranst; Melissa Benoit; Annelies Janssens; Rudi Vennekens; Pieter Vanden Berghe; Thierry Galli; Joris Vriens; Thomas Voets

The cation channel TRPM8 plays a central role in the somatosensory system, as a key sensor of innocuously cold temperatures and cooling agents. Although increased functional expression of TRPM8 has been implicated in various forms of pathological cold hypersensitivity, little is known about the cellular and molecular mechanisms that determine TRPM8 abundance at the plasma membrane. Here we demonstrate constitutive transport of TRPM8 towards the plasma membrane in atypical, non-acidic transport vesicles that contain lysosomal-associated membrane protein 1 (LAMP1), and provide evidence that vesicle-associated membrane protein 7 (VAMP7) mediates fusion of these vesicles with the plasma membrane. In line herewith, VAMP7-deficient mice exhibit reduced functional expression of TRPM8 in sensory neurons and concomitant deficits in cold avoidance and icilin-induced cold hypersensitivity. Our results uncover a cellular pathway that controls functional plasma membrane incorporation of a temperature-sensitive TRP channel, and thus regulates thermosensitivity in vivo.


Journal of Visualized Experiments | 2012

The use of cystometry in small rodents: a study of bladder chemosensation.

Pieter Uvin; Wouter Everaerts; Silvia Pinto; Yeranddy A. Alpizar; Mathieu Boudes; Thomas Gevaert; Thomas Voets; Bernd Nilius; Karel Talavera; Dirk De Ridder

The lower urinary tract (LUT) functions as a dynamic reservoir that is able to store urine and to efficiently expel it at a convenient time. While storing urine, however, the bladder is exposed for prolonged periods to waste products. By acting as a tight barrier, the epithelial lining of the LUT, the urothelium, avoids re-absorption of harmful substances. Moreover, noxious chemicals stimulate the bladders nociceptive innervation and initiate voiding contractions that expel the bladders contents. Interestingly, the bladders sensitivity to noxious chemicals has been used successfully in clinical practice, by intravesically infusing the TRPV1 agonist capsaicin to treat neurogenic bladder overactivity. This underscores the advantage of viewing the bladder as a chemosensory organ and prompts for further clinical research. However, ethical issues severely limit the possibilities to perform, in human subjects, the invasive measurements that are necessary to unravel the molecular bases of LUT clinical pharmacology. A way to overcome this limitation is the use of several animal models. Here we describe the implementation of cystometry in mice and rats, a technique that allows measuring the intravesical pressure in conditions of controlled bladder perfusion. After laparotomy, a catheter is implanted in the bladder dome and tunneled subcutaneously to the interscapular region. Then the bladder can be filled at a controlled rate, while the urethra is left free for micturition. During the repetitive cycles of filling and voiding, intravesical pressure can be measured via the implanted catheter. As such, the pressure changes can be quantified and analyzed. Moreover, simultaneous measurement of the voided volume allows distinguishing voiding contractions from non-voiding contractions. Importantly, due to the differences in micturition control between rodents and humans, cystometric measurements in these animals have only limited translational value. Nevertheless, they are quite instrumental in the study of bladder pathophysiology and pharmacology in experimental pre-clinical settings. Recent research using this technique has revealed the key role of novel molecular players in the mechano- and chemo-sensory properties of the bladder.


PLOS ONE | 2013

Crucial Role of TRPC1 and TRPC4 in Cystitis-Induced Neuronal Sprouting and Bladder Overactivity

Mathieu Boudes; Pieter Uvin; Silvia Pinto; Marc Freichel; Lutz Birnbaumer; Thomas Voets; Dirk De Ridder; Rudi Vennekens

Purpose During cystitis, increased innervation of the bladder by sensory nerves may contribute to bladder overactivity and pain. The mechanisms whereby cystitis leads to hyperinnervation of the bladder are, however, poorly understood. Since TRP channels have been implicated in the guidance of growth cones and survival of neurons, we investigated their involvement in the increases in bladder innervation and bladder activity in rodent models of cystitis. Materials and Methods To induce bladder hyperactivity, we chronically injected cyclophosphamide in rats and mice. All experiments were performed a week later. We used quantitative transcriptional analysis and immunohistochemistry to determine TRP channel expression on retrolabelled bladder sensory neurons. To assess bladder function and referred hyperalgesia, urodynamic analysis, detrusor strip contractility and Von Frey filament experiments were done in wild type and knock-out mice. Results Repeated cyclophosphamide injections induce a specific increase in the expression of TRPC1 and TRPC4 in bladder-innervating sensory neurons and the sprouting of sensory fibers in the bladder mucosa. Interestingly, cyclophosphamide-treated Trpc1/c4−/− mice no longer exhibited increased bladder innervations, and, concomitantly, the development of bladder overactivity was diminished in these mice. We did not observe a difference neither in bladder contraction features of double knock-out animals nor in cyclophosphamide-induced referred pain behavior. Conclusions Collectively, our data suggest that TRPC1 and TRPC4 are involved in the sprouting of sensory neurons following bladder cystitis, which leads to overactive bladder disease.


Nature | 2018

A TRP channel trio mediates acute noxious heat sensing

Ine Vandewauw; Katrien De Clercq; Marie Mulier; Katharina Held; Silvia Pinto; Nele Van Ranst; Andrei Segal; Thierry Voet; Rudi Vennekens; Katharina Zimmermann; Joris Vriens; Thomas Voets

Acute pain represents a crucial alarm signal to protect us from injury. Whereas the nociceptive neurons that convey pain signals were described more than a century ago, the molecular sensors that detect noxious thermal or mechanical insults have yet to be fully identified. Here we show that acute noxious heat sensing in mice depends on a triad of transient receptor potential (TRP) ion channels: TRPM3, TRPV1, and TRPA1. We found that robust somatosensory heat responsiveness at the cellular and behavioural levels is observed only if at least one of these TRP channels is functional. However, combined genetic or pharmacological elimination of all three channels largely and selectively prevents heat responses in both isolated sensory neurons and rapidly firing C and Aδ sensory nerve fibres that innervate the skin. Strikingly, Trpv1−/−Trpm3−/−Trpa1−/− triple knockout (TKO) mice lack the acute withdrawal response to noxious heat that is necessary to avoid burn injury, while showing normal nociceptive responses to cold or mechanical stimuli and a preserved preference for moderate temperatures. These findings indicate that the initiation of the acute heat-evoked pain response in sensory nerve endings relies on three functionally redundant TRP channels, representing a fault-tolerant mechanism to avoid burn injury.


Pflügers Archiv: European Journal of Physiology | 2016

TRPM4-dependent post-synaptic depolarization is essential for the induction of NMDA receptor-dependent LTP in CA1 hippocampal neurons

Aurélie Menigoz; Tariq Ahmed; Victor Sabanov; Koenraad Philippaert; Silvia Pinto; Sara Kerselaers; Andrei Segal; Marc Freichel; Thomas Voets; Bernd Nilius; Rudi Vennekens; Detlef Balschun

TRPM4 is a calcium-activated but calcium-impermeable non-selective cation (CAN) channel. Previous studies have shown that TRPM4 is an important regulator of Ca2+-dependent changes in membrane potential in excitable and non-excitable cell types. However, its physiological significance in neurons of the central nervous system remained unclear. Here, we report that TRPM4 proteins form a CAN channel in CA1 neurons of the hippocampus and we show that TRPM4 is an essential co-activator of N-methyl-d-aspartate (NMDA) receptors (NMDAR) during the induction of long-term potentiation (LTP). Disrupting the Trpm4 gene in mice specifically eliminates NMDAR-dependent LTP, while basal synaptic transmission, short-term plasticity, and NMDAR-dependent long-term depression are unchanged. The induction of LTP in Trpm4−/− neurons was rescued by facilitating NMDA receptor activation or post-synaptic membrane depolarization. Accordingly, we obtained normal LTP in Trpm4−/− neurons in a pairing protocol, where post-synaptic depolarization was applied in parallel to pre-synaptic stimulation. Taken together, our data are consistent with a novel model of LTP induction in CA1 hippocampal neurons, in which TRPM4 is an essential player in a feed-forward loop that generates the post-synaptic membrane depolarization which is necessary to fully activate NMDA receptors during the induction of LTP but which is dispensable for the induction of long-term depression (LTD). These results have important implications for the understanding of the induction process of LTP and the development of nootropic medication.


European Urology | 2018

Intravesical Activation of the Cation Channel TRPV4 Improves Bladder Function in a Rat Model for Detrusor Underactivity

Yves Deruyver; Emmanuel Weyne; Karel Dewulf; Roma Rietjens; Silvia Pinto; Nele Van Ranst; Jan Franken; Matthias Vanneste; Maarten Albersen; Thomas Gevaert; Rudi Vennekens; Dirk De Ridder; Thomas Voets; Wouter Everaerts

BACKGROUND Improvement of bladder emptying by modulating afferent nerve activity is an attractive therapeutic strategy for detrusor underactivity. Transient receptor potential vanilloid 4 (TRPV4) is a sensory ion channel in urothelial cells that contribute to the detection of bladder filling. OBJECTIVE To investigate the potential benefit of intravesical TRPV4 agonists in a pelvic nerve injury rat model for detrusor underactivity. DESIGN, SETTING, AND PARTICIPANTS Female wild-type and Trpv4 knockout rats underwent sham surgery or bilateral pelvic nerve injury (bPNI). Four weeks later, rats underwent cystometry with infusion of the TRPV4 agonist GSK1016790A. Bladders were harvested for in vitro pharmacological studies, quantitative reverse polymerase chain reaction and immunohistochemistry. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Data are expressed as median ± interquartile range. Statistical comparisons were made using the Mann-Witney U test and Wilcoxon signed rank test as appropriate. RESULTS AND LIMITATIONS Rats with bPNI showed a phenotype characteristic of detrusor underactivity with lower-amplitude voiding contractions, decreased voiding frequency, and increased postvoid residual. Intravesical application of GSK1016790A increased voiding frequency and reduced postvoid residual in wild-type, but not Trpv4-/-, rats. In isolated bladder strips, GSK1016790A did not induce relevant contractions, indicating that the observed improvements in bladder function are the result of increased afferent signalling through TRPV4 activation, rather than a local effect on the detrusor. The altered urinary phenotype of Trpv4-/- mice was not apparent in the Trpv4-/- rat model, suggesting species-related functional variations. Our results are limited to the preclinical setting in rodents. CONCLUSIONS Intravesical activation of TRPV4 improves bladder dysfunction after bPNI by increasing afferent signalling. PATIENT SUMMARY We demonstrate that the sensory protein transient receptor potential vanilloid 4 (TRPV4) can be targeted to improve bladder function in animals that have iatrogenic injury to the nerves innervating the bladder. Further research is required to determine whether these results can be translated to patients with an underactive bladder.


EJNMMI research | 2015

(18F)FDG-PET brain imaging during the micturition cycle in rats detects regions involved in bladder afferent signalling

Yves Deruyver; Roma Rietjens; Jan Franken; Silvia Pinto; Ann Van Santvoort; Cindy Casteels; Thomas Voets; Dirk De Ridder

BackgroundThis feasibility study established an experimental protocol to evaluate brain activation patterns using fluorodeoxyglucose positron emission tomography ((18F)FDG-PET) during volume-induced voiding and isovolumetric bladder contractions in rats.MethodsFemale Sprague-Dawley rats were anaesthetized with urethane and underwent either volume-induced voiding cystometry or isovolumetric cystometry and simultaneous functional PET brain imaging after injection of (18F)FDG in the tail vein. Brain glucose metabolism in both groups was compared to their respective control conditions (empty bladder). Relative glucose metabolism images were anatomically standardized to Paxinos space and analysed voxel-wise using Statistical Parametric Mapping 12 (SPM12).ResultsDuring volume-induced voiding, glucose hypermetabolism was observed in the insular cortex while uptake was decreased in a cerebellar cluster and the dorsal midbrain. Relative glucose metabolism during isovolumetric bladder contractions increased in the insular and cingulate cortices and decreased in the cerebellum.ConclusionsOur findings demonstrate that volume-induced voiding as well as isovolumetric bladder contractions in rats provokes changes in brain metabolism, including activation of the insular and cingulate cortices, which is consistent with their role in the mapping of bladder afferent activity. These findings are in line with human studies. Our results provide a basis for further research into the brain control of the lower urinary tract in small laboratory animals.

Collaboration


Dive into the Silvia Pinto's collaboration.

Top Co-Authors

Avatar

Thomas Voets

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Rudi Vennekens

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Dirk De Ridder

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Pieter Uvin

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Jan Franken

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Mathieu Boudes

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Wouter Everaerts

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Andrei Segal

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Roma Rietjens

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Yves Deruyver

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge