40 results on '"Mätlik K"'
Search Results
2. Nicotinamide riboside alleviates Parkinson's disease symptoms but downregulates dopamine metabolism
- Author
-
Turconi, G., primary, Alam, F., additional, SenGupta, T., additional, Pirnes-Karhu, S., additional, Olfat, S., additional, Schmidt, M., additional, Mätlik, K., additional, Montaño-Rodriguez, A., additional, Heiskanen, V., additional, Piepponen, P., additional, Brenner, C., additional, Holmberg, C., additional, Nilsen, H., additional, Andressoo, J.-O., additional, and Pirinen, E., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Gene Knock Up via 3’UTR editing to study gene function in vivo
- Author
-
Schweizer N, Cowlishaw Mc, Giorgio Turconi, Olfat S, Garton Dr, Jaan-Olle Andressoo, Mätlik K, Johan Jakobsson, Anne Panhelainen, Jaakko Kopra, Ana R. Montaño-Rodríguez, Fu-Ping Zhang, Timo Petteri Piepponen, Lauriina L. Porokuokka, and Sipilä P
- Subjects
0303 health sciences ,biology ,Three prime untranslated region ,Endogeny ,Phenotype ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Glial cell line-derived neurotrophic factor ,biology.protein ,Copy-number variation ,Gene ,030217 neurology & neurosurgery ,Function (biology) ,030304 developmental biology - Abstract
Currently available genetic tools do not allow researchers to upregulate (‘Knock Up’) the levels of a given protein while retaining its cell-type-specific regulation. As a result, we have limited ability to develop overexpression-related disease models, to study the contribution of single genes in diseases caused by copy number variations and to identify disease pathways for drug targets. Here we develop two approaches for endogenous gene upregulation:conditionalKnockUp (cKU) utilizing the Cre/lox system, and CRISPR-Cas9 mediated geneKnockUp (KU) in wild-type mouse embryos and human cells. Using glial cell line derived neurotrophic factor (GDNF) as a proof of concept, we show that both approaches resulted in upregulation of endogenous GDNF levels without disturbingGdnf’s expression pattern. Furthermore, CNS-specific GDNF cKU resulted in dopaminergic abnormalities and schizophrenia-like phenotypes. Our results suggest that gene Knock Up can reveal unknown gene functions and provide novel entry points for studying neurological disease.
- Published
- 2019
- Full Text
- View/download PDF
4. Role of two sequence motifs of mesencephalic astrocyte-derived neurotrophic factor in its survival-promoting activity
- Author
-
Mätlik, K, primary, Yu, Li-ying, additional, Eesmaa, A, additional, Hellman, M, additional, Lindholm, P, additional, Peränen, J, additional, Galli, E, additional, Anttila, J, additional, Saarma, M, additional, Permi, P, additional, Airavaara, M, additional, and Arumäe, U, additional
- Published
- 2015
- Full Text
- View/download PDF
5. L1 antisense promoter drives tissue-specific transcription of human genes.
- Author
-
Mätlik K, Redik K, and Speek M
- Published
- 2006
- Full Text
- View/download PDF
6. Combination of native and denaturing PAGE for the detection of protein binding regions in long fragments of genomic DNA
- Author
-
Metsis Madis, Mätlik Kert, Kaer Kristel, and Speek Mart
- Subjects
Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Background In a traditional electrophoresis mobility shift assay (EMSA) a 32P-labeled double-stranded DNA oligonucleotide or a restriction fragment bound to a protein is separated from the unbound DNA by polyacrylamide gel electrophoresis (PAGE) in nondenaturing conditions. An extension of this method uses the large population of fragments derived from long genomic regions (approximately 600 kb) for the identification of fragments containing protein binding regions. With this method, genomic DNA is fragmented by restriction enzymes, fragments are amplified by PCR, radiolabeled, incubated with nuclear proteins and the resulting DNA-protein complexes are separated by two-dimensional PAGE. Shifted DNA fragments containing protein binding sites are identified by using additional procedures, i. e. gel elution, PCR amplification, cloning and sequencing. Although the method allows simultaneous analysis of a large population of fragments, it is relatively laborious and can be used to detect only high affinity protein binding sites. Here we propose an alternative and straightforward strategy which is based on a combination of native and denaturing PAGE. This strategy allows the identification of DNA fragments containing low as well as high affinity protein binding regions, derived from genomic DNA ( Results We have combined an EMSA-based selection step with subsequent denaturing PAGE for the localization of protein binding regions in long (up to10 kb) fragments of genomic DNA. Our strategy consists of the following steps: digestion of genomic DNA with a 4-cutter restriction enzyme (AluI, BsuRI, TruI, etc), separation of low and high molecular weight fractions of resultant DNA fragments, 32P-labeling with Klenow polymerase, traditional EMSA, gel elution and identification of the shifted bands (or smear) by denaturing PAGE. The identification of DNA fragments containing protein binding sites is carried out by running the gel-eluted fragments alongside with the full "spectrum" of initial restriction fragments of known size. Here the strategy is used for the identification of protein-binding regions in the 5' region of the rat p75 neurotrophin receptor (p75NTR) gene. Conclusion The developed strategy is based on a combination of traditional EMSA and denaturing PAGE for the identification of protein binding regions in long fragments of genomic DNA. The identification is straightforward and can be applied to shifted bands corresponding to stable DNA-protein complexes as well as unstable complexes, which undergo dissociation during electrophoresis.
- Published
- 2008
- Full Text
- View/download PDF
7. Mice lacking Astn2 have ASD-like behaviors and altered cerebellar circuit properties.
- Author
-
Hanzel M, Fernando K, Maloney SE, Horn Z, Gong S, Mätlik K, Zhao J, Pasolli HA, Heissel S, Dougherty JD, Hull C, and Hatten ME
- Subjects
- Animals, Mice, Behavior, Animal physiology, Nerve Tissue Proteins metabolism, Nerve Tissue Proteins genetics, Disease Models, Animal, Membrane Proteins metabolism, Membrane Proteins genetics, Male, Mice, Knockout, Autism Spectrum Disorder metabolism, Autism Spectrum Disorder genetics, Autism Spectrum Disorder physiopathology, Purkinje Cells metabolism, Cerebellum metabolism
- Abstract
Astrotactin 2 (ASTN2) is a transmembrane neuronal protein highly expressed in the cerebellum that functions in receptor trafficking and modulates cerebellar Purkinje cell (PC) synaptic activity. Individuals with ASTN2 mutations exhibit neurodevelopmental disorders, including autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), learning difficulties, and language delay. To provide a genetic model for the role of the cerebellum in ASD-related behaviors and study the role of ASTN2 in cerebellar circuit function, we generated global and PC-specific conditional Astn2 knockout (KO and cKO, respectively) mouse lines. Astn2 KO mice exhibit strong ASD-related behavioral phenotypes, including a marked decrease in separation-induced pup ultrasonic vocalization calls, hyperactivity, repetitive behaviors, altered behavior in the three-chamber test, and impaired cerebellar-dependent eyeblink conditioning. Hyperactivity and repetitive behaviors are also prominent in Astn2 cKO animals, but they do not show altered behavior in the three-chamber test. By Golgi staining, Astn2 KO PCs have region-specific changes in dendritic spine density and filopodia numbers. Proteomic analysis of Astn2 KO cerebellum reveals a marked upregulation of ASTN2 family member, ASTN1, a neuron-glial adhesion protein. Immunohistochemistry and electron microscopy demonstrate a significant increase in Bergmann glia volume in the molecular layer of Astn2 KO animals. Electrophysiological experiments indicate a reduced frequency of spontaneous excitatory postsynaptic currents (EPSCs), as well as increased amplitudes of both spontaneous EPSCs and inhibitory postsynaptic currents in the Astn2 KO animals, suggesting that pre- and postsynaptic components of synaptic transmission are altered. Thus, ASTN2 regulates ASD-like behaviors and cerebellar circuit properties., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
8. Nicotinamide riboside first alleviates symptoms but later downregulates dopamine metabolism in proteasome inhibition mouse model of Parkinson's disease.
- Author
-
Turconi G, Alam F, SenGupta T, Pirnes-Karhu S, Olfat S, Schmidt MS, Mätlik K, Montaño-Rodriguez A, Heiskanen V, Garton D, Piepponen PT, Brenner C, Holmberg CI, Nilsen H, Pirinen E, and Andressoo JO
- Abstract
Parkinson's disease (PD) is associated with a reduction in 26/20S proteasome and mitochondrial function and depletion of dopamine. Activation of mitochondrial function with the NAD
+ precursor nicotinamide riboside (NR) is a potential therapeutic for PD. However, despite recently started clinical trials, analysis of NR in mammalian animal PD models is lacking and data in simpler PD models is limited. We analyzed the effect of NR in C. elegans and in mouse 26/20S proteasome inhibition models of PD. In C. elegans , NR rescued α-synuclein overexpression induced phenotypes likely by activating the mitochondrial unfolded protein response. However, in a proteasome inhibitor-induced mouse model of PD, NR first partially rescued behavioural dysfunction, but later resulted in decrease in dopamine and its related gene expression in the substantia nigra. Our results suggest that reduction in 26/20S function with long term NR treatment may increase risk for developing reduced nigrostriatal DA function., Competing Interests: The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Charles Brenner reports a relationship with ChromaDex Inc that includes: consulting or advisory and equity or stocks. Charles Brenner reports a relationship with Athena Therapeutics that includes: equity or stocks. Charles Brenner reports a relationship with Juvenis that includes: equity or stocks. Eija Pirinen reports a relationship with ChromaDex Inc that includes: speaking and lecture fees. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2024 Published by Elsevier Ltd.)- Published
- 2024
- Full Text
- View/download PDF
9. Selective vulnerability of layer 5a corticostriatal neurons in Huntington's disease.
- Author
-
Pressl C, Mätlik K, Kus L, Darnell P, Luo JD, Paul MR, Weiss AR, Liguore W, Carroll TS, Davis DA, McBride J, and Heintz N
- Subjects
- Animals, Neurons metabolism, Pyramidal Cells metabolism, Cerebral Cortex metabolism, Solitary Nucleus metabolism, Disease Models, Animal, Huntingtin Protein genetics, Huntingtin Protein metabolism, Huntington Disease metabolism
- Abstract
The properties of the cell types that are selectively vulnerable in Huntington's disease (HD) cortex, the nature of somatic CAG expansions of mHTT in these cells, and their importance in CNS circuitry have not been delineated. Here, we employed serial fluorescence-activated nuclear sorting (sFANS), deep molecular profiling, and single-nucleus RNA sequencing (snRNA-seq) of motor-cortex samples from thirteen predominantly early stage, clinically diagnosed HD donors and selected samples from cingulate, visual, insular, and prefrontal cortices to demonstrate loss of layer 5a pyramidal neurons in HD. Extensive mHTT CAG expansions occur in vulnerable layer 5a pyramidal cells, and in Betz cells, layers 6a and 6b neurons that are resilient in HD. Retrograde tracing experiments in macaque brains identify layer 5a neurons as corticostriatal pyramidal cells. We propose that enhanced somatic mHTT CAG expansion and altered synaptic function act together to cause corticostriatal disconnection and selective neuronal vulnerability in HD cerebral cortex., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Cell-type-specific CAG repeat expansions and toxicity of mutant Huntingtin in human striatum and cerebellum.
- Author
-
Mätlik K, Baffuto M, Kus L, Deshmukh AL, Davis DA, Paul MR, Carroll TS, Caron MC, Masson JY, Pearson CE, and Heintz N
- Subjects
- Humans, Animals, Cerebellum metabolism, Disease Models, Animal, Corpus Striatum, Huntington Disease genetics
- Abstract
Brain region-specific degeneration and somatic expansions of the mutant Huntingtin (mHTT) CAG tract are key features of Huntington's disease (HD). However, the relationships among CAG expansions, death of specific cell types and molecular events associated with these processes are not established. Here, we used fluorescence-activated nuclear sorting (FANS) and deep molecular profiling to gain insight into the properties of cell types of the human striatum and cerebellum in HD and control donors. CAG expansions arise at mHTT in striatal medium spiny neurons (MSNs), cholinergic interneurons and cerebellar Purkinje neurons, and at mutant ATXN3 in MSNs from SCA3 donors. CAG expansions in MSNs are associated with higher levels of MSH2 and MSH3 (forming MutSβ), which can inhibit nucleolytic excision of CAG slip-outs by FAN1. Our data support a model in which CAG expansions are necessary but may not be sufficient for cell death and identify transcriptional changes associated with somatic CAG expansions and striatal toxicity., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
11. MANF protein expression is upregulated in immune cells in the ischemic human brain and systemic recombinant MANF delivery in rat ischemic stroke model demonstrates anti-inflammatory effects.
- Author
-
Anttila JE, Mattila OS, Liew HK, Mätlik K, Mervaala E, Lindholm P, Lindahl M, Lindsberg PJ, Tseng KY, and Airavaara M
- Subjects
- Humans, Rats, Mice, Animals, Rats, Sprague-Dawley, Brain metabolism, Nerve Growth Factors genetics, Nerve Growth Factors metabolism, Nerve Growth Factors therapeutic use, Cerebral Infarction metabolism, Inflammation metabolism, Ischemic Stroke metabolism, Stroke metabolism
- Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) has cytoprotective effects on various injuries, including cerebral ischemia, and it can promote recovery even when delivered intracranially several days after ischemic stroke. In the uninjured rodent brain, MANF protein is expressed almost exclusively in neurons, but post-ischemic MANF expression has not been characterized. We aimed to investigate how endogenous cerebral MANF protein expression evolves in infarcted human brains and rodent ischemic stroke models. During infarct progression, the cerebral MANF expression pattern both in human and rat brains shifted drastically from neurons to expression in inflammatory cells. Intense MANF immunoreactivity took place in phagocytic microglia/macrophages in the ischemic territory, peaking at two weeks post-stroke in human and one-week post-stroke in rat ischemic cortex. Using double immunofluorescence and mice lacking MANF gene and protein from neuronal stem cells, neurons, astrocytes, and oligodendrocytes, we verified that MANF expression was induced in microglia/macrophage cells in the ischemic hemisphere. Embarking on the drastic expression transition towards inflammatory cells and the impact of blood-borne inflammation in stroke, we hypothesized that exogenously delivered MANF protein can modulate tissue recovery processes. In an attempt to enhance recovery, we designed a set of proof-of-concept studies using systemic delivery of recombinant MANF in a rat model of cortical ischemic stroke. Intranasal recombinant MANF treatment decreased infarct volume and reduced the severity of neurological deficits. Intravenous recombinant MANF treatment decreased the levels of pro-inflammatory cytokines and increased the levels of anti-inflammatory cytokine IL-10 in the infarcted cortex one-day post-stroke. In conclusion, MANF protein expression is induced in activated microglia/macrophage cells in infarcted human and rodent brains, and this could implicate MANF's involvement in the regulation of post-stroke inflammation in patients and experimental animals. Moreover, systemic delivery of recombinant MANF shows promising immunomodulatory effects and therapeutic potential in experimental ischemic stroke., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. Selective Vulnerability of Layer 5a Corticostriatal Neurons in Huntington's Disease.
- Author
-
Pressl C, Mätlik K, Kus L, Darnell P, Luo JD, Paul MR, Weiss AR, Liguore W, Carroll TS, Davis DA, McBride J, and Heintz N
- Abstract
The properties of the cell types that are selectively vulnerable in Huntington's disease (HD) cortex, the nature of somatic CAG expansions of mHTT in these cells, and their importance in CNS circuitry have not been delineated. Here we employed serial fluorescence activated nuclear sorting (sFANS), deep molecular profiling, and single nucleus RNA sequencing (snRNAseq) to demonstrate that layer 5a pyramidal neurons are vulnerable in primary motor cortex and other cortical areas of HD donors. Extensive mHTT -CAG expansions occur in vulnerable layer 5a pyramidal cells, and in Betz cells, layer 6a, layer 6b neurons that are resilient in HD. Retrograde tracing experiments in macaque brains identify the vulnerable layer 5a neurons as corticostriatal pyramidal cells. We propose that enhanced somatic mHTT -CAG expansion and altered synaptic function act together to cause corticostriatal disconnection and selective neuronal vulnerability in the HD cerebral cortex.
- Published
- 2023
- Full Text
- View/download PDF
13. Altered chromatin occupancy of patient-associated H4 mutants misregulate neuronal differentiation.
- Author
-
Feng L, Barrows D, Zhong L, Mätlik K, Porter EG, Djomo AM, Yau I, Soshnev AA, Carroll TS, Wen D, Hatten ME, Garcia BA, and Allis CD
- Abstract
Chromatin is a crucial regulator of gene expression and tightly controls development across species. Mutations in only one copy of multiple histone genes were identified in children with developmental disorders characterized by microcephaly, but their mechanistic roles in development remain unclear. Here we focus on dominant mutations affecting histone H4 lysine 91. These H4K91 mutants form aberrant nuclear puncta at specific heterochromatin regions. Mechanistically, H4K91 mutants demonstrate enhanced binding to the histone variant H3.3, and ablation of H3.3 or the H3.3-specific chaperone DAXX diminishes the mutant localization to chromatin. Our functional studies demonstrate that H4K91 mutant expression increases chromatin accessibility, alters developmental gene expression through accelerating pro-neural differentiation, and causes reduced mouse brain size in vivo , reminiscent of the microcephaly phenotypes of patients. Together, our studies unveil a distinct molecular pathogenic mechanism from other known histone mutants, where H4K91 mutants misregulate cell fate during development through abnormal genomic localization., Competing Interests: Declaration of Interests The authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
14. Histone bivalency regulates the timing of cerebellar granule cell development.
- Author
-
Mätlik K, Govek EE, Paul MR, Allis CD, and Hatten ME
- Subjects
- Animals, Mice, Transcriptional Activation, Cell Differentiation genetics, Histones metabolism, Epigenesis, Genetic
- Abstract
Developing neurons undergo a progression of morphological and gene expression changes as they transition from neuronal progenitors to mature neurons. Here we used RNA-seq and H3K4me3 and H3K27me3 ChIP-seq to analyze how chromatin modifications control gene expression in a specific type of CNS neuron: the mouse cerebellar granule cell (GC). We found that in proliferating GC progenitors (GCPs), H3K4me3/H3K27me3 bivalency is common at neuronal genes and undergoes dynamic changes that correlate with gene expression during migration and circuit formation. Expressing a fluorescent sensor for bivalent domains revealed subnuclear bivalent foci in proliferating GCPs. Inhibiting H3K27 methyltransferases EZH1 and EZH2 in vitro and in organotypic cerebellar slices dramatically altered the expression of bivalent genes, induced the down-regulation of migration-related genes and up-regulation of synaptic genes, inhibited glial-guided migration, and accelerated terminal differentiation. Thus, histone bivalency is required to regulate the timing of the progression from progenitor cells to mature neurons., (© 2023 Mätlik et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2023
- Full Text
- View/download PDF
15. Cell Type Specific CAG Repeat Expansions and Toxicity of Mutant Huntingtin in Human Striatum and Cerebellum.
- Author
-
Mätlik K, Baffuto M, Kus L, Deshmukh AL, Davis DA, Paul MR, Carroll TS, Caron MC, Masson JY, Pearson CE, and Heintz N
- Abstract
Brain region-specific degeneration and somatic expansions of the mutant Huntingtin (mHTT) CAG tract are key features of Huntington's disease (HD). However, the relationships between CAG expansions, death of specific cell types, and molecular events associated with these processes are not established. Here we employed fluorescence-activated nuclear sorting (FANS) and deep molecular profiling to gain insight into the properties of cell types of the human striatum and cerebellum in HD and control donors. CAG expansions arise in striatal medium spiny neurons (MSNs) and cholinergic interneurons, in cerebellar Purkinje neurons, and at mATXN3 in MSNs from SCA3 donors. CAG expansions in MSNs are associated with higher levels of MSH2 and MSH3 (forming MutSβ), which can inhibit nucleolytic excision of CAG slip-outs by FAN1 in a concentration-dependent manner. Our data indicate that ongoing CAG expansions are not sufficient for cell death, and identify transcriptional changes associated with somatic CAG expansions and striatal toxicity.
- Published
- 2023
- Full Text
- View/download PDF
16. In vivo modulation of endogenous gene expression via CRISPR/Cas9-mediated 3'UTR editing.
- Author
-
Mätlik K, Olfat S, Cowlishaw MC, Moreno ED, Ollila S, and Andressoo JO
- Abstract
The 3' untranslated regions (UTRs) modulate gene expression levels by regulating mRNA stability and translation. We previously showed that the replacement of the negative regulatory elements from the 3'UTR of glial cell line-derived neurotrophic factor (GDNF) resulted in increased endogenous GDNF expression while retaining its normal spatiotemporal expression pattern. Here, we have developed a methodology for the generation of in vivo hyper- and hypomorphic alleles via 3'UTR targeting using the CRISPR/Cas9 system. We demonstrate that CRISPR/Cas9-mediated excision of a long inhibitory sequence from Gdnf native 3'UTR in mouse zygotes increases the levels of endogenous GDNF with similar phenotypic alterations in embryonic kidney development as we described in GDNF constitutive and conditional hypermorphic mice. Furthermore, we show that CRISPR/Cas9-mediated targeting of 3'UTRs in vivo allows the modulation of the expression levels of two other morphogens, Gdf11 and Bdnf . Together, our work demonstrates the power of in vivo 3'UTR editing using the CRISPR/Cas9 system to create hyper- and hypomorphic alleles, suggesting wide applicability in studies on gene function and potentially, in gene therapy., Competing Interests: The authors declare that they do not have competing interests., (© 2023 Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
17. Increased Physiological GDNF Levels Have No Effect on Dopamine Neuron Protection and Restoration in a Proteasome Inhibition Mouse Model of Parkinson's Disease.
- Author
-
Olfat S, Mätlik K, Kopra JJ, Garton DR, Iivanainen VH, Bhattacharya D, Jakobsson J, Piepponen TP, and Andressoo JO
- Subjects
- Mice, Animals, Glial Cell Line-Derived Neurotrophic Factor metabolism, Dopamine metabolism, Neuroprotection, Proteasome Endopeptidase Complex metabolism, Dopaminergic Neurons metabolism, Disease Models, Animal, Parkinson Disease drug therapy, Parkinson Disease metabolism, Neurodegenerative Diseases metabolism
- Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease that comprises a range of motor and nonmotor symptoms. Glial cell line-derived neurotrophic factor (GDNF) promotes the survival of dopamine neurons in vitro and in vivo , and intracranial delivery of GDNF has been tested in six clinical trials for treating PD. However, clinical trials with ectopic GDNF have yielded variable results, which could in part result from abnormal expression site and levels caused by ectopic overexpression. Therefore, an important open question is whether an increase in endogenous GDNF expression could be potent in reversing PD progression. Here, we tested the therapeutic potential of endogenous GDNF using mice in which endogenous GDNF can be conditionally upregulated specifically in cells that express GDNF naturally (conditional GDNF hypermorphic mice; Gdnf
cHyper ). We analyzed the impact of endogenous GDNF upregulation in both neuroprotection and neurorestoration procedures, and for both motor and nonmotor symptoms in the proteasome inhibitor lactacystin (LC) model of PD. Our results showed that upregulation of endogenous GDNF in the adult striatum is not protective in LC-induced PD model in mice. Since age is the largest risk factor for PD, we also analyzed the effect of deletion of endogenous GDNF in aged Gdnf conditional knock-out mice. We found that GDNF deletion does not increase susceptibility to LC-induced damage. We conclude that endogenous GDNF does not impact the outcome in the LC-induced proteasome inhibition mouse model of Parkinson's disease., Competing Interests: The authors declare no competing financial interests., (Copyright © 2023 Olfat et al.)- Published
- 2023
- Full Text
- View/download PDF
18. The effects of chronic high-dose morphine on microgliosis and the microglial transcriptome in rat spinal cord.
- Author
-
Ahlström FH, Viisanen H, Karhinen L, Mätlik K, Blomqvist KJ, Lilius TO, Sidorova YA, Palada V, Rauhala PV, and Kalso EA
- Subjects
- Rats, Male, Female, Animals, Microglia, Transcriptome genetics, Analgesics pharmacology, Pain metabolism, Spinal Cord metabolism, Morphine therapeutic use, Analgesics, Opioid pharmacology, Analgesics, Opioid therapeutic use
- Abstract
Background: Opioids are efficacious and safe analgesic drugs in short-term use for acute pain but chronic use can lead to tolerance and dependence. Opioid-induced microglial activation may contribute to the development of tolerance and this process may differ between males and females. A link is suggested between this microglial activation and inflammation, disturbances of circadian rhythms, and neurotoxic effects. We set out to further delineate the effects of chronic morphine on pain behaviour, microglial and neuronal staining, and the transcriptome of spinal microglia, to better understand the role of microglia in the consequences of long-term high-dose opioid administration. Experimental Approach: In two experiments, we administered increasing subcutaneous doses of morphine hydrochloride or saline to male and female rats. Thermal nociception was assessed with the tail flick and hot plate tests. In Experiment I, spinal cord (SC) samples were prepared for immunohistochemical staining for microglial and neuronal markers. In Experiment II, the transcriptome of microglia from the lumbar SC was analysed. Key Results: Female and male rats had similar antinociceptive responses to morphine and developed similar antinociceptive tolerance to thermal stimuli following chronic increasing high doses of s.c. morphine. The area of microglial IBA1-staining in SC decreased after 2 weeks of morphine administration in both sexes. Following morphine treatment, the differentially expressed genes identified in the microglial transcriptome included ones related to the circadian rhythm , apoptosis, and immune system processes. Conclusions: Female and male rats showed similar pain behaviour following chronic high doses of morphine. This was associated with decreased staining of spinal microglia, suggesting either decreased activation or apoptosis. High-dose morphine administration also associated with several changes in gene expression in SC microglia, e.g., those related to the circadian rhythm ( Per2, Per3, Dbp ). These changes should be considered in the clinical consequences of long-term high-dose administration of opioids.
- Published
- 2023
- Full Text
- View/download PDF
19. Elevated endogenous GDNF induces altered dopamine signalling in mice and correlates with clinical severity in schizophrenia.
- Author
-
Mätlik K, Garton DR, Montaño-Rodríguez AR, Olfat S, Eren F, Casserly L, Damdimopoulos A, Panhelainen A, Porokuokka LL, Kopra JJ, Turconi G, Schweizer N, Bereczki E, Piehl F, Engberg G, Cervenka S, Piepponen TP, Zhang FP, Sipilä P, Jakobsson J, Sellgren CM, Erhardt S, and Andressoo JO
- Subjects
- Animals, Mice, Glial Cell Line-Derived Neurotrophic Factor metabolism, Glial Cell Line-Derived Neurotrophic Factor pharmacology, Corpus Striatum metabolism, Signal Transduction, Dopamine metabolism, Schizophrenia metabolism
- Abstract
Presynaptic increase in striatal dopamine is the primary dopaminergic abnormality in schizophrenia, but the underlying mechanisms are not understood. Here, we hypothesized that increased expression of endogenous GDNF could induce dopaminergic abnormalities that resemble those seen in schizophrenia. To test the impact of GDNF elevation, without inducing adverse effects caused by ectopic overexpression, we developed a novel in vivo approach to conditionally increase endogenous GDNF expression. We found that a 2-3-fold increase in endogenous GDNF in the brain was sufficient to induce molecular, cellular, and functional changes in dopamine signalling in the striatum and prefrontal cortex, including increased striatal presynaptic dopamine levels and reduction of dopamine in prefrontal cortex. Mechanistically, we identified adenosine A2a receptor (A
2A R), a G-protein coupled receptor that modulates dopaminergic signalling, as a possible mediator of GDNF-driven dopaminergic abnormalities. We further showed that pharmacological inhibition of A2A R with istradefylline partially normalised striatal GDNF and striatal and cortical dopamine levels in mice. Lastly, we found that GDNF levels are increased in the cerebrospinal fluid of first episode psychosis patients, and in post-mortem striatum of schizophrenia patients. Our results reveal a possible contributor for increased striatal dopamine signalling in a subgroup of schizophrenia patients and suggest that GDNF-A2A R crosstalk may regulate dopamine function in a therapeutically targetable manner., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
20. Antagonism of peripheral opioid receptors by methylnaltrexone does not prevent morphine tolerance in rats.
- Author
-
Blomqvist KJ, Dudek KA, Viisanen H, Mätlik K, Ahlström FHG, Laitila J, Kalso EA, Rauhala PV, and Lilius TO
- Subjects
- Analgesics, Opioid pharmacology, Animals, Dose-Response Relationship, Drug, Drug Tolerance, Male, Narcotic Antagonists pharmacology, Quaternary Ammonium Compounds, Rats, Rats, Sprague-Dawley, Receptors, Opioid, Receptors, Opioid, mu, Morphine pharmacology, Naltrexone analogs & derivatives, Naltrexone pharmacology
- Abstract
Opioids are effective analgesics in the management of severe pain. However, tolerance, leading to dose escalation and adverse effects are significant limiting factors in their use. The role of peripheral opioid receptors in analgesia has been discussed especially under inflammatory conditions. The results from pharmacological and conditional knockout studies together do not provide a clear picture of the contribution of peripheral opioid receptors on antinociceptive tolerance and this needs to be evaluated. Therefore, we studied whether the peripherally restricted opioid receptor antagonist, methylnaltrexone (MNTX), could prevent morphine tolerance without attenuating the antinociceptive effect of morphine. Male Sprague-Dawley rats were treated for 7 days with increasing subcutaneous doses of morphine (5-30 mg/kg) and were coadministered saline, MNTX (0.5 or 2 mg/kg), or naltrexone (NTX; 2 mg/kg). Nociception was assessed with tail-flick, hotplate, and von Frey tests. Morphine, MNTX, and NTX concentrations in the plasma, brain, and spinal cord were measured by liquid chromatography-tandem mass spectrometry. In acute coadministration, NTX, but not MNTX, abolished the acute antinociceptive effects of morphine in all nociceptive tests. The antinociceptive tolerance after repeated morphine administration was also prevented by NTX but not by MNTX. MNTX penetrated to the spinal cord and the brain to some extent after repeated administration. The results do not support the use of MNTX for preventing opioid tolerance and also suggest that morphine tolerance is mediated by central rather than peripheral opioid receptors in the rat., (© 2020 The Authors. Journal of Neuroscience Research published by Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
21. Increased Endogenous GDNF in Mice Protects Against Age-Related Decline in Neuronal Cholinergic Markers.
- Author
-
Mitra S, Turconi G, Darreh-Shori T, Mätlik K, Aquilino M, Eriksdotter M, and Andressoo JO
- Abstract
Gradual decline in cholinergic transmission and cognitive function occurs during normal aging, whereas pathological loss of cholinergic function is a hallmark of different types of dementia, including Alzheimer's disease (AD), Lewy body dementia (LBD), and Parkinson's disease dementia (PDD). Glial cell line-derived neurotrophic factor (GDNF) is known to modulate and enhance the dopamine system. However, how endogenous GDNF influences brain cholinergic transmission has remained elusive. In this study, we explored the effect of a twofold increase in endogenous GDNF (Gdnf hypermorphic mice, Gdnf
wt/hyper ) on cholinergic markers and cognitive function upon aging. We found that Gdnfwt/hyper mice resisted an overall age-associated decline in the cholinergic index observed in the brain of Gdnfwt/wt animals. Biochemical analysis revealed that the level of nerve growth factor (NGF), which is important for survival and function of central cholinergic neurons, was significantly increased in several brain areas of old Gdnfwt/hyper mice. Analysis of expression of genes involved in cholinergic transmission in the cortex and striatum confirmed modulation of cholinergic pathways by GDNF upon aging. In line with these findings, Gdnfwt/hyper mice did not undergo an age-related decline in cognitive function in the Y-maze test, as observed in the wild type littermates. Our results identify endogenous GDNF as a potential modulator of cholinergic transmission and call for future studies on endogenous GDNF function in neurodegenerative disorders characterized by cognitive impairments, including AD, LBD, and PDD., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Mitra, Turconi, Darreh-Shori, Mätlik, Aquilino, Eriksdotter and Andressoo.)- Published
- 2021
- Full Text
- View/download PDF
22. Postnatal prolongation of mammalian nephrogenesis by excess fetal GDNF.
- Author
-
Li H, Kurtzeborn K, Kupari J, Gui Y, Siefker E, Lu B, Mätlik K, Olfat S, Montaño-Rodríguez AR, Huh SH, Costantini F, Andressoo JO, and Kuure S
- Subjects
- Animals, Axin Protein metabolism, Cell Differentiation genetics, Glial Cell Line-Derived Neurotrophic Factor genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Stem Cells cytology, Wnt Proteins metabolism, Glial Cell Line-Derived Neurotrophic Factor metabolism, Nephrons embryology, Nephrons growth & development, Organogenesis genetics, Wnt Signaling Pathway genetics
- Abstract
Nephron endowment, defined during the fetal period, dictates renal and related cardiovascular health throughout life. We show here that, despite its negative effects on kidney growth, genetic increase of GDNF prolongs the nephrogenic program beyond its normal cessation. Multi-stage mechanistic analysis revealed that excess GDNF maintains nephron progenitors and nephrogenesis through increased expression of its secreted targets and augmented WNT signaling, leading to a two-part effect on nephron progenitor maintenance. Abnormally high GDNF in embryonic kidneys upregulates its known targets but also Wnt9b and Axin2, with concomitant deceleration of nephron progenitor proliferation. Decline of GDNF levels in postnatal kidneys normalizes the ureteric bud and creates a permissive environment for continuation of the nephrogenic program, as demonstrated by morphologically and molecularly normal postnatal nephron progenitor self-renewal and differentiation. These results establish that excess GDNF has a bi-phasic effect on nephron progenitors in mice, which can faithfully respond to GDNF dosage manipulation during the fetal and postnatal period. Our results suggest that sensing the signaling activity level is an important mechanism through which GDNF and other molecules contribute to nephron progenitor lifespan specification., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2021. Published by The Company of Biologists Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
23. Mitoxantrone, pixantrone and mitoxantrone (2-hydroxyethyl)piperazine are toll-like receptor 4 antagonists, inhibit NF-κB activation, and decrease TNF-alpha secretion in primary microglia.
- Author
-
Rinne M, Mätlik K, Ahonen T, Vedovi F, Zappia G, Moreira VM, Yli-Kauhaluoma J, Leino S, Salminen O, Kalso E, Airavaara M, and Xhaard H
- Subjects
- Microglia, Mitoxantrone pharmacology, Tumor Necrosis Factor-alpha drug effects, Tumor Necrosis Factor-alpha metabolism, Isoquinolines pharmacology, NF-kappa B drug effects, Piperazine pharmacology, Toll-Like Receptor 4 antagonists & inhibitors
- Abstract
Toll-like receptor 4 (TLR4) recognizes various endogenous and microbial ligands and is an essential part in the innate immune system. TLR4 signaling initiates transcription factor NF-κB and production of proinflammatory cytokines. TLR4 contributes to the development or progression of various diseases including stroke, neuropathic pain, multiple sclerosis, rheumatoid arthritis and cancer, and better therapeutics are currently sought for these conditions. In this study, a library of 140 000 compounds was virtually screened and a resulting hit-list of 1000 compounds was tested using a cellular reporter system. The topoisomerase II inhibitor mitoxantrone and its analogues pixantrone and mitoxantrone (2-hydroxyethyl)piperazine were identified as inhibitors of TLR4 and NF-κB activation. Mitoxantrone was shown to bind directly to the TLR4, and pixantrone and mitoxantrone (2-hydroxyethyl)piperazine were shown to inhibit the production of proinflammatory cytokines such as tumor necrosis factor alpha (TNFα) in primary microglia. The inhibitory effect on NF-κB activation or on TNFα production was not mediated through cytotoxity at ≤ 1 µM concentration for pixantrone and mitoxantrone (2-hydroxyethyl)piperazine treated cells, as assessed by ATP counts. This study thus identifies a new mechanism of action for mitoxantrone, pixantrone, and mitoxantrone (2-hydroxyethyl)piperazine through the TLR4., Competing Interests: Declarations of Competing Interest None., (Copyright © 2020. Published by Elsevier B.V.)
- Published
- 2020
- Full Text
- View/download PDF
24. Molecular profile of the rat peri-infarct region four days after stroke: Study with MANF.
- Author
-
Teppo J, Vaikkinen A, Stratoulias V, Mätlik K, Anttila JE, Smolander OP, Pöhö P, Harvey BK, Kostiainen R, and Airavaara M
- Subjects
- Animals, Brain Ischemia genetics, Brain Ischemia metabolism, Brain Ischemia pathology, Cerebral Infarction metabolism, Cerebral Infarction pathology, Gene Transfer Techniques, Male, Metabolomics methods, Nerve Growth Factors administration & dosage, Rats, Rats, Sprague-Dawley, Stroke metabolism, Stroke pathology, Time Factors, Cerebral Infarction genetics, Nerve Growth Factors genetics, Nerve Growth Factors metabolism, Proteomics methods, Stroke genetics, Transcriptome genetics
- Abstract
The peri-infarct region after ischemic stroke is the anatomical location for many of the endogenous recovery processes; however, -the molecular events in the peri-infarct region remain poorly characterized. In this study, we examine the molecular profile of the peri-infarct region on post-stroke day four, a time when reparative processes are ongoing. We used a multiomics approach, involving RNA sequencing, and mass spectrometry-based proteomics and metabolomics to characterize molecular changes in the peri-infarct region. We also took advantage of our previously developed method to express transgenes in the peri-infarct region where self-complementary adeno-associated virus (AAV) vectors were injected into the brain parenchyma on post-stroke day 2. We have previously used this method to show that mesencephalic astrocyte-derived neurotrophic factor (MANF) enhances functional recovery from stroke and recruits phagocytic cells to the peri-infarct region. Here, we first analyzed the effects of stroke to the peri-infarct region on post-stroke day 4 in comparison to sham-operated animals, finding that strokeinduced changes in 3345 transcripts, 341 proteins, and 88 metabolites. We found that after stroke, genes related to inflammation, proliferation, apoptosis, and regeneration were upregulated, whereas genes encoding neuroactive ligand receptors and calcium-binding proteins were downregulated. In proteomics, we detected upregulation of proteins related to protein synthesis and downregulation of neuronal proteins. Metabolomic studies indicated that in after stroke tissue there is an increase in saccharides, sugar phosphates, ceramides and free fatty acids and a decrease of adenine, hypoxantine, adenosine and guanosine. We then compared the effects of post-stroke delivery of AAV1-MANF to AAV1-eGFP (enhanced green fluorescent protein). MANF administration increased the expression of 77 genes, most of which were related to immune response. In proteomics, MANF administration reduced S100A8 and S100A9 protein levels. In metabolomics, no significant differences between MANF and eGFP treatment were detected, but relative to sham surgery group, most of the changes in lipids were significant in the AAV-eGFP group only. This work describes the molecular profile of the peri-infarct region during recovery from ischemic stroke, and establishes a resource for further stroke studies. These results provide further support for parenchymal MANF as a modulator of phagocytic function., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
25. Cerebral Dopamine Neurotrophic Factor Diffuses Around the Brainstem and Does Not Undergo Anterograde Transport After Injection to the Substantia Nigra.
- Author
-
Albert K, Renko JM, Mätlik K, Airavaara M, and Voutilainen MH
- Abstract
Cerebral dopamine neurotrophic factor (CDNF) has shown therapeutic potential in rodent and non-human primate models of Parkinson's disease by protecting the dopamine neurons from degeneration and even restoring their phenotype and function. Previously, neurorestorative efficacy of CDNF in the 6-hydroxydopamine (6-OHDA) model of Parkinson's disease as well as diffusion of the protein in the striatum (STR) has been demonstrated and studied. Here, experiments were performed to characterize the diffusion and transport of supra-nigral CDNF in non-lesioned rats. We injected recombinant human CDNF to the substantia nigra (SN) of naïve male Wistar rats and analyzed the brains 2, 6, and 24 h after injections. We performed immunohistochemical stainings using an antibody specific to human CDNF and radioactivity measurements after injecting iodinated CDNF. Unlike the previously reported striatonigral retrograde transport seen after striatal injection, active anterograde transport of CDNF to the STR could not be detected after nigral injection. There was, however, clear diffusion of CDNF to the brain areas surrounding the SN, and CDNF colocalized with tyrosine hydroxylase (TH)-positive neurons. Overall, our results provide insight on how CDNF injected to the SN may act in this region of the brain.
- Published
- 2019
- Full Text
- View/download PDF
26. Two-fold elevation of endogenous GDNF levels in mice improves motor coordination without causing side-effects.
- Author
-
Mätlik K, Võikar V, Vilenius C, Kulesskaya N, and Andressoo JO
- Subjects
- Animals, Corpus Striatum metabolism, Dopamine metabolism, Dopamine Plasma Membrane Transport Proteins metabolism, Dopaminergic Neurons metabolism, Female, Male, Mesencephalon metabolism, Mice, Parkinson Disease metabolism, Behavior, Animal physiology, Glial Cell Line-Derived Neurotrophic Factor metabolism, Motor Activity physiology
- Abstract
Glial cell line-derived neurotrophic factor (GDNF) promotes the survival of dopaminergic neurons in vitro and in vivo. For this reason, GDNF is currently in clinical trials for the treatment of Parkinson's disease (PD). However, how endogenous GDNF influences dopamine system function and animal behavior is not fully understood. We recently generated GDNF hypermorphic mice that express increased levels of endogenous GDNF from the native locus, resulting in augmented function of the nigrostriatal dopamine system. Specifically, Gdnf
wt/hyper mice have a mild increase in striatal and midbrain dopamine levels, increased dopamine transporter activity, and 15% increased numbers of midbrain dopamine neurons and striatal dopaminergic varicosities. Since changes in the dopamine system are implicated in several neuropsychiatric diseases, including schizophrenia, attention deficit hyperactivity disorder (ADHD) and depression, and ectopic GDNF delivery associates with side-effects in PD models and clinical trials, we further investigated Gdnfwt/hyper mice using 20 behavioral tests. Despite increased dopamine levels, dopamine release and dopamine transporter activity, there were no differences in psychiatric disease related phenotypes. However, compared to controls, male Gdnfwt/hyper mice performed better in tests measuring motor function. Therefore, a modest elevation of endogenous GDNF levels improves motor function but does not induce adverse behavioral outcomes.- Published
- 2018
- Full Text
- View/download PDF
27. Poststroke delivery of MANF promotes functional recovery in rats.
- Author
-
Mätlik K, Anttila JE, Kuan-Yin T, Smolander OP, Pakarinen E, Lehtonen L, Abo-Ramadan U, Lindholm P, Zheng C, Harvey B, Arumäe U, Lindahl M, and Airavaara M
- Subjects
- Animals, Behavior, Animal, Brain Ischemia complications, Dependovirus genetics, Disease Models, Animal, Gene Expression, Genetic Vectors genetics, Humans, Magnetic Resonance Imaging, Male, Nerve Growth Factors metabolism, Rats, Stroke diagnosis, Stroke etiology, Transduction, Genetic, Transgenes, Astrocytes metabolism, Nerve Growth Factors genetics, Stroke metabolism, Stroke Rehabilitation
- Abstract
Stroke is the most common cause of adult disability in developed countries, largely because spontaneous recovery is often incomplete, and no pharmacological means to hasten the recovery exist. It was recently shown that mesencephalic astrocyte-derived neurotrophic factor (MANF) induces alternative or M2 activation of immune cells after retinal damage in both fruit fly and mouse and mediates retinal repair. Therefore, we set out to study whether poststroke MANF administration would enhance brain tissue repair and affect behavioral recovery of rats after cerebral ischemic injury. We used the distal middle cerebral artery occlusion (dMCAo) model of ischemia-reperfusion injury and administered MANF either as a recombinant protein or via adeno-associated viral (AAV) vector. We discovered that, when MANF was administered to the peri-infarct region 2 or 3 days after stroke, it promoted functional recovery of the animals without affecting the lesion volume. Further, AAV7-MANF treatment transiently increased the number of phagocytic macrophages in the subcortical peri-infarct regions. In addition, the analysis of knockout mice revealed the neuroprotective effects of endogenous MANF against ischemic injury, although endogenous MANF had no effect on immune cell-related gene expression. The beneficial effect of MANF treatment on the reversal of stroke-induced behavioral deficits implies that MANF-based therapies could be used for the repair of brain tissue after stroke.
- Published
- 2018
- Full Text
- View/download PDF
28. Synthesis of 7β-hydroxy-8-ketone opioid derivatives with antagonist activity at mu- and delta-opioid receptors.
- Author
-
Ahonen TJ, Rinne M, Grutschreiber P, Mätlik K, Airavaara M, Schaarschmidt D, Lang H, Reiss D, Xhaard H, Gaveriaux-Ruff C, Yli-Kauhaluoma J, and Moreira VM
- Subjects
- Analgesics, Opioid chemical synthesis, HEK293 Cells, Humans, Ketones chemical synthesis, Ketones chemistry, Ketones pharmacology, Models, Molecular, Narcotic Antagonists chemical synthesis, Receptors, Opioid, delta metabolism, Receptors, Opioid, mu metabolism, Structure-Activity Relationship, Analgesics, Opioid chemistry, Analgesics, Opioid pharmacology, Narcotic Antagonists chemistry, Narcotic Antagonists pharmacology, Receptors, Opioid, delta antagonists & inhibitors, Receptors, Opioid, mu antagonists & inhibitors
- Abstract
Despite extensive years of research, the direct oxidation of the 7,8-double bond of opioids has so far received little attention and knowledge about the effects of this modification on activity at the different opioid receptors is scarce. We herein report that potassium permanganate supported on iron(II) sulfate heptahydrate can be used as a convenient oxidant in the one-step, heterogeneous conversion of Δ
7,8 -opioids to the corresponding 7β-hydroxy-8-ketones. Details of the reaction mechanism are given and the effects of the substituent at position 6 of several opioids on the reaction outcome is discussed. The opioid hydroxy ketones prepared are antagonists at the mu- and delta-opioid receptors. Docking simulations and detailed structure-activity analysis revealed that the presence of the 7β-hydroxy-8-ketone functionality in the prepared compounds can be used to gain activity towards the delta opioid receptor. The 7β-hydroxy-8-ketones prepared with this method can also be regarded as versatile intermediates for the synthesis of other opioids of interest., (Copyright © 2018 Elsevier Masson SAS. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
29. Post-stroke Intranasal (+)-Naloxone Delivery Reduces Microglial Activation and Improves Behavioral Recovery from Ischemic Injury.
- Author
-
Anttila JE, Albert K, Wires ES, Mätlik K, Loram LC, Watkins LR, Rice KC, Wang Y, Harvey BK, and Airavaara M
- Subjects
- Administration, Intranasal, Animals, Behavior, Animal drug effects, Disease Models, Animal, Naloxone administration & dosage, Narcotic Antagonists administration & dosage, Rats, Rats, Sprague-Dawley, Brain Ischemia drug therapy, Cerebral Cortex drug effects, Corpus Striatum drug effects, Macrophage Activation drug effects, Microglia drug effects, Naloxone pharmacology, Narcotic Antagonists pharmacology, Stroke drug therapy, Thalamus drug effects
- Abstract
Ischemic stroke is the leading cause of disability, and effective therapeutic strategies are needed to promote complete recovery. Neuroinflammation plays a significant role in stroke pathophysiology, and there is limited understanding of how it affects recovery. The aim of this study was to characterize the spatiotemporal expression profile of microglial activation and whether dampening microglial/macrophage activation post-stroke facilitates the recovery. For dampening microglial/macrophage activation, we chose intranasal administration of naloxone, a drug that is already in clinical use for opioid overdose and is known to decrease microglia/macrophage activation. We characterized the temporal progression of microglia/macrophage activation following cortical ischemic injury in rat and found the peak activation in cortex 7 d post-stroke. Unexpectedly, there was a chronic expression of phagocytic cells in the thalamus associated with neuronal loss. (+)-Naloxone, an enantiomer that reduces microglial activation without antagonizing opioid receptors, was administered intranasally starting 1 d post-stroke and continuing for 7 d. (+)-Naloxone treatment decreased microglia/macrophage activation in the striatum and thalamus, promoted behavioral recovery during the 14-d monitoring period, and reduced neuronal death in the lesioned cortex and ipsilateral thalamus. Our results are the first to show that post-stroke intranasal (+)-naloxone administration promotes short-term functional recovery and reduces microglia/macrophage activation. Therefore, (+)-naloxone is a promising drug for the treatment of ischemic stroke, and further studies should be conducted.
- Published
- 2018
- Full Text
- View/download PDF
30. Differential Spinal and Supraspinal Activation of Glia in a Rat Model of Morphine Tolerance.
- Author
-
Jokinen V, Sidorova Y, Viisanen H, Suleymanova I, Tiilikainen H, Li Z, Lilius TO, Mätlik K, Anttila JE, Airavaara M, Tian L, Rauhala PV, and Kalso EA
- Subjects
- Animals, Brain metabolism, Brain pathology, Drug Tolerance, Hyperalgesia metabolism, Hyperalgesia pathology, Male, Models, Animal, Neuroglia metabolism, Neuroglia pathology, Nociceptive Pain drug therapy, Nociceptive Pain metabolism, Nociceptive Pain pathology, Rats, Sprague-Dawley, Spinal Cord metabolism, Spinal Cord pathology, Transcriptome drug effects, Analgesics, Opioid pharmacology, Brain drug effects, Morphine pharmacology, Neuroglia drug effects, Spinal Cord drug effects
- Abstract
Development of tolerance is a well known pharmacological characteristic of opioids and a major clinical problem. In addition to the known neuronal mechanisms of opioid tolerance, activation of glia has emerged as a potentially significant new mechanism. We studied activation of microglia and astrocytes in morphine tolerance and opioid-induced hyperalgesia in rats using immunohistochemistry, flow cytometry and RNA sequencing in spinal- and supraspinal regions. Chronic morphine treatment that induced tolerance and hyperalgesia also increased immunoreactivity of spinal microglia in the dorsal and ventral horns. Flow cytometry demonstrated that morphine treatment increased the proportion of M2-polarized spinal microglia, but failed to impact the number or the proportion of M1-polarized microglia. In the transcriptome of microglial cells isolated from the spinal cord (SC), morphine treatment increased transcripts related to cell activation and defense response. In the studied brain regions, no activation of microglia or astrocytes was detected by immunohistochemistry, except for a decrease in the number of microglial cells in the substantia nigra. In flow cytometry, morphine caused a decrease in the number of microglial cells in the medulla, but otherwise no change was detected for the count or the proportion of M1- and M2-polarized microglia in the medulla or sensory cortex. No evidence for the activation of glia in the brain was seen. Our results suggest that glial activation associated with opioid tolerance and opioid-induced hyperalgesia occurs mainly at the spinal level. The transcriptome data suggest that the microglial activation pattern after chronic morphine treatment has similarities with that of neuropathic pain., (Copyright © 2018 IBRO. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
31. Intrastriatally Infused Exogenous CDNF Is Endocytosed and Retrogradely Transported to Substantia Nigra.
- Author
-
Mätlik K, Vihinen H, Bienemann A, Palgi J, Voutilainen MH, Booms S, Lindahl M, Jokitalo E, Saarma M, Huttunen HJ, Airavaara M, and Arumäe U
- Subjects
- Adrenergic Agents pharmacology, Animals, Corpus Striatum drug effects, Dopaminergic Neurons metabolism, Dopaminergic Neurons ultrastructure, Endocytosis drug effects, Humans, Male, Mice, Microscopy, Immunoelectron, Nerve Growth Factors metabolism, Nerve Growth Factors ultrastructure, Neural Pathways drug effects, Neural Pathways physiology, Oxidopamine pharmacology, Phosphopyruvate Hydratase metabolism, Protein Transport drug effects, Protein Transport genetics, Rats, Rats, Transgenic, Rats, Wistar, Substantia Nigra drug effects, Time Factors, Tyrosine 3-Monooxygenase metabolism, Corpus Striatum metabolism, Endocytosis physiology, Nerve Growth Factors administration & dosage, Substantia Nigra metabolism
- Abstract
Cerebral dopamine neurotrophic factor (CDNF) protects the nigrostriatal dopaminergic (DA) neurons in rodent models of Parkinson's disease and restores DA circuitry when delivered after these neurons have begun to degenerate. These DA neurons have been suggested to transport striatal CDNF retrogradely to the substantia nigra (SN). However, in cultured cells the binding and internalization of extracellular CDNF has not been reported. The first aim of this study was to examine the cellular localization and pharmacokinetic properties of recombinant human CDNF (rhCDNF) protein after its infusion into rat brain parenchyma. Second, we aimed to study whether the transport of rhCDNF from the striatum to the SN results from its retrograde transport via DA neurons or from its anterograde transport via striatal GABAergic projection neurons. We show that after intrastriatal infusion, rhCDNF diffuses rapidly and broadly, and is cleared with a half-life of 5.5 h. Confocal microscopy analysis of brain sections at 2 and 6 h after infusion of rhCDNF revealed its widespread unspecific internalization by cortical and striatal neurons, exhibiting different patterns of subcellular rhCDNF distribution. Electron microscopy analysis showed that rhCDNF is present inside the endosomes and multivesicular bodies. In addition, we present data that after intrastriatal infusion the rhCDNF found in the SN is almost exclusively localized to the DA neurons, thus showing that it is retrogradely transported.
- Published
- 2017
- Full Text
- View/download PDF
32. Developing therapeutically more efficient Neurturin variants for treatment of Parkinson's disease.
- Author
-
Runeberg-Roos P, Piccinini E, Penttinen AM, Mätlik K, Heikkinen H, Kuure S, Bespalov MM, Peränen J, Garea-Rodríguez E, Fuchs E, Airavaara M, Kalkkinen N, Penn R, and Saarma M
- Subjects
- Amphetamine pharmacology, Animals, CHO Cells, Cricetulus, Disease Models, Animal, Humans, Macaca fascicularis, Male, Models, Molecular, Neurturin genetics, Oxidopamine toxicity, Parkinson Disease complications, Parkinson Disease etiology, Proto-Oncogene Proteins c-ret genetics, Proto-Oncogene Proteins c-ret metabolism, Rats, Rats, Wistar, Stereotyped Behavior drug effects, Sympatholytics toxicity, Tyrosine 3-Monooxygenase metabolism, Drug Design, Genetic Variation genetics, Neurturin chemistry, Neurturin metabolism, Parkinson Disease drug therapy, Parkinson Disease metabolism
- Abstract
In Parkinson's disease midbrain dopaminergic neurons degenerate and die. Oral medications and deep brain stimulation can relieve the initial symptoms, but the disease continues to progress. Growth factors that might support the survival, enhance the activity, or even regenerate degenerating dopamine neurons have been tried with mixed results in patients. As growth factors do not pass the blood-brain barrier, they have to be delivered intracranially. Therefore their efficient diffusion in brain tissue is of crucial importance. To improve the diffusion of the growth factor neurturin (NRTN), we modified its capacity to attach to heparan sulfates in the extracellular matrix. We present four new, biologically fully active variants with reduced heparin binding. Two of these variants are more stable than WT NRTN in vitro and diffuse better in rat brains. We also show that one of the NRTN variants diffuses better than its close homolog GDNF in monkey brains. The variant with the highest stability and widest diffusion regenerates dopamine fibers and improves the conditions of rats in a 6-hydroxydopamine model of Parkinson's disease more potently than GDNF, which previously showed modest efficacy in clinical trials. The new NRTN variants may help solve the major problem of inadequate distribution of NRTN in human brain tissue., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
33. From microRNA target validation to therapy: lessons learned from studies on BDNF.
- Author
-
Varendi K, Mätlik K, and Andressoo JO
- Subjects
- 3' Untranslated Regions, Animals, Gene Expression Regulation, Humans, MicroRNAs analysis, MicroRNAs genetics, RNA, Messenger analysis, RNA, Messenger genetics, Brain-Derived Neurotrophic Factor genetics, MicroRNAs metabolism, RNA, Messenger metabolism
- Abstract
During the past decade, the identification of microRNA (miR) targets has become common laboratory practice, and various strategies are now used to detect interactions between miRs and their mRNA targets. However, the current lack of a standardized identification process often leads to incomplete and/or conflicting results. Here, we review the problems most commonly encountered when verifying miR-mRNA interactions, and we propose a workflow for future studies. To illustrate the challenges faced when validating a miR target, we discuss studies in which the regulation of brain-derived neurotrophic factor by miRs was investigated, and we highlight several controversies that emerged from these studies. Finally, we discuss the therapeutic use of miR inhibitors, and we discuss several questions that should be addressed before proceeding to preclinical testing.
- Published
- 2015
- Full Text
- View/download PDF
34. AAV-mediated targeting of gene expression to the peri-infarct region in rat cortical stroke model.
- Author
-
Mätlik K, Abo-Ramadan U, Harvey BK, Arumäe U, and Airavaara M
- Subjects
- Animals, Brain pathology, Brain virology, Brain Ischemia pathology, Brain Ischemia physiopathology, Brain Ischemia therapy, Brain Ischemia virology, Disease Models, Animal, Genetic Therapy methods, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Immunohistochemistry, Infarction, Middle Cerebral Artery, Magnetic Resonance Imaging, Male, Metal Nanoparticles, Nerve Growth Factors genetics, Nerve Growth Factors metabolism, Rats, Sprague-Dawley, Stroke pathology, Stroke therapy, Stroke virology, Transduction, Genetic methods, Brain physiopathology, Dependovirus genetics, Gene Expression, Genetic Vectors, Stroke physiopathology
- Abstract
Background: For stroke patients the recovery of cognitive and behavioral functions is often incomplete. Functional recovery is thought to be mediated largely by connectivity rearrangements in the peri-infarct region. A method for manipulating gene expression in this region would be useful for identifying new recovery-enhancing treatments., New Method: We have characterized a way of targeting adeno-associated virus (AAV) vectors to the peri-infarct region of cortical ischemic lesion in rats 2days after middle cerebral artery occlusion (MCAo)., Results: We used magnetic resonance imaging (MRI) to show that the altered properties of post-ischemic brain tissue facilitate the spreading of intrastriatally injected nanoparticles toward the infarct. We show that subcortical injection of green fluorescent protein-encoding dsAAV7-GFP resulted in transduction of cells in and around the white matter tract underlying the lesion, and in the cortex proximal to the lesion. A similar result was achieved with dsAAV7 vector encoding the cerebral dopamine neurotrophic factor (CDNF), a protein with therapeutic potential., Comparison With Existing Methods: Viral vector-mediated intracerebral gene delivery has been used before in rodent models of ischemic injury. However, the method of targeting gene expression to the peri-infarct region, after the initial phase of ischemic cell death, has not been described before., Conclusions: We demonstrate a straightforward and robust way to target AAV vector-mediated over-expression of genes to the peri-infarct region in a rat stroke model. This method will be useful for studying the action of specific proteins in peri-infarct region during the recovery process., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
35. Glia cell line-derived neurotrophic factor mediates survival of murine sympathetic precursors.
- Author
-
Heermann S, Mätlik K, Hinz U, Fey J, Arumae U, and Krieglstein K
- Subjects
- Animals, Cell Survival, Cells, Cultured, Embryo, Mammalian, Mice, Reverse Transcriptase Polymerase Chain Reaction, Adrenergic Fibers, Glial Cell Line-Derived Neurotrophic Factor metabolism, Neural Stem Cells cytology, Neurogenesis physiology
- Abstract
During embryonic development, neurons are first produced in excess, and final numbers are adjusted by apoptosis at later stages. Crucial to this end is the amount of target-derived growth factor available for the neurons. By this means, the target size correctly matches the innervating neuron number. This target-derived survival has been well studied for sympathetic neurons, and nerve growth factor (NGF) was identified to be the crucial factor for maintaining sympathetic neurons at late embryonic and early postnatal stages, with a virtual complete loss of sympathetic neurons in NGF knockout (KO) mice. This indicates that all sympathetic neurons are dependent on NGF. However, also different glia cell line-derived neurotrophic factor (GDNF) KO mice consistently presented a loss of sympathetic neurons. This was the rationale for investigating the role of GDNF for sympathetic precursor/neuron survival. Here we show that GDNF is capable of promoting survival of 30% sympathetic precursors dissociated at E13. This is in line with data from GDNF KOs in which a comparable sympathetic neuron loss was observed at late embryonic stages, although the onset of the phenotype was unclear. We further present data showing that GDNF ligand and canonical receptors are expressed in sympathetic neurons especially at embryonic stages, raising the possibility of an autocrine/paracrine GDNF action. Finally, we show that GDNF also maintained neonatal sympathetic neurons (40%) cultured for 2 days. However, the GDNF responsiveness was lost at 5 days in vitro., (Copyright © 2013 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
36. PCSK9 regulates neuronal apoptosis by adjusting ApoER2 levels and signaling.
- Author
-
Kysenius K, Muggalla P, Mätlik K, Arumäe U, and Huttunen HJ
- Subjects
- Animals, Caspase 3 metabolism, HEK293 Cells, Human Umbilical Vein Endothelial Cells, Humans, LDL-Receptor Related Proteins genetics, Mice, Phosphorylation, Potassium metabolism, Proprotein Convertase 9, Proprotein Convertases genetics, Proto-Oncogene Proteins c-jun metabolism, RNA Interference, Reelin Protein, Serine Endopeptidases genetics, Signal Transduction, Apoptosis physiology, LDL-Receptor Related Proteins metabolism, Neurons cytology, Proprotein Convertases physiology, Serine Endopeptidases physiology
- Abstract
The secreted protease proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to low-density lipid (LDL) receptor family members LDLR, very low density lipoprotein receptor (VLDLR) and apolipoprotein receptor 2 (ApoER2), and promotes their degradation in intracellular acidic compartments. In the liver, LDLR is a major controller of blood LDL levels, whereas VLDLR and ApoER2 in the brain mediate Reelin signaling, a critical pathway for proper development of the nervous system. Expression level of PCSK9 in the brain is highest in the cerebellum during perinatal development, but is also increased in the adult brain after ischemia. The mechanism of PCSK9 function and its involvement in neuronal apoptosis is poorly understood. We show here that RNAi-mediated knockdown of PCSK9 significantly reduced the death of potassium-deprived cerebellar granule neurons (CGN), as shown by reduced levels of nuclear phosphorylated c-Jun and activated caspase-3, as well as condensed apoptotic nuclei. ApoER2 protein levels were increased in PCSK9 RNAi cells. Knockdown of ApoER2 but not of VLDLR was sufficient to reverse the protection provided by PCSK9 RNAi, suggesting that proapoptotic signaling of PCSK9 is mediated by altered ApoER2 function. Pharmacological inhibition of signaling pathways associated with lipoprotein receptors suggested that PCSK9 regulates neuronal apoptosis independently of NMDA receptor function but in concert with ERK and JNK signaling pathways. PCSK9 RNAi also reduced staurosporine-induced CGN apoptosis and axonal degeneration in the nerve growth factor-deprived dorsal root ganglion neurons. We conclude that PCSK9 potentiates neuronal apoptosis via modulation of ApoER2 levels and related anti-apoptotic signaling pathways.
- Published
- 2012
- Full Text
- View/download PDF
37. Acetylcholinesterase inhibitors rapidly activate Trk neurotrophin receptors in the mouse hippocampus.
- Author
-
Autio H, Mätlik K, Rantamäki T, Lindemann L, Hoener MC, Chao M, Arumäe U, and Castrén E
- Subjects
- Analysis of Variance, Animals, Donepezil, Enzyme-Linked Immunosorbent Assay, Gene Expression Regulation drug effects, Immunoprecipitation, Male, Mice, Mice, Inbred C57BL, Phosphorylation drug effects, Receptor, trkA genetics, Receptor, trkB genetics, Signal Transduction drug effects, Time Factors, Tyrosine metabolism, Cholinesterase Inhibitors pharmacology, Galantamine pharmacology, Hippocampus drug effects, Indans pharmacology, Piperidines pharmacology, Receptor, trkA metabolism, Receptor, trkB metabolism
- Abstract
Acetylcholinesterase inhibitors are first-line therapies for Alzheimer's disease. These drugs increase cholinergic tone in the target areas of the cholinergic neurons of the basal forebrain. Basal forebrain cholinergic neurons are dependent upon trophic support by nerve growth factor (NGF) through its neurotrophin receptor, TrkA. In the present study, we investigated whether the acetylcholinesterase inhibitors donepezil and galantamine could influence neurotrophin receptor signaling in the brain. Acute administration of donepezil (3 mg/kg, i.p.) led to the rapid autophosphorylation of TrkA and TrkB neurotrophin receptors in the adult mouse hippocampus. Similarly, galantamine dose-dependently (3, 9 mg/kg, i.p.) increased TrkA and TrkB phosphorylation in the mouse hippocampus. Both treatments also increased the phosphorylation of transcription factor CREB and tended to increase the phosphorylation of AKT kinase but did not alter the activity of MAPK42/44. Chronic treatment with galantamine (3 mg/kg, i.p., 14 days), did not induce changes in hippocampal NGF and BDNF synthesis or protein levels. Our findings show that acetylcholinesterase inhibitors are capable of rapidly activating hippocampal neurotrophin signaling and thus suggest that therapies targeting Trk signaling may already be in clinical use in the treatment of AD., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
38. Persephin signaling through GFRalpha1: the potential for the treatment of Parkinson's disease.
- Author
-
Sidorova YA, Mätlik K, Paveliev M, Lindahl M, Piranen E, Milbrandt J, Arumäe U, Saarma M, and Bespalov MM
- Subjects
- Animals, Biological Assay methods, Cell Line, Glial Cell Line-Derived Neurotrophic Factor metabolism, Glial Cell Line-Derived Neurotrophic Factor Receptors genetics, Humans, Mice, Models, Molecular, Nerve Growth Factors chemistry, Nerve Growth Factors genetics, Nerve Growth Factors metabolism, Nerve Tissue Proteins genetics, Neurons physiology, Protein Conformation, Proto-Oncogene Proteins c-ret genetics, Proto-Oncogene Proteins c-ret metabolism, Rats, Rats, Wistar, Glial Cell Line-Derived Neurotrophic Factor Receptors metabolism, Nerve Tissue Proteins metabolism, Parkinson Disease therapy, Signal Transduction physiology
- Abstract
Neurotrophic factors promote survival, proliferation and differentiation of neurons inducing intracellular signaling via specific receptors. The conventional biochemical methods often fail to reveal full repertoire of neurotrophic factor-receptor interactions because of their limited sensitivity. We evaluated several approaches to study signaling of Glial cell line-Derived Neurotrophic Factor (GDNF) family ligands and found that reporter-gene systems possess exceptionally high sensitivity and a heuristic power to identify novel biologically relevant growth factor-receptor interactions. We identified persephin, a GDNF family member, as a novel ligand for GFRalpha1/RET receptor complex. We confirmed this finding by several independent methods, including neurite outgrowth assay from the explants of sympathetic ganglia expressing Gfralpha1 and Ret mRNA but not persephin's conventional receptor GFRalpha4. As the activation of GFRalpha1/RET was shown to rescue dopaminergic neurons, our results suggest the potential of persephin for the treatment of Parkinson's disease.
- Published
- 2010
- Full Text
- View/download PDF
39. Combination of native and denaturing PAGE for the detection of protein binding regions in long fragments of genomic DNA.
- Author
-
Kaer K, Mätlik K, Metsis M, and Speek M
- Subjects
- Animals, Base Sequence, DNA Restriction Enzymes metabolism, Electrophoretic Mobility Shift Assay, Molecular Sequence Data, Nucleic Acid Denaturation, PC12 Cells, Protein Binding, Rats, Receptors, Nerve Growth Factor genetics, Receptors, Nerve Growth Factor metabolism, DNA genetics, DNA metabolism, DNA Fragmentation, DNA-Binding Proteins metabolism, Electrophoresis, Gel, Two-Dimensional methods, Genome genetics
- Abstract
Background: In a traditional electrophoresis mobility shift assay (EMSA) a 32P-labeled double-stranded DNA oligonucleotide or a restriction fragment bound to a protein is separated from the unbound DNA by polyacrylamide gel electrophoresis (PAGE) in nondenaturing conditions. An extension of this method uses the large population of fragments derived from long genomic regions (approximately 600 kb) for the identification of fragments containing protein binding regions. With this method, genomic DNA is fragmented by restriction enzymes, fragments are amplified by PCR, radiolabeled, incubated with nuclear proteins and the resulting DNA-protein complexes are separated by two-dimensional PAGE. Shifted DNA fragments containing protein binding sites are identified by using additional procedures, i. e. gel elution, PCR amplification, cloning and sequencing. Although the method allows simultaneous analysis of a large population of fragments, it is relatively laborious and can be used to detect only high affinity protein binding sites. Here we propose an alternative and straightforward strategy which is based on a combination of native and denaturing PAGE. This strategy allows the identification of DNA fragments containing low as well as high affinity protein binding regions, derived from genomic DNA (<10 kb) of known sequence., Results: We have combined an EMSA-based selection step with subsequent denaturing PAGE for the localization of protein binding regions in long (up to 10 kb) fragments of genomic DNA. Our strategy consists of the following steps: digestion of genomic DNA with a 4-cutter restriction enzyme (AluI, BsuRI, TruI, etc), separation of low and high molecular weight fractions of resultant DNA fragments, 32P-labeling with Klenow polymerase, traditional EMSA, gel elution and identification of the shifted bands (or smear) by denaturing PAGE. The identification of DNA fragments containing protein binding sites is carried out by running the gel-eluted fragments alongside with the full "spectrum" of initial restriction fragments of known size. Here the strategy is used for the identification of protein-binding regions in the 5' region of the rat p75 neurotrophin receptor (p75NTR) gene., Conclusion: The developed strategy is based on a combination of traditional EMSA and denaturing PAGE for the identification of protein binding regions in long fragments of genomic DNA. The identification is straightforward and can be applied to shifted bands corresponding to stable DNA-protein complexes as well as unstable complexes, which undergo dissociation during electrophoresis.
- Published
- 2008
- Full Text
- View/download PDF
40. Many human genes are transcribed from the antisense promoter of L1 retrotransposon.
- Author
-
Nigumann P, Redik K, Mätlik K, and Speek M
- Subjects
- 5' Flanking Region genetics, Alternative Splicing, Base Sequence, Databases, Nucleic Acid, Expressed Sequence Tags, Gene Expression Regulation, Genes genetics, Humans, Molecular Sequence Data, DNA, Antisense genetics, Promoter Regions, Genetic genetics, Retroelements genetics, Transcription, Genetic genetics
- Abstract
Human L1 retrotransposon has two transcription-regulatory regions: an internal or sense promoter driving transcription of the full-length L1, and an antisense promoter (ASP) driving transcription in the opposite direction into adjacent cellular sequences yielding chimeric transcripts. Both promoters are located in the 5'-untranslated region (5'-UTR) of L1. Chimeric transcripts derived from the L1 ASP are highly represented in expressed-sequence tag (EST) databases. Using a bioinformatics approach, we have characterized 10 chimeric ESTs (cESTs) derived from the EST division of GenBank. These cESTs contained 3' regions similar or identical to known cellular mRNA sequences. They were accurately spliced and preferentially expressed in tumor cell lines. Analysis of the hundreds of cESTs suggests that the L1 ASP-driven transcription is a common phenomenon not only for tumor cells but also for normal ones and may involve transcriptional interference or epigenetic control of different cellular genes.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.