89 results on '"Igor Jakovcevski"'
Search Results
2. The Neuroprotective Effect of Neural Cell Adhesion Molecule L1 in the Hippocampus of Aged Alzheimer’s Disease Model Mice
- Author
-
Miljana Aksic, Igor Jakovcevski, Mohammad I. K. Hamad, Vladimir Jakovljevic, Sanja Stankovic, and Maja Vulovic
- Subjects
adhesion molecule L1 ,Alzheimer’s disease ,APP/PS1 mice ,GABAergic interneurons ,hippocampus ,synapses ,Biology (General) ,QH301-705.5 - Abstract
Alzheimer’s disease (AD) is a severe neurodegenerative disorder and the most common form of dementia, causing the loss of cognitive function. Our previous study has shown, using a doubly mutated mouse model of AD (APP/PS1), that the neural adhesion molecule L1 directly binds amyloid peptides and decreases plaque load and gliosis when injected as an adeno-associated virus construct (AAV-L1) into APP/PS1 mice. In this study, we microinjected AAV-L1, using a Hamilton syringe, directly into the 3-month-old APP/PS1 mouse hippocampus and waited for a year until significant neurodegeneration developed. We stereologically counted the principal neurons and parvalbumin-positive interneurons in the hippocampus, estimated the density of inhibitory synapses around principal cells, and compared the AAV-L1 injection models with control injections of green fluorescent protein (AAV-GFP) and the wild-type hippocampus. Our results show that there is a significant loss of granule cells in the dentate gyrus of the APP/PS1 mice, which was improved by AAV-L1 injection, compared with the AAV-GFP controls (p < 0.05). There is also a generalized loss of parvalbumin-positive interneurons in the hippocampus of APP/PS1 mice, which is ameliorated by AAV-L1 injection, compared with the AAV-GFP controls (p < 0.05). Additionally, AAV-L1 injection promotes the survival of inhibitory synapses around the principal cells compared with AAV-GFP controls in all three hippocampal subfields (p < 0.01). Our results indicate that L1 promotes neuronal survival and protects the synapses in an AD mouse model, which could have therapeutic implications.
- Published
- 2024
- Full Text
- View/download PDF
3. Mice Mutated in the First Fibronectin Domain of Adhesion Molecule L1 Show Brain Malformations and Behavioral Abnormalities
- Author
-
Viviana Granato, Ludovica Congiu, Igor Jakovcevski, Ralf Kleene, Benjamin Schwindenhammer, Luciana Fernandes, Sandra Freitag, Melitta Schachner, and Gabriele Loers
- Subjects
L1CAM ,mutation ,locomotion ,open field ,social interactions ,circadian rhythm ,Microbiology ,QR1-502 - Abstract
The X-chromosome-linked cell adhesion molecule L1 (L1CAM), a glycoprotein mainly expressed by neurons in the central and peripheral nervous systems, has been implicated in many neural processes, including neuronal migration and survival, neuritogenesis, synapse formation, synaptic plasticity and regeneration. L1 consists of extracellular, transmembrane and cytoplasmic domains. Proteolytic cleavage of L1’s extracellular and transmembrane domains by different proteases generates several L1 fragments with different functions. We found that myelin basic protein (MBP) cleaves L1’s extracellular domain, leading to enhanced neuritogenesis and neuronal survival in vitro. To investigate in vivo the importance of the MBP-generated 70 kDa fragment (L1-70), we generated mice with an arginine to alanine substitution at position 687 (L1/687), thereby disrupting L1’s MBP cleavage site and obliterating L1-70. Young adult L1/687 males showed normal anxiety and circadian rhythm activities but enhanced locomotion, while females showed altered social interactions. Older L1/687 males were impaired in motor coordination. Furthermore, L1/687 male and female mice had a larger hippocampus, with more neurons in the dentate gyrus and more proliferating cells in the subgranular layer, while the thickness of the corpus callosum and the size of lateral ventricles were normal. In summary, subtle mutant morphological changes result in subtle behavioral changes.
- Published
- 2024
- Full Text
- View/download PDF
4. The Role of Tenascin-C on the Structural Plasticity of Perineuronal Nets and Synaptic Expression in the Hippocampus of Male Mice
- Author
-
Ana Jakovljević, Vera Stamenković, Joko Poleksić, Mohammad I. K. Hamad, Gebhard Reiss, Igor Jakovcevski, and Pavle R. Andjus
- Subjects
tenascin-C ,perineuronal nets ,enriched environment ,hippocampus ,synaptic plasticity ,Microbiology ,QR1-502 - Abstract
Neuronal plasticity is a crucial mechanism for an adapting nervous system to change. It is shown to be regulated by perineuronal nets (PNNs), the condensed forms of the extracellular matrix (ECM) around neuronal bodies. By assessing the changes in the number, intensity, and structure of PNNs, the ultrastructure of the PNN mesh, and the expression of inhibitory and excitatory synaptic inputs on these neurons, we aimed to clarify the role of an ECM glycoprotein, tenascin-C (TnC), in the dorsal hippocampus. To enhance neuronal plasticity, TnC-deficient (TnC-/-) and wild-type (TnC+/+) young adult male mice were reared in an enriched environment (EE) for 8 weeks. Deletion of TnC in TnC-/- mice showed an ultrastructural reduction of the PNN mesh and an increased inhibitory input in the dentate gyrus (DG), and an increase in the number of PNNs with a rise in the inhibitory input in the CA2 region. EE induced an increased inhibitory input in the CA2, CA3, and DG regions; in DG, the change was also followed by an increased intensity of PNNs. No changes in PNNs or synaptic expression were found in the CA1 region. We conclude that the DG and CA2 regions emerged as focal points of alterations in PNNs and synaptogenesis with EE as mediated by TnC.
- Published
- 2024
- Full Text
- View/download PDF
5. The long-term effects of maternal deprivation on the number and size of inhibitory interneurons in the rat amygdala and nucleus accumbens
- Author
-
Dubravka Aleksic, Joko Poleksic, Gorana Agatonovic, Vuk Djulejic, Maja Vulovic, Miljana Aksic, Gebhard Reiss, Mohammad I. K. Hamad, Igor Jakovcevski, and Milan Aksic
- Subjects
amygdala ,GABAergic interneurons ,nucleus accumbens ,maternal deprivation ,parvalbumin ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
IntroductionThere is an increasing evidence supporting the hypothesis that traumatic experiences during early developmental periods might be associated with psychopathology later in life. Maternal deprivation (MD) in rodents has been proposed as an animal model for certain aspects of neuropsychiatric disorders.MethodsTo determine whether early-life stress leads to changes in GABAergic, inhibitory interneurons in the limbic system structures, specifically the amygdala and nucleus accumbens, 9-day-old Wistar rats were exposed to a 24 h MD. On postnatal day 60 (P60), the rats were sacrificed for morphometric analysis and their brains were compared to the control group.ResultsResults show that MD affect GABAergic interneurons, leading to the decrease in density and size of the calcium-binding proteins parvalbumin-, calbindin-, and calretinin-expressing interneurons in the amygdala and nucleus accumbens.DiscussionThis study indicates that early stress in life leads to changes in the number and morphology of the GABAergic, inhibitory interneurons in the amygdala and nucleus accumbens, most probably due to the loss of neurons during postnatal development and it further contributes to understanding the effects of maternal deprivation on brain development.
- Published
- 2023
- Full Text
- View/download PDF
6. Mice lacking perforin have improved regeneration of the injured femoral nerve
- Author
-
Igor Jakovcevski, Monika von Düring, David Lutz, Maja Vulović, Mohammad Hamad, Gebhard Reiss, Eckart Förster, and Melitta Schachner
- Subjects
blood-nerve barrier ,femoral nerve injury ,locomotor recovery ,lymphocytes ,myelination ,nk-cells ,perforin ,reinnervation ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
The role that the immune system plays after injury of the peripheral nervous system is still not completely understood. Perforin, a natural killer cell- and T-lymphocyte-derived enzyme that mediates cytotoxicity, plays important roles in autoimmune diseases, infections and central nervous system trauma, such as spinal cord injury. To dissect the roles of this single component of the immune response to injury, we tested regeneration after femoral nerve injury in perforin-deficient (Pfp–/–) and wild-type control mice. Single frame motion analysis showed better motor recovery in Pfp–/– mice compared with control mice at 4 and 8 weeks after injury. Retrograde tracing of the motoneuron axons regrown into the motor nerve branch demonstrated more correctly projecting motoneurons in the spinal cord of Pfp–/– mice compared with wild-types. Myelination of regrown axons measured by g-ratio was more extensive in Pfp–/– than in wild-type mice in the motor branch of the femoral nerve. Pfp–/– mice displayed more cholinergic synaptic terminals around cell bodies of spinal motoneurons after injury than the injured wild-types. We histologically analyzed lymphocyte infiltration 10 days after surgery and found that in Pfp–/– mice the number of lymphocytes in the regenerating nerves was lower than in wild-types, suggesting a closed blood-nerve barrier in Pfp–/– mice. We conclude that perforin restricts motor recovery after femoral nerve injury owing to decreased survival of motoneurons and reduced myelination.
- Published
- 2022
- Full Text
- View/download PDF
7. Editorial: Extracellular matrix in development and disorders of the nervous system
- Author
-
Igor Jakovcevski, Pavle R. Andjus, and Eckart Förster
- Subjects
extracellular matrix (ECM) ,nervous system ,regeneration ,perineuronal nets (PNNs) ,synapses ,Biology (General) ,QH301-705.5 - Published
- 2023
- Full Text
- View/download PDF
8. The impact of early life maternal deprivation on the perineuronal nets in the prefrontal cortex and hippocampus of young adult rats
- Author
-
Ana Jakovljevic, Gorana Agatonovic, Dubravka Aleksic, Milan Aksic, Gebhard Reiss, Eckart Förster, Antonios Stamatakis, Igor Jakovcevski, and Joko Poleksic
- Subjects
perineuronal nets ,interneurons ,maternal deprivation ,early life stress ,prefrontal cortex ,hippocampus ,Biology (General) ,QH301-705.5 - Abstract
Early life stress negatively impacts brain development and affects structure and function of parvalbumin immunopositive (PV+) inhibitory neurons. Main regulators of PV+ interneurons activity and plasticity are perineuronal nets (PNNs), an extracellular matrix formation that enwraps PV+ interneurons mainly in the neocortex and hippocampus. To experimentally address the impact of early life stress on the PNNs and PV+ interneurons in the medial prefrontal cortex and dorsal hippocampus in rats, we employed a 24 h maternal deprivation protocol. We show that maternal deprivation in the medial prefrontal cortex of adult rats caused a decrease in density of overall PNNs and PNNs that enwrap PV+ interneurons in the rostral cingulate cortex. Furthermore, a staining intensity decrease of overall PNNs and PNN+/PV+ cells was found in the prelimbic cortex. Finally, a decrease in both intensity and density of overall PNNs and PNNs surrounding PV+ cells was observed in the infralimbic cortex, together with increase in the intensity of VGAT inhibitory puncta. Surprisingly, maternal deprivation did not cause any changes in the density of PV+ interneurons in the mPFC, neither had it affected PNNs and PV+ interneurons in the hippocampus. Taken together, our findings indicate that PNNs, specifically the ones enwrapping PV+ interneurons in the medial prefrontal cortex, are affected by early life stress.
- Published
- 2022
- Full Text
- View/download PDF
9. The impact of hyperpolarization-activated cyclic nucleotide-gated (HCN) and voltage-gated potassium KCNQ/Kv7 channels on primary microglia function
- Author
-
Sabine Ulrike Vay, Lea Jessica Flitsch, Monika Rabenstein, Helena Monière, Igor Jakovcevski, Pavle Andjus, Dunja Bijelic, Stefan Blaschke, Helene Luise Walter, Gereon Rudolf Fink, Michael Schroeter, and Maria Adele Rueger
- Subjects
Neuroinflammation ,Cerebral ischemia ,Ion channel ,Ih-current ,ZD7288 ,XE-991 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Microglia are essential to maintain cell homeostasis in the healthy brain and are activated after brain injury. Upon activation, microglia polarize towards different phenotypes. The course of microglia activation is complex and depends on signals in the surrounding milieu. Recently, it has been suggested that microglia respond to ion currents, as a way of regulating their activity and function. Methods and results Under the hypothesis that HCN and KCNQ/Kv7 channels impact on microglia, we studied primary rat microglia in the presence or absence of specific pharmacological blockade or RNA silencing. Primary microglia expressed the subunits HCN1-4, Kv7.2, Kv7.3, and Kv7.5. The expression of HCN2, as well as Kv7.2 and Kv7.3, varied among different microglia phenotypes. The pharmacological blockade of HCN channels by ZD7288 resulted in cell depolarization with slowly rising intracellular calcium levels, leading to enhanced survival and reduced proliferation rates of resting microglia. Furthermore, ZD7288 treatment, as well as knockdown of HCN2 RNA by small interfering RNA, resulted in an attenuation of later microglia activation—both towards the anti- and pro-inflammatory phenotype. However, HCN channel inhibition enhanced the phagocytic capacity of IL4-stimulated microglia. Blockade of Kv7/KCNQ channel by XE-991 exclusively inhibited the migratory capacity of resting microglia. Conclusion These observations suggest that the HCN current contributes to various microglia functions and impacts on the course of microglia activation, while the Kv7/KCNQ channels affect microglia migration. Characterizing the role of HCN channels in microglial functioning may offer new therapeutic approaches for targeted modulation of neuroinflammation as a hallmark of various neurological disorders.
- Published
- 2020
- Full Text
- View/download PDF
10. Mice Mutated in the Third Fibronectin Domain of L1 Show Enhanced Hippocampal Neuronal Cell Death, Astrogliosis and Alterations in Behavior
- Author
-
Ludovica Congiu, Viviana Granato, Igor Jakovcevski, Ralf Kleene, Luciana Fernandes, Sandra Freitag, Matthias Kneussel, Melitta Schachner, and Gabriele Loers
- Subjects
L1CAM ,hippocampus ,neuronal cell death ,astrogliosis ,behavior ,Microbiology ,QR1-502 - Abstract
Adhesion molecules play major roles in cell proliferation, migration, survival, neurite outgrowth and synapse formation during nervous system development and in adulthood. The neural cell adhesion molecule L1 contributes to these functions during development and in synapse formation and synaptic plasticity after trauma in adulthood. Mutations of L1 in humans result in L1 syndrome, which is associated with mild-to-severe brain malformations and mental disabilities. Furthermore, mutations in the extracellular domain were shown to cause a severe phenotype more often than mutations in the intracellular domain. To explore the outcome of a mutation in the extracellular domain, we generated mice with disruption of the dibasic sequences RK and KR that localize to position 858RKHSKR863 in the third fibronectin type III domain of murine L1. These mice exhibit alterations in exploratory behavior and enhanced marble burying activity. Mutant mice display higher numbers of caspase 3-positive neurons, a reduced number of principle neurons in the hippocampus, and an enhanced number of glial cells. Experiments suggest that disruption of the dibasic sequence in L1 results in subtle impairments in brain structure and functions leading to obsessive-like behavior in males and reduced anxiety in females.
- Published
- 2023
- Full Text
- View/download PDF
11. Maternal Deprivation in Rats Decreases the Expression of Interneuron Markers in the Neocortex and Hippocampus
- Author
-
Milan Aksic, Joko Poleksic, Dubravka Aleksic, Natasa Petronijevic, Nevena V. Radonjic, Maja Jakovcevski, Slobodan Kapor, Nevena Divac, Branislav R. Filipovic, and Igor Jakovcevski
- Subjects
cerebral cortex ,hippocampus ,interneurons ,maternal deprivation ,schizophrenia ,synapses ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Human anatomy ,QM1-695 - Abstract
Early life stress has profound effects on the development of the central nervous system. We exposed 9-day-old rat pups to a 24 h maternal deprivation (MD) and sacrificed them as young adults (60-day-old), with the aim to study the effects of early stress on forebrain circuitry. We estimated numbers of various immunohistochemically defined interneuron subpopulations in several neocortical regions and in the hippocampus. MD rats showed reduced numbers of parvalbumin-expressing interneurons in the CA1 region of the hippocampus and in the prefrontal cortex, compared with controls. Numbers of reelin-expressing and calretinin-expressing interneurons were also reduced in the CA1 and CA3 hippocampal areas, but unaltered in the neocortex of MD rats. The number of calbinin-expressing interneurons in the neocortex was similar in the MD rats compared with controls. We analyzed cell death in 15-day-old rats after MD and found no difference compared to control rats. Thus, our results more likely reflect the downregulation of markers than the actual loss of interneurons. To investigate synaptic activity in the hippocampus we immunostained for glutamatergic and inhibitory vesicular transporters. The number of inhibitory synapses was decreased in the CA1 and CA3 regions of the hippocampus in MD rats, with the normal number of excitatory synapses. Our results indicate complex, cell type-specific, and region-specific alterations in the inhibitory circuitry induced by maternal deprivation. Such alterations may underlie symptoms of MD at the behavioral level and possibly contribute to mechanisms by which early life stress causes neuropsychiatric disorders, such as schizophrenia.
- Published
- 2021
- Full Text
- View/download PDF
12. Cell Adhesion Molecule Close Homolog of L1 (CHL1) Guides the Regrowth of Regenerating Motor Axons and Regulates Synaptic Coverage of Motor Neurons
- Author
-
Daria Guseva, Igor Jakovcevski, Andrey Irintchev, Iryna Leshchyns’ka, Vladimir Sytnyk, Evgeni Ponimaskin, and Melitta Schachner
- Subjects
cell adhesion molecule close homolog of L1 (CHL1) ,femoral nerve ,peripheral nerve regeneration ,motor neuron ,mouse ,preferential motor re-innervation ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The close homolog of L1 (CHL1) is a cell adhesion molecule involved in regulation of neuronal differentiation and survival, neurite outgrowth and axon guidance during development. In the mature nervous system, CHL1 regulates synaptic activity and plasticity. The aim of the present study was to evaluate the influence of CHL1 on peripheral nerve regeneration after trauma. Using the established model of mouse femoral nerve regeneration, CHL1 knock-out mice were investigated in comparison to the wild type littermates. First, non-injured mice of both genotypes were compared regarding the synaptic phenotypes in the corresponding spinal cord segment. While no differences in phenotypes were detectable in the femoral nerve, corresponding segments in the spinal cord were observed to differ in that inhibitory perisomatic innervation of motor neurons was increased in CHL1-deficient mice, and numbers of perisomatic cholinergic synapses on motor neuronal somata were reduced. Regarding the femoral nerve after injury, CHL1-deficient mice demonstrated preferential motor axon regrowth into the saphenous vs. quadriceps branch after nerve transection upstream of the nerve bifurcation by 8 weeks after transection, indicating decreased preferential motor re-innervation. Furthermore, in injured wild-type mice, enhanced CHL1 expression was observed in regenerating axons in the proximal nerve stump upstream of the bifurcation at days 1, 3, 5, 7 and 14, and in the distal stump at days 7 and 14 after injury, when compared to non-injured mice. Injury-related upregulation of CHL1 expression was more pronounced in axons than in Schwann cells. Despite a more pronounced capacity for preferential motor axon regrowth in wild-type vs. mutant mice, only a tendency for difference in recovery of motor functions was observed between genotypes, without statistical significance Taken together, these results indicate that CHL1 is involved in peripheral nerve regeneration, because it guides regrowing axons into the appropriate nerve branch and regulates synaptic coverage in the spinal cord.
- Published
- 2018
- Full Text
- View/download PDF
13. Confocal Synaptology: Synaptic Rearrangements in Neurodegenerative Disorders and upon Nervous System Injury
- Author
-
Maja Vulovic, Nevena Divac, and Igor Jakovcevski
- Subjects
Alzheimer’s disease ,cholinergic synapses ,femoral nerve ,hippocampus ,spinal cord injury ,vesicular glutamate transporters ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Human anatomy ,QM1-695 - Abstract
The nervous system is a notable exception to the rule that the cell is the structural and functional unit of tissue systems and organs. The functional unit of the nervous system is the synapse, the contact between two nerve cells. As such, synapses are the foci of investigations of nervous system organization and function, as well as a potential readout for the progression of various disorders of the nervous system. In the past decade the development of antibodies specific to presynaptic terminals has enabled us to assess, at the optical, laser scanning microscopy level, these subcellular structures, and has provided a simple method for the quantification of various synapses. Indeed, excitatory (glutamatergic) and inhibitory synapses can be visualized using antibodies against the respective vesicular transporters, and choline-acetyl transferase (ChAT) immunoreactivity identifies cholinergic synapses throughout the central nervous system. Here we review the results of several studies in which these methods were used to estimate synaptic numbers as the structural equivalent of functional outcome measures in spinal cord and femoral nerve injuries, as well as in genetic mouse models of neurodegeneration, including Alzheimer’s disease (AD). The results implicate disease- and brain region-specific changes in specific types of synapses, which correlate well with the degree of functional deficit caused by the disease process. Additionally, results are reproducible between various studies and experimental paradigms, supporting the reliability of the method. To conclude, this quantitative approach enables fast and reliable estimation of the degree of the progression of neurodegenerative changes and can be used as a parameter of recovery in experimental models.
- Published
- 2018
- Full Text
- View/download PDF
14. Diversity of Cortical Interneurons in Primates: The Role of the Dorsal Proliferative Niche
- Author
-
Nevena V. Radonjić, Albert E. Ayoub, Fani Memi, Xiaojing Yu, Asif Maroof, Igor Jakovcevski, Stewart A. Anderson, Pasko Rakic, and Nada Zecevic
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Evolutionary elaboration of tissues starts with changes in the genome and location of the stem cells. For example, GABAergic interneurons of the mammalian neocortex are generated in the ventral telencephalon and migrate tangentially to the neocortex, in contrast to the projection neurons originating in the ventricular/subventricular zone (VZ/SVZ) of the dorsal telencephalon. In human and nonhuman primates, evidence suggests that an additional subset of neocortical GABAergic interneurons is generated in the cortical VZ and a proliferative niche, the outer SVZ. The origin, magnitude, and significance of this species-specific difference are not known. We use a battery of assays applicable to the human, monkey, and mouse organotypic cultures and supravital tissue to identify neuronal progenitors in the cortical VZ/SVZ niche that produce a subset of GABAergic interneurons. Our findings suggest that these progenitors constitute an evolutionary novelty contributing to the elaboration of higher cognitive functions in primates.
- Published
- 2014
- Full Text
- View/download PDF
15. Adhesion molecule L1 binds to amyloid beta and reduces Alzheimer's disease pathology in mice
- Author
-
Nevena Djogo, Igor Jakovcevski, Christian Müller, Hyun Joon Lee, Jin-Chong Xu, Mira Jakovcevski, Sebastian Kügler, Gabriele Loers, and Melitta Schachner
- Subjects
Adeno-associated virus serotype 5 ,Alzheimer's disease ,Amyloid precursor protein ,Amyloid beta peptides ,APPPS1 mice ,Astrogliosis ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder and the most common cause of elderly dementia. In an effort to contribute to the potential of molecular approaches to reduce degenerative processes we have tested the possibility that the neural adhesion molecule L1 ameliorates some characteristic cellular and molecular parameters associated with the disease in a mouse model of AD. Three-month-old mice overexpressing mutated forms of amyloid precursor protein and presenilin-1 under the control of a neuron-specific promoter received an injection of adeno-associated virus encoding the neuronal isoform of full-length L1 (AAV-L1) or, as negative control, green fluorescent protein (AAV-GFP) into the hippocampus and occipital cortex. Four months after virus injection, the mice were analyzed for histological and biochemical parameters of AD. AAV-L1 injection decreased the Aβ plaque load, levels of Aβ42, Aβ42/40 ratio and astrogliosis compared with AAV-GFP controls. AAV-L1 injected mice also had increased densities of inhibitory synaptic terminals on pyramidal cells in the hippocampus when compared with AAV-GFP controls. Numbers of microglial cells/macrophages were similar in both groups, but numbers of microglial cells/macrophages per plaque were increased in AAV-L1 injected mice. To probe for a molecular mechanism that may underlie these effects, we analyzed whether L1 would directly and specifically interact with Aβ. In a label-free binding assay, concentration dependent binding of the extracellular domain of L1, but not of the close homolog of L1 to Aβ40 and Aβ42 was seen, with the fibronectin type III homologous repeats 1–3 of L1 mediating this effect. Aggregation of Aβ42 in vitro was reduced in the presence of the extracellular domain of L1. The combined observations indicate that L1, when overexpressed in neurons and glia, reduces several histopathological hallmarks of AD in mice, possibly by reduction of Aβ aggregation. L1 thus appears to be a candidate molecule to ameliorate the pathology of AD, when applied in therapeutically viable treatment schemes.
- Published
- 2013
- Full Text
- View/download PDF
16. Embryonic stem cell-derived L1 overexpressing neural aggregates enhance recovery after spinal cord injury in mice.
- Author
-
Yi-Fang Cui, Jin-Chong Xu, Gunnar Hargus, Igor Jakovcevski, Melitta Schachner, and Christian Bernreuther
- Subjects
Medicine ,Science - Abstract
An obstacle to early stem cell transplantation into the acutely injured spinal cord is poor survival of transplanted cells. Transplantation of embryonic stem cells as substrate adherent embryonic stem cell-derived neural aggregates (SENAs) consisting mainly of neurons and radial glial cells has been shown to enhance survival of grafted cells in the injured mouse brain. In the attempt to promote the beneficial function of these SENAs, murine embryonic stem cells constitutively overexpressing the neural cell adhesion molecule L1 which favors axonal growth and survival of grafted and imperiled cells in the inhibitory environment of the adult mammalian central nervous system were differentiated into SENAs and transplanted into the spinal cord three days after compression lesion. Mice transplanted with L1 overexpressing SENAs showed improved locomotor function when compared to mice injected with wild-type SENAs. L1 overexpressing SENAs showed an increased number of surviving cells, enhanced neuronal differentiation and reduced glial differentiation after transplantation when compared to SENAs not engineered to overexpress L1. Furthermore, L1 overexpressing SENAs rescued imperiled host motoneurons and parvalbumin-positive interneurons and increased numbers of catecholaminergic nerve fibers distal to the lesion. In addition to encouraging the use of embryonic stem cells for early therapy after spinal cord injury L1 overexpression in the microenvironment of the lesioned spinal cord is a novel finding in its functions that would make it more attractive for pre-clinical studies in spinal cord regeneration and most likely other diseases of the nervous system.
- Published
- 2011
- Full Text
- View/download PDF
17. Oligodendrocyte development and the onset of myelination in the human fetal brain
- Author
-
Igor Jakovcevski, Radmila Filipovic, Zhicheng Mo, Sonja Rakic, and Nada Zecevic
- Subjects
Chemokines ,Immunohistochemistry ,Transcription Factors ,PSA-NCAM ,human brain development ,Myelination ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Human anatomy ,QM1-695 - Abstract
Oligodendrocytes are cells that myelinate axons, providing saltatory conduction of action potentials and proper function of the central nervous system. Myelination begins prenatally in the human, and the sequence of oligodendrocyte development and the onset of myelination are not thoroughly investigated. This knowledge is important to better understand human diseases, such as periventricular leukomalacia, one of the leading causes of motor deficit in premature babies, and demyelinating disorders such as multiple sclerosis (MS). In this review we discuss the spatial and temporal progression of oligodendrocyte lineage characterized by the expression of specific markers and transcription factors in the human fetal brain from the early embryonic period (5 gestational weeks, gw) until midgestation (24 gw). Our in vitro evidence indicated that a subpopulation of human oligodendrocytes may have dorsal origin, from cortical radial glia cells, in addition to their ventral telencephalic origin. Furthermore, we demonstrated that the regulation of myelination in the human fetal brain includes positive and negative regulators. Chemokines, such as CXCL1, abundant in proliferative zones during brain development and in regions of remyelination in adult, are discussed in the view of their potential roles in stimulating oligodendrocyte development. Other signals are inhibitory and may include, but are not limited to, polysialic acid modification of the neural cell adhesion molecule on axons. Overall, important differences in temporal and spatial distribution and regulatory signals for oligodendrocyte differentiation exist in the human brain. Those differences may underlie the unique susceptibility of humans to demyelinating diseases, such as MS.
- Published
- 2009
- Full Text
- View/download PDF
18. Inhibitory, But Not Excitatory Synapses Are Reduced in the Hippocampus of the Six-Month-Old Alzheimer’s Disease Model Mouse
- Author
-
Miljana Aksic, Ivona Bankovic, Igor Jakovcevski, Andrea Mojsoska, Sanja Stankovic, and Maja Vulovic
- Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder and the most common cause of elderly dementia. One of the main features of AD diseased brain are amyloid plaques, pathological depositions made of β-amyloid peptide, derived from β- amyloid precursor protein (APP). To assess how AD pathology affects synapses in the hippocampus, brain region to be one of the earliest with obvious pathological changes, we examined APPPS1 mice, transgeneticaly expressing human APP mutation (“Swedish mutation”) and human presenilin-1 mutation under the neuron-specific promoter, which develop AD symptoms early in life. We analyzed inhibitory and excitatory synapses using immunoflourescent staining and laser scanning confocal microscopy. In APPPS1 mice, inhibitory synaptic terminals labeled with vesicular inhibitory transmitter transporter (VGAT) were reduced in CA1 and CA3 regions of the hippocampus in APPPS1 mice compared to controls. This was true for both parvalbumin-positive and parvalbumin-negative terminals. On the other hand, excitatory synapses, coming from either hippocampal or entorhinal projections were similar between the genotypes. We conclude that first changes in the hippocampus caused by amyloid pathology affect inhibitory, but not excitatory synapses.
- Published
- 2023
- Full Text
- View/download PDF
19. Mice lacking perforin have improved regeneration of the injured femoral nerve
- Author
-
Melitta Schachner, Igor Jakovcevski, Monika von Düring, David Lutz, Maja Vulović, Mohammad Hamad, Gebhard Reiss, and Eckart Förster
- Subjects
Developmental Neuroscience ,blood-nerve barrier ,femoral nerve injury ,locomotor recovery ,lymphocytes ,myelination ,nk-cells ,perforin ,reinnervation ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
The role that the immune system plays after injury of the peripheral nervous system is still not completely understood. Perforin, a natural killer cell- and T-lymphocyte-derived enzyme that mediates cytotoxicity, plays important roles in autoimmune diseases, infections and central nervous system trauma, such as spinal cord injury. To dissect the roles of this single component of the immune response to injury, we tested regeneration after femoral nerve injury in perforin-deficient (Pfp–/–) and wild-type control mice. Single frame motion analysis showed better motor recovery in Pfp–/– mice compared with control mice at 4 and 8 weeks after injury. Retrograde tracing of the motoneuron axons regrown into the motor nerve branch demonstrated more correctly projecting motoneurons in the spinal cord of Pfp–/– mice compared with wild-types. Myelination of regrown axons measured by g-ratio was more extensive in Pfp–/– than in wild-type mice in the motor branch of the femoral nerve. Pfp–/– mice displayed more cholinergic synaptic terminals around cell bodies of spinal motoneurons after injury than the injured wild-types. We histologically analyzed lymphocyte infiltration 10 days after surgery and found that in Pfp–/– mice the number of lymphocytes in the regenerating nerves was lower than in wild-types, suggesting a closed blood-nerve barrier in Pfp–/– mice. We conclude that perforin restricts motor recovery after femoral nerve injury owing to decreased survival of motoneurons and reduced myelination.
- Published
- 2022
20. Histone H1 improves regeneration after mouse spinal cord injury and changes shape and gene expression of cultured astrocytes
- Author
-
Gabriele Loers, Ralf Kleene, Melitta Schachner, Igor Jakovcevski, and Bibhudatta Mishra
- Subjects
030506 rehabilitation ,Gene Expression ,Glial scar ,Histones ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Developmental Neuroscience ,Histone H1 ,Cell Movement ,Gene expression ,medicine ,Animals ,Axon ,Cells, Cultured ,Spinal Cord Injuries ,Motor Neurons ,biology ,Recovery of Function ,medicine.disease ,Axons ,Nerve Regeneration ,Cell biology ,Astrogliosis ,Disease Models, Animal ,Histone ,medicine.anatomical_structure ,nervous system ,Neurology ,Astrocytes ,Sialic Acids ,biology.protein ,Neurology (clinical) ,0305 other medical science ,Neuroglia ,Locomotion ,030217 neurology & neurosurgery ,Reinnervation ,Astrocyte - Abstract
Background We have shown that histone H1 is a binding partner for polysialic acid (PSA) and that it improves functional recovery, axon regrowth/sprouting, and target reinnervation after mouse femoral nerve injury. Objective Here, we analyzed whether histone H1 affects functional recovery, axon regrowth/sprouting, and target reinnervation after spinal cord injury of adult mice. Furthermore, we tested in vitro histone H1's effect on astrocytic gene expression, cell shape and migration as well as on cell survival of cultured motoneurons. Methods We applied histone H1 to compressed spinal cord and determined functional recovery and number of fibrillary acidic protein (GFAP)- and neuron-glial antigen 2 (NG2)- positive glial cells, which contribute to glial scarring. Histone H1's effect on migration of astrocytes, astrocytic gene expression and motoneuronal survival was determined using scratch-wounded astroglial monolayer cultures, astrocyte cultures for microarray analysis, and motoneuron cell culture under oxidative stress conditions, respectively. Results Histone H1 application improves locomotor functions and enhances monoaminergic and cholinergic reinnervation of the spinal cord. Expression levels of GFAP and NG2 around the lesion site were decreased in histone H1-treated mice relative to vehicle-treated mice six weeks after injury. Histone H1 reduced astrocytic migration, changed the shape of GFAP- and NG2-positive glial cells and altered gene expression. Gene ontology enrichment analysis indicated that in particular genes coding for proteins involved in proliferation, differentiation, migration and apoptosis are dysregulated. The up- and down-regulation of distinct genes was confirmed by qPCR and Western blot analysis. Moreover, histone H1 reduced hydrogen peroxide-induced cell death of cultured motoneurons. Conclusions The combined observations indicate that histone H1 locally applied to the lesion site, improves regeneration after spinal cord injury. Some of these beneficial functions of histone H1 in vivo and in vitro can be attributed to its interaction with PSA-carrying neural cell adhesion molecule.
- Published
- 2019
- Full Text
- View/download PDF
21. Pharmacology of repair after peripheral nerve injury
- Author
-
Nevena Divac, Milan Aksić, Lukas Rasulić, Maja Jakovcevski, Milos Basailovic, and Igor Jakovcevski
- Subjects
Pharmacology ,business.industry ,Calcium channel ,Regeneration (biology) ,Central nervous system ,Bioinformatics ,Functional recovery ,law.invention ,Peripheral ,Nerve Regeneration ,medicine.anatomical_structure ,Molecular level ,Randomized controlled trial ,law ,Peripheral Nerve Injuries ,Peripheral nerve injury ,medicine ,Humans ,Pharmacology (medical) ,Peripheral Nerves ,business ,Immunosuppressive Agents - Abstract
Peripheral nerve injuries are common and present with a broad spectrum of symptoms, some of which may be the cause of life-long disabilities. The peripheral nerves show a far greater capacity for regeneration than those in the central nervous system, and the process of nerve regeneration resembles developmental processes to a certain degree. The regeneration of peripheral nerves does not always lead to a full functional recovery. That is why surgical methods are still the most reliable therapeutic options after injuries of peripheral nerves. However, there is an array of potential pharmacological options that could enhance the repair processes after surgery. This review gives a summary of the recent literature relevant to different classes of pharmacologically active substances that are used either as supplements or off-label as potential enhancers of peripheral nerve repair. Antioxidants, vitamins, calcium channel blockers, immunosuppressive drugs, growth factors, and neuroactive glycans are among the most researched in this field. More research is necessary to understand their mechanisms of action at the cellular and molecular level, and randomized clinical trials in order to establish their efficacy and safety, as well as possible synergistic or adverse interactions among them.
- Published
- 2021
22. Developmental HCN channelopathy results in decreased neural progenitor proliferation and microcephaly in mice
- Author
-
Malte Stockebrand, Rafael Campos-Martin, Marta Florio, Achim Tresch, Niklas Kleinenkuhnen, Steffi Sandke, Igor Jakovcevski, Sabine Ulrike Vay, Maria Adele Rueger, Jochen Roeper, Anna Katharina Schlusche, Wieland B. Huttner, and Dirk Isbrandt
- Subjects
Microcephaly ,medicine.anatomical_structure ,Cell division ,Cerebral cortex ,Precursor cell ,medicine ,Cell fate determination ,Progenitor cell ,Biology ,medicine.disease ,Embryonic stem cell ,Neural stem cell ,Cell biology - Abstract
The development of the cerebral cortex relies on the controlled division of neural stem and progenitor cells. The requirement for precise spatiotemporal control of proliferation and cell fate places a high demand on the cell division machinery, and defective cell division can cause microcephaly and other brain malformations. Cell-extrinsic and intrinsic factors govern the capacity of cortical progenitors to produce large numbers of neurons and glia within a short developmental time window. In particular, ion channels shape the intrinsic biophysical properties of precursor cells and neurons and control their membrane potential throughout the cell cycle. We found that hyperpolarization-activated cyclic nucleotide-gated cation (HCN)-channel subunits are expressed in mouse, rat, and human neural progenitors. Loss of HCN-channel function in rat neural stem cells impaired their proliferation by affecting the cell-cycle progression, causing G1 accumulation and dysregulation of genes associated with human microcephaly. Transgene-mediated, dominant-negative loss of HCN-channel function in the embryonic mouse telencephalon resulted in pronounced microcephaly. Together, our findings suggest a novel role for HCN-channel subunits as a part of a general mechanism influencing cortical development in mammals.Significance StatementImpaired cell cycle regulation of neural stem and progenitor cells can affect cortical development and cause microcephaly. During cell cycle progression, the cellular membrane potential changes through the activity of ion channels and tends to be more depolarized in proliferating cells. HCN channels, which mediate a depolarizing current in neurons and cardiac cells, are linked to neurodevelopmental diseases, also contribute to the control of cell-cycle progression and proliferation of neuronal precursor cells. In this study, HCN-channel deficiency during embryonic and fetal brain development resulted in marked microcephaly of mice designed to be deficient in HCN-channel function in dorsal forebrain progenitors. The findings suggest that HCN-channel subunits are part of a general mechanism influencing cortical development in mammals.
- Published
- 2021
- Full Text
- View/download PDF
23. Corrigendum: Different Functions of Recombinantly Expressed Domains of Tenascin-C in Glial Scar Formation
- Author
-
Mina Peric, Eckart Förster, Igor Jakovcevski, Dunja Bijelić, Pavle R. Andjus, Melitta Schachner, and Marija Adžić
- Subjects
lcsh:Immunologic diseases. Allergy ,biology ,Chemistry ,Immunology ,Tenascin C ,medicine.disease ,microglia/macrophages ,spinal cord injury ,Microglia macrophages ,Cell biology ,Glial scar ,astrocyte ,medicine.anatomical_structure ,biology.protein ,medicine ,Immunology and Allergy ,glial scar ,tenascin-C ,lcsh:RC581-607 ,Spinal cord injury ,Astrocyte - Published
- 2021
- Full Text
- View/download PDF
24. Corrigendum: Different Functions of Recombinantly Expressed Domains of Tenascin-C in Glial Scar Formation
- Author
-
Marija Adžić, Melitta Schachner, Igor Jakovcevski, Eckart Förster, Dunja Bijelić, Pavle R. Andjus, and Mina Peric
- Subjects
0301 basic medicine ,Gene isoform ,lcsh:Immunologic diseases. Allergy ,medicine.medical_treatment ,Immunology ,microglia/macrophages ,Glial scar ,Extracellular matrix ,03 medical and health sciences ,Cicatrix ,Mice ,0302 clinical medicine ,astrocyte ,Protein Domains ,medicine ,Animals ,Immunology and Allergy ,Spinal Cord Injuries ,Original Research ,Mice, Knockout ,biology ,Microglia ,Cell growth ,Chemistry ,Tenascin C ,Correction ,Tenascin ,musculoskeletal system ,spinal cord injury ,Cell biology ,Alternative Splicing ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Astrocytes ,biology.protein ,glial scar ,tenascin-C ,lcsh:RC581-607 ,030217 neurology & neurosurgery ,Astrocyte - Abstract
Extracellular matrix glycoprotein tenascin-C (TnC) is highly expressed in vertebrates during embryonic development and thereafter transiently in tissue niches undergoing extensive remodeling during regeneration after injury. TnC’s different functions can be attributed to its multimodular structure represented by distinct domains and alternatively spliced isoforms. Upon central nervous system injury, TnC is upregulated and secreted into the extracellular matrix mainly by astrocytes. The goal of the present study was to elucidate the role of different TnC domains in events that take place after spinal cord injury (SCI). Astrocyte cultures prepared from TnC-deficient (TnC-/-) and wild-type (TnC+/+) mice were scratched and treated with different recombinantly generated TnC fragments. Gap closure, cell proliferation and expression of GFAP and cytokines were determined in these cultures. Gap closurein vitrowas found to be delayed by TnC fragments, an effect mainly mediated by decreasing proliferation of astrocytes. The most potent effects were observed with fragments FnD, FnA and their combination. TnC-/- astrocyte cultures exhibited higher GFAP protein and mRNA expression levels, regardless of the type of fragment used for treatment. Application of TnC fragments induced also pro-inflammatory cytokine production by astrocytesin vitro.In vivo, however, the addition of FnD or Fn(D+A) led to a difference between the two genotypes, with higher levels of GFAP expression in TnC+/+ mice. FnD treatment of injured TnC-/- mice increased the density of activated microglia/macrophages in the injury region, while overall cell proliferation in the injury site was not affected. We suggest that altogether these results may explain how the reaction of astrocytes is delayed while their localization is restricted to the border of the injury site to allow microglia/macrophages to form a lesion core during the first stages of glial scar formation, as mediated by TnC and, in particular, the alternatively spliced FnD domain.
- Published
- 2021
- Full Text
- View/download PDF
25. Developmental HCN channelopathy results in decreased neural progenitor proliferation and microcephaly in mice
- Author
-
Jochen Roeper, Niklas Kleinenkuhnen, Malte Stockebrand, Marta Florio, Anna Katharina Schlusche, Igor Jakovcevski, Sabine Ulrike Vay, Steffi Sandke, Dirk Isbrandt, Achim Tresch, Wieland B. Huttner, Rafael Campos-Martin, and Maria Adele Rueger
- Subjects
Microcephaly ,etiology [Channelopathies] ,Cell division ,brain development ,metabolism [Neural Stem Cells] ,cytology [Cerebral Cortex] ,Mice ,Neural Stem Cells ,metabolism [Embryonic Stem Cells] ,Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels ,physiology [Neural Stem Cells] ,microcephaly ,Cells, Cultured ,Cerebral Cortex ,Multidisciplinary ,biology ,Cell Death ,Cell Cycle ,Biological Sciences ,physiology [Neurogenesis] ,Neural stem cell ,physiology [Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels] ,Cell biology ,etiology [Microcephaly] ,medicine.anatomical_structure ,Cerebral cortex ,embryology [Cerebral Cortex] ,ddc:500 ,Neurogenesis ,antagonists & inhibitors [Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels] ,Mice, Transgenic ,Cell fate determination ,genetics [Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels] ,HCN channel ,medicine ,Animals ,Humans ,Progenitor cell ,Embryonic Stem Cells ,Cell Proliferation ,physiology [Cell Proliferation] ,embryology [Channelopathies] ,medicine.disease ,Embryonic stem cell ,physiology [Embryonic Stem Cells] ,Rats ,metabolism [Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels] ,embryology [Microcephaly] ,HCN channelopathy ,biology.protein ,Channelopathies - Abstract
The development of the cerebral cortex relies on the controlled division of neural stem and progenitor cells. The requirement for precise spatiotemporal control of proliferation and cell fate places a high demand on the cell division machinery, and defective cell division can cause microcephaly and other brain malformations. Cell-extrinsic and -intrinsic factors govern the capacity of cortical progenitors to produce large numbers of neurons and glia within a short developmental time window. In particular, ion channels shape the intrinsic biophysical properties of precursor cells and neurons and control their membrane potential throughout the cell cycle. We found that hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channel subunits are expressed in mouse, rat, and human neural progenitors. Loss of HCN channel function in rat neural stem cells impaired their proliferation by affecting the cell-cycle progression, causing G1 accumulation and dysregulation of genes associated with human microcephaly. Transgene-mediated, dominant-negative loss of HCN channel function in the embryonic mouse telencephalon resulted in pronounced microcephaly. Together, our findings suggest a role for HCN channel subunits as a part of a general mechanism influencing cortical development in mammals.
- Published
- 2021
- Full Text
- View/download PDF
26. Impact of Depletion of Microglia/Macrophages on Regeneration after Spinal Cord Injury
- Author
-
Gebhard Reiss, Eckart Förster, Melitta Schachner, and Igor Jakovcevski
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Central nervous system ,Mice, Transgenic ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Medicine ,Animals ,Spinal cord injury ,Spinal Cord Injuries ,Microglia ,biology ,business.industry ,General Neuroscience ,Regeneration (biology) ,Macrophages ,Recovery of Function ,medicine.disease ,Mice, Inbred C57BL ,Lumbar Spinal Cord ,030104 developmental biology ,medicine.anatomical_structure ,Integrin alpha M ,Spinal Cord ,biology.protein ,Cholinergic ,business ,030217 neurology & neurosurgery ,Astrocyte - Abstract
Microglia/macrophages play important functional roles in regeneration after central nervous system injury. Infiltration of circulating macrophages and proliferation of resident microglia occur within minutes following spinal cord injury. Activated microglia/macrophages clear tissue debris, but activation over time may hamper repair. To study the role of these cells in regeneration after spinal cord injury we used CD11b-herpes simplex virus thymidine kinase (HSVTK) (TK) transgenic mice, in which viral thymidine kinase activates ganciclovir toxicity in CD11b-expressing myeloid cells, including macrophages and microglia. A severe reduction in number of these cells was seen in TK versus wild-type littermate mice at 1 week and 5 weeks after injury, and numbers of Mac-2 expressing activated microglia/macrophages were almost completely reduced at these time points. One week after injury TK mice showed better locomotor recovery, but recovery was similar to wild-type mice as measured weekly up to 5 weeks thereafter. At 5 weeks after injury, numbers of axons at the lesion site and neurons in the lumbar spinal cord did not differ between groups. Also, catecholaminergic innervation of spinal motoneurons was similar. However, cholinergic innervation was lower and glial scarring was increased in TK mice compared to wild-type mice. We conclude that reducing numbers of CD11b-expressing cells improves locomotor recovery in the early phase after spinal cord injury, but does not affect recovery in the following 4 weeks. These observations point to differences in outcomes of astrocytic response and cholinergic innervation under CD11b cell ablation, which are, however, not reflected in the locomotor parameters analyzed at 5 weeks after injury.
- Published
- 2020
27. Perforin affects regeneration in a mouse spinal cord injury model
- Author
-
Melitta Schachner and Igor Jakovcevski
- Subjects
0301 basic medicine ,Male ,Pore Forming Cytotoxic Proteins ,Natural killer cell ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Cholinergic synapse ,Spinal cord injury ,Spinal Cord Injuries ,biology ,Microglia ,Behavior, Animal ,General Neuroscience ,Regeneration (biology) ,General Medicine ,medicine.disease ,Cell biology ,Nerve Regeneration ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Perforin ,biology.protein ,030217 neurology & neurosurgery ,Locomotion ,Astrocyte - Abstract
Locomotor outcomes in perforin-deficient (Pfp-/-) mice and wild-type littermate controls were measured after severe compression injury of the lower thoracic spinal cord up to six weeks after injury.According to both the Basso mouse scale score and single frame motion analysis, motor recovery of Pfp-/- mice was similar to wild-type controls. Interestingly, immunohistochemical analysis of cell types at six weeks after injury showed enhanced cholinergic reinnervation of spinal motor neurons caudal to the lesion site and neurofilament-positive structures at the injury site in Pfp-/- mice, whereas numbers of microglia/macrophages and astrocytes were decreased compared with controls.We conclude that, although, loss of perforin does not change the locomotor outcome after injury, it beneficially affects diverse cellular features, such as number of axons, cholinergic synapses, astrocytes and microglia/macrophages at or caudal to the lesion site. Perforin's ability to contribute to Rag2's influence on locomotion was observed in mice doubly deficient in perforin and Rag2 which recovered better than Rag2-/- or Pfp-/- mice, suggesting that natural killer cells can cooperate with T- and B-cells in spinal cord injury.
- Published
- 2020
28. The Na+/H+ exchanger Nhe1 modulates network excitability via GABA release
- Author
-
Hartmut T. Bocker, Julia Preobraschenski, Theresa Heinrich, Lutz Liebmann, Eric Seemann, Reinhard Jahn, Michael M. Kessels, J. Christopher Hennings, Melanie Gerth, Igor Jakovcevski, Christian A. Hübner, Britta Qualmann, Martin Westermann, Dirk Isbrandt, and Publica
- Subjects
Male ,Vesicular Inhibitory Amino Acid Transport Proteins ,physiology [Sodium-Hydrogen Exchanger 1] ,Membrane Potentials ,metabolism [Vesicular Inhibitory Amino Acid Transport Proteins] ,0302 clinical medicine ,GABAergic Neurons ,physiology [GABAergic Neurons] ,gamma-Aminobutyric Acid ,Sodium-Hydrogen Exchanger 1 ,Chemistry ,05 social sciences ,Cell biology ,metabolism [Presynaptic Terminals] ,Excitatory postsynaptic potential ,GABAergic ,Female ,Cognitive Neuroscience ,Presynaptic Terminals ,Glutamic Acid ,physiology [Interneurons] ,Mice, Transgenic ,physiology [Presynaptic Terminals] ,physiopathology [Epilepsy] ,Epilepsie ,Neurotransmission ,Inhibitory postsynaptic potential ,Synaptic vesicle ,050105 experimental psychology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Glutamatergic ,Interneurons ,metabolism [Vesicular Glutamate Transport Protein 1] ,mental disorders ,physiology [gamma-Aminobutyric Acid] ,Animals ,0501 psychology and cognitive sciences ,ddc:610 ,ion homeostasis ,metabolism [gamma-Aminobutyric Acid] ,CA1 Region, Hippocampal ,physiology [CA1 Region, Hippocampal] ,Epilepsy ,metabolism [Glutamic Acid] ,synaptic pH regulation ,Mice, Inbred C57BL ,Electrophysiology ,Ion homeostasis ,nervous system ,Vesicular Glutamate Transport Protein 1 ,Lichtenstein-Knorr syndrome ,Ataxie ,030217 neurology & neurosurgery - Abstract
Brain functions are extremely sensitive to pH changes because of the pH-dependence of proteins involved in neuronal excitability and synaptic transmission. Here, we show that the Na+/H+ exchanger Nhe1, which uses the Na+ gradient to extrude H+, is expressed at both inhibitory and excitatory presynapses. We disrupted Nhe1 specifically in mice either in Emx1-positive glutamatergic neurons or in parvalbumin-positive cells, mainly GABAergic interneurons. While Nhe1 disruption in excitatory neurons had no effect on overall network excitability, mice with disruption of Nhe1 in parvalbumin-positive neurons displayed epileptic activity. From our electrophysiological analyses in the CA1 of the hippocampus, we conclude that the disruption in parvalbumin-positive neurons impairs the release of GABA-loaded vesicles, but increases the size of GABA quanta. The latter is most likely an indirect pH-dependent effect, as Nhe1 was not expressed in purified synaptic vesicles itself. Conclusively, our data provide first evidence that Nhe1 affects network excitability via modulation of inhibitory interneurons.
- Published
- 2019
- Full Text
- View/download PDF
29. Tenascin-C deficiency protects mice from experimental autoimmune encephalomyelitis
- Author
-
Vera Stamenković, Đorđe Miljković, Melitta Schachner, Miljana Momčilović, Pavle R. Andjus, Milos Jovanovic, and Igor Jakovcevski
- Subjects
0301 basic medicine ,metabolism [Encephalomyelitis, Autoimmune, Experimental] ,medicine.medical_treatment ,Tenascin-C ,Mice ,metabolism [Spleen] ,0302 clinical medicine ,immunology [Th1 Cells] ,Immunology and Allergy ,Cells, Cultured ,Mice, Knockout ,Toll-like receptor ,immunology [Tenascin] ,Experimental autoimmune encephalomyelitis ,Myelin-associated glycoprotein ,Interleukin-17 ,Tenascin C ,deficiency [Tenascin] ,Tenascin ,musculoskeletal system ,Cytokine ,Neurology ,prevention & control [Encephalomyelitis, Autoimmune, Experimental] ,Female ,Interleukin 17 ,Encephalomyelitis, Autoimmune, Experimental ,Immunology ,metabolism [Th1 Cells] ,Biology ,immunology [Spleen] ,Myelin oligodendrocyte glycoprotein ,Multiple sclerosis ,T helper cells ,Interferon-gamma ,03 medical and health sciences ,Immune system ,medicine ,Animals ,ddc:610 ,cytology [Spleen] ,Th1 Cells ,immunology [Encephalomyelitis, Autoimmune, Experimental] ,immunology [Th17 Cells] ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,biology.protein ,Th17 Cells ,Neurology (clinical) ,metabolism [Th17 Cells] ,Spleen ,030217 neurology & neurosurgery - Abstract
The extracellular matrix glycoprotein tenascin-C (TnC) has been increasingly appreciated as a molecule susceptibly reacting to abnormalities in the mammalian immune system. TnC expression is elevated in inflamed tissues outside the immune system, but also in lymphoid organs. It participates in the promotion of inflammatory responses. Here, the role of TnC in a paradigm of CNS autoimmunity was investigated. Experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, was induced in mice deficient in TnC (TnC−/− mice). Amelioration of EAE was observed in these mice in comparison to their wild-type (TnC+/+) littermates. Since T helper (Th)1 and Th17 cells play a dominant role in the pathogenesis of EAE, these cells were investigated in addition to analyzing locomotor functions and pro-inflammatory cytokine levels. Smaller numbers of interferon-gamma-producing Th1 cells and reduced ability of Th17 cells to produce interleukin-17 were observed in spleens of TnC−/− mice challenged by immunization with the myelin associated glycoprotein (MOG) when compared to TnC+/+ mice. There was no difference in Th1 and Th17 responses in non-immunized TnC−/− and TnC+/+ mice, thus excluding generalized immunosuppression in TnC−/− mice. These results show that TnC is important for the pathogenesis of CNS autoimmunity and that its deficiency interferes with Th1 and Th17 encephalitogenic potentials.
- Published
- 2017
- Full Text
- View/download PDF
30. Impact of neural cell adhesion molecule deletion on regeneration after mouse spinal cord injury
- Author
-
Melitta Schachner, Gabriele Loers, Igor Jakovcevski, Vedangana Saini, and Gurcharan Kaur
- Subjects
0301 basic medicine ,Genetically modified mouse ,Nervous system ,medicine.medical_specialty ,Genotype ,metabolism [Axons] ,Biology ,metabolism [Spinal Cord Injuries] ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Injury Site ,Cell Movement ,Internal medicine ,medicine ,genetics [Neural Cell Adhesion Molecules] ,Animals ,ddc:610 ,Neural Cell Adhesion Molecules ,Spinal cord injury ,Cells, Cultured ,Spinal Cord Injuries ,physiology [Axons] ,metabolism [Astrocytes] ,Glial fibrillary acidic protein ,genetics [Spinal Cord Injuries] ,General Neuroscience ,physiology [Astrocytes] ,medicine.disease ,Spinal cord ,Axons ,Nerve Regeneration ,Mice, Inbred C57BL ,metabolism [Neural Cell Adhesion Molecules] ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,nervous system ,Astrocytes ,biology.protein ,Female ,Neural cell adhesion molecule ,Neuroscience ,Locomotion ,030217 neurology & neurosurgery ,Astrocyte - Abstract
The neural cell adhesion molecule (NCAM) plays important functional roles in development of the nervous system. We investigated the influence of a constitutive ablation of NCAM on the outcome of spinal cord injury. Transgenic mice lacking NCAM (NCAM-/-) were subjected to severe compression injury of the lower thoracic spinal cord using wild-type (NCAM+/+) littermates as controls. According to the single-frame motion analysis, the NCAM-/- mice showed reduced locomotor recovery in comparison to control mice at 3 and 6 weeks after injury, indicating an overall positive impact of NCAM on recovery after injury. Also the Basso Mouse Scale score was lower in NCAM-/- mice at 3 weeks after injury, whereas at 6 weeks after injury the difference between genotypes was not statistically significant. Worse locomotor function was associated with decreased monoaminergic and cholinergic innervation of the spinal cord caudal to the injury site and decreased axonal regrowth/sprouting at the site of injury. Astrocytic scar formation at the injury site, as assessed by immunohistology for glial fibrillary acidic protein at and around the lesion site was increased in NCAM-/- compared with NCAM+/+ mice. Migration of cultured monolayer astrocytes from NCAM-/- mice was reduced as assayed by scratch wounding. Numbers of Iba-1 immunopositive microglia were not different between genotypes. We conclude that constitutive NCAM deletion in young adult mice reduces recovery after spinal cord injury, validating the hypothesized beneficial role of this molecule in recovery after injury.
- Published
- 2016
- Full Text
- View/download PDF
31. Treatment during a vulnerable developmental period rescues a genetic epilepsy
- Author
-
Anton Ivanov, Vu Thao Quyen Le-Schulte, Christophe Bernard, Fabio Morellini, Ronny Eichler, Axel Neu, Stephan Lawrence Marguet, Igor Jakovcevski, Ileana L. Hanganu-Opatz, Dirk Isbrandt, and Andrea Merseburg
- Subjects
therapeutic use [Bumetanide] ,Male ,Nervous system ,drug effects [Body Weight] ,Time Factors ,pathology [Nerve Net] ,Hippocampus ,Hippocampal formation ,Bioinformatics ,pathology [Epilepsy] ,Epilepsy ,Cognition ,drug effects [Behavior, Animal] ,Solute Carrier Family 12, Member 2 ,Premovement neuronal activity ,Bumetanide ,Neurons ,Behavior, Animal ,KCNQ Potassium Channels ,genetics [KCNQ Potassium Channels] ,Electroencephalography ,pathology [CA1 Region, Hippocampal] ,metabolism [Solute Carrier Family 12, Member 2] ,General Medicine ,medicine.anatomical_structure ,metabolism [Neurons] ,drug effects [Cognition] ,drug effects [Growth and Development] ,Female ,Growth and Development ,medicine.symptom ,Proto-Oncogene Proteins c-fos ,medicine.drug ,drug effects [Embryo, Mammalian] ,medicine.medical_specialty ,pathology [Embryo, Mammalian] ,genetics [Epilepsy] ,genetics [Mutation] ,Inflammation ,drug therapy [Epilepsy] ,Biology ,metabolism [RNA, Messenger] ,General Biochemistry, Genetics and Molecular Biology ,genetics [RNA, Messenger] ,Internal medicine ,drug effects [Nerve Net] ,medicine ,drug effects [Neurons] ,Animals ,ddc:610 ,RNA, Messenger ,pharmacology [Bumetanide] ,metabolism [Embryo, Mammalian] ,CA1 Region, Hippocampal ,pathology [Inflammation] ,metabolism [KCNQ Potassium Channels] ,Body Weight ,Antagonist ,Embryo, Mammalian ,medicine.disease ,Mice, Inbred C57BL ,Endocrinology ,Animals, Newborn ,metabolism [Proto-Oncogene Proteins c-fos] ,Mutation ,Nerve Net - Abstract
The nervous system is vulnerable to perturbations during specific developmental periods. Insults during such susceptible time windows can have long-term consequences, including the development of neurological diseases such as epilepsy. Here we report that a pharmacological intervention timed during a vulnerable neonatal period of cortical development prevents pathology in a genetic epilepsy model. By using mice with dysfunctional Kv7 voltage-gated K(+) channels, which are mutated in human neonatal epilepsy syndromes, we demonstrate the safety and efficacy of the sodium-potassium-chloride cotransporter NKCC1 antagonist bumetanide, which was administered during the first two postnatal weeks. In Kv7 current-deficient mice, which normally display epilepsy, hyperactivity and stereotypies as adults, transient bumetanide treatment normalized neonatal in vivo cortical network and hippocampal neuronal activity, prevented structural damage in the hippocampus and restored wild-type adult behavioral phenotypes. Furthermore, bumetanide treatment did not adversely affect control mice. These results suggest that in individuals with disease susceptibility, timing prophylactically safe interventions to specific windows during development may prevent or arrest disease progression.
- Published
- 2015
- Full Text
- View/download PDF
32. Age-dependent loss of parvalbumin-expressing hippocampal interneurons in mice deficient in CHL1, a mental retardation and schizophrenia susceptibility gene
- Author
-
Giorgi Papashvili, Fang Kuang, Vladimir Sytnyk, Igor Jakovcevski, Barbara Schmalbach, Alexander G. Nikonenko, Iryna Leshchyns'ka, Eka Lepsveridze, Melitta Schachner, Nevena Djogo, and Alexander Dityatev
- Subjects
Aging ,Cerebellum ,Patch-Clamp Techniques ,metabolism [Interleukin-6] ,Hippocampus ,Hippocampal formation ,genetics [Gene Expression Regulation] ,metabolism [Parvalbumins] ,Biochemistry ,Chl1 protein, mouse ,Mice ,genetics [Excitatory Postsynaptic Potentials] ,genetics [Cell Adhesion Molecules] ,metabolism [Interneurons] ,metabolism [S100 Proteins] ,biology ,musculoskeletal, neural, and ocular physiology ,Microfilament Proteins ,S100 Proteins ,Long-term potentiation ,deficiency [Cell Adhesion Molecules] ,Parvalbumins ,medicine.anatomical_structure ,Excitatory postsynaptic potential ,Enzyme-Linked Immunosorbent Assay ,Mice, Transgenic ,In Vitro Techniques ,Inhibitory postsynaptic potential ,Cellular and Molecular Neuroscience ,Interneurons ,medicine ,Animals ,ddc:610 ,metabolism [Phosphopyruvate Hydratase] ,metabolism [Interleukin-3] ,Aif1 protein, mouse ,Interleukin-6 ,Calcium-Binding Proteins ,Excitatory Postsynaptic Potentials ,metabolism [Microfilament Proteins] ,metabolism [Synapses] ,metabolism [Calcium-Binding Proteins] ,Microscopy, Electron ,Gene Expression Regulation ,nervous system ,cytology [Hippocampus] ,Phosphopyruvate Hydratase ,ultrastructure [Synapses] ,Synapses ,Synaptic plasticity ,biology.protein ,Interleukin-3 ,Cell Adhesion Molecules ,Neuroscience ,Parvalbumin - Abstract
In humans, deletions/mutations in the CHL1/CALL gene are associated with mental retardation and schizophrenia. Juvenile CHL1-deficient (CHL1(-/-) ) mice have been shown to display abnormally high numbers of parvalbumin-expressing (PV(+) ) hippocampal interneurons and, as adults, display behavioral traits observed in neuropsychiatric disorders. Here, we addressed the question whether inhibitory interneurons and synaptic plasticity in the CHL1(-/-) mouse are affected during brain maturation and in adulthood. We found that hippocampal, but not neocortical, PV(+) interneurons were reduced with age in CHL1(-/-) mice, from a surplus of +27% at 1 month to a deficit of -20% in adulthood compared with wild-type littermates. This loss occurred during brain maturation, correlating with microgliosis and enhanced interleukin-6 expression. In parallel with the loss of PV(+) interneurons, the inhibitory input to adult CA1 pyramidal cells was reduced and a deficit in short- and long-term potentiation developed at CA3-CA1 excitatory synapses between 2 and 9 months of age in CHL1(-/-) mice. This deficit could be abrogated by a GABAA receptor agonist. We propose that region-specific aberrant GABAergic synaptic connectivity resulting from the mutation and a subsequently enhanced synaptic elimination during brain maturation lead to microgliosis, increase in pro-inflammatory cytokine levels, loss of interneurons, and impaired synaptic plasticity. Close homolog of L1-deficient (CHL1(-/-) ) mice have abnormally high numbers of parvalbumin (PV)-expressing hippocampal interneurons in juvenile animals, but in adult animals a loss of these cells is observed. This loss correlates with an increased density of microglia (M), enhanced interleukin-6 (IL6) production and a deficit in short- and long-term potentiation at CA3-CA1 excitatory synapses. Furthermore, adult CHL1(-/-) mice display behavioral traits similar to those observed in neuropsychiatric disorders of humans.
- Published
- 2015
- Full Text
- View/download PDF
33. The Subventricular Zone: A Key Player in Human Neocortical Development
- Author
-
Igor Jakovcevski, Radmila Filipovic, J. Alberto Ortega, Nevena V. Radonjić, Nada Zecevic, Inseyah Bagasrawala, and Fani Memi
- Subjects
0301 basic medicine ,Cell type ,Interneuron ,animal diseases ,Subventricular zone ,Neocortex ,Biology ,cytology [Neocortex] ,03 medical and health sciences ,physiology [Stem Cell Niche] ,Neural Stem Cells ,ddc:150 ,Lateral Ventricles ,medicine ,physiology [Neural Stem Cells] ,Animals ,Humans ,Progenitor cell ,Stem Cell Niche ,cytology [Neural Stem Cells] ,physiology [Neocortex] ,General Neuroscience ,Neurogenesis ,growth & development [Neocortex] ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Cerebral cortex ,Cerebral ventricle ,Neurology (clinical) ,Neuroscience - Abstract
One of the main characteristics of the developing brain is that all neurons and the majority of macroglia originate first in the ventricular zone (VZ), next to the lumen of the cerebral ventricles, and later on in a secondary germinal area above the VZ, the subventricular zone (SVZ). The SVZ is a transient compartment mitotically active in humans for several gestational months. It serves as a major source of cortical projection neurons as well as an additional source of glial cells and potentially some interneuron subpopulations. The SVZ is subdivided into the smaller inner (iSVZ) and the expanded outer SVZ (oSVZ). The enlargement of the SVZ and, in particular, the emergence of the oSVZ are evolutionary adaptations that were critical to the expansion and unique cellular composition of the primate cerebral cortex. In this review, we discuss the cell types and organization of the human SVZ during the first half of the 40 weeks of gestation that comprise intrauterine development. We focus on this period as it is when the bulk of neurogenesis in the human cerebral cortex takes place. We consider how the survival and fate of SVZ cells depend on environmental influences, by analyzing the results from in vitro experiments with human cortical progenitor cells. This in vitro model is a powerful tool to better understand human neocortex formation and the etiology of neurodevelopmental disorders, which in turn will facilitate the design of targeted preventive and/or therapeutic strategies.
- Published
- 2018
- Full Text
- View/download PDF
34. Hippocampal insulin resistance links maternal obesity with impaired neuronal plasticity in adult offspring
- Author
-
Eva Hucklenbruch-Rother, Inga Bae-Gartz, Jörg Dötsch, Christina Vohlen, Sarah Appel, Andrea Mesaros, Regina Ensenauer, Igor Jakovcevski, Ruth Janoschek, Marion Handwerk, Rebecca Kuglin, and Lisa Schmitz
- Subjects
0301 basic medicine ,Male ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,metabolism [Hippocampus] ,Hippocampal formation ,Hippocampus ,Mice ,0302 clinical medicine ,Endocrinology ,Pregnancy ,Hyperinsulinemia ,physiology [Neuronal Plasticity] ,Insulin ,Neurons ,Neuronal Plasticity ,Psychiatry and Mental health ,Prenatal Exposure Delayed Effects ,Female ,Signal Transduction ,medicine.medical_specialty ,Offspring ,Neurogenesis ,physiology [Insulin Resistance] ,Biology ,Cell Line ,03 medical and health sciences ,Insulin resistance ,Internal medicine ,Hyperinsulinism ,metabolism [Obesity] ,medicine ,Animals ,ddc:610 ,Obesity ,Biological Psychiatry ,Endocrine and Autonomic Systems ,medicine.disease ,metabolism [Prenatal Exposure Delayed Effects] ,Doublecortin ,metabolism [Insulin] ,Mice, Inbred C57BL ,Insulin receptor ,Disease Models, Animal ,030104 developmental biology ,metabolism [Hyperinsulinism] ,Synaptic plasticity ,biology.protein ,Insulin Resistance ,030217 neurology & neurosurgery - Abstract
Objective Maternal obesity and a disturbed metabolic environment during pregnancy and lactation have been shown to result in many long-term health consequences for the offspring. Among them, impairments in neurocognitive development and performance belong to the most dreaded ones. So far, very few mechanistic approaches have aimed to determine the responsible molecular events. Methods In a mouse model of maternal diet-induced obesity and perinatal hyperinsulinemia, we assessed adult offspring’s hippocampal insulin signaling as well as concurrent effects on markers of hippocampal neurogenesis, synaptic plasticity and function using western blotting and immunohistochemistry. In search for a potential link between neuronal insulin resistance and hippocampal plasticity, we additionally quantified protein expression of key molecules of synaptic plasticity in an in vitro model of acute neuronal insulin resistance. Results Maternal obesity and perinatal hyperinsulinemia result in adult hippocampal insulin resistance with subsequently reduced hippocampal mTor signaling and altered expression of markers of neurogenesis (doublecortin), synaptic plasticity (FoxO1, pSynapsin) and function (vGlut, vGAT) in the offspring. The observed effects are independent of the offspring’s adult metabolic phenotype and can be associated with multiple previously reported behavioral abnormalities. Additionally, we demonstrate that induction of insulin resistance in cultured hippocampal neurons reduces mTor signaling, doublecortin and vGAT protein expression. Conclusions Hippocampal insulin resistance might play a key role in mediating the long-term effects of maternal obesity and perinatal hyperinsulinemia on hippocampal plasticity and the offspring’s neurocognitive outcome.
- Published
- 2017
- Full Text
- View/download PDF
35. Tegaserod, a small compound mimetic of polysialic acid, promotes functional recovery after spinal cord injury in mice
- Author
-
Gabriele Loers, Igor Jakovcevski, Yan-Qin Shen, Melitta Schachner, and Hong-Chao Pan
- Subjects
Agonist ,Serotonin ,Indoles ,Tegaserod ,Tyrosine 3-Monooxygenase ,Neurite ,Cell Survival ,medicine.drug_class ,Cell Count ,Nerve Tissue Proteins ,Motor Activity ,Pharmacology ,Cicatrix ,Glial Fibrillary Acidic Protein ,Animals ,Medicine ,Gliosis ,Spinal cord injury ,Spinal Cord Injuries ,Neurons ,Glial fibrillary acidic protein ,biology ,Tyrosine hydroxylase ,business.industry ,Polysialic acid ,General Neuroscience ,Recovery of Function ,medicine.disease ,Spinal cord ,Axons ,Hindlimb ,Mice, Inbred C57BL ,Disease Models, Animal ,Neuroprotective Agents ,medicine.anatomical_structure ,Spinal Cord ,Astrocytes ,biology.protein ,Female ,business ,Neuroscience ,medicine.drug - Abstract
In a previous study, we have shown that the small organic compound tegaserod, a drug approved for clinical application in an unrelated condition, is a mimic of the regeneration-beneficial glycan polysialic acid (PSA) in a mouse model of femoral nerve injury. Several independent observations have shown positive effects of PSA and its mimetic peptides in different paradigms of injury of the central and peripheral mammalian nervous systems. Since small organic compounds generally have advantages over metabolically rapidly degraded glycans and the proteolytically vulnerable mimetic peptides, a screen for a small PSA mimetic compound was successfully carried out, and the identified molecule proved to be beneficial in neurite outgrowth in vitro, independent of its originally described function as a 5-HT4 receptor agonist. In the present study, a mouse spinal cord compression device was used to elicit severe compression injury. We show that tegaserod promotes hindlimb motor function at 6 weeks after spinal cord injury compared to the control group receiving vehicle only. Immunohistology of the spinal cord rostral and caudal to the lesion site showed increased numbers of neurons, and a reduced area and intensity of glial fibrillary acidic protein immunoreactivity. Quantification of regrowth/sprouting of axons immunoreactive for tyrosine hydroxylase and serotonin showed increased axonal density rostral and caudal to the injury site in the ventral horns of mice treated with tegaserod. The combined observations suggest that tegaserod has the potential for treatment of spinal cord injuries in higher vertebrates.
- Published
- 2014
- Full Text
- View/download PDF
36. Second-Generation Antipsychotics and Extrapyramidal Adverse Effects
- Author
-
Milica Prostran, Igor Jakovcevski, Nevena Divac, and Natasa Cerovac
- Subjects
medicine.medical_specialty ,Pediatrics ,Drug-Related Side Effects and Adverse Reactions ,medicine.medical_treatment ,lcsh:Medicine ,Context (language use) ,Review Article ,General Biochemistry, Genetics and Molecular Biology ,Benzodiazepines ,Basal Ganglia Diseases ,Extrapyramidal symptoms ,medicine ,Humans ,Psychiatry ,Adverse effect ,Antipsychotic ,Clozapine ,Risperidone ,General Immunology and Microbiology ,Receptors, Dopamine D2 ,business.industry ,lcsh:R ,General Medicine ,medicine.disease ,Comorbidity ,3. Good health ,Tolerability ,Schizophrenia ,medicine.symptom ,business ,Antipsychotic Agents ,medicine.drug - Abstract
Antipsychotic-induced extrapyramidal adverse effects are well recognized in the context of first-generation antipsychotic drugs. However, the introduction of second-generation antipsychotics, with atypical mechanism of action, especially lower dopamine receptors affinity, was met with great expectations among clinicians regarding their potentially lower propensity to cause extrapyramidal syndrome. This review gives a brief summary of the recent literature relevant to second-generation antipsychotics and extrapyramidal syndrome. Numerous studies have examined the incidence and severity of extrapyramidal syndrome with first- and second-generation antipsychotics. The majority of these studies clearly indicate that extrapyramidal syndrome does occur with second-generation agents, though in lower rates in comparison with first generation. Risk factors are the choice of a particular second-generation agent (with clozapine carrying the lowest risk and risperidone the highest), high doses, history of previous extrapyramidal symptoms, and comorbidity. Also, in comparative studies, the choice of a first-generation comparator significantly influences the results. Extrapyramidal syndrome remains clinically important even in the era of second-generation antipsychotics. The incidence and severity of extrapyramidal syndrome differ amongst these antipsychotics, but the fact is that these drugs have not lived up to the expectation regarding their tolerability.
- Published
- 2014
- Full Text
- View/download PDF
37. Prefrontal Cortical Dysfunction After Overexpression of Histone Deacetylase 1
- Author
-
David P. Gavin, Mira Jakovcevski, Amanda C. Mitchell, Guangping Gao, Schahram Akbarian, Igor Jakovcevski, Rahul Bharadwaj, and Juerg R. Straubhaar
- Subjects
medicine.medical_specialty ,Memory, Long-Term ,Microarray ,Genes, MHC Class II ,Down-Regulation ,Prefrontal Cortex ,Histone Deacetylase 1 ,Mice, Transgenic ,Biology ,Article ,Mice ,Internal medicine ,Gene expression ,medicine ,Haloperidol ,Animals ,Prefrontal cortex ,Clozapine ,Biological Psychiatry ,Neurons ,Working memory ,Histocompatibility Antigens Class II ,HDAC1 ,Up-Regulation ,Memory, Short-Term ,Endocrinology ,Astrocytes ,Exploratory Behavior ,Protein deacetylase ,Stereotyped Behavior ,Transcriptome ,Neuroscience ,medicine.drug - Abstract
Background Postmortem brain studies have shown that HDAC1 —a lysine deacetylase with broad activity against histones and nonhistone proteins—is frequently expressed at increased levels in prefrontal cortex (PFC) of subjects diagnosed with schizophrenia and related disease. However, it remains unclear whether upregulated expression of Hdac1 in the PFC could affect cognition and behavior. Methods Using adeno-associated virus, an Hdac1 transgene was expressed in young adult mouse PFC, followed by behavioral assays for working and long-term memory, repetitive activity, and response to novelty. Prefrontal cortex transcriptomes were profiled by microarray. Antipsychotic drug effects were explored in mice treated for 21 days with haloperidol or clozapine. Results Hdac1 overexpression in PFC neurons and astrocytes resulted in robust impairments in working memory, increased repetitive behaviors, and abnormal locomotor response profiles in novel environments. Long-term memory remained intact. Over 300 transcripts showed subtle but significant changes in Hdac1 -overexpressing PFC. Major histocompatibility complex class II (MHC II)-related transcripts, including HLA-DQA1/H2-Aa , HLA-DQB1/H2-Ab1, and HLA-DRB1/H2-Eb1, located in the chromosome 6p21.3-22.1 schizophrenia and bipolar disorder risk locus, were among the subset of genes with a more robust (>1.5-fold) downregulation in expression. Hdac1 levels declined during the course of normal PFC development. Antipsychotic drug treatment, including the atypical clozapine, did not affect Hdac1 levels in PFC but induced expression of multiple MHC II transcripts. Conclusions Excessive HDAC1 activity, due to developmental defects or other factors, is associated with behavioral alterations and dysregulated expression of MHC II and other gene transcripts in the PFC.
- Published
- 2013
- Full Text
- View/download PDF
38. Transgenic overexpression of the cell adhesion molecule L1 in neurons facilitates recovery after mouse spinal cord injury
- Author
-
Igor Jakovcevski, Emanuela Szpotowicz, Melitta Schachner, L.S. Hölters, and Nevena Djogo
- Subjects
medicine.medical_specialty ,Central nervous system ,Mice, Transgenic ,Neural Cell Adhesion Molecule L1 ,Glial scar ,Mice ,Spinal cord compression ,Internal medicine ,medicine ,Animals ,Spinal cord injury ,Spinal Cord Injuries ,Neurons ,Glial fibrillary acidic protein ,biology ,General Neuroscience ,Recovery of Function ,Spinal cord ,medicine.disease ,Immunohistochemistry ,Nerve Regeneration ,Disease Models, Animal ,Lumbar Spinal Cord ,medicine.anatomical_structure ,Endocrinology ,nervous system ,Corticospinal tract ,biology.protein ,Neuroscience - Abstract
It has been shown that the X-chromosome-linked neural cell adhesion molecule L1 plays a beneficial role in regeneration after spinal cord injury (SCI) in young adult rodents when applied in various molecular and cellular forms. In an attempt to further characterize the multiple functions of L1 after severe SCI we analyzed locomotor functions and measured axonal regrowth/sprouting and sparing, glial scarring, and synaptic remodeling at 6 weeks after severe spinal cord compression injury at the T7-9 levels of L1-deficient mice (L1-/y) and their wild-type (L1+/y) littermates, as well as mice that overexpress L1 under the control of the neuron-specific Thy-1 promoter (L1tg) and their wild-type littermates (L1+/+). No differences were found in the locomotor scale score and single frame motion analysis between L1-/y and L1+/y mice during 6 weeks after SCI, most likely due to the very low expression of L1 in the adult spinal cord of wild-type mice. L1tg mice, however, showed better locomotor recovery than their L1+/+ littermates, being associated with enhanced numbers of catecholaminergic axons in the lumbar spinal cord, but not of cholinergic, GABAergic or glutamatergic terminals around motoneuron cell bodies in the lumbar spinal cord. Additionally, no difference between L1tg and L1+/+ mice was detectable in dieback of corticospinal tract axons. Neuronal L1 overexpression did not influence the size of the glial fibrillary acidic protein-immunoreactive astrocytic scar 6 weeks after injury. We conclude that neuronal overexpression of L1 improves functional recovery from SCI by increasing catecholaminergic axonal regrowth/sprouting and/or sparing of severed axons without affecting the glial scar size.
- Published
- 2013
- Full Text
- View/download PDF
39. The extracellular matrix glycoprotein tenascin-C and matrix metalloproteinases modify cerebellar structural plasticity by exposure to an enriched environment
- Author
-
Lidija Radenovic, Leszek Kaczmarek, Pavle R. Andjus, Milos Jovanovic, Tomasz Jaworski, Stefan Stamenković, Igor Jakovcevski, Maciej Gawlak, Grzegorz M. Wilczynski, Melitta Schachner, and Vera Stamenković
- Subjects
0301 basic medicine ,Male ,Cerebellum ,metabolism [Purkinje Cells] ,Extracellular matrix ,Purkinje Cells ,Mice ,0302 clinical medicine ,genetics [Matrix Metalloproteinase 9] ,Mice, Knockout ,Neuronal Plasticity ,biology ,General Neuroscience ,Perineuronal net ,Tenascin C ,physiology [Purkinje Cells] ,Tenascin ,metabolism [Cerebellum] ,metabolism [Tenascin] ,musculoskeletal system ,Cell biology ,metabolism [Matrix Metalloproteinase 9] ,medicine.anatomical_structure ,Matrix Metalloproteinase 9 ,Gelatinases ,Matrix Metalloproteinase 2 ,Anatomy ,Histology ,physiology [Matrix Metalloproteinase 9] ,Environment ,Inhibitory postsynaptic potential ,03 medical and health sciences ,genetics [Tenascin] ,medicine ,Animals ,ddc:610 ,physiology [Tenascin] ,metabolism [Gelatinases] ,physiology [Cerebellum] ,Colocalization ,metabolism [Synapses] ,Mmp2 protein, mouse ,Mice, Inbred C57BL ,Mmp9 protein, mouse ,030104 developmental biology ,Synapses ,Synaptic plasticity ,biology.protein ,Neuroscience ,030217 neurology & neurosurgery - Abstract
The importance of the extracellular matrix (ECM) glycoprotein tenascin-C (TnC) and the ECM degrading enzymes, matrix metalloproteinases (MMPs) -2 and -9, in cerebellar histogenesis is well established. This study aimed to examine whether there is a functional relationship between these molecules in regulating structural plasticity of the lateral deep cerebellar nucleus. To this end, starting from postnatal day 21, TnC- or MMP-9-deficient mice were exposed to an enriched environment (EE). We show that 8 weeks of exposure to EE leads to reduced lectin-based staining of perineuronal nets (PNNs), reduction in the size of GABAergic and increase in the number and size of glutamatergic synaptic terminals in wild-type mice. Conversely, TnC-deficient mice showed reduced staining of PNNs compared to wild-type mice maintained under standard conditions, and exposure to EE did not further reduce, but even slightly increased PNN staining. EE did not affect the densities of the two types of synaptic terminals in TnC-deficient mice, while the size of inhibitory, but not excitatory synaptic terminals was increased. In the time frame of 4-8 weeks, MMP-9, but not MMP-2, was observed to influence PNN remodeling and cerebellar synaptic plasticity as revealed by measurement of MMP-9 activity and colocalization with PNNs and synaptic markers. These findings were supported by observations on MMP-9-deficient mice. The present study suggests that TnC contributes to the regulation of structural plasticity in the cerebellum and that interactions between TnC and MMP-9 are likely to be important for these processes to occur.
- Published
- 2017
- Full Text
- View/download PDF
40. Perinatal phencyclidine administration decreases the density of cortical interneurons and increases the expression of neuregulin-1
- Author
-
Igor Jakovcevski, Vladimir Bumbasirevic, Nataša Petronijević, and Nevena V. Radonjić
- Subjects
Male ,Time Factors ,Interneuron ,Neuregulin-1 ,Phencyclidine ,Hippocampus ,Biology ,Hippocampal formation ,Receptors, N-Methyl-D-Aspartate ,Interneurons ,Pregnancy ,Cortex (anatomy) ,medicine ,Animals ,Reelin ,Cerebral Cortex ,Pharmacology ,Dentate gyrus ,Rats ,Reelin Protein ,medicine.anatomical_structure ,Animals, Newborn ,Gene Expression Regulation ,nervous system ,biology.protein ,NMDA receptor ,Female ,Excitatory Amino Acid Antagonists ,Neuroscience ,medicine.drug - Abstract
Perinatal phencyclidine (PCP) administration in rat blocks the N-methyl d-aspartate receptor (NMDAR) and causes symptoms reminiscent of schizophrenia in human. A growing body of evidence suggests that alterations in γ-aminobutyric acid (GABA) interneuron neurotransmission may be associated with schizophrenia. Neuregulin-1 (NRG-1) is a trophic factor important for neurodevelopment, synaptic plasticity, and wiring of GABA circuits. The aim of this study was to determine the long-term effects of perinatal PCP administration on the projection and local circuit neurons and NRG-1 expression in the cortex and hippocampus. Rats were treated on postnatal day 2 (P2), P6, P9, and P12 with either PCP (10 mg/kg) or saline. Morphological studies and determination of NRG-1 expression were performed at P70. We demonstrate reduced densities of principal neurons in the CA3 and dentate gyrus (DG) subregions of the hippocampus and a reduction of major interneuronal populations in all cortical and hippocampal regions studied in PCP-treated rats compared with controls. For the first time, we show the reduced density of reelin- and somatostatin-positive cells in the cortex and hippocampus of animals perinatally treated with PCP. Furthermore, an increase in the numbers of perisomatic inhibitory terminals around the principal cells was observed in the motor cortex and DG. We also show that perinatal PCP administration leads to an increased NRG-1 expression in the cortex and hippocampus. Taken together, our findings demonstrate that perinatal PCP administration increases NRG-1 expression and reduces the number of projecting and local circuit neurons, revealing complex consequences of NMDAR blockade.
- Published
- 2013
- Full Text
- View/download PDF
41. Tenascins and inflammation in disorders of the nervous system
- Author
-
Melitta Schachner, Pavle R. Andjus, Igor Jakovcevski, and Djordje Miljković
- Subjects
Nervous system ,Cell type ,biology ,Regeneration (biology) ,Organic Chemistry ,Clinical Biochemistry ,Tenascin C ,Tenascin ,musculoskeletal system ,Biochemistry ,medicine.anatomical_structure ,Immune system ,Synaptic plasticity ,Immunology ,biology.protein ,medicine ,Animals ,Humans ,Tenascin-R ,Nervous System Diseases ,Neuroscience ,Neuroinflammation - Abstract
In vitro and in vivo studies on the role of tenascins have shown that the two paradigmatic glycoproteins of the tenascin family, tenascin-C (TnC) and tenascin-R (TnR) play important roles in cell proliferation and migration, fate determination, axonal pathfinding, myelination, and synaptic plasticity. As components of the extracellular matrix, both molecules show distinct, but also overlapping dual functions in inhibiting and promoting cell interactions depending on the cell type, developmental stage and molecular microenvironment. They are expressed by neurons and glia as well as, for TnC, by cells of the immune system. The functional relationship between neural and immune cells becomes relevant in acute and chronic nervous system disorders, in particular when the blood brain and blood peripheral nerve barriers are compromised. In this review, we will describe the functional parameters of the two molecules in cell interactions during development and, in the adult, in synaptic activity and plasticity, as well as regeneration after injury, with TnC being conducive for regeneration and TnR being inhibitory for functional recovery. Although not much is known about the role of tenascins in neuroinflammation, we will describe emerging knowledge on the interplay between neural and immune cells in autoimmune diseases, such as multiple sclerosis and polyneuropathies. We will attempt to point out the directions of experimental approaches that we envisage would help gaining insights into the complex interplay of TnC and TnR with the cells that express them in pathological conditions of nervous and immune systems.
- Published
- 2012
- Full Text
- View/download PDF
42. Heterozygosity for the mutated X-chromosome-linked L1 cell adhesion molecule gene leads to increased numbers of neurons and enhanced metabolism in the forebrain of female carrier mice
- Author
-
Konstantin-Alexander Hossmann, Janinne Sylvie Schmid, Günter Mies, Alexander G. Nikonenko, Zhang Ling, Christian Bernreuther, Igor Jakovcevski, and Melitta Schachner
- Subjects
Heterozygote ,Histology ,Blotting, Western ,Apoptosis ,Neural Cell Adhesion Molecule L1 ,Biology ,Loss of heterozygosity ,Mice ,Prosencephalon ,Species Specificity ,Genes, X-Linked ,Interneurons ,Image Processing, Computer-Assisted ,medicine ,Animals ,Humans ,In Situ Hybridization ,Mice, Knockout ,Analysis of Variance ,Neocortex ,Cell growth ,General Neuroscience ,Immunohistochemistry ,Null allele ,Molecular biology ,Cortex (botany) ,Phenotype ,medicine.anatomical_structure ,Bromodeoxyuridine ,nervous system ,Forebrain ,Immunology ,biology.protein ,Autoradiography ,Female ,Anatomy ,Parvalbumin - Abstract
Mutations in the X-chromosomal L1CAM gene lead to severe neurological deficits. In this study, we analyzed brains of female mice heterozygous for L1 (L1+/-) to gain insights into the brain structure of human females carrying one mutated L1 allele. From postnatal day 7 onward into adulthood, L1+/- female mice show an increased density of neurons in the neocortex and basal ganglia in comparison to wild-type (L1+/+) mice, correlating with enhanced metabolic parameters as measured in vivo. The densities of astrocytes and parvalbumin immunoreactive interneurons were not altered. No significant differences between L1+/- and L1+/+ mice were seen for cell proliferation in the cortex during embryonic days 11.5-15.5. Neuronal differentiation as estimated by analysis of doublecortin-immunoreactive cortical cells of embryonic brains was similar in L1+/- and L1+/+ mice. Interestingly, at postnatal days 3 and 5, apoptosis was reduced in L1+/- compared to L1+/+ mice. We suggest that reduced apoptosis leads to increased neuronal density in adult L1+/- mice. In conclusion, L1+/- mice display an unexpected phenotype that is not an intermediate between L1+/+ mice and mice deficient in L1 (L1-/y), but a novel phenotype which is challenging to understand regarding its underlying molecular and cellular mechanisms.
- Published
- 2012
- Full Text
- View/download PDF
43. Improved regeneration after spinal cord injury in mice lacking functional T- and B-lymphocytes
- Author
-
Bin Wu, Nevena Djogo, Dragana Matic, Emanuela Szpotowicz, Melitta Schachner, and Igor Jakovcevski
- Subjects
Genetically modified mouse ,Pathology ,medicine.medical_specialty ,T-Lymphocytes ,Blotting, Western ,Mice, Transgenic ,Motor Activity ,Biology ,Ventral column ,Mice ,Immune system ,Developmental Neuroscience ,medicine ,Animals ,Spinal cord injury ,Spinal Cord Injuries ,B-Lymphocytes ,Microglia ,Recovery of Function ,medicine.disease ,Spinal cord ,Immunohistochemistry ,Nerve Regeneration ,Mice, Inbred C57BL ,Disease Models, Animal ,Lumbar Spinal Cord ,medicine.anatomical_structure ,Neurology ,Female ,Neuroscience ,Astrocyte - Abstract
It is widely accepted that the immune system plays important functional roles in regeneration after injury to the spinal cord. Immune response towards injury involves a complex interplay of immune system cells, such as neutrophils, macrophages and microglia, T- and B-lymphocytes. We investigated the influence of the lymphocyte component of the immune system on the locomotor outcome of severe spinal cord injury in a genetic mouse model of immune suppression. Transgenic mice lacking mature T- and B-lymphocytes due to the recombination activating gene 2 gene deletion (RAG2-/- mice) were subjected to severe compression of the lower thoracic spinal cord, with the wild-type mice of the same inbred background serving as controls. According to both the Basso Mouse Scale score and single frame motion analysis, the RAG2-/- mice showed improved recovery in comparison to control mice at six weeks after injury. Better locomotor function was associated with enhanced catecholaminergic and cholinergic reinnervation of the spinal cord caudal to injury and increased axonal regrowth/sprouting at the site of injury. Myelination of axons in the ventral column measured as g-ratio was more extensive in RAG2-/- than in control mice 6weeks after injury. Additionally, the number of microglia/macrophages was decreased in the lumbar spinal cord of RAG2-/- mice after injury, whereas the number of astrocytes was increased compared with controls. We conclude that T- and B-lymphocytes restrict functional recovery from spinal cord injury by increasing numbers of microglia/macrophages as well as decreasing axonal sprouting and myelination.
- Published
- 2012
- Full Text
- View/download PDF
44. Adhesion molecule L1 overexpressed under the control of the neuronal Thy-1 promoter improves myelination after peripheral nerve injury in adult mice
- Author
-
Andrey Irintchev, Daria Guseva, Meike Zerwas, Melitta Schachner, Meifang Xiao, and Igor Jakovcevski
- Subjects
Male ,Blotting, Western ,Motor nerve ,Mice, Transgenic ,Neural Cell Adhesion Molecule L1 ,Biology ,Mice ,Developmental Neuroscience ,medicine ,Animals ,Axon ,Promoter Regions, Genetic ,Myelin Sheath ,Motor Neurons ,Neurons ,Motor neuron ,Nerve injury ,Axons ,Nerve Regeneration ,Cell biology ,Saphenous nerve ,medicine.anatomical_structure ,nervous system ,Neurology ,Peripheral nervous system ,Peripheral nerve injury ,Thy-1 Antigens ,Schwann Cells ,medicine.symptom ,Neuroscience ,Femoral Nerve ,Reinnervation - Abstract
L1 is an adhesion molecule favorably influencing the functional and anatomical recoveries after central nervous system (CNS) injuries. Its roles in peripheral nervous system (PNS) regeneration are less well understood. Studies using knockout mice have surprisingly revealed that L1 has a negative impact on functional nerve regeneration by inhibiting Schwann cell proliferation. To further elucidate the roles of L1 in PNS regeneration, here we used a novel transgenic mouse overexpressing L1 in neurons, but not in PNS or CNS glial cells, under the control of a neuron-specific Thy-1 promoter. Without nerve injury, the transgene expression, as compared to wild-type mice, had no effect on femoral nerve function, numbers of quadriceps motoneurons and myelinated axons in the femoral nerve but resulted in slightly reduced myelination in the sensory saphenous nerve and increased neurofilament density in myelinated axons of the quadriceps motor nerve branch. After femoral nerve injury, L1 overexpression had no impact on the time course and degree of functional recovery. Unaffected were also numbers of regenerated quadriceps motoneurons, precision of muscle reinnervation, axon numbers and internodal lengths in the regenerated nerves. Despite the lack of functional effects, myelination in the motor and sensory femoral nerve branches was significantly improved and loss of perisomatic inhibitory terminals on motoneurons was attenuated in the transgenic mice. Our results indicate that L1 is a regulator of myelination in the injured PNS and warrant studies aiming to improve function in demyelinating PNS and CNS disorders using exogenous L1.
- Published
- 2011
- Full Text
- View/download PDF
45. Interneurons in the developing human neocortex
- Author
-
Igor Jakovcevski, Nada Zecevic, and Frances Hu
- Subjects
education.field_of_study ,Neocortex ,Interneuron ,biology ,Population ,Subventricular zone ,Cellular and Molecular Neuroscience ,Corticogenesis ,medicine.anatomical_structure ,nervous system ,Developmental Neuroscience ,Cerebral cortex ,medicine ,biology.protein ,Calretinin ,education ,Neuroscience ,Parvalbumin - Abstract
Cortical interneurons play a crucial role in the functioning of cortical microcircuitry as they provide inhibitory input to projection (pyramidal) neurons. Despite their involvement in various neurological and psychiatric disorders, our knowledge about their development in human cerebral cortex is still incomplete. Here we demonstrate that at the beginning of corticogenesis, at embryonic 5 gestation weeks (gw, Carnegie stage 16) in human, early neurons could be labeled with calretinin, calbindin, and GABA antibodies. These immunolabeled cells show a gradient from the ganglionic eminences (GE) toward the neocortex, suggesting that GE is a well conserved source of early born cortical interneurons from rodents to human. At mid-term (20 gw), however, a subset of calretinin+ cells proliferates in the cortical subventricular zone (SVZ), suggesting a second set of interneuron progenitors that have neocortical origin. Neuropeptide Y, somatostatin, or parvalbumin cells are sparse in mid-term cerebral cortex. In addition to the early source of cortical interneurons in the GE and later in the neocortical SVZ, other regions, such as the subpial granular layer, may also contribute to the population of human cortical interneurons. In conclusion, our findings from cryosections and previous in vitro results suggest that cortical interneuron progenitor population is more complex in humans relative to rodents. The increased complexity of progenitors is probably evolutionary adaptation necessary for development of the higher brain functions characteristic to humans. © 2010 Wiley Periodicals, Inc. Develop Neurobiol 71: 18–33, 2011
- Published
- 2010
- Full Text
- View/download PDF
46. The Extracellular Matrix Glycoprotein Tenascin-C Is Beneficial for Spinal Cord Regeneration
- Author
-
Andrey Irintchev, Hsing-Yin Liu, Sally Meiners, Neha Bhagat, Melitta Schachner, Hyun Joon Lee, Gabriele Loers, Igor Jakovcevski, Grit Taschenberger, Sebastian Kügler, Jian Chen, and Ronak Shah
- Subjects
Spinal Cord Regeneration ,Neurite ,Blotting, Western ,Genetic Vectors ,Central nervous system ,Tenascin ,Biology ,H-Reflex ,Lesion ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,medicine ,Genetics ,Animals ,Spinal cord injury ,Molecular Biology ,Spinal Cord Injuries ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Tenascin C ,Anatomy ,Dependovirus ,medicine.disease ,musculoskeletal system ,Immunohistochemistry ,Cell biology ,Lumbar Spinal Cord ,medicine.anatomical_structure ,biology.protein ,Molecular Medicine ,Female ,Original Article ,medicine.symptom ,Locomotion ,030217 neurology & neurosurgery - Abstract
Tenascin-C (TNC), a major component of the extracellular matrix, is strongly upregulated after injuries of the central nervous system (CNS) but its role in tissue repair is not understood. Both regeneration promoting and inhibiting roles of TNC have been proposed considering its abilities to both support and restrict neurite outgrowth in vitro. Here, we show that spontaneous recovery of locomotor functions after spinal cord injury is impaired in adult TNC-deficient (TNC(-/-)) mice in comparison to wild-type (TNC(+/+)) mice. The impaired recovery was associated with attenuated excitability of the plantar Hoffmann reflex (H-reflex), reduced glutamatergic input, reduced sprouting of monaminergic axons in the lumbar spinal cord and enhanced post-traumatic degeneration of corticospinal axons. The degeneration of corticospinal axons in TNC(-/-) mice was normalized to TNC(+/+) levels by application of the alternatively spliced TNC fibronectin type III homologous domain D (fnD). Finally, overexpression of TNC-fnD via adeno-associated virus in wild-type mice improved locomotor recovery, increased monaminergic axons sprouting, and reduced lesion scar volume after spinal cord injury. The functional efficacy of the viral-mediated TNC indicates a potentially useful approach for treatment of spinal cord injury.
- Published
- 2010
- Full Text
- View/download PDF
47. Cellular Form of Prion Protein Inhibits Reelin-Mediated Shedding of Caspr from the Neuronal Cell Surface to Potentiate Caspr-Mediated Inhibition of Neurite Outgrowth
- Author
-
Igor Jakovcevski, Hyun Joon Lee, Vasudharani Devanathan, Elior Peles, Shen Li, Antonella Santuccione, Vladimir Sytnyk, Melitta Schachner, and Iryna Leshchyns'ka
- Subjects
Time Factors ,Cell ,Mice ,Catecholamines ,Cerebellum ,Cricetinae ,Serine ,Reelin ,Cells, Cultured ,Mice, Knockout ,Neurons ,Extracellular Matrix Proteins ,biology ,medicine.diagnostic_test ,Cell adhesion molecule ,General Neuroscience ,Chinese hamster ovary cell ,Serine Endopeptidases ,Brain ,Articles ,Protein Transport ,medicine.anatomical_structure ,Spinal Cord ,Female ,Cricetulus ,Locomotion ,Subcellular Fractions ,Neurite ,Prions ,Cell Adhesion Molecules, Neuronal ,Proteolysis ,Growth Cones ,Enzyme-Linked Immunosorbent Assay ,Nerve Tissue Proteins ,CHO Cells ,Transfection ,Inhibitory postsynaptic potential ,Neurites ,medicine ,Animals ,Immunoprecipitation ,Biotinylation ,Spinal Cord Injuries ,Recovery of Function ,biology.organism_classification ,Mice, Inbred C57BL ,Disease Models, Animal ,Reelin Protein ,Gene Expression Regulation ,nervous system ,biology.protein ,Cell Adhesion Molecules ,Neuroscience - Abstract
Extension of axonal and dendritic processes in the CNS is tightly regulated by outgrowth-promoting and -inhibitory cues to assure precision of synaptic connections. We identify a novel role for contactin-associated protein (Caspr) as an inhibitory cue that reduces neurite outgrowth from CNS neurons. We show that proteolysis of Caspr at the cell surface is regulated by the cellular form of prion protein (PrP), which directly binds to Caspr. PrP inhibits Reelin-mediated shedding of Caspr from the cell surface, thereby increasing surface levels of Caspr and potentiating the inhibitory effect of Caspr on neurite outgrowth. PrP deficiency results in reduced levels of Caspr at the cell surface, enhanced neurite outgrowth in vitro, and more efficient regeneration of axons in vivo following spinal cord injury. Thus, we reveal a previously unrecognized role for Caspr and PrP in inhibitory modulation of neurite outgrowth in CNS neurons, which is counterbalanced by the proteolytic activity of Reelin.
- Published
- 2010
- Full Text
- View/download PDF
48. Improved Reversal Learning and Working Memory and Enhanced Reactivity to Novelty in Mice with Enhanced GABAergic Innervation in the Dentate Gyrus
- Author
-
Igor Jakovcevski, Elena Sivukhina, Luminita Stoenica, Alexander Dityatev, Melitta Schachner, Olena Bukalo, Elena Oulianova, Andrey Irintchev, and Fabio Morellini
- Subjects
Male ,Cognitive Neuroscience ,Inhibitory postsynaptic potential ,gamma-Aminobutyric acid ,Extracellular matrix ,Mice ,Cellular and Molecular Neuroscience ,Cognition ,Interneurons ,Memory ,medicine ,Animals ,Learning ,gamma-Aminobutyric Acid ,Mice, Knockout ,Working memory ,Chemistry ,Dentate gyrus ,Neural Inhibition ,Tenascin ,Mice, Inbred C57BL ,nervous system ,Dentate Gyrus ,Synaptic plasticity ,Excitatory postsynaptic potential ,GABAergic ,Neuroscience ,medicine.drug - Abstract
The balance between excitation and inhibition controls fundamental aspects of the hippocampal function. Here, we report an increase in the ratio of inhibitory to excitatory neurons in the dentate gyrus, accompanied by γ-aminobutyric acid(A) (GABA(A)) receptor-dependent impairment of synaptic plasticity and enhancement of activity-dependent changes in excitability in anesthetized adult mice deficient for the extracellular matrix glycoprotein tenascin-R (TNR). TNR-deficient mice showed faster reversal learning, improved working memory, and enhanced reactivity to novelty than wild-type littermates. Remarkably, in wild-type and TNR-deficient mice, faster reversal learning rates correlated at the individual animal level with ratios of parvalbumin-positive interneurons to granule cells and densities of parvalbumin-positive terminals on somata of granule cells. Our data demonstrate that modification of the extracellular matrix by ablation of TNR leads to a new structural and functional design of the dentate gyrus, with enhanced GABAergic innervation, that is, enhanced ratio of inhibitory to excitatory cells, and altered plasticity, promoting working memory and reversal learning. In wild-type mice, the enhanced ratio of inhibitory to excitatory cells in the dentate gyrus also positively correlated with reversal learning, indicating that level of inhibition regulates specific aspects of learning independent of the TNR gene.
- Published
- 2010
- Full Text
- View/download PDF
49. Lewisxand α2,3-Sialyl Glycans and Their Receptors TAG-1, Contactin, and L1 Mediate CD24-Dependent Neurite Outgrowth
- Author
-
Ralf Kleene, Melitta Schachner, Domna Karagogeos, Gabriele Loers, Barbara Ranscht, Frauke Splittstoesser, Nainesh Katagihallimath, Annika Lieberoth, and Igor Jakovcevski
- Subjects
Glycosylation ,Neurite ,Cell Adhesion Molecules, Neuronal ,Lewis X Antigen ,Enzyme-Linked Immunosorbent Assay ,Fibronectin type III domain ,Biology ,Transfection ,Mice ,Cricetulus ,Dorsal root ganglion ,Contactins ,Cerebellum ,Cricetinae ,Ganglia, Spinal ,Contactin 2 ,Neurites ,medicine ,Animals ,Immunoprecipitation ,skin and connective tissue diseases ,Receptor ,Structural motif ,Cells, Cultured ,Spinal Cord Injuries ,Mice, Knockout ,Neurons ,Binding Sites ,Dose-Response Relationship, Drug ,Cell adhesion molecule ,General Neuroscience ,CD24 Antigen ,Articles ,Recovery of Function ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Animals, Newborn ,Sialic Acids ,Neural cell adhesion molecule ,Signal transduction ,Peptides ,Leukocyte L1 Antigen Complex ,Locomotion ,Protein Binding - Abstract
Although carbohydrates have been implicated in cell interactions in the nervous system, the molecular bases of their functions have remained largely obscure. Here, we show that promotion or inhibition of neurite outgrowth of cerebellar or dorsal root ganglion neurons, respectively, induced by the mucin-type adhesion molecule CD24 depends on α2,3-linked sialic acid and Lewisxpresent on glia-specific CD24 glycoforms. α2,3-Sialyl residues of CD24 bind to a structural motif in the first fibronectin type III domain of the adhesion molecule L1. Following the observation that the adhesion molecules TAG-1 and Contactin show sequence homologies with fucose-specific lectins, we obtained evidence that TAG-1 and Contactin mediate Lewisx-dependent CD24-induced effects on neurite outgrowth. Thus, L1, TAG-1, and Contactin function as lectin-like neuronal receptors. Theircisinteractions with neighboring adhesion molecules, e.g., Caspr1 and Caspr2, and with their triggered signal transduction pathways elicit cell type-specific promotion or inhibition of neurite outgrowth induced by glial CD24 in a glycan-dependenttransinteraction.
- Published
- 2009
- Full Text
- View/download PDF
50. Close homologue of adhesion molecule L1 promotes survival of Purkinje and granule cells and granule cell migration during murine cerebellar development
- Author
-
Nada Zecevic, Melitta Schachner, Andrey Irintchev, Janina Siering, Shengming Yin, Laura Hoelters, Nicole Karl, Gunnar Hargus, Igor Jakovcevski, and Nevena Djogo
- Subjects
Male ,Cerebellum ,Purkinje cell ,Cell Count ,Biology ,Polymerase Chain Reaction ,Mice ,Purkinje Cells ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Interneurons ,Pregnancy ,Precursor cell ,medicine ,Animals ,Cells, Cultured ,030304 developmental biology ,Mice, Knockout ,Neurons ,0303 health sciences ,Cell Death ,Cell growth ,Cell adhesion molecule ,General Neuroscience ,Cell Differentiation ,Cell migration ,Dendrites ,Embryo, Mammalian ,Granule cell ,Immunohistochemistry ,Cell biology ,Mice, Inbred C57BL ,CXCL3 ,medicine.anatomical_structure ,Animals, Newborn ,nervous system ,Female ,Cell Adhesion Molecules ,Neuroglia ,030217 neurology & neurosurgery - Abstract
Several L1-related adhesion molecules, expressed in a well-coordinated temporospatial pattern during development, are important for fine tuning of specific cerebellar circuitries. We tested the hypothesis that CHL1, the close homologue of L1, abundantly expressed in the developing and adult cerebellum, is also required for normal cerebellar histogenesis. We found that constitutive ablation of CHL1 in mice caused significant loss (20–23%) of Purkinje and granule cells in the mature 2-month-old cerebellum. The ratio of stellate/basket interneurons to Purkinje cells was abnormally high (+38%) in CHL1-deficient (CHL1−/−) mice compared with wild-type (CHL1+/+) littermates, but the γ-aminobutyric acid (GABA)ergic synaptic inputs to Purkinje cell bodies and dendrites were normal, as were numbers of Golgi interneurons, microglia, astrocytes, and Bergmann glia. Purkinje cell loss occurred before the first postnatal week and was associated with enhanced apoptosis, presumably as a consequence of CHL1 deficiency in afferent axons. In contrast, generation of granule cells, as indicated by in vivo analyses of cell proliferation and death, was unaffected in 1-week-old CHL1−/− mice, but numbers of migrating granule cells in the molecular layer were increased. This increase was likely related to retarded cell migration because CHL1−/− granule cells migrated more slowly than CHL1+/+ cells in vitro, and Bergmann glial processes guiding migration in vivo expressed CHL1 in wild-type mice. Granule cell deficiency in adult CHL1−/− mice appeared to result from decreased precursor cell proliferation after the first postnatal week. Our results indicate that CHL1 promotes Purkinje and granule cell survival and granule cell migration during cerebellar development. J. Comp. Neurol. 513:496–510, 2009. © 2009 Wiley-Liss, Inc.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.