Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 172
Filtrar
1.
Cardiovasc Res ; 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38626311

RESUMEN

AIMS: Potential loss-of-function variants of ATP13A3, the gene encoding a P5B-type transport ATPase of undefined function, were recently identified in pulmonary arterial hypertension (PAH) patients. ATP13A3 is implicated in polyamine transport but its function has not been fully elucidated. Here, we sought to determine the biological function of ATP13A3 in vascular endothelial cells and how PAH-associated variants may contribute to disease pathogenesis. METHODS AND RESULTS: We studied the impact of ATP13A3 deficiency and overexpression in endothelial cell (EC) models (human pulmonary ECs, blood outgrowth ECs (BOECs) and HMEC-1 cells), including a PAH patient-derived BOEC line harbouring an ATP13A3 variant (LK726X). We also generated mice harbouring an Atp13a3 variant analogous to a human disease-associated variant to establish whether these mice develop PAH.ATP13A3 localised to the recycling endosomes of human ECs. Knockdown of ATP13A3 in ECs generally reduced the basal polyamine content and altered the expression of enzymes involved in polyamine metabolism. Conversely, overexpression of wild-type ATP13A3 increased polyamine uptake. Functionally, loss of ATP13A3 was associated with reduced EC proliferation, increased apoptosis in serum starvation and increased monolayer permeability to thrombin. Assessment of five PAH-associated missense ATP13A3 variants (L675V, M850I, V855M, R858H, L956P) confirmed loss-of-function phenotypes represented by impaired polyamine transport and dysregulated EC function. Furthermore, mice carrying a heterozygous germ-line Atp13a3 frameshift variant representing a human variant spontaneously developed a PAH phenotype, with increased pulmonary pressures, right ventricular remodelling and muscularisation of pulmonary vessels. CONCLUSION: We identify ATP13A3 as a polyamine transporter controlling polyamine homeostasis in ECs, deficiency of which leads to EC dysfunction and predisposes to PAH. This suggests a need for targeted therapies to alleviate the imbalances in polyamine homeostasis and EC dysfunction in PAH.

2.
Biochim Biophys Acta Mol Cell Res ; 1871(2): 119652, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38086447

RESUMEN

Pathogenic ATP10B variants have been described in patients with Parkinson's disease and dementia with Lewy body disease, and we previously established ATP10B as a late endo-/lysosomal lipid flippase transporting both phosphatidylcholine (PC) and glucosylceramide (GluCer) from the lysosomal exoplasmic to cytoplasmic membrane leaflet. Since several other lipid flippases regulate cellular lipid uptake, we here examined whether also ATP10B impacts cellular lipid uptake. Transient co-expression of ATP10B with its obligatory subunit CDC50A stimulated the uptake of fluorescently (NBD-) labeled PC in HeLa cells. This uptake is dependent on the transport function of ATP10B, is impaired by disease-associated variants and appears specific for NBD-PC. Uptake of non-ATP10B substrates, such as NBD-sphingomyelin or NBD-phosphatidylethanolamine is not increased. Remarkably, in stable cell lines co-expressing ATP10B/CDC50A we only observed increased NBD-PC uptake following treatment with rotenone, a mitochondrial complex I inhibitor that induces transport-dependent ATP10B phenotypes. Conversely, Im95m and WM-115 cells with endogenous ATP10B expression, present a decreased NBD-PC uptake following ATP10B knockdown, an effect that is exacerbated under rotenone stress. Our data show that the endo-/lysosomal lipid flippase ATP10B contributes to cellular PC uptake under specific cell stress conditions.


Asunto(s)
Rotenona , Humanos , Células HeLa , Rotenona/farmacología , Transporte Biológico , Membrana Celular/metabolismo
3.
Cell Death Dis ; 14(7): 436, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37454104

RESUMEN

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common genetic cause of Parkinson's disease (PD), with growing importance also for Crohn's disease and cancer. LRRK2 is a large and complex protein possessing both GTPase and kinase activity. Moreover, LRRK2 activity and function can be influenced by its phosphorylation status. In this regard, many LRRK2 PD-associated mutants display decreased phosphorylation of the constitutive phosphorylation cluster S910/S935/S955/S973, but the role of these changes in phosphorylation status with respect to LRRK2 physiological functions remains unknown. Here, we propose that the S910/S935/S955/S973 phosphorylation sites act as key regulators of LRRK2-mediated autophagy under both basal and starvation conditions. We show that quadruple LRRK2 phosphomutant cells (4xSA; S910A/S935A/S955A/S973A) have impaired lysosomal functionality and fail to induce and proceed with autophagy during starvation. In contrast, treatment with the specific LRRK2 kinase inhibitors MLi-2 (100 nM) or PF-06447475 (150 nM), which also led to decreased LRRK2 phosphorylation of S910/S935/S955/S973, did not affect autophagy. In explanation, we demonstrate that the autophagy impairment due to the 4xSA LRRK2 phospho-dead mutant is driven by its enhanced LRRK2 kinase activity. We show mechanistically that this involves increased phosphorylation of LRRK2 downstream targets Rab8a and Rab10, as the autophagy impairment in 4xSA LRRK2 cells is counteracted by expression of phosphorylation-deficient mutants T72A Rab8a and T73A Rab10. Similarly, reduced autophagy and decreased LRRK2 phosphorylation at the constitutive sites were observed in cells expressing the pathological R1441C LRRK2 PD mutant, which also displays increased kinase activity. These data underscore the relation between LRRK2 phosphorylation at its constitutive sites and the importance of increased LRRK2 kinase activity in autophagy regulation and PD pathology.


Asunto(s)
Autofagia , Proteínas de Unión al GTP rab , Fosforilación/fisiología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Mutación , Autofagia/genética , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
4.
Acta Neuropathol ; 146(2): 245-261, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37289222

RESUMEN

The link between the gut and the brain in Parkinson's disease (PD) pathogenesis is currently a subject of intense research. Indeed, gastrointestinal dysfunction is known as an early symptom in PD and inflammatory bowel disease (IBD) has recently been recognised as a risk factor for PD. The leucine-rich repeat kinase 2 (LRRK2) is a PD- and IBD-related protein with highest expression in immune cells. In this study, we provide evidence for a central role of LRRK2 in gut inflammation and PD. The presence of the gain-of-function G2019S mutation significantly increases the disease phenotype and inflammatory response in a mouse model of experimental colitis based on chronic dextran sulphate sodium (DSS) administration. Bone marrow transplantation of wild-type cells into G2019S knock-in mice fully rescued this exacerbated response, proving the key role of mutant LRRK2 in immune cells in this experimental colitis model. Furthermore, partial pharmacological inhibition of LRRK2 kinase activity also reduced the colitis phenotype and inflammation. Moreover, chronic experimental colitis also induced neuroinflammation and infiltration of peripheral immune cells into the brain of G2019S knock-in mice. Finally, combination of experimental colitis with overexpression of α-synuclein in the substantia nigra aggravated motor deficits and dopaminergic neurodegeneration in G2019S knock-in mice. Taken together, our results link LRRK2 with the immune response in colitis and provide evidence that gut inflammation can impact brain homeostasis and contribute to neurodegeneration in PD.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Enfermedad de Parkinson , Animales , Ratones , Colitis/inducido químicamente , Colitis/genética , Inmunidad , Inflamación , Enfermedades Inflamatorias del Intestino/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Ratones Transgénicos , Mutación/genética , Enfermedad de Parkinson/patología
5.
Biomolecules ; 13(6)2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37371498

RESUMEN

Polyamine homeostasis is disturbed in several human diseases, including cancer, which is hallmarked by increased intracellular polyamine levels and an upregulated polyamine transport system (PTS). Thus far, the polyamine transporters contributing to the elevated levels of polyamines in cancer cells have not yet been described, despite the fact that polyamine transport inhibitors are considered for cancer therapy. Here, we tested whether the upregulation of candidate polyamine transporters of the P5B transport ATPase family is responsible for the increased PTS in the well-studied breast cancer cell line MCF7 compared to the non-tumorigenic epithelial breast cell line MCF10A. We found that MCF7 cells presented elevated expression of a previously uncharacterized P5B-ATPase, ATP13A4, which was responsible for the elevated polyamine uptake activity. Furthermore, MCF7 cells were more sensitive to polyamine cytotoxicity, as demonstrated by cell viability, cell death and clonogenic assays. Importantly, the overexpression of ATP13A4 WT in MCF10A cells induced a MCF7 polyamine phenotype, with significantly higher uptake of BODIPY-labeled polyamines and increased sensitivity to polyamine toxicity. In conclusion, we established ATP13A4 as a new polyamine transporter in the human PTS and showed that ATP13A4 may play a major role in the increased polyamine uptake of breast cancer cells. ATP13A4 therefore emerges as a candidate therapeutic target for anticancer drugs that block the PTS.


Asunto(s)
Neoplasias de la Mama , Poliaminas , Femenino , Humanos , Adenosina Trifosfatasas/genética , Transporte Biológico , Neoplasias de la Mama/metabolismo , Células MCF-7 , Proteínas de Transporte de Membrana/metabolismo , Poliaminas/metabolismo , Regulación hacia Arriba
6.
Neurotherapeutics ; 20(1): 67-82, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-37052776

RESUMEN

Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA) are termed synucleinopathies, disorders that are characterized by the intracellular aggregation of the protein ɑ-synuclein. The cellular tropism of synuclein pathology in these syndromes is notably distinct since in the Lewy disorders, PD and DLB, ɑSyn forms aggregates in neurons whereas in MSA ɑSyn forms aggregates in oligodendrocytes. Studies examining ɑSyn pathology in experimental models and in human brain have now identified fibrillar ɑSyn with unique but distinct molecular signatures, suggesting that the structure of these ɑSyn fibrils might be closely tied to their cellular ontogeny. In contrast to the native structural heterogeneity of ɑSyn in vitro, the conformational landscape of fibrillar ɑSyn in human brain and in vivo transmission models appears to be remarkably uniform. Here, we review the studies by which we propose a hypothesis that the cellular host environment might be in part responsible for how ɑSyn filaments assemble into phenotype-specific strains. We postulate that the maturation of ɑSyn strains develops as a function of their in vivo transmission routes and cell-specific risk factors. The impact of the cellular environment on the structural diversity of ɑSyn might have important implications for the design of preclinical studies and their use for the development of ɑSyn-based biomarkers and therapeutic strategies. By combining phenotype-specific fibrils and relevant synucleinopathy transmission models, preclinical models might more closely reflect unique disease phenotypes.


Asunto(s)
Atrofia de Múltiples Sistemas , Enfermedad de Parkinson , Sinucleinopatías , Humanos , alfa-Sinucleína/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Sinucleinopatías/metabolismo , Neuronas/metabolismo
7.
NPJ Parkinsons Dis ; 9(1): 44, 2023 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-36973269

RESUMEN

In Parkinson's disease (PD) misfolded alpha-synuclein (aSyn) accumulates in the substantia nigra, where dopaminergic neurons are progressively lost. The mechanisms underlying aSyn pathology are still unclear, but they are hypothesized to involve the autophagy-lysosome pathway (ALP). LRRK2 mutations are a major cause of familial and sporadic PD, and LRRK2 kinase activity has been shown to be involved in pS129-aSyn inclusion modulation. We observed selective downregulation of the novel PD risk factor RIT2 in vitro and in vivo. Rit2 overexpression in G2019S-LRRK2 cells rescued ALP abnormalities and diminished aSyn inclusions. In vivo, viral mediated overexpression of Rit2 operated neuroprotection against AAV-A53T-aSyn. Furthermore, Rit2 overexpression prevented the A53T-aSyn-dependent increase of LRRK2 kinase activity in vivo. On the other hand, reduction of Rit2 levels leads to defects in the ALP, similar to those induced by the G2019S-LRRK2 mutation. Our data indicate that Rit2 is required for correct lysosome function, inhibits overactive LRRK2 to ameliorate ALP impairment, and counteracts aSyn aggregation and related deficits. Targeting Rit2 could represent an effective strategy to combat neuropathology in familial and idiopathic PD.

8.
Acta Neuropathol ; 145(5): 541-559, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36991261

RESUMEN

Symptoms in the urogenital organs are common in multiple system atrophy (MSA), also in the years preceding the MSA diagnosis. It is unknown how MSA is triggered and these observations in prodromal MSA led us to hypothesize that synucleinopathy could be triggered by infection of the genitourinary tract causing ɑ-synuclein (ɑSyn) to aggregate in peripheral nerves innervating these organs. As a first proof that peripheral infections could act as a trigger in MSA, this study focused on lower urinary tract infections (UTIs), given the relevance and high frequency of UTIs in prodromal MSA, although other types of infection might also be important triggers of MSA. We performed an epidemiological nested-case control study in the Danish population showing that UTIs are associated with future diagnosis of MSA several years after infection and that it impacts risk in both men and women. Bacterial infection of the urinary bladder triggers synucleinopathy in mice and we propose a novel role of ɑSyn in the innate immune system response to bacteria. Urinary tract infection with uropathogenic E. coli results in the de novo aggregation of ɑSyn during neutrophil infiltration. During the infection, ɑSyn is released extracellularly from neutrophils as part of their extracellular traps. Injection of MSA aggregates into the urinary bladder leads to motor deficits and propagation of ɑSyn pathology to the central nervous system in mice overexpressing oligodendroglial ɑSyn. Repeated UTIs lead to progressive development of synucleinopathy with oligodendroglial involvement in vivo. Our results link bacterial infections with synucleinopathy and show that a host response to environmental triggers can result in ɑSyn pathology that bears semblance to MSA.


Asunto(s)
Atrofia de Múltiples Sistemas , Sinucleinopatías , Infecciones Urinarias , Ratones , Femenino , Animales , Sinucleinopatías/patología , Estudios de Casos y Controles , Escherichia coli , Ratones Transgénicos , alfa-Sinucleína , Atrofia de Múltiples Sistemas/complicaciones , Atrofia de Múltiples Sistemas/patología , Infecciones Urinarias/complicaciones , Inmunidad Innata
9.
Int J Mol Sci ; 24(3)2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36768321

RESUMEN

Autosomal dominant mutations in the gene encoding α-synuclein (SNCA) were the first to be linked with hereditary Parkinson's disease (PD). Duplication and triplication of SNCA has been observed in PD patients, together with mutations at the N-terminal of the protein, among which A30P and A53T influence the formation of fibrils. By overexpressing human α-synuclein in the neuronal system of Drosophila, we functionally validated the ability of IP3K2, an ortholog of the GWAS identified risk gene, Inositol-trisphosphate 3-kinase B (ITPKB), to modulate α-synuclein toxicity in vivo. ITPKB mRNA and protein levels were also increased in SK-N-SH cells overexpressing wild-type α-synuclein, A53T or A30P mutants. Kinase overexpression was detected in the cytoplasmatic and in the nuclear compartments in all α-synuclein cell types. By quantifying mRNAs in the cortex of PD patients, we observed higher levels of ITPKB mRNA when SNCA was expressed more (p < 0.05), compared to controls. A positive correlation was also observed between SNCA and ITPKB expression in the cortex of patients, which was not seen in the controls. We replicated this observation in a public dataset. Our data, generated in SK-N-SH cells and in cortex from PD patients, show that the expression of α-synuclein and ITPKB is correlated in pathological situations.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Humanos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Mutación , Neuronas/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo
10.
Brain ; 146(1): 237-251, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-35170728

RESUMEN

Multiple system atrophy is a progressive neurodegenerative disease with prominent autonomic and motor features. During early stages, different subtypes of the disease are distinguished by their predominant parkinsonian or cerebellar symptoms, reflecting its heterogeneous nature. The pathognomonic feature of multiple system atrophy is the presence of α-synuclein (αSyn) protein deposits in oligodendroglial cells. αSyn can assemble in specific cellular or disease environments and form αSyn strains with unique structural features, but the ability of αSyn strains to propagate in oligodendrocytes remains elusive. Recently, it was shown that αSyn strains with related conformations exist in the brains of patients. Here, we investigated whether different αSyn strains can influence multiple system atrophy progression in a strain-dependent manner. To this aim, we injected two recombinant αSyn strains (fibrils and ribbons) in multiple system atrophy transgenic mice and found that they determined disease severity in multiple system atrophy via host-restricted and cell-specific pathology in vivo. αSyn strains significantly impact disease progression in a strain-dependent way via oligodendroglial, neurotoxic and immune-related mechanisms. Neurodegeneration and brain atrophy were accompanied by unique microglial and astroglial responses and the recruitment of central and peripheral immune cells. The differential activation of microglial cells correlated with the structural features of αSyn strains both in vitro and in vivo. Spectral analysis showed that ribbons propagated oligodendroglial inclusions that were structurally distinct from those of fibrils, with resemblance to oligodendroglial inclusions, in the brains of patients with multiple system atrophy. This study, therefore, shows that the multiple system atrophy phenotype is governed by both the nature of the αSyn strain and the host environment and that by injecting αSyn strains into an animal model of the disease, a more comprehensive phenotype can be established.


Asunto(s)
Atrofia de Múltiples Sistemas , alfa-Sinucleína , Ratones , Animales , alfa-Sinucleína/metabolismo , Atrofia de Múltiples Sistemas/patología , Modelos Animales de Enfermedad , Ratones Transgénicos , Gravedad del Paciente , Encéfalo/patología
11.
Neurobiol Dis ; 172: 105806, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35781002

RESUMEN

Parkinson's disease (PD) is currently considered a multisystemic disorder rather than a pure brain disease, in line with the multiple hit hypothesis from Braak. However, despite increasing evidence that the pathology might originate in the periphery, multiple unknown aspects and contradictory data on the pathological processes taking place in the periphery jeopardize the interpretation and therapeutic targeting of PD. Mutations in the leucine-rich-repeat kinase 2 (LRRK2) gene have been widely linked with familial and sporadic PD cases. However, the actual role of LRRK2 in PD pathophysiology is far from understood. There is evidence that LRRK2 may be involved in alpha-synuclein (α-synuclein) pathology and immune cell regulation, but it has also been associated with inflammatory diseases such as inflammatory bowel disease, tuberculosis, leprosy, and several other bacterial infections. In this review, we focus on the different roles of LRRK2 in the periphery. More specifically, we discuss the involvement of LRRK2 in the propagation of α-synuclein pathology and its regulatory role in peripheral inflammation. A deeper understanding of the multidimensional functions of LRRK2 will pave the way for more accurate characterization of PD pathophysiology and its association with other inflammatory diseases.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Mutación , Enfermedad de Parkinson/patología , alfa-Sinucleína/genética
12.
Small ; 18(18): e2200205, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35355419

RESUMEN

Optical interrogation of cellular electrical activity has proven itself essential for understanding cellular function and communication in complex networks. Voltage-sensitive dyes are important tools for assessing excitability but these highly lipophilic sensors may affect cellular function. Label-free techniques offer a major advantage as they eliminate the need for these external probes. In this work, it is shown that endogenous second-harmonic generation (SHG) from live cells is highly sensitive to changes in transmembrane potential (TMP). Simultaneous electrophysiological control of a living human embryonic kidney (HEK293T) cell, through a whole-cell voltage-clamp reveals a linear relation between the SHG intensity and membrane voltage. The results suggest that due to the high ionic strengths and fast optical response of biofluids, membrane hydration is not the main contributor to the observed field sensitivity. A conceptual framework is further provided that indicates that the SHG voltage sensitivity reflects the electric field within the biological asymmetric lipid bilayer owing to a nonzero χeff(2) tensor. Changing the TMP without surface modifications such as electrolyte screening offers high optical sensitivity to membrane voltage (≈40% per 100 mV), indicating the power of SHG for label-free read-out. These results hold promise for the design of a non-invasive label-free read-out tool for electrogenic cells.


Asunto(s)
Microscopía de Generación del Segundo Armónico , Colorantes , Células HEK293 , Humanos , Potenciales de la Membrana
13.
Curr Opin Neurobiol ; 72: 55-62, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34628360

RESUMEN

Genes associated with endolysosomal function have been recently associated with familial Parkinson's disease and described as risk factors for sporadic cases. This indicates that deficits in this pathway predispose to parkinsonism. To better understand the role of these genes in disease development, rodent models have been created by targeting genes playing a role in endolysosomal function, such as LRRK2, DNAJC6, SYNJ1, VPS35, GBA1, ATP13A2 and TMEM175. Here, we review the latest findings describing parkinsonian features in these animal models secondary to endolysosomal dysfunction. Also, we provide suggestions for further development and application of these animal models to better understand the contribution of endolysosomal dysfunction in Parkinson's disease and provide novel models for testing therapeutic approaches.


Asunto(s)
Enfermedad de Parkinson , Animales , Endosomas , Lisosomas/genética , Lisosomas/metabolismo , Enfermedad de Parkinson/genética , Roedores
14.
Exp Neurol ; 347: 113900, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34695425

RESUMEN

During the pathogenesis of Parkinson's disease (PD), aggregation of alpha-synuclein (αSyn) induces a vicious cycle of cellular impairments that lead to neurodegeneration. Consequently, removing toxic αSyn aggregates constitutes a plausible strategy against PD. In this work, we tested whether stimulating the autolysosomal degradation of αSyn aggregates through the Ras-related in brain 7 (Rab7) pathway can reverse αSyn-induced cellular impairment and prevent neurodegeneration in vivo. The disease-related A53T mutant of αSyn was expressed in primary neurons and in dopaminergic neurons of the rat brain simultaneously with wild type (WT) Rab7 or the T22N mutant as negative control. The cellular integrity was quantified by morphological and biochemical analyses. In primary neurons, WT Rab7 rescued the αSyn-induced loss of neurons and neurites. Furthermore, Rab7 decreased the amount of reactive oxygen species and the amount of Triton X-100 insoluble αSyn. In rat brain, WT Rab7 reduced αSyn-induced loss of dopaminergic axon terminals in the striatum and the loss of dopaminergic dendrites in the substantia nigra pars reticulata. Further, WT Rab7 lowered αSyn pathology as quantified by phosphorylated αSyn staining. Finally, WT Rab7 attenuated αSyn-induced DNA damage in primary neurons and rat brain. In brief, Rab7 reduced αSyn-induced pathology, ameliorated αSyn-induced neuronal degeneration, oxidative stress and DNA damage. These findings indicate that Rab7 is able to disrupt the vicious cycle of cellular impairment, αSyn pathology and neurodegeneration present in PD. Stimulation of Rab7 and the autolysosomal degradation pathway could therefore constitute a beneficial strategy for PD.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , alfa-Sinucleína/biosíntesis , alfa-Sinucleína/toxicidad , Proteínas de Unión a GTP rab7/biosíntesis , Proteínas de Unión a GTP rab7/farmacología , Animales , Células Cultivadas , Daño del ADN/efectos de los fármacos , Daño del ADN/fisiología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
15.
J Neurochem ; 158(5): 1186-1198, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34338310

RESUMEN

During adult rodent life, newborn neurons are added to the olfactory bulb (OB) in a tightly controlled manner. Upon arrival in the OB, input synapses from the local bulbar network and the higher olfactory cortex precede the formation of functional output synapses, indicating a possible role for these regions in newborn neuron survival. An interplay between the environment and the piriform cortex in the regulation of newborn neuron survival has been suggested. However, the specific network and the neuronal cell types responsible for this effect have not been elucidated. Furthermore, the role of the other olfactory cortical areas in this process is not known. Here we demonstrate that pyramidal neurons in the mouse anterior olfactory nucleus, the first cortical area for odor processing, have a key role in the survival of newborn neurons. Using DREADD (Designer Receptors Exclusively Activated by Designer Drugs) technology, we applied chronic stimulation to the anterior olfactory nucleus and observed a decrease in newborn neurons in the OB through induction of apoptosis. These findings provide further insight into the network regulating neuronal survival in adult neurogenesis and strengthen the importance of the surrounding network for sustained integration of new neurons.


Asunto(s)
Neurogénesis/fisiología , Neuronas/fisiología , Bulbo Olfatorio/citología , Bulbo Olfatorio/fisiología , Corteza Olfatoria/citología , Corteza Olfatoria/fisiología , Factores de Edad , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Femenino , Ratones , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Odorantes , Bulbo Olfatorio/efectos de los fármacos , Corteza Olfatoria/efectos de los fármacos , Vías Olfatorias/citología , Vías Olfatorias/efectos de los fármacos , Vías Olfatorias/fisiología , Olfato/fisiología
16.
Neurobiol Dis ; 157: 105426, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34144124

RESUMEN

LRRK2 is a highly phosphorylated multidomain protein and mutations in the gene encoding LRRK2 are a major genetic determinant of Parkinson's disease (PD). Dephosphorylation at LRRK2's S910/S935/S955/S973 phosphosite cluster is observed in several conditions including in sporadic PD brain, in several disease mutant forms of LRRK2 and after pharmacological LRRK2 kinase inhibition. However, the mechanism of LRRK2 dephosphorylation is poorly understood. We performed a phosphatome-wide reverse genetics screen to identify phosphatases involved in the dephosphorylation of the LRRK2 phosphosite S935. Candidate phosphatases selected from the primary screen were tested in mammalian cells, Xenopus oocytes and in vitro. Effects of PP2A on endogenous LRRK2 phosphorylation were examined via expression modulation with CRISPR/dCas9. Our screening revealed LRRK2 phosphorylation regulators linked to the PP1 and PP2A holoenzyme complexes as well as CDC25 phosphatases. We showed that dephosphorylation induced by different kinase inhibitor triggered relocalisation of phosphatases PP1 and PP2A in LRRK2 subcellular compartments in HEK-293 T cells. We also demonstrated that LRRK2 is an authentic substrate of PP2A both in vitro and in Xenopus oocytes. We singled out the PP2A holoenzyme PPP2CA:PPP2R2 as a powerful phosphoregulator of pS935-LRRK2. Furthermore, we demonstrated that this specific PP2A holoenzyme induces LRRK2 relocalization and triggers LRRK2 ubiquitination, suggesting its involvement in LRRK2 clearance. The identification of the PPP2CA:PPP2R2 complex regulating LRRK2 S910/S935/S955/S973 phosphorylation paves the way for studies refining PD therapeutic strategies that impact LRRK2 phosphorylation.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 2/metabolismo , Animales , Células HEK293 , Holoenzimas/metabolismo , Humanos , Técnicas In Vitro , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteínas del Tejido Nervioso/metabolismo , Oocitos/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Proteínas de Xenopus/metabolismo , Xenopus laevis
17.
Hum Gene Ther ; 32(11-12): 616-627, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34006117

RESUMEN

Several neurodegenerative disorders are characterized by oligodendroglial pathology and myelin loss. Oligodendrogliopathies are a group of rare diseases for which there currently is no therapy. Gene delivery through viral vectors to oligodendrocytes is a potential strategy to deliver therapeutic molecules to oligodendrocytes for disease modification. However, targeting oligodendroglial cells in vivo is challenging due to their widespread distribution in white and gray matter. In this study, we aimed to address several of these difficulties by designing and testing different oligodendroglial targeting vectors in rat and mouse brain, utilizing different promoters, serotypes, and delivery routes. We found that different oligodendroglial promoters (myelin basic protein [MBP], cytomegalovirus-enhanced MBP, and myelin-associated glycoprotein [MAG]) vary considerably in their ability to drive oligodendroglial transgene expression and different viral vector serotypes (rAAV2/7, rAAV2/8, and rAAV2/9) exhibit varying efficacies in transducing oligodendrocytes. Different administration routes through intracerebral or intraventricular injection allow widespread targeting of mature oligodendrocytes. Delivery of rAAV2/9-MAG-GFP into the cerebrospinal fluid results in GFP expression along the entire rostrocaudal axis of the spinal cord. Collectively, these results show that oligodendrocytes can be targeted with high specificity and widespread expression, which will be useful for gene therapeutic interventions or disease modeling purposes.


Asunto(s)
Oligodendroglía , Roedores , Animales , Encéfalo , Vectores Genéticos/genética , Ratones , Ratas , Transgenes
18.
Int J Mol Sci ; 22(5)2021 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-33799982

RESUMEN

ATP13A2, a late endo-/lysosomal polyamine transporter, is implicated in a variety of neurodegenerative diseases, including Parkinson's disease and Kufor-Rakeb syndrome, an early-onset atypical form of parkinsonism. Loss-of-function mutations in ATP13A2 result in lysosomal deficiency as a consequence of impaired lysosomal export of the polyamines spermine/spermidine. Furthermore, accumulating evidence suggests the involvement of ATP13A2 in regulating the fate of α-synuclein, such as cytoplasmic accumulation and external release. However, no consensus has yet been reached on the mechanisms underlying these effects. Here, we aimed to gain more insight into how ATP13A2 is linked to α-synuclein biology in cell models with modified ATP13A2 activity. We found that loss of ATP13A2 impairs lysosomal membrane integrity and induces α-synuclein multimerization at the membrane, which is enhanced in conditions of oxidative stress or exposure to spermine. In contrast, overexpression of ATP13A2 wildtype (WT) had a protective effect on α-synuclein multimerization, which corresponded with reduced αsyn membrane association and stimulation of the ubiquitin-proteasome system. We also found that ATP13A2 promoted the secretion of α-synuclein through nanovesicles. Interestingly, the catalytically inactive ATP13A2 D508N mutant also affected polyubiquitination and externalization of α-synuclein multimers, suggesting a regulatory function independent of the ATPase and transport activity. In conclusion, our study demonstrates the impact of ATP13A2 on α-synuclein multimerization via polyamine transport dependent and independent functions.


Asunto(s)
ATPasas de Translocación de Protón/metabolismo , alfa-Sinucleína/metabolismo , Línea Celular Tumoral , Exocitosis , Humanos , Membranas Intracelulares/metabolismo , Lisosomas/metabolismo , Mutación , Estrés Oxidativo , Complejo de la Endopetidasa Proteasomal/metabolismo , Multimerización de Proteína , ATPasas de Translocación de Protón/genética , Espermina/metabolismo , Ubiquitina/metabolismo
19.
Epilepsia ; 62(3): 659-670, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33570167

RESUMEN

OBJECTIVE: One third of epilepsy patients do not become seizure-free using conventional medication. Therefore, there is a need for alternative treatments. Preclinical research using designer receptors exclusively activated by designer drugs (DREADDs) has demonstrated initial success in suppressing epileptic activity. Here, we evaluated whether long-term chemogenetic seizure suppression could be obtained in the intraperitoneal kainic acid rat model of temporal lobe epilepsy, when DREADDs were selectively expressed in excitatory hippocampal neurons. METHODS: Epileptic male Sprague Dawley rats received unilateral hippocampal injections of adeno-associated viral vector encoding the inhibitory DREADD hM4D(Gi), preceded by a cell-specific promotor targeting excitatory neurons. The effect of clozapine-mediated DREADD activation on dentate gyrus evoked potentials and spontaneous electrographic seizures was evaluated. Animals were systemically treated with single (.1 mg/kg/24 h) or repeated (.1 mg/kg/6 h) injections of clozapine. In addition, long-term continuous release of clozapine and olanzapine (2.8 mg/kg/7 days) using implantable minipumps was evaluated. All treatments were administered during the chronic epileptic phase and between 1.5 and 13.5 months after viral transduction. RESULTS: In the DREADD group, dentate gyrus evoked potentials were inhibited after clozapine treatment. Only in DREADD-expressing animals, clozapine reduced seizure frequency during the first 6 h postinjection. When administered repeatedly, seizures were suppressed during the entire day. Long-term treatment with clozapine and olanzapine both resulted in significant seizure-suppressing effects for multiple days. Histological analysis revealed DREADD expression in both hippocampi and some cortical regions. However, lesions were also detected at the site of vector injection. SIGNIFICANCE: This study shows that inhibition of the hippocampus using chemogenetics results in potent seizure-suppressing effects in the intraperitoneal kainic acid rat model, even 1 year after viral transduction. Despite a need for further optimization, chemogenetic neuromodulation represents a promising treatment prospect for temporal lobe epilepsy.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Clozapina/uso terapéutico , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Olanzapina/uso terapéutico , Receptores de Neurotransmisores/genética , Animales , Giro Dentado/efectos de los fármacos , Giro Dentado/fisiopatología , Modelos Animales de Enfermedad , Potenciales Evocados/fisiología , Quinasas de Receptores Acoplados a Proteína-G/efectos de los fármacos , Quinasas de Receptores Acoplados a Proteína-G/genética , Edición Génica/métodos , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de Neurotransmisores/efectos de los fármacos , Convulsiones/prevención & control
20.
Neurotherapeutics ; 18(2): 949-961, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33594532

RESUMEN

The development of disease-modifying therapies for Parkinson's disease is a major challenge which would be facilitated by a better understanding of the pathogenesis. Leucine-rich repeat kinase 2 (LRRK2) and α-synuclein are key players in Parkinson's disease, but their relationship remains incompletely resolved. Previous studies investigating the effect of LRRK2 on α-synuclein-induced neurotoxicity and neuroinflammation in preclinical Parkinson's disease models have reported conflicting results. Here, we aimed to further explore the functional interaction between α-synuclein and LRRK2 and to evaluate the therapeutic potential of targeting physiological LRRK2 levels. We studied the effects of total LRRK2 protein loss as well as pharmacological LRRK2 kinase inhibition in viral vector-mediated α-synuclein-based Parkinson's disease models developing early- and late-stage neurodegeneration. Surprisingly, total LRRK2 ablation or in-diet treatment with the LRRK2 kinase inhibitor MLi-2 did not significantly modify α-synuclein-induced motor deficits, dopaminergic cell loss, or α-synuclein pathology. Interestingly, we found a significant effect on α-synuclein-induced neuroinflammatory changes in the absence of LRRK2, with a reduced microglial activation and CD4+ and CD8+ T cell infiltration. This observed lack of protection against α-synuclein-induced toxicity should be well considered in light of the ongoing therapeutic development of LRRK2 kinase inhibitors for idiopathic Parkinson's disease. Future studies will be crucial to understand the link between these neuroinflammatory processes and disease progression as well as the role of α-synuclein and LRRK2 in these pathological events.


Asunto(s)
Indazoles/administración & dosificación , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/antagonistas & inhibidores , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neuroinflamatorias/enzimología , Pirimidinas/administración & dosificación , alfa-Sinucleína/toxicidad , Animales , Masculino , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/patología , Enfermedades Neuroinflamatorias/inducido químicamente , Enfermedades Neuroinflamatorias/patología , Ratas , Ratas Long-Evans , Ratas Transgénicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...