Schematic representation of the mechanisms responsible of rotenone, MPP + and 6-OHDA toxicity in β-cells. (a) Rotenone, highly lipophilic, enters the cell freely, it reduces GSIS and it strongly inhibits mitochondrial Complex I, causing an increase of ROS production that is responsible of cell distress. MPP + enters the cell through DAT and other transporters and then it has a mechanism similar to rotenone. 6-OHDA enters the cell through DAT transporter and then it transforms to 6-OHDA quinones responsible of the formation of H 2 O 2 and other oxidative species. It also reduces mitochondrial complex I and IV activity but it does not affect GSIS. (b) Table summarizing the IC 50 of the three neurotoxins in the different assays and morphological changes analyzed with electronic microscopy.

Schematic representation of the mechanisms responsible of rotenone, MPP + and 6-OHDA toxicity in β-cells. (a) Rotenone, highly lipophilic, enters the cell freely, it reduces GSIS and it strongly inhibits mitochondrial Complex I, causing an increase of ROS production that is responsible of cell distress. MPP + enters the cell through DAT and other transporters and then it has a mechanism similar to rotenone. 6-OHDA enters the cell through DAT transporter and then it transforms to 6-OHDA quinones responsible of the formation of H 2 O 2 and other oxidative species. It also reduces mitochondrial complex I and IV activity but it does not affect GSIS. (b) Table summarizing the IC 50 of the three neurotoxins in the different assays and morphological changes analyzed with electronic microscopy.

Source publication
Article
Full-text available
Neurotoxins such as rotenone, 1-methyl-4-phenylpyridinium (MPP+) and 6-hydroxydopamine (6-OHDA) are well known for their high toxicity on dopaminergic neurons and are associated with Parkinson’s disease (PD) in murine models and humans. In addition, PD patients often have glucose intolerance and may develop type 2 diabetes (T2D), whereas T2D patien...

Contexts in source publication

Context 1
... in other cell types, such as microglial cells, breast cancer cells and fibroblasts, rotenone was toxic but at higher concentrations, showing a similarity between β-cells and dopaminergic cell lines in terms of high vulnerability to rotenone [42][43][44]. Regarding the mechanism of action, rotenone acts as mitochondrial complex I inhibitor and it has been used as a PD model for both in vitro and in vivo studies [25] (Figure 6). This inhibitory effect is responsible for mitochondrial dysfunction that causes an increase of ROS production and a reduction of cellular bioenergetics. ...
Context 2
... together, these data confirm the inhibitory action of rotenone on β-cells mitochondria, involving complex I, altering cellular bioenergetics and increasing ROS production that is responsible for cell death. For MPP + , we can postulate a similar mechanism as it is a potent inhibitor of the mitochondrial complex I [29], whereas for 6-OHDA the generation of H 2 O 2 and other oxidative species through its chemical transformation should be responsible for its toxicity together with the inhibition of mitochondrial complex I and IV [21] (Figure 6). The connection between mitochondrial inhibition, determined by redox activity or ATP production and cell death is not simple [45]. ...

Citations

... Interestingly, ROT prevented this effect, suggesting that ROT works independently upon its action on VD3R. Several earlier and more recent studies [59][60][61][62][63] on cell lines (SK-N-MC human neuroblastoma cells, differentiated dopaminergic neuroblastoma SH-SY5Y cells, HL-60 cells, pancreatic β-cell lines) and primary dopaminergic cultures, as well, showed that ROT alters mitochondrial structure and bioenergetics, decreasing mitochondrial membrane potential and increasing ROS production. Thus, we demonstrated that, in the PC12 cells model, ROT induces all those changes, and the oxidative stress produced leads to cell death. ...
Article
Full-text available
Parkinson’s disease (PD) is characterized by dopaminergic cell loss in the substantia nigra, and PD brains show neuroinflammation, oxidative stress, and mitochondrial dysfunction. The study evaluated the neuroprotective activity of 1α,25-dihydroxy vitamin D3 (VD3), on the rotenone (ROT)-induced cytotoxicity in PC12 cells. The viability parameters were assessed by the MTT and flow cytometry, on cells treated or not with VD3 and/or ROT. Besides, ROS production, cell death, mitochondrial transmembrane potential, reduced GSH, superoxide accumulation, molecular docking (TH and Keap1-Nrf2), and TH, Nrf2, NF-kB, and VD3 receptor protein contents by western blot were evaluated. VD3 was shown to improve the viability of ROT-exposed cells. Cells exposed to ROT showed increased production of ROS and superoxide, which decreased after VD3. ROT decrease in the mitochondrial transmembrane potential was prevented, after VD3 treatment and, VD3 was shown to interact with tyrosine hydroxylase (TH) and Nrf2. While ROT decreased TH, Nrf2, and NF-kB expressions, these effects were reversed by VD3. In addition, VD3 also increased VD3 receptor protein contents and values went back to those of controls after ROT exposure. VD3 protects PC12 cells against ROT damage, by decreasing oxidative stress and improving mitochondrial function. One target seems to be the TH molecule and possibly an indirect Nrf2 activation could also justify its neuroprotective actions on this PC12 cell model of PD.
Article
Full-text available
Nanometric scale size oscillations seem to be a fundamental feature of all living organisms on Earth. Their detection usually requires complex and very sensitive devices. However, some recent studies demonstrated that very simple optical microscopes and dedicated image processing software can also fulfill this task. This novel technique, termed as optical nanomotion detection (ONMD), was recently successfully used on yeast cells to conduct rapid antifungal sensitivity tests. In this study, we demonstrate that the ONMD method can monitor motile sub-cellular organelles, such as mitochondria. Here, mitochondrial isolates (from HEK 293 T and Jurkat cells) undergo predictable motility when viewed by ONMD and triggered by mitochondrial toxins, citric acid intermediates, and dietary and bacterial fermentation products (short-chain fatty acids) at various doses and durations. The technique has superior advantages compared to classical methods since it is rapid, possesses a single organelle sensitivity, and is label-and attachment-free.
Article
About 10% of the adult population is living with type 2 diabetes mellitus (T2DM) and 1% of the population over 60 years of age is suffering from Parkinson’s disease (PD). A school of thought firmly believes that T2DM, an age-related disease, augments PD risk. Such relationship is reflected from the severity of PD symptoms in drug naive subjects possessing T2DM. Onset of Parkinsonian feature in case controls possessing T2DM corroborates the role of hyperglycemia in PD. A few cohort, meta-analysis and animal studies have shown an increased PD risk owing to insulin resistance. High fat diet and role of insulin signaling in the regulation of sugar metabolism, oxidative stress, α-synuclein aggregation and accumulation, inflammatory response and mitochondrial function in PD models and sporadic PD further connect the two. Although little is reported about the implication of PD in hyperglycemia and T2DM, a few studies have also contradicted. Ameliorative effect of anti-diabetic drugs on Parkinsonian symptoms and vague outcome of anti-PD medications in T2DM patients also suggest a link. The article reviews the literature supporting augmented risk of one by the other, analysis of proof of the concept, facts, challenges, future possibilities and standpoint on the subject.
Article
Insulin-secreting β-cells in the pancreatic islets are exposed to various endogenous and exogenous stressing conditions, which may lead to β-cell dysfunction or apoptosis and ultimately to diabetes mellitus. However, the detailed molecular mechanisms underlying β-cell's inability to survive under severe stresses remain to be explored. This study used two common chemical stressors, thapsigargin and rotenone, to induce endoplasmic reticulum (ER) and mitochondria stress in a rat insuloma INS-1 832/13 β-cell line, mimicking the conditions experienced by dysfunctional β-cells. Proteomic changes of cells upon treatment with stressors at IC50 were profiled with TMT-based quantitative proteomics and further verified using label-free quantitive proteomics. The differentially expressed proteins under stress conditions were selected for in-depth bioinformatic analysis. Thapsigargin treatment specifically perturbed unfolded protein response (UPR) related pathways; in addition, 58 proteins not previously linked to the UPR related pathways were identified with consistent upregulation under stress induced by thapsigargin. Conversely, rotenone treatment resulted in significant proteome changes in key mitochondria regulatory pathways such as fatty acid β-oxidation, cellular respiration, citric acid cycle, and respiratory electron transport. Our data also demonstrated that both stressors increased reactive oxygen species production and depleted adenosine triphosphate synthesis, resulting in significant dysregulation of oxidative phosphorylation signaling pathways. These novel dysregulated proteins may suggest an alternative mechanism of action in β-cell dysfunction and provide potential targets for probing ER- and mitochondria stress-induced β-cell death.