1
|
Cervera-Juanes RP, Zimmerman KD, Wilhelm LJ, Lowe CC, Gonzales SW, Carlson T, Hitzemann R, Ferguson BM, Grant KA. Pre-existing DNA methylation signatures in the prefrontal cortex of alcohol-naïve nonhuman primates define neural vulnerability for future risky ethanol consumption. Neurobiol Dis 2025; 209:106886. [PMID: 40139280 PMCID: PMC12044430 DOI: 10.1016/j.nbd.2025.106886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/13/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Alcohol use disorder (AUD) is a highly prevalent, complex, multifactorial and heterogeneous disorder, with 11 % and 30 % of adults meeting criteria for past-year and lifetime AUD, respectively. Identification of the molecular mechanisms underlying risk for AUD would facilitate effective deployment of personalized interventions. Studies using rhesus monkeys and rats, have demonstrated that individuals with low cognitive flexibility and a predisposition towards habitual behaviors show an increased risk for future heavy drinking. Further, low cognitive flexibility is associated with reduced dorsolateral prefrontal cortex (dlPFC) function in rhesus monkeys. To explore the underlying unique molecular signatures that increase risk for chronic heavy drinking, a genome-wide DNA methylation (DNAm) analysis of the alcohol-naïve dlPFC-A46 biopsy prior to chronic alcohol self-administration was conducted. The DNAm profile provides a molecular snapshot of the alcohol-naïve dlPFC, with mapped genes and associated signaling pathways that vary across individuals. The analysis identified 1,463 differentially methylated regions (DMRs) related to unique genes that were strongly associated with average ethanol intake consumed over 6 months of voluntary self-administration. These findings translate behavioral phenotypes into neural markers of risk for AUD, and hold promise for parallel discoveries in risk for other disorders involving impaired cognitive flexibility. SIGNIFICANCE: Alcohol use disorder (AUD) is a highly prevalent and heterogeneous disorder. Prevention strategies to accurately identify individuals with a high risk for AUD, would help reduce the prevalence, and severity of AUD. Our novel epigenomic analysis of the alcohol-naïve nonhuman primate cortex provides a molecular snapshot of the vulnerable brain, pointing to circuitry and molecular mechanisms associated with cortical development, synaptic functions, glutamatergic signaling and coordinated signaling pathways. With a complex disorder like AUD, having the ability to identify the molecular mechanisms underlying AUD risk is critical for better development of personalized effective treatments.
Collapse
Affiliation(s)
- Rita P Cervera-Juanes
- Department of Translational Neuroscience, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, United States of America; Center for Precision Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, United States of America.
| | - Kip D Zimmerman
- Center for Precision Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, United States of America; Department of Internal Medicine, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157, United States of America
| | - Larry J Wilhelm
- Department of Translational Neuroscience, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, United States of America
| | - Clara Christine Lowe
- Department of Translational Neuroscience, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, United States of America
| | - Steven W Gonzales
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States of America
| | - Tim Carlson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States of America
| | - Robert Hitzemann
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States of America; Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, United States of America
| | - Betsy M Ferguson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States of America; Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States of America
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States of America; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States of America; Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, United States of America
| |
Collapse
|
2
|
Zeppilli S, Gurrola AO, Demetci P, Brann DH, Pham TM, Attey R, Zilkha N, Kimchi T, Datta SR, Singh R, Tosches MA, Crombach A, Fleischmann A. Single-cell genomics of the mouse olfactory cortex reveals contrasts with neocortex and ancestral signatures of cell type evolution. Nat Neurosci 2025; 28:937-948. [PMID: 40200010 DOI: 10.1038/s41593-025-01924-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 02/19/2025] [Indexed: 04/10/2025]
Abstract
Understanding the molecular logic of cortical cell-type diversity can illuminate cortical circuit function and evolution. Here, we performed single-nucleus transcriptome and chromatin accessibility analyses to compare neurons across three- to six-layered cortical areas of adult mice and across tetrapod species. We found that, in contrast to the six-layered neocortex, glutamatergic neurons of the three-layered mouse olfactory (piriform) cortex displayed continuous rather than discrete variation in transcriptomic profiles. Subsets of piriform and neocortical glutamatergic cells with conserved transcriptomic profiles were distinguished by distinct, area-specific epigenetic states. Furthermore, we identified a prominent population of immature neurons in piriform cortex and observed that, in contrast to the neocortex, piriform cortex exhibited divergence between glutamatergic cells in laboratory versus wild-derived mice. Finally, we showed that piriform neurons displayed greater transcriptomic similarity to cortical neurons of turtles, lizards and salamanders than to those of the neocortex. In summary, despite over 200 million years of coevolution alongside the neocortex, olfactory cortex neurons retain molecular signatures of ancestral cortical identity.
Collapse
Affiliation(s)
- Sara Zeppilli
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Alonso O Gurrola
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Pinar Demetci
- Department of Computer Science, Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - David H Brann
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Tuan M Pham
- Department of Computer Science, Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - Robin Attey
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Noga Zilkha
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Kimchi
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Sandeep R Datta
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Ritambhara Singh
- Department of Computer Science, Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - Maria A Tosches
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Anton Crombach
- Inria Centre de Lyon, Villeurbanne, France.
- INSA-Lyon, CNRS, UCBL, LIRIS, UMR5205, Villeurbanne, France.
- INSA-Lyon, CITI, UR3720, Villeurbanne, France.
| | - Alexander Fleischmann
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| |
Collapse
|
3
|
Sepúlveda-Cuéllar RD, Soria-Medina DA, Cañedo-Solares I, Gómez-Chávez F, Molina-López LM, Cruz-Martínez MY, Correa D. Controversies and insights into cytokine regulation of neurogenesis and behavior in adult rodents. Front Immunol 2025; 16:1550660. [PMID: 40352932 PMCID: PMC12061686 DOI: 10.3389/fimmu.2025.1550660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
Adult learning, memory, and social interaction partially depend on neurogenesis in two regions: the hippocampus and the subventricular zone. There is evidence that the immune system is important for these processes in pathological situations, but there is no review of its role in non-pathological or near-physiological conditions. Although further research is warranted in this area, some conclusions can be drawn. Intrusive LyC6hi monocytes and autoreactive CD4+ T cells have a positive impact on neurogenesis and behavior, but the latter are deleterious if specific to external antigens. Mildly activated microglia play a crucial role in promoting these processes, by eliminating apoptotic neuronal progenitors and producing low levels of interleukins, which increase if the cells are activated, leading to inhibition of neurogenesis. Chemokines are poorly studied, but progenitor cells and neurons express their receptors, which appear important for migration and maturation. The few works that jointly analyzed neurogenesis and behavior showed congruent effects of immune cells and cytokines. In conclusion, the immune system components -mostly local- seem of utmost importance for the control of behavior under non-pathological conditions.
Collapse
Affiliation(s)
- Rodrigo Daniel Sepúlveda-Cuéllar
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| | - Diego Alberto Soria-Medina
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
- Facultad de Psicología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Irma Cañedo-Solares
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría (INP), Secretaría de Salud, Ciudad de México, Mexico
| | - Fernando Gómez-Chávez
- Laboratorio de Enfermedades Osteoarticulares e Inmunológicas, Sección de Estudios de Posgrado e Investigación, Escuela Nacional de Medicina y Homeopatía (ENMyH), Instituto Politécnico Nacional (IPN), Ciudad de México, Mexico
| | - Liliana Monserrat Molina-López
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| | - María Yolanda Cruz-Martínez
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| | - Dolores Correa
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México, Huixquilucan, EdoMex, Mexico
| |
Collapse
|
4
|
Zhang Q, Yi Y, Chen T, Ai Y, Chen Z, Liu G, Tang Z, Chen J, Xu T, Chen X, Liu J, Xia Y. M2 microglia-derived small extracellular vesicles modulate NSC fate after ischemic stroke via miR-25-3p/miR-93-5p-TGFBR/PTEN/FOXO3 axis. J Nanobiotechnology 2025; 23:311. [PMID: 40270025 PMCID: PMC12020034 DOI: 10.1186/s12951-025-03390-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Endogenous neurogenesis could promote stroke recovery. Furthermore, anti-inflammatory phenotypical microglia (M2-microglia) could facilitate Neural Stem Cell (NSC)-mediated neurogenesis following Ischemic Stroke (IS). Nonetheless, the mechanisms through which M2 microglia influence NSC-mediated neurogenesis post-IS remain unclear. On the other hand, M2 microglia-derived small Extracellular Vesicles (M2-sEVs) could exert phenomenal biological effects and play significant roles in cell-to-cell interactions, highlighting their potential involvement in NSC-mediated neurogenesis post-IS, forming the basis of this study. METHODS M2-sEVs were first isolated from IL-4-stimulated microglia. For in vivo tests, M2-sEVs were intravenously injected into mice every day for 14 days after transient Middle Cerebral Artery Occlusion (tMCAO). Following that, the infarct volume and neurological function, as well as NSC proliferation in the Subventricular Zone and dentate gyrus, migration, and differentiation in the infarct area, were examined. For in vitro tests, M2-sEVs were administered to NSC subjected to Oxygen-Glucose Deprivation (OGD) and then reoxygenation, after which NSC proliferation and differentiation were assessed. Finally, M2-sEVs were subjected to microRNA sequencing to explore the regulatory mechanisms. RESULTS Our findings revealed that M2-sEVs reduced the infarct volume and increased the neurological score in mice post-tMCAO. Furthermore, M2-sEV treatment promoted NSC proliferation and neuronal differentiation both in vivo and in vitro. Additionally, microRNA sequencing revealed miR-93-5p and miR-25-3p enrichment in M2-sEVs. Inhibitors of these miRNAs prevented TGFBR, PTEN, and FOXO3 downregulation in NSC, reversing M2-sEVs' beneficial effects on neurogenesis and sensorimotor recovery. CONCLUSIONS M2-sEVs increased NSC proliferation and neuronal differentiation, and protected against IS, at least partially, via delivering miR-25-3p and miR-93-5p to downregulate TGFBR, PTEN, and FOXO3 expression in NSC.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
| | - Yan Yi
- Reproductive Medicine Center, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
| | - Tiange Chen
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
| | - Ying Ai
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
| | - Ziyang Chen
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
| | - Ganzhi Liu
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China
| | - Zexuan Tang
- School of Graduate Studies, Biomedical Science - Dental Scholars Track Program, Rutgers Biomedical and Health Sciences, Rutgers University, Newark, NJ, 07103, USA
| | - Jianwei Chen
- Bio-Intelligent Manufacturing and Living Matter Bioprinting Center, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen, China
| | - Tao Xu
- Bio-Intelligent Manufacturing and Living Matter Bioprinting Center, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen, China
| | - Xin Chen
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China.
| | - Jinfang Liu
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China.
| | - Yuguo Xia
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, CN, 410008, China.
| |
Collapse
|
5
|
Santos M, Moreira JAF, Santos SS, Solá S. Sustaining Brain Youth by Neural Stem Cells: Physiological and Therapeutic Perspectives. Mol Neurobiol 2025:10.1007/s12035-025-04774-z. [PMID: 39985708 DOI: 10.1007/s12035-025-04774-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
In the last two decades, stem cells (SCs) have attracted considerable interest for their research value and therapeutic potential in many fields, namely in neuroscience. On the other hand, the discovery of adult neurogenesis, the process by which new neurons are generated in the adult brain, challenged the traditional view that the brain is a static structure after development. The recent findings showing that adult neurogenesis has a significant role in brain plasticity, learning and memory, and emotional behavior, together with the fact that it is strongly dependent on several external and internal factors, have sparked more interest in this area. The mechanisms of adult neural stem cell (NSC) regulation, the physiological role of NSC-mediated neuroplasticity throughout life, and the most recent NSC-based therapeutic applications will be concisely reviewed. Noteworthy, due to their multipotency, self-renewal potential, and ability to secrete growth and immunomodulatory factors, NSCs have been mainly suggested for (1) transplantation, (2) neurotoxicology tests, and (3) drug screening approaches. The clinical trials of NSC-based therapy for different neurologic conditions are, nonetheless, mostly in the early phases and have not yet demonstrated conclusive efficacy or safety. Here, we provide an outlook of the major challenges and limitations, as well as some promising directions that could help to move toward stem cell widespread use in the treatment and prevention of several neurological disorders.
Collapse
Affiliation(s)
- Matilde Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - João A Ferreira Moreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - Sónia Sá Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal.
| |
Collapse
|
6
|
Zhang F, Fu Y, Jimenez-Cyrus D, Zhao T, Shen Y, Sun Y, Zhang Z, Wang Q, Kawaguchi R, Geschwind DH, He C, Ming GL, Song H. m 6A/YTHDF2-mediated mRNA decay targets TGF-β signaling to suppress the quiescence acquisition of early postnatal mouse hippocampal NSCs. Cell Stem Cell 2025; 32:144-156.e8. [PMID: 39476834 PMCID: PMC11698649 DOI: 10.1016/j.stem.2024.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/26/2024] [Accepted: 10/02/2024] [Indexed: 01/06/2025]
Abstract
Quiescence acquisition of proliferating neural stem cells (NSCs) is required to establish the adult NSC pool. The underlying molecular mechanisms are not well understood. Here, we showed that conditional deletion of the m6A reader Ythdf2, which promotes mRNA decay, in proliferating NSCs in the early postnatal mouse hippocampus elevated quiescence acquisition in a cell-autonomous fashion with decreased neurogenesis. Multimodal profiling of m6A modification, YTHDF2 binding, and mRNA decay in hippocampal NSCs identified shared targets in multiple transforming growth factor β (TGF-β)-signaling-pathway components, including TGF-β ligands, maturation factors, receptors, transcription regulators, and signaling regulators. Functionally, Ythdf2 deletion led to TGF-β-signaling activation in NSCs, suppression of which rescued elevated quiescence acquisition of proliferating hippocampal NSCs. Our study reveals the dynamic nature and critical roles of mRNA decay in establishing the quiescent adult hippocampal NSC pool and uncovers a distinct mode of epitranscriptomic control via co-regulation of multiple components of the same signaling pathway.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; School of Life Sciences, Nanjing University, Nanjing, PRC
| | - Yao Fu
- Department of Biology, School of Art and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennisse Jimenez-Cyrus
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ting Zhao
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yachen Shen
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yusha Sun
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qing Wang
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Riki Kawaguchi
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA; The Epigenetics Institute, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Neurosurgery, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Bedolla A, Wegman E, Weed M, Stevens MK, Ware K, Paranjpe A, Alkhimovitch A, Ifergan I, Taranov A, Peter JD, Gonzalez RMS, Robinson JE, McClain L, Roskin KM, Greig NH, Luo Y. Adult microglial TGFβ1 is required for microglia homeostasis via an autocrine mechanism to maintain cognitive function in mice. Nat Commun 2024; 15:5306. [PMID: 38906887 PMCID: PMC11192737 DOI: 10.1038/s41467-024-49596-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/11/2024] [Indexed: 06/23/2024] Open
Abstract
While TGF-β signaling is essential for microglial function, the cellular source of TGF-β1 ligand and its spatial regulation remains unclear in the adult CNS. Our data supports that microglia but not astrocytes or neurons are the primary producers of TGF-β1 ligands needed for microglial homeostasis. Microglia-Tgfb1 KO leads to the activation of microglia featuring a dyshomeostatic transcriptome that resembles disease-associated, injury-associated, and aged microglia, suggesting microglial self-produced TGF-β1 ligands are important in the adult CNS. Astrocytes in MG-Tgfb1 inducible (i)KO mice show a transcriptome profile that is closely aligned with an LPS-associated astrocyte profile. Additionally, using sparse mosaic single-cell microglia KO of TGF-β1 ligand we established an autocrine mechanism for signaling. Here we show that MG-Tgfb1 iKO mice present cognitive deficits, supporting that precise spatial regulation of TGF-β1 ligand derived from microglia is required for the maintenance of brain homeostasis and normal cognitive function in the adult brain.
Collapse
Affiliation(s)
- Alicia Bedolla
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Elliot Wegman
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Max Weed
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | | | - Kierra Ware
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Aditi Paranjpe
- Information Services for Research, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Anastasia Alkhimovitch
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Igal Ifergan
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Aleksandr Taranov
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Joshua D Peter
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Rosa Maria Salazar Gonzalez
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, US
| | - J Elliott Robinson
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, US
| | - Lucas McClain
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Krishna M Roskin
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, US
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA.
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA.
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
8
|
Ma W, Yang J, Zhang J, He R, Luo Y, Li C, Zhao F, Tao F, Fan J, Yin L, Zhu K, Niu S, Li L. Cerebral protective effect of in situ and remote ischemic postconditioning on ischemic stroke rat via the TGFβ1-Smad2/3 signaling pathway. Brain Res 2024; 1824:148685. [PMID: 38006988 DOI: 10.1016/j.brainres.2023.148685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 11/27/2023]
Abstract
Patients with acute ischemic stroke achieve inadequate benefit due to the short therapeutic window for thrombolysis and the risk of ischemia/reperfusion (IR) injury. Ischemic postconditioning induces endogenous cerebral protection for acute ischemic stroke, although the protective mechanisms associated with ischemic postconditioning haven't been well clarified. In present study, the rat models of ischemic cerebral stroke with in situ and remote ischemic postconditioning (ISP and RIP) were established successfully. The Zea Longa and the modified neurological severity scoring (mNSS) were carried out to evaluate neurological function in the rats, while the open field test was explored to estimate their autonomic athletic ability. The 2,3,5-riphenyltetrazolium chloride (TTC) staining method was used to measure the size of the infarcts. TUNEL and Nissl's staining were used to detect the apoptosis rate of cells in the ischemic penumbra, with the expression of TGFβ1, Smad2, and Smad3 in the ischemic penumbra and serum detected by immunohistochemical staining, qRT-PCR, Western blots, and ELISA analysis. We showed that application of both types of ischemic postconditioning had cerebral protective effects for the ischemic stroke rats, that included effective reduction in the volume of cerebral infarction, alleviation of apoptosis and inflammation in the ischemic penumbra, and promotion of recovery of neurological function. These effects included significantly enriched gene ontology (GO) terms after RIP intervention that were related to TGFβ1, increased protein levels of TGFβ1 and decreased levels of p-Smad2/3 and smad3 following RIP intervention. We showed that the TGFβ1-Smad2/3 signaling pathway was associated with the cerebral protection of ischemic postconditioning.
Collapse
Affiliation(s)
- Wei Ma
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Jinwei Yang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China; Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Jinfen Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Rui He
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Yi Luo
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Chunyan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China; Department of Neurology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Feng Zhao
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Fengping Tao
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Jingjing Fan
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Luwei Yin
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Kewei Zhu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Shourui Niu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Liyan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
9
|
Medoro A, Davinelli S, Milella L, Willcox BJ, Allsopp RC, Scapagnini G, Willcox DC. Dietary Astaxanthin: A Promising Antioxidant and Anti-Inflammatory Agent for Brain Aging and Adult Neurogenesis. Mar Drugs 2023; 21:643. [PMID: 38132964 PMCID: PMC10744637 DOI: 10.3390/md21120643] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Decreased adult neurogenesis, or the gradual depletion of neural stem cells in adult neurogenic niches, is considered a hallmark of brain aging. This review provides a comprehensive overview of the intricate relationship between aging, adult neurogenesis, and the potential neuroregenerative properties of astaxanthin, a carotenoid principally extracted from the microalga Haematococcus pluvialis. The unique chemical structure of astaxanthin enables it to cross the blood-brain barrier and easily reach the brain, where it may positively influence adult neurogenesis. Astaxanthin can affect molecular pathways involved in the homeostasis, through the activation of FOXO3-related genetic pathways, growth, and regeneration of adult brain neurons, enhancing cell proliferation and the potency of stem cells in neural progenitor cells. Furthermore, astaxanthin appears to modulate neuroinflammation by suppressing the NF-κB pathway, reducing the production of pro-inflammatory cytokines, and limiting neuroinflammation associated with aging and chronic microglial activation. By modulating these pathways, along with its potent antioxidant properties, astaxanthin may contribute to the restoration of a healthy neurogenic microenvironment, thereby preserving the activity of neurogenic niches during both normal and pathological aging.
Collapse
Affiliation(s)
- Alessandro Medoro
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (A.M.); (S.D.)
| | - Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (A.M.); (S.D.)
| | - Luigi Milella
- Department of Science, University of Basilicata, V. le Ateneo Lucano 10, 85100 Potenza, Italy;
| | - Bradley J. Willcox
- Center of Biomedical Research Excellence for Translational Research on Aging, Kuakini Medical Center, Honolulu, HI 96817, USA; (B.J.W.); (R.C.A.); (D.C.W.)
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96822, USA
| | - Richard C. Allsopp
- Center of Biomedical Research Excellence for Translational Research on Aging, Kuakini Medical Center, Honolulu, HI 96817, USA; (B.J.W.); (R.C.A.); (D.C.W.)
- Institute for Biogenesis Research, University of Hawaii, Honolulu, HI 96822, USA
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (A.M.); (S.D.)
| | - Donald Craig Willcox
- Center of Biomedical Research Excellence for Translational Research on Aging, Kuakini Medical Center, Honolulu, HI 96817, USA; (B.J.W.); (R.C.A.); (D.C.W.)
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96822, USA
- Department of Human Welfare, Okinawa International University, Ginowan 901-2211, Japan
| |
Collapse
|
10
|
Bedolla A, Wegman E, Weed M, Paranjpe A, Alkhimovitch A, Ifergan I, McClain L, Luo Y. Microglia-derived TGF-β1 ligand maintains microglia homeostasis via autocrine mechanism and is critical for normal cognitive function in adult mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547814. [PMID: 37461569 PMCID: PMC10349967 DOI: 10.1101/2023.07.05.547814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
While TGF-β signaling is essential for microglial function, the cellular source of TGF-β ligand and its spatial regulation remains unclear in the adult CNS. Our data support that microglia, not astrocytes or neurons, are the primary producers of TGF-β1 ligands needed for microglial homeostasis. Microglia (MG)-Tgfb1 inducible knockout (iKO) leads to the activation of microglia featuring a dyshomeostatic transcriptomic profile that resembles disease-associated microglia (DAMs), injury-associated microglia, and aged microglia, suggesting that microglial self-produced TGF-β1 ligands are important in the adult CNS. Interestingly, astrocytes in MG-Tgfb1 iKO mice show a transcriptome profile that closely aligns with A1-like astrocytes. Additionally, using sparse mosaic single-cell microglia iKO of TGF-β1 ligand, we established an autocrine mechanism for TGF-β signaling. Importantly MG-Tgfb1 iKO mice show cognitive deficits, supporting that precise spatial regulation of TGF-β1 ligand derived from microglia is critical for the maintenance of brain homeostasis and normal cognitive function in the adult brain.
Collapse
Affiliation(s)
- Alicia Bedolla
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Elliot Wegman
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Max Weed
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Aditi Paranjpe
- Information Services, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Anastasia Alkhimovitch
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Igal Ifergan
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Lucas McClain
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
11
|
Davinelli S, Medoro A, Ali S, Passarella D, Intrieri M, Scapagnini G. Dietary Flavonoids and Adult Neurogenesis: Potential Implications for Brain Aging. Curr Neuropharmacol 2023; 21:651-668. [PMID: 36321225 PMCID: PMC10207917 DOI: 10.2174/1570159x21666221031103909] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/27/2022] [Accepted: 08/19/2022] [Indexed: 02/10/2023] Open
Abstract
Adult neurogenesis deficiency has been proposed to be a common hallmark in different age-related neurodegenerative diseases. The administration of flavonoids is currently reported as a potentially beneficial strategy for preventing brain aging alterations, including adult neurogenesis decline. Flavonoids are a class of plant-derived dietary polyphenols that have drawn attention for their neuroprotective and pro-cognitive effects. Although they undergo extensive metabolism and localize in the brain at low concentrations, flavonoids are now believed to improve cerebral vasculature and interact with signal transduction cascades involved in the regulation of adult neurogenesis. Furthermore, many dietary flavonoids have been shown to reduce oxidative stress and neuroinflammation, improving the neuronal microenvironment where adult neurogenesis occurs. The overall goal of this review is to summarize the evidence supporting the role of flavonoids in modulating adult neurogenesis as well as to highlight how these dietary agents may be promising candidates in restoring healthy brain function during physiological and pathological aging.
Collapse
Affiliation(s)
- Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso 86100, Italy
| | - Alessandro Medoro
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso 86100, Italy
| | - Sawan Ali
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso 86100, Italy
| | - Daniela Passarella
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso 86100, Italy
| | - Mariano Intrieri
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso 86100, Italy
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso 86100, Italy
| |
Collapse
|
12
|
Bindu GSS, Thekkekkara D, Narayanan TL, Narayanan J, Chalasani SH, Manjula SN. The Role of TGF-β in Cognitive Decline Associated with Radiotherapy in Brain Tumor. J Pharmacol Pharmacother 2022. [DOI: 10.1177/0976500x221107503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cognitive decline is a late adverse event in brain tumor survivors. The patients receiving radiation treatment exhibit a wide range of damage and impairment in attention, memory, and executive function compared to the untreated group. After radiation treatment, various changes are observed in astrocytes, oligodendrocytes, white matter, and vasculature. The major affected areas are the hippocampus and prefrontal cortex. Neurogenesis impairment is one of the primary mechanisms responsible for cognitive dysfunction. Various cytokines and growth factors are responsible for inducing apoptosis of neural cells, which results in impaired neurogenesis in response to radiotherapy. Transforming growth factor (TGF-β) is one of the key cytokines released in response to radiation. TGF-β plays a major role in neuronal apoptosis through various pathways such as the MAP kinase pathway, JAK/STAT pathway, and protein kinase pathway. In contrast, activation of the ALK5 pathway via TGF-β improves neurogenesis. So, the current review article focuses on the detailed effects of TGF-β on neuronal cells concerning radiation exposure. This in-depth knowledge will help researchers focus more on the TGF-β pathway and come up with new treatment schedules which will help reduce cognitive dysfunctions in brain tumor patients produced as a result of radiation therapy.
Collapse
Affiliation(s)
- G. S. Swarna Bindu
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, SS Nagar, Mysuru, Karnataka, India
| | - Dithu Thekkekkara
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, SS Nagar, Mysuru, Karnataka, India
| | - T. Lakshmi Narayanan
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, SS Nagar, Mysuru, Karnataka, India
| | - Jiju Narayanan
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, SS Nagar, Mysuru, Karnataka, India
| | - Sri Harsha Chalasani
- Department of Pharmacy Practice, JSS College of Pharmacy, JSSAHER, SS Nagar, Mysuru, Karnataka, India
| | - S. N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, SS Nagar, Mysuru, Karnataka, India
| |
Collapse
|
13
|
Endogenous Neural Stem Cell Mediated Oligodendrogenesis in the Adult Mammalian Brain. Cells 2022; 11:cells11132101. [PMID: 35805185 PMCID: PMC9265817 DOI: 10.3390/cells11132101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 02/08/2023] Open
Abstract
Oligodendrogenesis is essential for replacing worn-out oligodendrocytes, promoting myelin plasticity, and for myelin repair following a demyelinating injury in the adult mammalian brain. Neural stem cells are an important source of oligodendrocytes in the adult brain; however, there are considerable differences in oligodendrogenesis from neural stem cells residing in different areas of the adult brain. Amongst the distinct niches containing neural stem cells, the subventricular zone lining the lateral ventricles and the subgranular zone in the dentate gyrus of the hippocampus are considered the principle areas of adult neurogenesis. In addition to these areas, radial glia-like cells, which are the precursors of neural stem cells, are found in the lining of the third ventricle, where they are called tanycytes, and in the cerebellum, where they are called Bergmann glia. In this review, we will describe the contribution and regulation of each of these niches in adult oligodendrogenesis.
Collapse
|
14
|
Luo J. TGF-β as a Key Modulator of Astrocyte Reactivity: Disease Relevance and Therapeutic Implications. Biomedicines 2022; 10:1206. [PMID: 35625943 PMCID: PMC9138510 DOI: 10.3390/biomedicines10051206] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 02/06/2023] Open
Abstract
Astrocytes are essential for normal brain development and functioning. They respond to brain injury and disease through a process referred to as reactive astrogliosis, where the reactivity is highly heterogenous and context-dependent. Reactive astrocytes are active contributors to brain pathology and can exert beneficial, detrimental, or mixed effects following brain insults. Transforming growth factor-β (TGF-β) has been identified as one of the key factors regulating astrocyte reactivity. The genetic and pharmacological manipulation of the TGF-β signaling pathway in animal models of central nervous system (CNS) injury and disease alters pathological and functional outcomes. This review aims to provide recent understanding regarding astrocyte reactivity and TGF-β signaling in brain injury, aging, and neurodegeneration. Further, it explores how TGF-β signaling modulates astrocyte reactivity and function in the context of CNS disease and injury.
Collapse
Affiliation(s)
- Jian Luo
- Palo Alto Veterans Institute for Research, VAPAHCS, Palo Alto, CA 94304, USA
| |
Collapse
|
15
|
Gradari S, Herrera A, Tezanos P, Fontán-Lozano Á, Pons S, Trejo JL. The Role of Smad2 in Adult Neuroplasticity as Seen through Hippocampal-Dependent Spatial Learning/Memory and Neurogenesis. J Neurosci 2021; 41:6836-6849. [PMID: 34210778 PMCID: PMC8360684 DOI: 10.1523/jneurosci.2619-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 11/21/2022] Open
Abstract
Adult neural plasticity is an important and intriguing phenomenon in the brain, and adult hippocampal neurogenesis is directly involved in modulating neural plasticity by mechanisms that are only partially understood. We have performed gain-of-function and loss-of-function experiments to study Smad2, a transcription factor selected from genes that are demethylated after exercise through the analysis of an array of physical activity-induced factors, and their corresponding gene expression, and an efficient inducer of plasticity. In these studies, changes in cell number and morphology were analyzed in the hippocampal dentate gyrus (cell proliferation and survival, including regional distribution, and structural maturation/differentiation, including arborization, dendritic spines, and neurotransmitter-specific vesicles) of sedentary male mice, after evaluation in a battery of behavioral tests. As a result, we reveal a role for Smad2 in the balance of proliferation versus maturation of differentiating immature cells (Smad2 silencing increases both the proliferation and survival of cycling cells in the dentate granule cell layer), and in the plasticity of both newborn and mature neurons in mice (by decreasing dendritic arborization and dendritic spine number). Moreover, Smad2 silencing specifically compromises spatial learning in mice (through impairments of spatial tasks acquisition both in long-term learning and working memory). These data suggest that Smad2 participates in adult neural plasticity by influencing the proliferation and maturation of dentate gyrus neurons.SIGNIFICANCE STATEMENT Smad2 is one of the main components of the transforming growth factor-β (TGF-β) pathway. The commitment of cell fate in the nervous system is tightly coordinated by SMAD2 signaling, as are further differentiation steps (e.g., dendrite and axon growth, myelination, and synapse formation). However, there are no studies that have directly evaluated the role of Smad2 gene in hippocampus of adult animals. Modulation of these parameters in the adult hippocampus can affect hippocampal-dependent behaviors, which may shed light on the mechanisms that regulate adult neurogenesis and behavior. We demonstrate here a role for Smad2 in the maturation of differentiating immature cells and in the plasticity of mature neurons. Moreover, Smad2 silencing specifically compromises the spatial learning abilities of adult male mice.
Collapse
Affiliation(s)
- Simona Gradari
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
| | - Antonio Herrera
- Institute of Molecular Biology, Consejo Superior de Investigaciones Científicas, 08028 Barcelona, Spain
| | - Patricia Tezanos
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
| | - Ángela Fontán-Lozano
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
- Department of Physiology, School of Biology, University of Sevilla, 41004 Sevilla, Spain
| | - Sebastián Pons
- Institute of Molecular Biology, Consejo Superior de Investigaciones Científicas, 08028 Barcelona, Spain
| | - José Luis Trejo
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
| |
Collapse
|
16
|
Peters S, Kuespert S, Wirkert E, Heydn R, Jurek B, Johannesen S, Hsam O, Korte S, Ludwig FT, Mecklenburg L, Mrowetz H, Altendorfer B, Poupardin R, Petri S, Thal DR, Hermann A, Weishaupt JH, Weis J, Aksoylu IS, Lewandowski SA, Aigner L, Bruun TH, Bogdahn U. Reconditioning the Neurogenic Niche of Adult Non-human Primates by Antisense Oligonucleotide-Mediated Attenuation of TGFβ Signaling. Neurotherapeutics 2021; 18:1963-1979. [PMID: 33860461 PMCID: PMC8609055 DOI: 10.1007/s13311-021-01045-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2021] [Indexed: 02/04/2023] Open
Abstract
Adult neurogenesis is a target for brain rejuvenation as well as regeneration in aging and disease. Numerous approaches showed efficacy to elevate neurogenesis in rodents, yet translation into therapies has not been achieved. Here, we introduce a novel human TGFβ-RII (Transforming Growth Factor-Receptor Type II) specific LNA-antisense oligonucleotide ("locked nucleotide acid"-"NVP-13"), which reduces TGFβ-RII expression and downstream receptor signaling in human neuronal precursor cells (ReNcell CX® cells) in vitro. After we injected cynomolgus non-human primates repeatedly i.th. with NVP-13 in a preclinical regulatory 13-week GLP-toxicity program, we could specifically downregulate TGFβ-RII mRNA and protein in vivo. Subsequently, we observed a dose-dependent upregulation of the neurogenic niche activity within the hippocampus and subventricular zone: human neural progenitor cells showed significantly (up to threefold over control) enhanced differentiation and cell numbers. NVP-13 treatment modulated canonical and non-canonical TGFβ pathways, such as MAPK and PI3K, as well as key transcription factors and epigenetic factors involved in stem cell maintenance, such as MEF2A and pFoxO3. The latter are also dysregulated in clinical neurodegeneration, such as amyotrophic lateral sclerosis. Here, we provide for the first time in vitro and in vivo evidence for a novel translatable approach to treat neurodegenerative disorders by modulating neurogenesis.
Collapse
Affiliation(s)
- Sebastian Peters
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
- Velvio GmbH, Am Biopark 11, Regensburg, Germany
| | - Sabrina Kuespert
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
- Velvio GmbH, Am Biopark 11, Regensburg, Germany
| | - Eva Wirkert
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
- Velvio GmbH, Am Biopark 11, Regensburg, Germany
| | - Rosmarie Heydn
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
- Velvio GmbH, Am Biopark 11, Regensburg, Germany
| | - Benjamin Jurek
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | - Siw Johannesen
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
- Velvio GmbH, Am Biopark 11, Regensburg, Germany
| | - Ohnmar Hsam
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Sven Korte
- Covance Preclinical Services GmbH, Muenster, Germany
| | | | | | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria
- Institute of Experimental and Clinical Cell Therapy, Spinal Cord Injury and Tissue Regeneration Center (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria
- Institute of Experimental and Clinical Cell Therapy, Spinal Cord Injury and Tissue Regeneration Center (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Rodolphe Poupardin
- Institute of Experimental and Clinical Cell Therapy, Spinal Cord Injury and Tissue Regeneration Center (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Susanne Petri
- Department of Neurology, University Hospital MHH, Hannover, Germany
| | - Dietmar R Thal
- Department for Imaging and Pathology, Laboratory for Neuropathology, University of Leuven, Leuven, Belgium
- Laboratory of Neuropathology, Institute of Pathology, Ulm University, Ulm, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section "Albrecht-Kossel", Department of Neurology, University Medical Center Rostock, University of Rostock, and German Center for Neurodegenerative Diseases (DZNE) Rostock, Rostock, Germany
| | - Jochen H Weishaupt
- Department of Neurology, University Hospital Mannheim, Mannheim, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| | - Inci Sevval Aksoylu
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Sebastian A Lewandowski
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
- SciLifeLab, School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria
- Institute of Experimental and Clinical Cell Therapy, Spinal Cord Injury and Tissue Regeneration Center (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Tim-Henrik Bruun
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
- Velvio GmbH, Am Biopark 11, Regensburg, Germany
| | - Ulrich Bogdahn
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany.
- Velvio GmbH, Am Biopark 11, Regensburg, Germany.
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria.
| |
Collapse
|
17
|
TGF-β/Smad Signalling in Neurogenesis: Implications for Neuropsychiatric Diseases. Cells 2021; 10:cells10061382. [PMID: 34205102 PMCID: PMC8226492 DOI: 10.3390/cells10061382] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
TGF-β/Smad signalling has been the subject of extensive research due to its role in the cell cycle and carcinogenesis. Modifications to the TGF-β/Smad signalling pathway have been found to produce disparate effects on neurogenesis. We review the current research on canonical and non-canonical TGF-β/Smad signalling pathways and their functions in neurogenesis. We also examine the observed role of neurogenesis in neuropsychiatric disorders and the relationship between TGF-β/Smad signalling and neurogenesis in response to stressors. Overlapping mechanisms of cell proliferation, neurogenesis, and the development of mood disorders in response to stressors suggest that TGF-β/Smad signalling is an important regulator of stress response and is implicated in the behavioural outcomes of mood disorders.
Collapse
|
18
|
Audesse AJ, Webb AE. Mechanisms of enhanced quiescence in neural stem cell aging. Mech Ageing Dev 2020; 191:111323. [PMID: 32781077 DOI: 10.1016/j.mad.2020.111323] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/24/2022]
Abstract
The maintenance of neural stem cell function is vital to ensure neurogenesis throughout adulthood. During aging, there is a significant reduction in adult neurogenesis that correlates with a decline in cognitive function. Although recent studies have revealed novel extrinsic and intrinsic mechanisms that regulate the adult neural stem cell (NSC) pool and lineage progression, the precise molecular mechanisms that drive dysregulation of adult neurogenesis in the context of aging are only beginning to emerge. Recent studies have shed light on mechanisms that regulate the earliest step of adult neurogenesis, the activation of quiescent NSCs. Interestingly, the ability of NSCs to enter the cell cycle in the aged brain significantly declines suggesting a deepend state of quiescence. Given the likely contribution of adult neurogenesis to supporting cognitive function in humans, enhancing neurogenesis may be a strategy to combat age-related cognitive decline. This review highlights the mechanisms that regulate the NSC pool throughout adulthood and discusses how dysregulation of these processes may contribute to the decline in neurogenesis and cognitive function throughout aging.
Collapse
Affiliation(s)
- Amanda J Audesse
- Graduate Program in Neuroscience, Brown University, USA; Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA; Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA; Center on the Biology of Aging, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
19
|
Kandasamy M, Anusuyadevi M, Aigner KM, Unger MS, Kniewallner KM, de Sousa DMB, Altendorfer B, Mrowetz H, Bogdahn U, Aigner L. TGF-β Signaling: A Therapeutic Target to Reinstate Regenerative Plasticity in Vascular Dementia? Aging Dis 2020; 11:828-850. [PMID: 32765949 PMCID: PMC7390515 DOI: 10.14336/ad.2020.0222] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/22/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular dementia (VaD) is the second leading form of memory loss after Alzheimer's disease (AD). Currently, there is no cure available. The etiology, pathophysiology and clinical manifestations of VaD are extremely heterogeneous, but the impaired cerebral blood flow (CBF) represents a common denominator of VaD. The latter might be the result of atherosclerosis, amyloid angiopathy, microbleeding and micro-strokes, together causing blood-brain barrier (BBB) dysfunction and vessel leakage, collectively originating from the consequence of hypertension, one of the main risk factors for VaD. At the histopathological level, VaD displays abnormal vascular remodeling, endothelial cell death, string vessel formation, pericyte responses, fibrosis, astrogliosis, sclerosis, microglia activation, neuroinflammation, demyelination, white matter lesions, deprivation of synapses and neuronal loss. The transforming growth factor (TGF) β has been identified as one of the key molecular factors involved in the aforementioned various pathological aspects. Thus, targeting TGF-β signaling in the brain might be a promising therapeutic strategy to mitigate vascular pathology and improve cognitive functions in patients with VaD. This review revisits the recent understanding of the role of TGF-β in VaD and associated pathological hallmarks. It further explores the potential to modulate certain aspects of VaD pathology by targeting TGF-β signaling.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi, India.
| | - Muthuswamy Anusuyadevi
- Molecular Gerontology Group, Department of Biochemistry, School of Life Sciences, Bharathidhasan University, Tiruchirappalli, Tamil Nadu, India.
| | - Kiera M Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Michael S Unger
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Kathrin M Kniewallner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Diana M Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Ulrich Bogdahn
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Velvio GmbH, Regensburg, Germany.
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
20
|
Kato T, Sekine Y, Nozaki H, Uemura M, Ando S, Hirokawa S, Onodera O. Excessive Production of Transforming Growth Factor β1 Causes Mural Cell Depletion From Cerebral Small Vessels. Front Aging Neurosci 2020; 12:151. [PMID: 32581764 PMCID: PMC7283554 DOI: 10.3389/fnagi.2020.00151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 05/05/2020] [Indexed: 12/14/2022] Open
Abstract
It is increasingly becoming apparent that cerebrovascular dysfunction contributes to the pathogenic processes involved in vascular dementia, Alzheimer’s disease, and other neurodegenerative disorders. Under these pathologic conditions, the degeneration of cerebral blood vessels is frequently accompanied by a loss of mural cells from the vascular walls. Vascular mural cells play pivotal roles in cerebrovascular functions, such as regulation of cerebral blood flow and maintenance of the blood-brain barrier (BBB). Therefore, cerebrovascular mural cell impairment is involved in the pathophysiology of vascular-related encephalopathies, and protecting these cells is essential for maintaining brain health. However, our understanding of the molecular mechanism underlying mural cell abnormalities is incomplete. Several reports have indicated that dysregulated transforming growth factor β (TGFβ) signaling is involved in the development of cerebral arteriopathies. These studies have specifically suggested the involvement of TGFβ overproduction. Although cerebrovascular toxicity via vascular fibrosis by extracellular matrix accumulation or amyloid deposition is known to occur with enhanced TGFβ production, whether increased TGFβ results in the degeneration of vascular mural cells in vivo remains unknown. Here, we demonstrated that chronic TGFβ1 overproduction causes a dropout of mural cells and reduces their coverage on cerebral vessels in both smooth muscle cells and pericytes. Mural cell degeneration was also accompanied by vascular luminal dilation. TGFβ1 overproduction in astrocytes significantly increased TGFβ1 content in the cerebrospinal fluid (CSF) and increased TGFβ signaling-regulated gene expression in both pial arteries and brain capillaries. These results indicate that TGFβ is an important effector that mediates mural cell abnormalities under pathological conditions related to cerebral arteriopathies.
Collapse
Affiliation(s)
- Taisuke Kato
- Department of System Pathology for Neurological Disorders, Brain Science Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yumi Sekine
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroaki Nozaki
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masahiro Uemura
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Shoichiro Ando
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Sachiko Hirokawa
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
21
|
Fessel J. If ineffective levels of transforming growth factors and their receptor account for old age being a risk factor for Alzheimer's disease, then increasing TGFBR2 might be therapeutic. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12019. [PMID: 32382652 PMCID: PMC7202202 DOI: 10.1002/trc2.12019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/02/2020] [Accepted: 02/17/2020] [Indexed: 12/19/2022]
Abstract
If it is correct that ineffective levels of transforming growth factors beta and their receptor account for old age being a risk factor for Alzheimer's disease (AD), then increasing TGFBR2 might be therapeutic. Pacltaxel is a direct way to increase TGFBR2 levels. Indirect ways that will increase TGFBR2, include decreasing the levels of c-myc because that will lower the miRNA cluster 17-92, particularly its miR-17 and miR-20a components; and raising EGFR because that also will increase TGFBR2. Metformin and desferrioxamine are drugs that decrease c-myc; and statins increase levels of EGF. Clinical trials using those drugs, would demonstrate whether they decrease the progression from amnestic mild cognitive impairment to AD.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
| |
Collapse
|
22
|
Bobkova NV, Poltavtseva RA, Leonov SV, Sukhikh GT. Neuroregeneration: Regulation in Neurodegenerative Diseases and Aging. BIOCHEMISTRY (MOSCOW) 2020; 85:S108-S130. [PMID: 32087056 DOI: 10.1134/s0006297920140060] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It had been commonly believed for a long time, that once established, degeneration of the central nervous system (CNS) is irreparable, and that adult person merely cannot restore dead or injured neurons. The existence of stem cells (SCs) in the mature brain, an organ with minimal regenerative ability, had been ignored for many years. Currently accepted that specific structures of the adult brain contain neural SCs (NSCs) that can self-renew and generate terminally differentiated brain cells, including neurons and glia. However, their contribution to the regulation of brain activity and brain regeneration in natural aging and pathology is still a subject of ongoing studies. Since the 1970s, when Fuad Lechin suggested the existence of repair mechanisms in the brain, new exhilarating data from scientists around the world have expanded our knowledge on the mechanisms implicated in the generation of various cell phenotypes supporting the brain, regulation of brain activity by these newly generated cells, and participation of SCs in brain homeostasis and regeneration. The prospects of the SC research are truthfully infinite and hitherto challenging to forecast. Once researchers resolve the issues regarding SC expansion and maintenance, the implementation of the SC-based platform could help to treat tissues and organs impaired or damaged in many devastating human diseases. Over the past 10 years, the number of studies on SCs has increased exponentially, and we have already become witnesses of crucial discoveries in SC biology. Comprehension of the mechanisms of neurogenesis regulation is essential for the development of new therapeutic approaches for currently incurable neurodegenerative diseases and neuroblastomas. In this review, we present the latest achievements in this fast-moving field and discuss essential aspects of NSC biology, including SC regulation by hormones, neurotransmitters, and transcription factors, along with the achievements of genetic and chemical reprogramming for the safe use of SCs in vitro and in vivo.
Collapse
Affiliation(s)
- N V Bobkova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - R A Poltavtseva
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia. .,National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V. I. Kulakov, Ministry of Healthcare of Russian Federation, Moscow, 117997, Russia
| | - S V Leonov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia. .,Moscow Institute of Physics and Technology (National Research University), The Phystech School of Biological and Medical Physics, Dolgoprudny, Moscow Region, 141700, Russia
| | - G T Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V. I. Kulakov, Ministry of Healthcare of Russian Federation, Moscow, 117997, Russia.
| |
Collapse
|
23
|
Chen MB, Yang AC, Yousef H, Lee D, Chen W, Schaum N, Lehallier B, Quake SR, Wyss-Coray T. Brain Endothelial Cells Are Exquisite Sensors of Age-Related Circulatory Cues. Cell Rep 2020; 30:4418-4432.e4. [PMID: 32234477 PMCID: PMC7292569 DOI: 10.1016/j.celrep.2020.03.012] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 11/13/2019] [Accepted: 03/05/2020] [Indexed: 12/28/2022] Open
Abstract
Brain endothelial cells (BECs) are key constituents of the blood-brain barrier (BBB), protecting the brain from pathogens and restricting access of circulatory factors. Yet, because circulatory proteins have prominent age-related effects on adult neurogenesis, neuroinflammation, and cognitive function in mice, we wondered whether BECs receive and potentially relay signals between the blood and brain. Using single-cell RNA sequencing of hippocampal BECs, we discover that capillary BECs-compared with arterial and venous BECs-undergo the greatest transcriptional changes in normal aging, upregulating innate immunity and oxidative stress response pathways. Short-term infusions of aged plasma into young mice recapitulate key aspects of this aging transcriptome, and remarkably, infusions of young plasma into aged mice exert rejuvenation effects on the capillary transcriptome. Together, these findings suggest that the transcriptional age of BECs is exquisitely sensitive to age-related circulatory cues and pinpoint the BBB itself as a promising therapeutic target to treat brain disease.
Collapse
Affiliation(s)
- Michelle B Chen
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Andrew C Yang
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA; ChEM-H, Stanford University, Stanford, CA, USA
| | - Hanadie Yousef
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Davis Lee
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Winnie Chen
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Nicholas Schaum
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Benoit Lehallier
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub, Stanford, CA 94305, USA.
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA; ChEM-H, Stanford University, Stanford, CA, USA; Department of Veterans Affairs, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
24
|
Chompre G, Martinez-Orengo N, Cruz M, Porter JT, Noel RJ. TGFβRI antagonist inhibits HIV-1 Nef-induced CC chemokine family ligand 2 (CCL2) in the brain and prevents spatial learning impairment. J Neuroinflammation 2019; 16:262. [PMID: 31829243 PMCID: PMC6905066 DOI: 10.1186/s12974-019-1664-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 11/27/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND HIV-1-associated neurocognitive disorders (HAND) progression is related to continued inflammation despite undetectable viral loads and may be caused by early viral proteins expressed by latently infected cells. Astrocytes represent an HIV reservoir in the brain where the early viral neurotoxin negative factor (Nef) is produced. We previously demonstrated that astrocytic expression of Nef in the hippocampus of rats causes inflammation, macrophage infiltration, and memory impairment. Since these processes are affected by TGFβ signaling pathways, and TGFβ-1 is found at higher levels in the central nervous system of HIV-1+ individuals and is released by astrocytes, we hypothesized a role for TGFβ-1 in our model of Nef neurotoxicity. METHODS To test this hypothesis, we compared cytokine gene expression by cultured astrocytes expressing Nef or green fluorescent protein. To determine the role of Nef and a TGFβRI inhibitor on memory and learning, we infused astrocytes expressing Nef into the hippocampus of rats and then treated them daily with an oral dose of SD208 (10 mg/kg) or placebo for 7 days. During this time, locomotor activity was recorded in an open field and spatial learning tested in the novel location recognition paradigm. Postmortem tissue analyses of inflammatory and signaling molecules were conducted using immunohistochemistry and immunofluorescence. RESULTS TGFβ-1 was induced in cultures expressing Nef at 24 h followed by CCL2 induction which was prevented by blocking TGFβRI with SD208 (competitive inhibitor). Interestingly, Nef seems to change the TGFβRI localization as suggested by the distribution of the immunoreactivity. Nef caused a deficit in spatial learning that was recovered upon co-administration of SD208. Brain tissue from Nef-treated rats given SD208 showed reduced CCL2, phospho-SMAD2, cluster of differentiation 163 (CD163), and GFAP immunoreactivity compared to the placebo group. CONCLUSIONS Consistent with our previous findings, rats treated with Nef showed deficits in spatial learning and memory in the novel location recognition task. In contrast, rats treated with Nef + SD208 showed better spatial learning suggesting that Nef disrupts memory formation in a TGFβ-1-dependent manner. The TGFβRI inhibitor further reduced the induction of inflammation by Nef which was concomitant with decreased TGFβ signaling. Our findings suggest that TGFβ-1 signaling is an intriguing target to reduce neuroHIV.
Collapse
Affiliation(s)
- Gladys Chompre
- Biology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico
| | - Neysha Martinez-Orengo
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Medical School, Ponce Research Institute, P.O. Box 7004, Ponce, PR, 00731, USA
| | - Myrella Cruz
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Medical School, Ponce Research Institute, P.O. Box 7004, Ponce, PR, 00731, USA
| | - James T Porter
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Medical School, Ponce Research Institute, P.O. Box 7004, Ponce, PR, 00731, USA
| | - Richard J Noel
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Medical School, Ponce Research Institute, P.O. Box 7004, Ponce, PR, 00731, USA.
| |
Collapse
|
25
|
Fessel J. Ineffective levels of transforming growth factors and their receptor account for old age being a risk factor for Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:899-905. [PMID: 31890854 PMCID: PMC6926356 DOI: 10.1016/j.trci.2019.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
After the midninth decade of age, the incidence rates of Alzheimer's disease (AD) and the presence of active TGF-β1 show comparable increases. The hypothesis is proposed that the reason why advanced age is a major risk factor for AD is a progressive decrease with advancing age in the numbers of TGFR2 receptors in the brain, with the consequence of a decline in the neurotrophic efficacy of TGF-β1 and 2 despite their already increased levels in older persons. Alternative, possible reasons are discussed but rejected because either those reasons may also affect young persons or because they cannot be validated in a clinical trial. The proposed hypothesis may be validated in persons with aMCI after raising their brain levels of TGF-β1 and 2 by using a combination of three drugs, lithium, memantine, plus either glatiramer or venlafaxine, and then assessing their progression to AD.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Emeritus, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
26
|
TGF-β Signaling in Cellular Senescence and Aging-Related Pathology. Int J Mol Sci 2019; 20:ijms20205002. [PMID: 31658594 PMCID: PMC6834140 DOI: 10.3390/ijms20205002] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/27/2022] Open
Abstract
Aging is broadly defined as the functional decline that occurs in all body systems. The accumulation of senescent cells is considered a hallmark of aging and thought to contribute to the aging pathologies. Transforming growth factor-β (TGF-β) is a pleiotropic cytokine that regulates a myriad of cellular processes and has important roles in embryonic development, physiological tissue homeostasis, and various pathological conditions. TGF-β exerts potent growth inhibitory activities in various cell types, and multiple growth regulatory mechanisms have reportedly been linked to the phenotypes of cellular senescence and stem cell aging in previous studies. In addition, accumulated evidence has indicated a multifaceted association between TGF-β signaling and aging-associated disorders, including Alzheimer’s disease, muscle atrophy, and obesity. The findings regarding these diseases suggest that the impairment of TGF-β signaling in certain cell types and the upregulation of TGF-β ligands contribute to cell degeneration, tissue fibrosis, inflammation, decreased regeneration capacity, and metabolic malfunction. While the biological roles of TGF-β depend highly on cell types and cellular contexts, aging-associated changes are an important additional context which warrants further investigation to better understand the involvement in various diseases and develop therapeutic options. The present review summarizes the relationships between TGF-β signaling and cellular senescence, stem cell aging, and aging-related diseases.
Collapse
|
27
|
Trigiani LJ, Royea J, Tong XK, Hamel E. Comparative benefits of simvastatin and exercise in a mouse model of vascular cognitive impairment and dementia. FASEB J 2019; 33:13280-13293. [PMID: 31557051 PMCID: PMC6894065 DOI: 10.1096/fj.201901002r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Aerobic physical exercise (EX) and controlling cardiovascular risk factors in midlife can improve and protect cognitive function in healthy individuals and are considered to be effective at reducing late-onset dementia incidence. By investigating commonalities between these preventative approaches, we sought to identify possible targets for effective interventions. We compared the efficacy of EX and simvastatin (SV) pharmacotherapy to counteract cognitive deficits induced by a high-cholesterol diet (2%, HCD) in mice overexpressing TGF-β1 (TGF mice), a model of vascular cognitive impairment and dementia. Cognitive deficits were found in hypercholesterolemic mice for object recognition memory, and both SV and EX prevented this decline. EX improved stimulus-evoked cerebral blood flow responses and was as effective as SV in normalizing endothelium-dependent vasodilatory responses in cerebral arteries. The up-regulation of galectin-3-positive microglial cells in white matter (WM) of HCD-fed TGF mice with cognitive deficits was significantly reduced by both SV and EX concurrently with cognitive recovery. Altered hippocampal neurogenesis, gray matter astrogliosis, or microgliosis did not correlate with cognitive deficits or benefits. Overall, results indicate that SV and EX prevented cognitive decline in hypercholesterolemic mice and that they share common sites of action in preventing endothelial cell dysfunction and reducing WM inflammation.-Trigiani, L. J., Royea, J., Tong, X.-K., Hamel, E. Comparative benefits of simvastatin and exercise in a mouse model of vascular cognitive impairment and dementia.
Collapse
Affiliation(s)
- Lianne J Trigiani
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jessika Royea
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Xin-Kang Tong
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
28
|
Ceci M, Mariano V, Romano N. Zebrafish as a translational regeneration model to study the activation of neural stem cells and role of their environment. Rev Neurosci 2019; 30:45-66. [PMID: 30067512 DOI: 10.1515/revneuro-2018-0020] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023]
Abstract
The review is an overview of the current knowledge of neuronal regeneration properties in mammals and fish. The ability to regenerate the damaged parts of the nervous tissue has been demonstrated in all vertebrates. Notably, fish and amphibians have the highest capacity for neurogenesis, whereas reptiles and birds are able to only regenerate specific regions of the brain, while mammals have reduced capacity for neurogenesis. Zebrafish (Danio rerio) is a promising model of study because lesions in the brain or complete cross-section of the spinal cord are followed by an effective neuro-regeneration that successfully restores the motor function. In the brain and the spinal cord of zebrafish, stem cell activity is always able to re-activate the molecular programs required for central nervous system regeneration. In mammals, traumatic brain injuries are followed by reduced neurogenesis and poor axonal regeneration, often insufficient to functionally restore the nervous tissue, while spinal injuries are not repaired at all. The environment that surrounds the stem cell niche constituted by connective tissue and stimulating factors, including pro-inflammation molecules, seems to be a determinant in triggering stem cell proliferation and/or the trans-differentiation of connective elements (mainly fibroblasts). Investigating and comparing the neuronal regeneration in zebrafish and mammals may lead to a better understanding of the mechanisms behind neurogenesis, and the failure of the regenerative response in mammals, first of all, the role of inflammation, considered the main inhibitor of the neuronal regeneration.
Collapse
Affiliation(s)
- Marcello Ceci
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| | - Vittoria Mariano
- Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nicla Romano
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| |
Collapse
|
29
|
Baror R, Neumann B, Segel M, Chalut KJ, Fancy SPJ, Schafer DP, Franklin RJM. Transforming growth factor-beta renders ageing microglia inhibitory to oligodendrocyte generation by CNS progenitors. Glia 2019; 67:1374-1384. [PMID: 30861188 PMCID: PMC6563458 DOI: 10.1002/glia.23612] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/17/2019] [Accepted: 02/22/2019] [Indexed: 01/26/2023]
Abstract
It is now well-established that the macrophage and microglial response to CNS demyelination influences remyelination by removing myelin debris and secreting a variety of signaling molecules that influence the behaviour of oligodendrocyte progenitor cells (OPCs). Previous studies have shown that changes in microglia contribute to the age-related decline in the efficiency of remyelination. In this study, we show that microglia increase their expression of the proteoglycan NG2 with age, and that this is associated with an altered micro-niche generated by aged, but not young, microglia that can divert the differentiation OPCs from oligodendrocytes into astrocytes in vitro. We further show that these changes in ageing microglia are generated by exposure to high levels of TGFβ. Thus, our findings suggest that the rising levels of circulating TGFβ known to occur with ageing contribute to the age-related decline in remyelination by impairing the ability of microglia to promote oligodendrocyte differentiation from OPCs, and therefore could be a potential therapeutic target to promote remyelination.
Collapse
Affiliation(s)
- Roey Baror
- Wellcome‐MRC Stem Cell InstituteUniversity of CambridgeCambridgeUnited Kingdom
- Department of PaediatricsUniversity California San FranciscoSan FranciscoCalifornia
| | - Björn Neumann
- Wellcome‐MRC Stem Cell InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Michael Segel
- Wellcome‐MRC Stem Cell InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Kevin J. Chalut
- Wellcome‐MRC Stem Cell InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Stephen P. J. Fancy
- Department of PaediatricsUniversity California San FranciscoSan FranciscoCalifornia
| | - Dorothy P. Schafer
- Department of Neurobiology and the Brudnik Neuropsychiatric InstituteUniversity of Massachusetts Medical SchoolWorcesterMassachusetts
| | | |
Collapse
|
30
|
Collins RT, Flor JM, Tang X, Bange JM, Zarate YA. Parental-reported neurodevelopmental issues in Loeys-Dietz syndrome. RESEARCH IN DEVELOPMENTAL DISABILITIES 2018; 83:153-159. [PMID: 30212788 DOI: 10.1016/j.ridd.2018.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/21/2018] [Accepted: 08/06/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Loeys-Dietz syndrome (LDS) is a congenital multisystem disorder affecting the cardiovascular and musculoskeletal system. Limited data have reported neurodevelopmental (ND) issues in LDS. AIMS To determine the extent of ND issues in patients with LDS. METHODS A prospective study was performed of LDS patients or their caregivers. The study included data collected via an online survey of age-specific questions. Standard statistical methods were used for baseline and demographic characteristics, as well as group comparisons. OUTCOMES Data were obtained from 67 patients with LDS (54% female). Median age was 14.9 years. Gene mutations included TGFBR1 (39%), TGFBR2 (40%), SMAD3 (7%), and unknown (14%). Motor delays (30%, 18/61) and hypotonia (63%, 37/60) occurred frequently. Physical (62%, 39/62), occupational (41%, 23/56), and speech therapies (34%, 20/58) were common. Feeding issues were common (41%, 23/56). TGFBR1 mutations were more frequent among those with motor delays and feeding issues. CONCLUSIONS Patients with LDS and/or their caregivers report at least one ND problem in most cases, and many require therapies. These data suggest ND disorders should be considered to be part of the phenotype.
Collapse
Affiliation(s)
- R T Collins
- University of Arkansas for Medical Sciences Department of Internal Medicine and Department of Pediatrics, Division of Cardiology, and Arkansas Children's Hospital, Little Rock, AR.
| | - J M Flor
- University of Arkansas for Medical Sciences Department of Pediatrics, Division of Developmental Pediatrics, and Arkansas Children's Hospital, Little Rock, AR
| | - X Tang
- University of Arkansas for Medical Sciences Department of Pediatrics, and Arkansas Children's Hospital, Little Rock, AR
| | - J M Bange
- Louisiana State University Health Sciences Center School of Allied Health, Shreveport, LA
| | - Y A Zarate
- University of Arkansas for Medical Sciences Department of Pediatrics, Section of Genetics and Metabolism, and Arkansas Children's Hospital, Little Rock, AR
| |
Collapse
|
31
|
Schepanski S, Buss C, Hanganu-Opatz IL, Arck PC. Prenatal Immune and Endocrine Modulators of Offspring's Brain Development and Cognitive Functions Later in Life. Front Immunol 2018; 9:2186. [PMID: 30319639 PMCID: PMC6168638 DOI: 10.3389/fimmu.2018.02186] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/04/2018] [Indexed: 12/17/2022] Open
Abstract
Milestones of brain development in mammals are completed before birth, which provide the prerequisite for cognitive and intellectual performances of the offspring. Prenatal challenges, such as maternal stress experience or infections, have been linked to impaired cognitive development, poor intellectual performances as well as neurodevelopmental and psychiatric disorders in the offspring later in life. Fetal microglial cells may be the target of such challenges and could be functionally modified by maternal markers. Maternal markers can cross the placenta and reach the fetus, a phenomenon commonly referred to as “vertical transfer.” These maternal markers include hormones, such as glucocorticoids, and also maternal immune cells and cytokines, all of which can be altered in response to prenatal challenges. Whilst it is difficult to discriminate between the maternal or fetal origin of glucocorticoids and cytokines in the offspring, immune cells of maternal origin—although low in frequency—can be clearly set apart from offspring's cells in the fetal and adult brain. To date, insights into the functional role of these cells are limited, but it is emergingly recognized that these maternal microchimeric cells may affect fetal brain development, as well as post-natal cognitive performances and behavior. Moreover, the inheritance of vertically transferred cells across generations has been proposed, yielding to the presence of a microchiome in individuals. Hence, it will be one of the scientific challenges in the field of neuroimmunology to identify the functional role of maternal microchimeric cells as well as the brain microchiome. Maternal microchimeric cells, along with hormones and cytokines, may induce epigenetic changes in the fetal brain. Recent data underpin that brain development in response to prenatal stress challenges can be altered across several generations, independent of a genetic predisposition, supporting an epigenetic inheritance. We here discuss how fetal brain development and offspring's cognitive functions later in life is modulated in the turnstile of prenatal challenges by introducing novel and recently emerging pathway, involving maternal hormones and immune markers.
Collapse
Affiliation(s)
- Steven Schepanski
- Laboratory of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Buss
- Institute of Medical Psychology, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Development, Health, and Disease Research Program, University of California, Irvine, Orange, CA, United States
| | - Ileana L Hanganu-Opatz
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra C Arck
- Laboratory of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
32
|
Choi SH, Bylykbashi E, Chatila ZK, Lee SW, Pulli B, Clemenson GD, Kim E, Rompala A, Oram MK, Asselin C, Aronson J, Zhang C, Miller SJ, Lesinski A, Chen JW, Kim DY, van Praag H, Spiegelman BM, Gage FH, Tanzi RE. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer's mouse model. Science 2018; 361:eaan8821. [PMID: 30190379 PMCID: PMC6149542 DOI: 10.1126/science.aan8821] [Citation(s) in RCA: 553] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 06/04/2018] [Accepted: 07/17/2018] [Indexed: 12/24/2022]
Abstract
Adult hippocampal neurogenesis (AHN) is impaired before the onset of Alzheimer's disease (AD) pathology. We found that exercise provided cognitive benefit to 5×FAD mice, a mouse model of AD, by inducing AHN and elevating levels of brain-derived neurotrophic factor (BDNF). Neither stimulation of AHN alone, nor exercise, in the absence of increased AHN, ameliorated cognition. We successfully mimicked the beneficial effects of exercise on AD mice by genetically and pharmacologically inducing AHN in combination with elevating BDNF levels. Suppressing AHN later led to worsened cognitive performance and loss of preexisting dentate neurons. Thus, pharmacological mimetics of exercise, enhancing AHN and elevating BDNF levels, may improve cognition in AD. Furthermore, applied at early stages of AD, these mimetics may protect against subsequent neuronal cell death.
Collapse
Affiliation(s)
- Se Hoon Choi
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Enjana Bylykbashi
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Zena K Chatila
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Star W Lee
- Laboratoy of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Benjamin Pulli
- Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Gregory D Clemenson
- Laboratoy of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Eunhee Kim
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Alexander Rompala
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Mary K Oram
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Caroline Asselin
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Jenna Aronson
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Can Zhang
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Sean J Miller
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Andrea Lesinski
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - John W Chen
- Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Henriette van Praag
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, and Brain Institute, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Fred H Gage
- Laboratoy of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
33
|
Kandasamy M, Aigner L. Reactive Neuroblastosis in Huntington's Disease: A Putative Therapeutic Target for Striatal Regeneration in the Adult Brain. Front Cell Neurosci 2018; 12:37. [PMID: 29593498 PMCID: PMC5854998 DOI: 10.3389/fncel.2018.00037] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/31/2018] [Indexed: 01/19/2023] Open
Abstract
The cellular and molecular mechanisms underlying the reciprocal relationship between adult neurogenesis, cognitive and motor functions have been an important focus of investigation in the establishment of effective neural replacement therapies for neurodegenerative disorders. While neuronal loss, reactive gliosis and defects in the self-repair capacity have extensively been characterized in neurodegenerative disorders, the transient excess production of neuroblasts detected in the adult striatum of animal models of Huntington's disease (HD) and in post-mortem brain of HD patients, has only marginally been addressed. This abnormal cellular response in the striatum appears to originate from the selective proliferation and ectopic migration of neuroblasts derived from the subventricular zone (SVZ). Based on and in line with the term "reactive astrogliosis", we propose to name the observed cellular event "reactive neuroblastosis". Although, the functional relevance of reactive neuroblastosis is unknown, we speculate that this process may provide support for the tissue regeneration in compensating the structural and physiological functions of the striatum in lieu of aging or of the neurodegenerative process. Thus, in this review article, we comprehend different possibilities for the regulation of striatal neurogenesis, neuroblastosis and their functional relevance in the context of HD.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi, India
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
34
|
Shohayeb B, Diab M, Ahmed M, Ng DCH. Factors that influence adult neurogenesis as potential therapy. Transl Neurodegener 2018; 7:4. [PMID: 29484176 PMCID: PMC5822640 DOI: 10.1186/s40035-018-0109-9] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 02/16/2018] [Indexed: 12/21/2022] Open
Abstract
Adult neurogenesis involves persistent proliferative neuroprogenitor populations that reside within distinct regions of the brain. This phenomenon was first described over 50 years ago and it is now firmly established that new neurons are continually generated in distinct regions of the adult brain. The potential of enhancing the neurogenic process lies in improved brain cognition and neuronal plasticity particularly in the context of neuronal injury and neurodegenerative disorders. In addition, adult neurogenesis might also play a role in mood and affective disorders. The factors that regulate adult neurogenesis have been broadly studied. However, the underlying molecular mechanisms of regulating neurogenesis are still not fully defined. In this review, we will provide critical analysis of our current understanding of the factors and molecular mechanisms that determine neurogenesis. We will further discuss pre-clinical and clinical studies that have investigated the potential of modulating neurogenesis as therapeutic intervention in neurodegeneration.
Collapse
Affiliation(s)
- Belal Shohayeb
- 1School of Biomedical Science, Faculty of Medicine, University of Queensland, St Lucia, QLD 4067 Australia
| | - Mohamed Diab
- 2Faculty of Pharmacy, Pharos University in Alexandria, P.O. Box Sidi Gaber, Alexandria, 21311 Egypt
| | - Mazen Ahmed
- 2Faculty of Pharmacy, Pharos University in Alexandria, P.O. Box Sidi Gaber, Alexandria, 21311 Egypt
| | - Dominic Chi Hiung Ng
- 1School of Biomedical Science, Faculty of Medicine, University of Queensland, St Lucia, QLD 4067 Australia
| |
Collapse
|
35
|
Tapella L, Cerruti M, Biocotino I, Stevano A, Rocchio F, Canonico PL, Grilli M, Genazzani AA, Lim D. TGF-β2 and TGF-β3 from cultured β-amyloid-treated or 3xTg-AD-derived astrocytes may mediate astrocyte-neuron communication. Eur J Neurosci 2018; 47:211-221. [DOI: 10.1111/ejn.13819] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 12/16/2017] [Accepted: 12/18/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Laura Tapella
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Matteo Cerruti
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Isabella Biocotino
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Alessio Stevano
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Francesca Rocchio
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Pier Luigi Canonico
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| |
Collapse
|
36
|
Waschek JA, Cohen JR, Chi GC, Proszynski TJ, Niewiadomski P. PACAP Promotes Matrix-Driven Adhesion of Cultured Adult Murine Neural Progenitors. ASN Neuro 2017; 9:1759091417708720. [PMID: 28523979 PMCID: PMC5439654 DOI: 10.1177/1759091417708720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
New neurons are born throughout the life of mammals in germinal zones of the brain known as neurogenic niches: the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus of the hippocampus. These niches contain a subpopulation of cells known as adult neural progenitor cells (aNPCs), which self-renew and give rise to new neurons and glia. aNPCs are regulated by many factors present in the niche, including the extracellular matrix (ECM). We show that the neuropeptide PACAP (pituitary adenylate cyclase-activating polypeptide) affects subventricular zone-derived aNPCs by increasing their surface adhesion. Gene array and reconstitution assays indicate that this effect can be attributed to the regulation of ECM components and ECM-modifying enzymes in aNPCs by PACAP. Our work suggests that PACAP regulates a bidirectional interaction between the aNPCs and their niche: PACAP modifies ECM production and remodeling, in turn the ECM regulates progenitor cell adherence. We speculate that PACAP may in this manner help restrict adult neural progenitors to the stem cell niche in vivo, with potential significance for aNPC function in physiological and pathological states.
Collapse
Affiliation(s)
- James A Waschek
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Joseph R Cohen
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Gloria C Chi
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Tomasz J Proszynski
- 2 Department of Cell Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel Niewiadomski
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,2 Department of Cell Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.,3 Centre of New Technologies, University of Warsaw, Warsaw, Poland
| |
Collapse
|
37
|
Mosher KI, Schaffer DV. Influence of hippocampal niche signals on neural stem cell functions during aging. Cell Tissue Res 2017; 371:115-124. [PMID: 29124394 DOI: 10.1007/s00441-017-2709-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 10/09/2017] [Indexed: 12/18/2022]
Abstract
The genesis of new neurons from neural stem cells in the adult brain offers the hope that this mechanism of plasticity can be harnessed for the treatment of brain injuries and diseases. However, neurogenesis becomes impaired during the normal course of aging; this is also the primary risk factor for most neurodegenerative diseases. The local microenvironment that regulates the function of resident neural stem cells (the "neurogenic niche") is a particularly complex network of various signaling mechanisms, rendering it especially challenging for the dissection of the control of these cells but offering the potential for the advancement of our understanding of the regulation/misregulation of neurogenesis. In this review, we examine the factors that control neurogenesis in an age-dependent manner, and we define these signals by the extrinsic mechanism through which they are presented to the neural stem cells. Secreted signals, cell-contact-dependent signals, and extracellular matrix cues all contribute to the regulation of the aging neurogenic niche and offer points of therapeutic intervention.
Collapse
Affiliation(s)
- Kira Irving Mosher
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA 94720, USA.
| | - David V Schaffer
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA 94720, USA.,Department of Chemical and Biomolecular Engineering, University of California at Berkeley, Berkeley, CA 94720, USA.,Department of Bioengineering, University of California at Berkeley, Berkeley, CA 94720, USA.,Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
38
|
Smith LK, White CW, Villeda SA. The systemic environment: at the interface of aging and adult neurogenesis. Cell Tissue Res 2017; 371:105-113. [PMID: 29124393 PMCID: PMC5748432 DOI: 10.1007/s00441-017-2715-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/09/2017] [Indexed: 02/06/2023]
Abstract
Aging results in impaired neurogenesis in the two neurogenic niches of the adult mammalian brain, the dentate gyrus of the hippocampus and the subventricular zone of the lateral ventricle. While significant work has characterized intrinsic cellular changes that contribute to this decline, it is increasingly apparent that the systemic environment also represents a critical driver of brain aging. Indeed, emerging studies utilizing the model of heterochronic parabiosis have revealed that immune-related molecular and cellular changes in the aging systemic environment negatively regulate adult neurogenesis. Interestingly, these studies have also demonstrated that age-related decline in neurogenesis can be ameliorated by exposure to the young systemic environment. While this burgeoning field of research is increasingly garnering interest, as yet, the precise mechanisms driving either the pro-aging effects of aged blood or the rejuvenating effects of young blood remain to be thoroughly defined. Here, we review how age-related changes in blood, blood-borne factors, and peripheral immune cells contribute to the age-related decline in adult neurogenesis in the mammalian brain, and posit both direct neural stem cell and indirect neurogenic niche-mediated mechanisms.
Collapse
Affiliation(s)
- Lucas K Smith
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, 94143, USA.,The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, 94143, USA.,Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Charles W White
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, 94143, USA.,The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, 94143, USA.,Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Saul A Villeda
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, 94143, USA. .,The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, 94143, USA. .,Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, 94143, USA. .,Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
39
|
Seth B, Yadav A, Agarwal S, Tiwari SK, Chaturvedi RK. Inhibition of the transforming growth factor-β/SMAD cascade mitigates the anti-neurogenic effects of the carbamate pesticide carbofuran. J Biol Chem 2017; 292:19423-19440. [PMID: 28982980 DOI: 10.1074/jbc.m117.798074] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 09/29/2017] [Indexed: 12/22/2022] Open
Abstract
The widely used carbamate pesticide carbofuran causes neurophysiological and neurobehavioral deficits in rodents and humans and therefore poses serious health hazards around the world. Previously, we reported that gestational carbofuran exposure has detrimental effects on hippocampal neurogenesis, the generation of new neurons from neural stem cells (NSC), in offspring. However, the underlying cellular and molecular mechanisms for carbofuran-impaired neurogenesis remain unknown. Herein, we observed that chronic carbofuran exposure from gestational day 7 to postnatal day 21 altered expression of genes and transcription factors and levels of proteins involved in neurogenesis and the TGF-β pathway (i.e. TGF-β; SMAD-2, -3, and -7; and SMURF-2) in the rat hippocampus. We found that carbofuran increases TGF-β signaling (i.e. increased phosphorylated SMAD-2/3 and reduced SMAD-7 expression) in the hippocampus, which reduced NSC proliferation because of increased p21 levels and reduced cyclin D1 levels. Moreover, the carbofuran-altered TGF-β signaling impaired neuronal differentiation (BrdU/DCX+ and BrdU/NeuN+ cells) and increased apoptosis and neurodegeneration in the hippocampus. Blockade of the TGF-β pathway with the specific inhibitor SB431542 and via SMAD-3 siRNA prevented carbofuran-mediated inhibition of neurogenesis in both hippocampal NSC cultures and the hippocampus, suggesting the specific involvement of this pathway. Of note, both in vitro and in vivo studies indicated that TGF-β pathway attenuation reverses carbofuran's inhibitory effects on neurogenesis and associated learning and memory deficits. These results suggest that carbofuran inhibits NSC proliferation and neuronal differentiation by altering TGF-β signaling. Therefore, we conclude that TGF-β may represent a potential therapeutic target against carbofuran-mediated neurotoxicity and neurogenesis disruption.
Collapse
Affiliation(s)
- Brashket Seth
- From the Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India.,the Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow 226001, Uttar Pradesh, India
| | - Anuradha Yadav
- From the Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India.,the Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow 226001, Uttar Pradesh, India
| | - Swati Agarwal
- From the Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India.,the Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, and
| | - Shashi Kant Tiwari
- From the Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India.,the Department of Pediatrics, University of California San Diego, La Jolla, California 92093
| | - Rajnish Kumar Chaturvedi
- From the Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India, .,the Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Lucknow Campus, Lucknow 226001, Uttar Pradesh, India
| |
Collapse
|
40
|
Zhu H, Gui Q, Hui X, Wang X, Jiang J, Ding L, Sun X, Wang Y, Chen H. TGF-β1/Smad3 Signaling Pathway Suppresses Cell Apoptosis in Cerebral Ischemic Stroke Rats. Med Sci Monit 2017; 23:366-376. [PMID: 28110342 PMCID: PMC5282965 DOI: 10.12659/msm.899195] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We desired to observe the changes of transforming growth factor-β1/drosophila mothers against decapentaplegic protein (TGF-β1/Smad3) signaling pathway in the hippocampus region of cerebral ischemic stroke rats so that the effects of this pathway on nerve cells can be investigated. MATERIAL AND METHODS The ischemic stroke models were built by middle cerebral artery occlusion (MCAO) in vivo and oxygen-glucose deprivation (OGD) in vitro. TGF-β1 and TGF-β1 inhibitors were injected into rat models while TGF-β1, TGF-β1 siRNA, Smad3, and Smad3 siRNA were transfected into cells. Infarct sizes were measured using triphenyltetrazolium chloride (TTC) staining, while the apoptosis rate of cells were calculated by Annexin V-fluorescein isothiocyanate/propidium iodide (Annexin V-FITC/PI) staining. Levels of TGF-β1, Smad3, and Bcl-2 were examined by real-time polymerase chain reaction (RT-PCR), immunohistochemical, and Western blot analysis. RESULTS The expressions of TGF-β1/Smad3 signal pathway were significantly increased in both model rats and BV2 cells, whereas the expression of Bcl-2 was down-regulated (P<0.05). The TGF-β1/Smad3 signal pathway exhibited protective effects, including the down-regulation of infarction size in cerebral tissues and the down-regulation of apoptosis rate of BV2 cells by increasing the expression of Bcl-2 (P<0.05). In addition, these effects could be antagonized by the corresponding inhibitors and siRNA (P<0.05). CONCLUSIONS The TGF-β1/Smad3 signaling pathway was up-regulated once cerebral ischemic stroke was simulated. TGF-β1 may activate the expression of Bcl-2 via Smad3 to suppress the apoptosis of neurons.
Collapse
Affiliation(s)
- Haiping Zhu
- Department of Neurosurgery, The First People's Hospital of Changshou City, Changshou, Jiangsu, China (mainland)
| | - Qunfeng Gui
- Department of Neurosurgery, Yancheng Third People's Hospital, The affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, Jiangsu, China (mainland)
| | - Xiaobo Hui
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Xiaodong Wang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Jian Jiang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Lianshu Ding
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Xiaoyang Sun
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Yanping Wang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Huaqun Chen
- Department of Neurosurgery, Yancheng Third People's Hospital, The affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, Jiangsu, China (mainland)
| |
Collapse
|
41
|
Chio CC, Lin MT, Chang CP, Lin HJ. A positive correlation exists between neurotrauma and TGF-β1-containing microglia in rats. Eur J Clin Invest 2016; 46:1063-1069. [PMID: 27759956 DOI: 10.1111/eci.12693] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 10/17/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Transforming growth factor-beta 1 (TGF-β1) regulates many processes after traumatic brain injury (TBI). Both Neuro AiD™ (MLC601) and astragaloside (AST) attenuate microglia activation in rats with TBI. The purpose of this study was to investigate whether MLC601 or AST improves output of TBI by affecting microglial expression of TGF-β1. MATERIALS AND METHODS Adult male Sprague-Dawley rats (120 in number) were used to investigate the contribution of TGF-β1-containing microglia in the MLC601-mediated or the AST-mediated neuroprotection in the brain trauma condition using lateral fluid percussion injury. RESULTS Pearson correlation analysis revealed that there was a positive correlation between brain injury (evidenced by both brain contused volume and neurological severity score) and the cortical numbers of TGF-β1-containing microglia for the rats (n = 12) 4 days post-TBI. MLC601 or AST significantly (P < 0·05) attenuated TBI-induced brain contused volume (119 ± 14 mm3 or 108 ± 11 mm3 vs. 160 ± 21 mm3 ), neurological severity score (7·8 ± 0·3 or 8·1 ± 0·4 vs. 10·2 ± 0·5) and numbers of TGF-β1-containing microglia (6% ± 2% or 11% ± 3% vs. 79% ± 7%) for the rats 4 days post-TBI. CONCLUSIONS There was a positive correlation between TBI and cortical numbers of TGF-β1-containing microglia which could be significantly attenuated by astragaloside or NeuroAiD™ (MLC601) in rats.
Collapse
Affiliation(s)
| | - Mao-Tsun Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan.,Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Hung-Jung Lin
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan.,Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| |
Collapse
|
42
|
Yousef H, Conboy MJ, Morgenthaler A, Schlesinger C, Bugaj L, Paliwal P, Greer C, Conboy IM, Schaffer D. Systemic attenuation of the TGF-β pathway by a single drug simultaneously rejuvenates hippocampal neurogenesis and myogenesis in the same old mammal. Oncotarget 2016; 6:11959-78. [PMID: 26003168 PMCID: PMC4494916 DOI: 10.18632/oncotarget.3851] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/24/2015] [Indexed: 12/18/2022] Open
Abstract
Stem cell function declines with age largely due to the biochemical imbalances in their tissue niches, and this work demonstrates that aging imposes an elevation in transforming growth factor β (TGF-β) signaling in the neurogenic niche of the hippocampus, analogous to the previously demonstrated changes in the myogenic niche of skeletal muscle with age. Exploring the hypothesis that youthful calibration of key signaling pathways may enhance regeneration of multiple old tissues, we found that systemically attenuating TGF-β signaling with a single drug simultaneously enhanced neurogenesis and muscle regeneration in the same old mice, findings further substantiated via genetic perturbations. At the levels of cellular mechanism, our results establish that the age-specific increase in TGF-β1 in the stem cell niches of aged hippocampus involves microglia and that such an increase is pro-inflammatory both in brain and muscle, as assayed by the elevated expression of β2 microglobulin (B2M), a component of MHC class I molecules. These findings suggest that at high levels typical of aged tissues, TGF-β1 promotes inflammation instead of its canonical role in attenuating immune responses. In agreement with this conclusion, inhibition of TGF-β1 signaling normalized B2M to young levels in both studied tissues.
Collapse
Affiliation(s)
- Hanadie Yousef
- Department of Molecular and Cellular Biology, UC Berkeley, Berkeley, CA, USA.,Current address: Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michael J Conboy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), UC Berkeley, Berkeley, CA, USA
| | - Adam Morgenthaler
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), UC Berkeley, Berkeley, CA, USA
| | - Christina Schlesinger
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), UC Berkeley, Berkeley, CA, USA
| | - Lukasz Bugaj
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), UC Berkeley, Berkeley, CA, USA
| | - Preeti Paliwal
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), UC Berkeley, Berkeley, CA, USA
| | - Christopher Greer
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), UC Berkeley, Berkeley, CA, USA
| | - Irina M Conboy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), UC Berkeley, Berkeley, CA, USA
| | - David Schaffer
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), UC Berkeley, Berkeley, CA, USA.,Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA, USA.,Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, CA, USA
| |
Collapse
|
43
|
Clausi MG, Kumari E, Levison SW. Unmasking the responses of the stem cells and progenitors in the subventricular zone after neonatal and pediatric brain injuries. Neural Regen Res 2016; 11:45-8. [PMID: 26981076 PMCID: PMC4774221 DOI: 10.4103/1673-5374.175041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
There is great interest in the regenerative potential of the neural stem cells and progenitors that populate the subventricular zone (SVZ). However, a comprehensive understanding of SVZ cell responses to brain injuries has been hindered by the lack of sensitive approaches to study the cellular composition of this niche. Here we review progress being made in deciphering the cells of the SVZ gleaned from the use of a recently designed flow cytometry panel that allows SVZ cells to be parsed into multiple subsets of progenitors as well as putative stem cells. We review how this approach has begun to unmask both the heterogeneity of SVZ cells as well as the dynamic shifts in cell populations with neonatal and pediatric brain injuries. We also discuss how flow cytometric analyses also have begun to reveal how specific cytokines, such as Leukemia inhibitory factor are coordinating SVZ responses to injury.
Collapse
Affiliation(s)
- Mariano Guardia Clausi
- Department of Pharmacology, Physiology and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Ekta Kumari
- Department of Pharmacology, Physiology and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Steven W Levison
- Department of Pharmacology, Physiology and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
44
|
Kempermann G. Activity Dependency and Aging in the Regulation of Adult Neurogenesis. Cold Spring Harb Perspect Biol 2015; 7:a018929. [PMID: 26525149 PMCID: PMC4632662 DOI: 10.1101/cshperspect.a018929] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Age and activity might be considered the two antagonistic key regulators of adult neurogenesis. Adult neurogenesis decreases with age but remains present, albeit at a very low level, even in the oldest individuals. Activity, be it physical or cognitive, increases adult neurogenesis and thereby seems to counteract age effects. It is, thus, proposed that activity-dependent regulation of adult neurogenesis might contribute to some sort of "neural reserve," the brain's ability to compensate functional loss associated with aging or neurodegeneration. Activity can have nonspecific and specific effects on adult neurogenesis. Mechanistically, nonspecific stimuli that largely affect precursor cell stages might be related by the local microenvironment, whereas more specific, survival-promoting effects take place at later stages of neuronal development and require the synaptic integration of the new cell and its particular synaptic plasticity.
Collapse
Affiliation(s)
- Gerd Kempermann
- German Center for Neurodegenerative Diseases (DZNE) Dresden and Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
45
|
Kuhn HG. Control of Cell Survival in Adult Mammalian Neurogenesis. Cold Spring Harb Perspect Biol 2015; 7:cshperspect.a018895. [PMID: 26511628 DOI: 10.1101/cshperspect.a018895] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The fact that continuous proliferation of stem cells and progenitors, as well as the production of new neurons, occurs in the adult mammalian central nervous system (CNS) raises several basic questions concerning the number of neurons required in a particular system. Can we observe continued growth of brain regions that sustain neurogenesis? Or does an elimination mechanism exist to maintain a constant number of cells? If so, are old neurons replaced, or are the new neurons competing for limited network access among each other? What signals support their survival and integration and what factors are responsible for their elimination? This review will address these and other questions regarding regulatory mechanisms that control cell-death and cell-survival mechanisms during neurogenesis in the intact adult mammalian brain.
Collapse
Affiliation(s)
- H Georg Kuhn
- Center for Brain Repair and Rehabilitation, Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg 413 90, Sweden
| |
Collapse
|
46
|
Capilla-Gonzalez V, Herranz-Pérez V, García-Verdugo JM. The aged brain: genesis and fate of residual progenitor cells in the subventricular zone. Front Cell Neurosci 2015; 9:365. [PMID: 26441536 PMCID: PMC4585225 DOI: 10.3389/fncel.2015.00365] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 09/03/2015] [Indexed: 12/12/2022] Open
Abstract
Neural stem cells (NSCs) persist in the adult mammalian brain through life. The subventricular zone (SVZ) is the largest source of stem cells in the nervous system, and continuously generates new neuronal and glial cells involved in brain regeneration. During aging, the germinal potential of the SVZ suffers a widespread decline, but the causes of this turn down are not fully understood. This review provides a compilation of the current knowledge about the age-related changes in the NSC population, as well as the fate of the newly generated cells in the aged brain. It is known that the neurogenic capacity is clearly disrupted during aging, while the production of oligodendroglial cells is not compromised. Interestingly, the human brain seems to primarily preserve the ability to produce new oligodendrocytes instead of neurons, which could be related to the development of neurological disorders. Further studies in this matter are required to improve our understanding and the current strategies for fighting neurological diseases associated with senescence.
Collapse
Affiliation(s)
- Vivian Capilla-Gonzalez
- Laboratory of Comparative Neurobiology, Department of Cell Biology, Instituto Cavanilles de Biodiversidad y Biología Evolutiva, University of Valencia, CIBERNED Valencia, Spain ; Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain
| | - Vicente Herranz-Pérez
- Laboratory of Comparative Neurobiology, Department of Cell Biology, Instituto Cavanilles de Biodiversidad y Biología Evolutiva, University of Valencia, CIBERNED Valencia, Spain ; Multiple Sclerosis and Neuroregeneration Mixed Unit, IIS Hospital La Fe Valencia, Spain
| | - Jose Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Department of Cell Biology, Instituto Cavanilles de Biodiversidad y Biología Evolutiva, University of Valencia, CIBERNED Valencia, Spain ; Multiple Sclerosis and Neuroregeneration Mixed Unit, IIS Hospital La Fe Valencia, Spain
| |
Collapse
|
47
|
Wang J, Xie L, Yang C, Ren C, Zhou K, Wang B, Zhang Z, Wang Y, Jin K, Yang GY. Activated regulatory T cell regulates neural stem cell proliferation in the subventricular zone of normal and ischemic mouse brain through interleukin 10. Front Cell Neurosci 2015; 9:361. [PMID: 26441532 PMCID: PMC4568339 DOI: 10.3389/fncel.2015.00361] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/28/2015] [Indexed: 11/23/2022] Open
Abstract
Recent studies have demonstrated that the depletion of Regulatory T cells (Tregs) inhibits neural progenitor cell migration after brain ischemia. However, whether Tregs affect neural stem/progenitor cell proliferation is unclear. We explored the effect of Tregs on neurogenesis in the subventricular zone (SVZ) after ischemia. Tregs were isolated and activated in vitro. Adult male C57BL/6 mice underwent 60 min transient middle cerebral artery occlusion (tMCAO). Then Tregs (1 × 105) were injected into the left lateral ventricle (LV) of normal and ischemic mouse brain. Neurogenesis was determined by immunostaining. The mechanism was examined by inhibiting interleukin 10 (IL-10) and transforming growth factor (TGF-β) signaling. We found that the number of BrdU+ cells in the SVZ was significantly increased in the activated Tregs-treated mice. Double immunostaining showed that these BrdU+ cells expressed Mash1. Blocking IL-10 reduced the number of Mash1+/BrdU+ cells, but increased the amount of GFAP+/BrdU+ cells. Here, we conclude that activated Tregs enhanced neural stem cell (NSC) proliferation in the SVZ of normal and ischemic mice; blockage of IL-10 abolished Tregs-mediated NSC proliferation in vivo and in vitro. Our results suggest that activated Tregs promoted NSC proliferation via IL-10, which provides a new therapeutic approach for ischemic stroke.
Collapse
Affiliation(s)
- Jixian Wang
- Department of Neurology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai, China ; Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Shanghai, China ; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Luokun Xie
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Chenqi Yang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Changhong Ren
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Kaijing Zhou
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Brian Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Zhijun Zhang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Shanghai, China
| | - Yongting Wang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Shanghai, China
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Guo-Yuan Yang
- Department of Neurology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai, China ; Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Shanghai, China
| |
Collapse
|
48
|
Addington CP, Roussas A, Dutta D, Stabenfeldt SE. Endogenous repair signaling after brain injury and complementary bioengineering approaches to enhance neural regeneration. Biomark Insights 2015; 10:43-60. [PMID: 25983552 PMCID: PMC4429653 DOI: 10.4137/bmi.s20062] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/20/2015] [Accepted: 03/24/2015] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) affects 5.3 million Americans annually. Despite the many long-term deficits associated with TBI, there currently are no clinically available therapies that directly address the underlying pathologies contributing to these deficits. Preclinical studies have investigated various therapeutic approaches for TBI: two such approaches are stem cell transplantation and delivery of bioactive factors to mitigate the biochemical insult affiliated with TBI. However, success with either of these approaches has been limited largely due to the complexity of the injury microenvironment. As such, this review outlines the many factors of the injury microenvironment that mediate endogenous neural regeneration after TBI and the corresponding bioengineering approaches that harness these inherent signaling mechanisms to further amplify regenerative efforts.
Collapse
Affiliation(s)
- Caroline P Addington
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Adam Roussas
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Dipankar Dutta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Sarah E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
49
|
|
50
|
Transforming growth factor-beta in the red nucleus plays antinociceptive effect under physiological and pathological pain conditions. Neuroscience 2015; 291:37-45. [PMID: 25662509 DOI: 10.1016/j.neuroscience.2015.01.059] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 01/17/2015] [Accepted: 01/27/2015] [Indexed: 02/07/2023]
Abstract
Previous studies have demonstrated that the red nucleus (RN) participates in the modulation of neuropathic pain and plays both a facilitated role by pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-1β (IL-1β), and an inhibitory role through the anti-inflammatory cytokine IL-10. In this study, we sought to investigate the expressions and roles of transforming growth factor-beta (TGF-β), a potent anti-inflammatory cytokine, as well as its type 1 receptor (TGF-β-R1) in the RN in normal and neuropathic pain rats. Immunohistochemistry showed that TGF-β and TGF-β-R1 were constitutively expressed in the RN of normal rats, and co-localized with neurons and all three glial cell types, astrocytes, microglia and oligodendrocytes. Following spared nerve injury (SNI), the expression levels of TGF-β and TGF-β-R1 were significantly down-regulated in the RN contralateral (but not ipsilateral) to the nerve injury side of rats at one week and reached the lowest level at two weeks after SNI, and both of them were co-localized with neurons and oligodendrocytes but not with astrocytes and microglia. Microinjection of different doses of anti-TGF-β antibody (250, 125, 50 ng) into the unilateral RN of normal rats dose-dependently decreased the mechanical withdrawal threshold of contralateral (but not ipsilateral) hind paw and induced significant mechanical hypersensitivity, which was similar to mechanical allodynia induced by peripheral nerve injury. In contrast, microinjection of different doses of recombinant rat TGF-β1 (500, 250, 100 ng) into the RN contralateral to the nerve injury side of SNI rats dose-dependently increased the paw withdrawal threshold and significantly alleviated mechanical allodynia induced by SNI. These results suggest that TGF-β in the RN participates in nociceptive processing and plays antinociceptive effects under normal physiological condition and in the development of neuropathic pain induced by SNI.
Collapse
|