Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Biomolecules ; 14(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38672445

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal motoneuron degenerative disease that is associated with demyelination. The Wobbler (WR) mouse exhibits motoneuron degeneration, gliosis and myelin deterioration in the cervical spinal cord. Since male WRs display low testosterone (T) levels in the nervous system, we investigated if T modified myelin-relative parameters in WRs in the absence or presence of the aromatase inhibitor, anastrozole (A). We studied myelin by using luxol-fast-blue (LFB) staining, semithin sections, electron microscopy and myelin protein expression, density of IBA1+ microglia and mRNA expression of inflammatory factors, and the glutamatergic parameters glutamine synthetase (GS) and the transporter GLT1. Controls and WR + T showed higher LFB, MBP and PLP staining, lower g-ratios and compact myelin than WRs and WR + T + A, and groups showing the rupture of myelin lamellae. WRs showed increased IBA1+ cells and mRNA for CD11b and inflammatory factors (IL-18, TLR4, TNFαR1 and P2Y12R) vs. controls or WR + T. IBA1+ cells, and CD11b were not reduced in WR + T + A, but inflammatory factors' mRNA remained low. A reduction of GS+ cells and GLT-1 immunoreactivity was observed in WRs and WR + T + A vs. controls and WR + T. Clinically, WR + T but not WR + T + A showed enhanced muscle mass, grip strength and reduced paw abnormalities. Therefore, T effects involve myelin protection, a finding of potential clinical translation.


Subject(s)
Amyotrophic Lateral Sclerosis , Disease Models, Animal , Myelin Sheath , Testosterone , Animals , Mice , Myelin Sheath/metabolism , Myelin Sheath/drug effects , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Male , Testosterone/pharmacology , Spinal Cord/metabolism , Spinal Cord/drug effects , Spinal Cord/pathology , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 2/genetics , Microglia/drug effects , Microglia/metabolism , Microglia/pathology
2.
Mol Neurobiol ; 61(1): 1-14, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37566177

ABSTRACT

Glucocorticoids exert antiinflammatory, antiproliferative and immunosupressive effects. Paradoxically they may also enhance inflammation particularly in the nervous system, as shown in Cushing´ syndrome and neurodegenerative disorders of humans and models of human diseases. ."The Wobbler mouse model of amyotrophic lateral sclerosis shows hypercorticoidism and neuroinflammation which subsided by treatment with the glucocorticoid receptor (GR) modulator Dazucorilant (CORT113176). This effect suggests that GR mediates the chronic glucocorticoid unwanted effects. We now tested this hypothesis using a chronic stress model resembling the condition of the Wobbler mouse Male NFR/NFR mice remained as controls or were subjected to a restraining / rotation stress protocol for 3 weeks, with a group of stressed mice receiving CORT113176 also for 3 weeks. We determined the mRNAS or reactive protein for the proinflamatory factors HMGB1, TLR4, NFkB, TNFα, markers of astrogliosis (GFAP, SOX9 and acquaporin 4), of microgliosis (Iba, CD11b, P2RY12 purinergic receptor) as well as serum IL1ß and corticosterone. We showed that chronic stress produced high levels of serum corticosterone and IL1ß, decreased body and spleen weight, produced microgliosis and astrogliosis and increased proinflammatory mediators. In stressed mice, modulation of the GR with CORT113176 reduced Iba + microgliosis, CD11b and P2RY12 mRNAs, immunoreactive HMGB1 + cells, GFAP + astrogliosis, SOX9 and acquaporin expression and TLR4 and NFkB mRNAs vs. stress-only mice. The effects of CORT113176 indicate that glucocorticoids are probably involved in neuroinflammation. Thus, modulation of the GR would become useful to dampen the inflammatory component of neurodegenerative disorders.


Subject(s)
HMGB1 Protein , Isoquinolines , Neurodegenerative Diseases , Pyrazoles , Male , Mice , Humans , Animals , Receptors, Glucocorticoid/metabolism , Corticosterone , HMGB1 Protein/metabolism , Neuroinflammatory Diseases , Gliosis/metabolism , Toll-Like Receptor 4/metabolism , Glucocorticoids/pharmacology , Spinal Cord/metabolism , Neurodegenerative Diseases/metabolism
3.
J Neuroendocrinol ; 36(2): e13362, 2024 02.
Article in English | MEDLINE | ID: mdl-38148478

ABSTRACT

Excessive consumption of sugary drinks negatively impacts the developing brain, producing long-lasting behavioral and metabolic disorders. Here, we study whether treatment with the antihyperglycemic agent metformin prevents some of the anxiety and memory alterations produced by chronic sucrose consumption. Male Sprague-Dawley rats had unrestricted access to water (control group) and a bottle containing a 10% sucrose solution (sucrose group, SUC) for 35 days. In parallel, a group of animals from SUC received metformin (25 mg/kg or 50 mg/kg, orally; MET 25 and MET 50 groups, respectively). After 2 weeks of metformin treatment, the animals weighed less than controls. SUC and MET 50 groups compensated for the caloric intake from the sugary solution by consuming less chow. In contrast, total energy intake in MET 25 was higher than the rest of the groups, but they still weighed less than control and SUC groups, suggesting that at this concentration, metformin delays body growth. The animals were then tested for the open field (OF), elevated plus maze (EPM) and novel object location (NOL) tests. In the OF, SUC animals spent more time in the central zone of the arena, evidenced by an increased number of entries and the distance traveled there. In the EPM, SUC animals spent more time in the open arms and less time in the central square. Metformin treatment prevented the decreased anxiety observed in SUC animals in the OF and EPM. In the NOL test, SUC animals showed less interest in novelty and metformin treatment did not improve this alteration. The preference for open spaces in the OF and EPM were associated with increased serum triglycerides (TG) and malondialdehyde levels in the medial prefrontal cortex (mPFC) and the hippocampus (HIP), while poor memory performance was associated with high basal blood glucose levels. In conclusion, the decreased anxiety-like behavior produced by chronic sucrose consumption was prevented by metformin treatment, through a mechanism that probably involves normalization of TG levels and decreased oxidative stress in mPFC and HIP.


Subject(s)
Metformin , Sucrose , Rats , Male , Animals , Rats, Sprague-Dawley , Metformin/pharmacology , Anxiety/drug therapy , Anxiety/metabolism , Brain
4.
Biomedicines ; 10(11)2022 Oct 27.
Article in English | MEDLINE | ID: mdl-36359243

ABSTRACT

We previously described that excessive consumption of sucrose during youth produces fear memory and anxiety-like behavior in adulthood. Here, we evaluated whether high cognitive function is also affected by studying early sucrose consumption in object recognition memory (NOR). Male Sprague Dawley rats were tested for short-term, long-term, and consolidated NOR after 25 days of unlimited sucrose access in juvenile (PD 25-50) or adult age (PD 75-100). All rats spent equal time exploring the two objects during the sample phase T1. When animals were exposed for 2, 24 h or 7 days later to a copy of the objects presented in T1 and a novel object, the sucrose-exposed juvenile group failed to distinguish between the familiar and the novel objects in contrast with the rest of the groups. Sucrose-exposed animals developed hypertriglyceridemia and glucose intolerance, but juvenile animals showed increased fasting glycemia and sustained the glucose intolerance longer. Moreover, sucrose decreased hippocampal proBDNF expression in juveniles while it was increased in adults, and sucrose also increased RAGE expression in adults. The NOR exploration ratio correlated negatively with basal glycemia and positively with proBDNF. Taken together, these data suggest that sucrose-induced alterations in glucose metabolism may contribute to a long-term decline in proBDNF and impaired recognition memory.

5.
Cell Mol Neurobiol ; 42(1): 23-40, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34138412

ABSTRACT

Progesterone regulates a number of processes in neurons and glial cells not directly involved in reproduction or sex behavior. Several neuroprotective effects are better observed under pathological conditions, as shown in the Wobbler mouse model of amyotrophic laterals sclerosis (ALS). Wobbler mice are characterized by forelimb atrophy due to motoneuron degeneration in the spinal cord, and include microgliosis and astrogliosis. Here we summarized current evidence on progesterone reversal of Wobbler neuropathology. We demonstrated that progesterone decreased motoneuron vacuolization with preservation of mitochondrial respiratory complex I activity, decreased mitochondrial expression and activity of nitric oxide synthase, increased Mn-dependent superoxide dismutase, stimulated brain-derived neurotrophic factor, increased the cholinergic phenotype of motoneurons, and enhanced survival with a concomitant decrease of death-related pathways. Progesterone also showed differential effects on glial cells, including increased oligodendrocyte density and downregulation of astrogliosis and microgliosis. These changes associate with reduced anti-inflammatory markers. The enhanced neurochemical parameters were accompanied by longer survival and increased muscle strength in tests of motor behavior. Because progesterone is locally metabolized to allopregnanolone (ALLO) in nervous tissues, we also studied neuroprotection by this derivative. Treatment of Wobbler mice with ALLO decreased oxidative stress and glial pathology, increased motoneuron viability and clinical outcome in a progesterone-like manner, suggesting that ALLO could mediate some progesterone effects in the spinal cord. In conclusion, the beneficial effects observed in different parameters support the versatile properties of progesterone and ALLO in a mouse model of motoneuron degeneration. The studies foresee future therapeutic opportunities with neuroactive steroids for deadly diseases like ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Neuroprotective Agents , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Mice , Motor Neurons , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Pregnanolone/metabolism , Pregnanolone/pharmacology , Pregnanolone/therapeutic use , Progesterone/metabolism , Progesterone/pharmacology , Progesterone/therapeutic use , Spinal Cord/metabolism
6.
Mol Neurobiol ; 58(5): 2088-2106, 2021 May.
Article in English | MEDLINE | ID: mdl-33411236

ABSTRACT

Patients suffering of amyotrophic lateral sclerosis (ALS) present motoneuron degeneration leading to muscle atrophy, dysphagia, and dysarthria. The Wobbler mouse, an animal model of ALS, shows a selective loss of motoneurons, astrocytosis, and microgliosis in the spinal cord. The incidence of ALS is greater in men; however, it increases in women after menopause, suggesting a role of sex steroids in ALS. Testosterone is a complex steroid that exerts its effects directly via androgen (AR) or Sigma-1 receptors and indirectly via estrogen receptors (ER) after aromatization into estradiol. Its reduced-metabolite 5α-dihydrotestosterone acts via AR. This study analyzed the effects of testosterone in male symptomatic Wobblers. Controls or Wobblers received empty or testosterone-filled silastic tubes for 2 months. The cervical spinal cord from testosterone-treated Wobblers showed (1) similar androgen levels to untreated control and (2) increased levels of testosterone, and its 5α-reduced metabolites, 5α- dihydrotestosterone, and 3ß-androstanediol, but (3) undetectable levels of estradiol compared to untreated Wobblers. Testosterone-treated controls showed comparable steroid concentrations to its untreated counterpart. In testosterone- treated Wobblers a reduction of AR, ERα, and aromatase and high levels of Sigma-1 receptor mRNAs was demonstrated. Testosterone treatment increased ChAT immunoreactivity and the antiinflammatory mediator TGFß, while it lessened vacuolated motoneurons, GFAP+ astrogliosis, the density of IBA1+ microgliosis, proinflammatory mediators, and oxidative/nitrosative stress. Clinically, testosterone treatment in Wobblers slowed the progression of paw atrophy and improved rotarod performance. Collectively, our findings indicate an antiinflammatory and protective effect of testosterone in the degenerating spinal cord. These results coincided with a high concentration of androgen-reduced derivatives after testosterone treatment suggesting that the steroid profile may have a beneficial role on disease progression.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Motor Neurons/drug effects , Neuroprotective Agents/therapeutic use , Testosterone/therapeutic use , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Aromatase/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Disease Models, Animal , Estrogen Receptor alpha/metabolism , Male , Mice , Motor Neurons/metabolism , Motor Neurons/pathology , Neuroprotective Agents/pharmacology , Receptors, Androgen/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Testosterone/metabolism , Testosterone/pharmacology , Treatment Outcome
7.
Int J Mol Sci ; 21(6)2020 Mar 20.
Article in English | MEDLINE | ID: mdl-32244957

ABSTRACT

Glucocorticoids are crucial for stress-coping, resilience, and adaptation. However, if the stress hormones become dysregulated, the vulnerability to stress-related diseases is enhanced. In this brief review, we discuss the role of glucocorticoids in the pathogenesis of neurodegenerative disorders in both human and animal models, and focus in particular on amyotrophic lateral sclerosis (ALS). For this purpose, we used the Wobbler animal model, which mimics much of the pathology of ALS including a dysfunctional hypothalamic-pituitary-adrenal axis. We discuss recent studies that demonstrated that the pathological cascade characteristic for motoneuron degeneration of ALS is mimicked in the genetically selected Wobbler mouse and can be attenuated by treatment with the selective glucocorticoid receptor antagonist (GRA) CORT113176. In long-term treatment (3 weeks) GRA attenuated progression of the behavioral, inflammatory, excitatory, and cell-death-signaling pathways while increasing the survival signal of serine-threonine kinase (pAkt). The action mechanism of the GRA may be either by interfering with GR deactivation or by restoring the balance between pro- and anti-inflammatory signaling pathways driven by the complementary mineralocorticoid receptor (MR)- and GR-mediated actions of corticosterone. Accordingly, GR antagonism may have clinical relevance for the treatment of neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases/drug therapy , Receptors, Glucocorticoid/metabolism , Animals , Corticosterone/blood , Corticosterone/chemistry , Disease Models, Animal , Humans , Inflammation/blood , Inflammation/complications , Models, Biological , Neurodegenerative Diseases/blood , Receptors, Glucocorticoid/antagonists & inhibitors
8.
J Steroid Biochem Mol Biol ; 192: 105385, 2019 09.
Article in English | MEDLINE | ID: mdl-31150830

ABSTRACT

The Wobbler mouse has been proposed as an experimental model of the sporadic form of amyotrophic lateral sclerosis (ALS). The administration of natural progesterone (PROG) to Wobbler mice attenuates neuropathology, inhibits oxidative stress, enhances the expression of genes involved in motoneuron function, increases survival and restores axonal transport. However, current pharmacological treatments for ALS patients are still partially effective. This encouraged us to investigate if the synthetic progestin norethindrone (NOR), showing higher potency than PROG and used for birth control and hormone therapy might also afford neuroprotection. Two-month-old Wobbler mice (wr/wr) were left untreated or received either a 20 mg pellet of PROG or a 1 mg pellet of NOR for 18 days. Untreated control NFR/NFR mice (background strain for Wobbler) were also employed. Wobblers showed typical clinical and spinal cord abnormalities, while these abnormalities were normalized with PROG treatment. Surprisingly, we found that NOR did not increase immunoreactivity and gene expression for choline-acetyltransferase, drastically decreased GFAP + astrogliosis, favored proinflammatory mediators, promoted the inflammatory phenotype of IBA1+ microglia, increased the receptor for advanced glycation end products (RAGE) mRNA and protein expression and the activity of nitric oxide synthase (NOS)/NADPH diaphorase in the cervical spinal cord. Additionally, NOR treatment produced atrophy of the thymus. The combined negative effects of NOR on clinical assessments (forelimb atrophy and rotarod performance) suggest a detrimental effect on muscle trophism and motor function. These findings reinforce the evidence that the type of progestin used for contraception, endometriosis or replacement therapy, may condition the outcome of preclinical and clinical studies targeting neurodegenerative diseases.


Subject(s)
Disease Models, Animal , Motor Neurons/drug effects , Neurodegenerative Diseases/drug therapy , Neuroprotection/drug effects , Norethindrone/pharmacology , Progesterone/pharmacology , Progestins/pharmacology , Animals , Contraceptives, Oral, Synthetic/pharmacology , Mice , Motor Neurons/pathology
9.
Cell Mol Neurobiol ; 39(4): 471-472, 2019 May.
Article in English | MEDLINE | ID: mdl-30941611

ABSTRACT

Steroids are complex molecules, exerting known and still unknown effects in the nervous system. Throughout this volume, the reader will find a wide spectrum of articles, giving an up-to-date account of the molecular, physiological, pharmacological, and clinical aspects of steroid action on the nervous system.


Subject(s)
Nervous System/drug effects , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Steroids/pharmacology , Animals , Humans , Mice , Neuroprotection/drug effects
10.
Neuroscience ; 384: 384-396, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29890290

ABSTRACT

Wobbler mice are experimental models for amyotrophic lateral sclerosis. As such they show motoneuron degeneration, motor deficits, and astrogliosis and microgliosis of the spinal cord. Additionally, Wobbler mice show increased plasma, spinal cord and brain corticosterone levels and focal adrenocortical hyperplasia, suggesting a pathogenic role for glucocorticoids in this disorder. Considering this endocrine background, we examined whether the glucocorticoid receptor (GR) modulator CORT 113176 prevents spinal cord neuropathology of Wobblers. CORT 113176 shows high affinity for the GR, with low or null affinity for other steroid receptors. We employed five-month-old genotyped Wobbler mice that received s.c. vehicle or 30 mg/kg/day for 4 days of CORT 113176 dissolved in sesame oil. The mice were used on the 4th day, 2 h after the last dose of CORT 113176. Vehicle-treated Wobbler mice presented vacuolated motoneurons, increased glial fibrillary acidic protein (GFAP)+ astrocytes and decreased glutamine synthase (GS)+ cells. There was strong neuroinflammation, shown by increased staining for IBA1+ microglia and CD11b mRNA, enhanced expression of tumor necrosis factor-α, its cognate receptor TNFR1, toll-like receptor 4, the inducible nitric oxide synthase, NFkB and the high-mobility group box 1 protein (HMGB1). Treatment of Wobbler mice with CORT 113176 reversed the abnormalities of motoneurons and down-regulated proinflammatory mediators and glial reactivity. Expression of glutamate transporters GLT1 and GLAST mRNAs and GLT1 protein was significantly enhanced over untreated Wobblers. In summary, antagonism of GR with CORT 113176 prevented neuropathology and showed anti-inflammatory and anti-glutamatergic effects in the spinal cord of Wobbler mice.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Inflammation/drug therapy , Isoquinolines/therapeutic use , Nerve Degeneration/drug therapy , Neuroprotective Agents/therapeutic use , Pyrazoles/therapeutic use , Receptors, Glucocorticoid/antagonists & inhibitors , Spinal Cord/drug effects , Amyotrophic Lateral Sclerosis/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Inflammation/metabolism , Inflammation/pathology , Isoquinolines/pharmacology , Mice , Mice, Neurologic Mutants , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neuroprotective Agents/pharmacology , Pyrazoles/pharmacology , Spinal Cord/metabolism , Spinal Cord/pathology , Treatment Outcome
11.
J Steroid Biochem Mol Biol ; 174: 201-216, 2017 11.
Article in English | MEDLINE | ID: mdl-28951257

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating disorder characterized by progressive death of motoneurons. The Wobbler (WR) mouse is a preclinical model sharing neuropathological similarities with human ALS. We have shown that progesterone (PROG) prevents the progression of motoneuron degeneration. We now studied if allopregnanolone (ALLO), a reduced metabolite of PROG endowed with gabaergic activity, also prevents WR neuropathology. Sixty-day old WRs remained untreated or received two steroid treatment regimens in order to evaluate the response of several parameters during early or prolonged steroid administration. ALLO was administered s.c. daily for 5days (4mg/kg) or every other day for 32days (3, 3mg/kg), while another group of WRs received a 20mg PROG pellet s.c. for 18 or 60days. ALLO administration to WRs increased ALLO serum levels without changing PROG and 5 alpha dihydroprogesterone (5α-DHP), whereas PROG treatment increased PROG, 5α-DHP and ALLO. Untreated WRs showed higher basal levels of serum 5α-DHP than controls. In the cervical spinal cord we studied markers of oxidative stress or associated to trophic responses. These included nitric oxide synthase (NOS) activity, motoneuron vacuolation, MnSOD immunoreactivity (IR), brain derived neurotrophic factor (BDNF) and TrkB mRNAs, p75 neurotrophin receptor (p75NTR) and, cell survival or death signals such as pAKT and the stress activated kinase JNK. Untreated WRs showed a reduction of MnSOD-IR and BDNF/TrkB mRNAs, associated to high p75NTR in motoneurons, neuronal and glial NOS hyperactivity and neuronal vacuolation. Also, low pAKT, mainly in young WRs, and a high pJNK in the old stage characterized WRs spinal cord. Except for MnSOD and BDNF, these alterations were prevented by an acute ALLO treatment, while short-term PROG elevated MnSOD. Moreover, after chronic administration both steroids enhanced MnSOD-IR and BDNF mRNA, while attenuated pJNK and NOS in glial cells. Long-term PROG also increased pAKT and reduced neuronal NOS, parameters not modulated by chronic ALLO. Clinically, both steroids improved muscle performance. Thus, ALLO was able to reduce neuropathology in this model. Since high oxidative stress activates p75NTR and pJNK in neurodegeneration, steroid reduction of these molecules may provide adequate neuroprotection. These data yield the first evidence that ALLO, a gabaergic neuroactive steroid, brings neuroprotection in a model of motoneuron degeneration.


Subject(s)
Nerve Degeneration/drug therapy , Neuroprotective Agents/therapeutic use , Pregnanolone/therapeutic use , Amyotrophic Lateral Sclerosis , Animals , Brain-Derived Neurotrophic Factor/genetics , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Female , Male , Mice , Motor Neurons/drug effects , Motor Neurons/pathology , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Neuroprotective Agents/blood , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase/metabolism , Pregnanolone/blood , Pregnanolone/pharmacology , Progesterone/blood , Progesterone/pharmacology , Progesterone/therapeutic use , Receptor, trkB/genetics , Receptors, Nerve Growth Factor/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase/metabolism
12.
Horm Metab Res ; 49(1): 16-22, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27813048

ABSTRACT

Despite several scientific and technological advances, there is no single neuroprotective treatment that can reverse the brain damage after acute ischemic stroke (AIS). Neuroactive steroids are cholesterol-derived hormones that have the ability to modulate the normal and pathologic nervous system employing genomic and nongenomic mechanisms. In this work, we first investigated if AIS affects the plasma concentration of 5 neuroactive steroids (cortisol, estradiol, progesterone, testosterone, and 3α-androstenediol glucuronide). Second, we studied if levels of circulating steroids associate with neurological, cognitive, and functional outcome in a cohort of 60- to 90 year-old male and female patients with AIS. For this purpose, we recruited patients who were hospitalized at the Emergency Room of the Central Military Hospital within the first 24 h after stroke onset. We designed 2 experimental groups, each one composed of 30 control subjects and 30 AIS patients, both males and females. The assessment of neurological deficit was performed with the NIHSS and the tests used for the functional and cognitive status were: (1) modified Rankin Scale; (2) Photo test, and (3) abbreviated Pfeiffer's mental status questionnaire. We observed a significant difference in plasma concentration of cortisol and estradiol between both experimental groups. In the AIS group, higher levels of these neuroactive steroids were associated with more pronounced neurological, cognitive and functional deficits in women compared to men. We propose that in elderly patients, high levels of circulating neuroactive steroids like cortisol and estradiol could potentiate AIS-mediated neuropathology in the ischemic and penumbra areas.


Subject(s)
Androstenediols/blood , Brain Ischemia/blood , Cognition/physiology , Gonadal Steroid Hormones/blood , Hydrocortisone/blood , Stroke , Aged , Aged, 80 and over , Brain Ischemia/diagnosis , Brain Ischemia/physiopathology , Brain Ischemia/psychology , Case-Control Studies , Female , Humans , Male , Middle Aged , Neurotransmitter Agents/blood , Prognosis , Recovery of Function , Stroke/blood , Stroke/diagnosis , Stroke/physiopathology , Stroke/psychology
13.
J Steroid Biochem Mol Biol ; 165(Pt B): 421-429, 2017 01.
Article in English | MEDLINE | ID: mdl-27597394

ABSTRACT

Previous studies of experimental autoimmune encephalomyelitis (EAE) have shown that progesterone decreases inflammatory cell infiltration and proinflammatory factors, increases myelination and attenuates clinical grade of EAE mice. To elucidate potential mediators of these effects, we analyzed the mRNA expression of neurosteroidogenic enzymes in the spinal cord, in view of the protective role of steroids in EAE. We also analyzed mitochondrial morphology and dynamics (fusion and fission proteins), considering the role of mitochondria in neurosteroidogenesis. EAE was induced in C57Bl6 mice using MOG40-54 and killed on day 16 after induction. Using qPCR, we found in steroid-untreated EAE mice decreased mRNAs for the steroidogenic acute regulatory protein (Star), voltage-dependent anion channel (VDAC), P450scc (cholesterol side-chain cleavage), 5α-reductase, 3α-hydroxysteroid dehydrogenase (3α-HSD) and aromatase, whereas levels of 3ß-hydroxysteroid dehydrogenase (3ß-HSD) showed a large intra-group variance. We also found increased mRNA expression of 18Kd translocator protein (TSPO), which likely resulted from the reactive microgliosis in this model. EAE mice also showed pathological mitochondrial morphology and reduced expression of fission and fusion protein mRNAs. Most importantly, pretreatment with progesterone a week before EAE induction increased Star,VDAC, P450scc, 5α-reductase type I, 3α-HSD and aromatase mRNAs and did not modify 3ß-HSD. TSPO mRNA was decreased, consequent with the inhibition of microgliosis. Mitochondrial morphology was improved and fission/fusion protein mRNAs were enhanced by progesterone treatment. Furthermore, progesterone protective effects on mitochondrial and endoplasmic reticulum may allow the recovery of neurosteroidogenesis. In this way, endogenously synthesized neurosteroids may reinforce the beneficial effects of exogenous progesterone previously shown in MS mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Multiple Sclerosis/metabolism , Neurotransmitter Agents/metabolism , Progesterone/metabolism , RNA, Messenger/metabolism , 17-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Cholestenone 5 alpha-Reductase/metabolism , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Mice , Mice, Inbred C57BL , Microscopy, Electron , Mitochondria/metabolism , Multiple Sclerosis/drug therapy , Phosphoproteins/metabolism , Spinal Cord/metabolism
14.
J Neuroimmunol ; 276(1-2): 89-97, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25200475

ABSTRACT

Progesterone plays a protective role in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS). Besides spinal cord neuropathology, MS patients present a dysfunctional hippocampus. In this work we studied the therapeutic effects of the progestin Nestorone in the brain of mice with chronic EAE. Nestorone decreased clinical grade and enhanced motor behavior. In addition, it increased cell proliferation and doublecortin positive neuroblasts in the hippocampus, increased GABAergic interneurons and attenuated the number of Iba1+ microglia/macrophages, events possibly linked to enhancement of neurogenesis. Therefore, Nestorone protected against hippocampus abnormalities and improved functional outcomes of EAE mice, suggesting its potential value for MS.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Norprogesterones/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Calcium-Binding Proteins/metabolism , Chronic Disease , Doublecortin Domain Proteins , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Freund's Adjuvant/toxicity , Glial Fibrillary Acidic Protein/metabolism , Ki-67 Antigen/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Microtubule-Associated Proteins/metabolism , Motor Activity/drug effects , Myelin Basic Protein/metabolism , Myelin-Oligodendrocyte Glycoprotein/toxicity , Neuropeptides/metabolism , Peptide Fragments/toxicity , Phosphopyruvate Hydratase/metabolism
15.
J Steroid Biochem Mol Biol ; 143: 40-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24565565

ABSTRACT

Mutant Wobbler mice are models for human amyotrophic lateral sclerosis (ALS). In addition to spinal cord degeneration, Wobbler mice show high levels of blood corticosterone, hyperactivity of the hypothalamic-pituitary-adrenal axis and abnormalities of the hippocampus. Hypersecretion of glucocorticoids increase hippocampus vulnerability, a process linked to an enriched content of glucocorticoid receptors (GR). Hence, we studied if a selective GR antagonist (CORT108297) with null affinity for other steroid receptors restored faulty hippocampus parameters of Wobbler mice. Three months old genotyped Wobbler mice received s.c. vehicle or CORT108297 during 4 days. We compared the response of doublecortin (DCX)+ neuroblasts in the subgranular layer of the dentate gyrus (DG), NeuN+ cells in the hilus of the DG, glial fibrillary acidic protein (GFAP)+ astrocytes and the phenotype of Iba1+ microglia in CORT108297-treated and vehicle-treated Wobblers. The number of DCX+ cells in Wobblers was lower than in control mice, whereas CORT108297 restored this parameter. After CORT108297 treatment, Wobblers showed diminished astrogliosis, and changed the phenotype of Iba1+ microglia from an activated to a quiescent form. These changes occurred without alterations in the hypercorticosteronemia or the number of NeuN+ cells of the Wobblers. In a separate experiment employing control NFR/NFR mice, treatment with corticosterone for 5 days reduced DCX+ neuroblasts and induced astrocyte hypertrophy, whereas treatment with CORT108297 antagonized these effects. Normalization of neuronal progenitors, astrogliosis and microglial phenotype by CORT108297 indicates the usefulness of this antagonist to normalize hippocampus parameters of Wobbler mice. Thus, CORT108297 opens new therapeutic options for the brain abnormalities of ALS patients and hyperadrenocorticisms.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Aza Compounds/pharmacology , Corticosterone/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Hippocampus/drug effects , Microtubule-Associated Proteins/physiology , Neuropeptides/physiology , Receptors, Glucocorticoid/antagonists & inhibitors , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Cells, Cultured , DNA-Binding Proteins , Disease Models, Animal , Doublecortin Domain Proteins , Doublecortin Protein , Female , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/abnormalities , Hippocampus/metabolism , Humans , Immunoenzyme Techniques , Mice , Mice, Neurologic Mutants , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nuclear Proteins/metabolism , Receptors, Glucocorticoid/metabolism
16.
J Neurochem ; 122(1): 185-95, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22486171

ABSTRACT

In the Wobbler mouse, a mutation of the Vps54 protein increases oxidative stress in spinal motoneurons, associated to toxic levels of nitric oxide and hyperactivity of nitric oxide synthase (NOS). Progesterone neuroprotection has been reported for several CNS diseases, including the Wobbler mouse neurodegeneration. In the present study, we analyzed progesterone effects on mitochondrial-associated parameters of symptomatic Wobbler mice. The activities of mitochondrial respiratory chain complexes I, II-III and IV and protein levels of mitochondrial and cytosolic NOS were determined in cervical and lumbar cords from control, Wobbler and Wobbler mice receiving a progesterone implant for 18 days. We found a significant reduction of complex I and II-III activities in mitochondria and increased protein levels of mitochondrial, but not cytosolic nNOS, in the cervical cord of Wobbler mice. Progesterone treatment prevented the reduction of complex I in the cervical region and the increased level of mitochondrial nNOS. Wobbler motoneurons also showed accumulation of amyloid precursor protein immunoreactivity and decreased activity and immunostaining of MnSOD. Progesterone treatment avoided these abnormalities. Therefore, administration of progesterone to clinically afflicted Wobblers (i) prevented the abnormal increase of mitochondrial nNOS and normalized respiratory complex I; (ii) decreased amyloid precursor protein accumulation, a sign of axonal degeneration, and (iii) increased superoxide dismutation. Thus, progesterone neuroprotection decreases mitochondriopathy of Wobbler mouse cervical spinal cord.


Subject(s)
Mitochondrial Diseases/pathology , Mitochondrial Diseases/prevention & control , Multienzyme Complexes/metabolism , Neuroprotective Agents/therapeutic use , Progesterone/therapeutic use , Spinal Cord/drug effects , Albinism, Oculocutaneous/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Drosophila Proteins , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Mice , Mice, Mutant Strains , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Diseases/genetics , Motor Neurons/drug effects , Motor Neurons/pathology , NADPH Dehydrogenase/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins , Spinal Cord/pathology , Spinal Cord/ultrastructure , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Superoxide Dismutase/metabolism , Vesicular Transport Proteins/genetics
17.
Neuroimmunomodulation ; 17(3): 146-9, 2010.
Article in English | MEDLINE | ID: mdl-20134188

ABSTRACT

In recent years, a growing list of publications point to the value of steroid hormones as an interesting option for the treatment of several type of lesions and diseases of the nervous system. Progesterone, well known for its role in pregnancy, has recently been shown to exert neuroprotective and promyelinating effects in both, the peripheral and central nervous system, including the injured spinal cord. Previous work from our laboratory has shown that progesterone actions in experimental models of spinal neurodegeneration or injury may involve the modulation of brain-derived neurotrophic factor, a neurotrophin with important implications in neuronal survival and axonal regeneration. The spinal cord is target for progesterone since neurons and glial cells express the intracellular receptors for this neuroactive steroid. However, the presence in the spinal cord of new membrane receptors and the enzymes involved in progesterone metabolism to its reduced derivatives, which modulate the activity of neurotransmitter receptors, suggest that progesterone actions involve pleiotropic mechanisms. Our recent data uncovering several molecular events may help to understand the protective and promyelinating actions of progesterone and further support the role of this steroid as a promising therapeutic agent for neurotrauma and/or neurodegenerative diseases.


Subject(s)
Neuroprotective Agents/pharmacology , Progesterone/pharmacology , Spinal Cord Injuries/drug therapy , Spinal Cord/drug effects , Animals , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Humans , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Neuroprotective Agents/metabolism , Neuroprotective Agents/therapeutic use , Progesterone/metabolism , Progesterone/therapeutic use , Spinal Cord/metabolism , Spinal Cord/physiopathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology
18.
Cell Mol Neurobiol ; 30(1): 123-35, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19693665

ABSTRACT

In the Wobbler mouse, a mutation in the Vps54 gene is accompanied by motoneuron degeneration and astrogliosis in the cervical spinal cord. Previous work has shown that these abnormalities are greatly attenuated by progesterone treatment of clinically afflicted Wobblers. However, whether progesterone is effective at all disease stages has not yet been tested. The present work used genotyped (wr/wr) Wobbler mice at three periods of the disease: early progressive (1-2 months), established (5-8 months) or late stages (12 months) and age-matched wildtype controls (NFR/NFR), half of which were implanted with a progesterone pellet (20 mg) for 18 days. In untreated Wobblers, degenerating vacuolated motoneurons were initially abundant, experienced a slight reduction at the established stage and dramatically diminished during the late period. In motoneurons, the cholinergic marker choline acetyltransferase (ChAT) was reduced at all stages of the Wobbler disease, whereas hyperexpression of the growth-associated protein (GAP43) mRNA preferentially occurred at the early progressive and established stages. Progesterone therapy significantly reduced motoneuron vacuolation, enhanced ChAT immunoreactive perikarya and reduced the hyperexpression of GAP43 during the early progressive and established stages. At all stage periods, untreated Wobblers showed high density of glial fibrillary acidic protein (GFAP)+ astrocytes and decreased number of glutamine synthase (GS) immunostained cells. Progesterone treatment down-regulated GFAP+ astrocytes and up-regulated GS+ cell number. These data reinforced the usefulness of progesterone to improve motoneuron and glial cell abnormalities of Wobbler mice and further showed that therapeutic benefit seems more effective at the early progressive and established periods, rather than on advance stages of spinal cord neurodegeneration.


Subject(s)
Motor Neurons/drug effects , Motor Neurons/pathology , Neuroglia/drug effects , Neuroglia/pathology , Progesterone/pharmacology , Spinal Cord Diseases/pathology , Spinal Cord/pathology , Animals , Anterior Horn Cells/drug effects , Anterior Horn Cells/enzymology , Anterior Horn Cells/pathology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Cell Count , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Female , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , Gene Expression Regulation/drug effects , Genotype , Glial Fibrillary Acidic Protein/metabolism , Glutamate-Ammonia Ligase/metabolism , Image Processing, Computer-Assisted , Male , Mice , Mice, Neurologic Mutants , Motor Neurons/enzymology , Neuroglia/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord Diseases/enzymology
19.
Brain Res ; 1283: 177-85, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19497309

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE), an induced model of Multiple Sclerosis presents spinal cord demyelination, axonal pathology and neuronal dysfunction. Previous work has shown that progesterone attenuated the clinical severity, demyelination and neuronal dysfunction of EAE mice (Garay et al., J. Steroid Biochem. Mol. Biol., 2008). Here we studied if progesterone also prevented axonal damage, a main cause of neurological disability. To this end, some axonal parameters were compared in EAE mice pretreated with progesterone a week before immunization with MOG(40-54) and in a group of steroid-free EAE mice. On day 16th after EAE induction, we determined in both groups and in control mice: a) axonal density in semithin sections of the spinal cord ventral funiculus; b) appearance of amyloid precursor protein (APP) immunopositive spheroids as an index of damaged axons; c) levels of the growth associated protein GAP43 mRNA and immunopositive cell bodies, as an index of aberrant axonal sprouting. Steroid-naive EAE mice showed decreased axonal density, shrunken axons, abundance of irregular vesicular structures, degenerating APP+ axons, increased expression of GAP43 mRNA and immunoreactive protein in motoneurons. Instead, EAE mice receiving progesterone treatment showed increased axonal counts, high proportion of small diameter axons, reduced APP+ profiles, and decreased GAP43 expression. In conclusion, progesterone enhanced axonal density, decreased axonal damage and prevented GAP43 hyperexpression in the spinal cord of EAE mice. Thus, progesterone also exerts protective effects on the axonal pathology developing in EAE mice.


Subject(s)
Axons/drug effects , Brain/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Multiple Sclerosis/drug therapy , Neuroprotective Agents/pharmacology , Progesterone/pharmacology , Amyloid beta-Protein Precursor/drug effects , Amyloid beta-Protein Precursor/metabolism , Animals , Axons/metabolism , Axons/pathology , Biomarkers/analysis , Biomarkers/metabolism , Brain/metabolism , Brain/pathology , Cell Count , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , GAP-43 Protein/genetics , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Neuroprotective Agents/metabolism , Progesterone/metabolism , Progestins/metabolism , Progestins/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Treatment Outcome , Wallerian Degeneration/drug therapy , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology
20.
Front Neuroendocrinol ; 30(2): 173-87, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19318112

ABSTRACT

Studies on the neuroprotective and promyelinating effects of progesterone in the nervous system are of great interest due to their potential clinical connotations. In peripheral neuropathies, progesterone and reduced derivatives promote remyelination, axonal regeneration and the recovery of function. In traumatic brain injury (TBI), progesterone has the ability to reduce edema and inflammatory cytokines, prevent neuronal loss and improve functional outcomes. Clinical trials have shown that short-and long-term progesterone treatment induces a significant improvement in the level of disability among patients with brain injury. In experimental spinal cord injury (SCI), molecular markers of functional motoneurons become impaired, including brain-derived neurotrophic factor (BDNF) mRNA, Na,K-ATPase mRNA, microtubule-associated protein 2 and choline acetyltransferase (ChAT). SCI also produces motoneuron chromatolysis. Progesterone treatment restores the expression of these molecules while chromatolysis subsided. SCI also causes oligodendrocyte loss and demyelination. In this case, a short progesterone treatment enhances proliferation and differentiation of oligodendrocyte progenitors into mature myelin-producing cells, whereas prolonged treatment increases a transcription factor (Olig1) needed to repair injury-induced demyelination. Progesterone neuroprotection has also been shown in motoneuron neurodegeneration. In Wobbler mice spinal cord, progesterone reverses the impaired expression of BDNF, ChAT and Na,K-ATPase, prevents vacuolar motoneuron degeneration and the development of mitochondrial abnormalities, while functionally increases muscle strength and the survival of Wobbler mice. Multiple mechanisms contribute to these progesterone effects, and the role played by classical nuclear receptors, extra nuclear receptors, membrane receptors, and the reduced metabolites of progesterone in neuroprotection and myelin formation remain an exciting field worth of exploration.


Subject(s)
Motor Neurons/drug effects , Motor Neurons/pathology , Nerve Degeneration , Neuroprotective Agents , Trauma, Nervous System , Animals , Brain Injuries/drug therapy , Brain Injuries/pathology , Brain Injuries/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Clinical Trials as Topic , Disease Models, Animal , Humans , Mice , Mice, Neurologic Mutants , Motor Neurons/cytology , Motor Neurons/metabolism , Myelin Sheath/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Receptors, Progesterone/metabolism , Sex Factors , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Trauma, Nervous System/drug therapy , Trauma, Nervous System/pathology , Trauma, Nervous System/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL