1
|
Al-Dhahi AS, Al-Kuraishy HM, Albuhadily AK, Al-Gareeb AI, Abdelaziz AM, Alexiou A, Papadakis M, Alruwaili M, El-Saber Batiha G. The possible role of neurogenesis activators in temporal lobe epilepsy: State of art and future perspective. Eur J Pharmacol 2025:177646. [PMID: 40258399 DOI: 10.1016/j.ejphar.2025.177646] [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: 12/12/2024] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/23/2025]
Abstract
Neurogenesis is a complex process by which the neurons and supporting cells of the central nervous system (CNS) are generated by neural stem cells. Adult hippocampal neurogenesis (AHN) in the human brain is an active process during life and plays a critical role in the regulation of memory, cognition, and mood. It has been shown that epilepsy is linked with dysregulation of AHN. Of note, AHN is very sensitive to the pathological electrical stimuli during epileptic seizures, which result in the induction of neurogenesis in acute epilepsy and inhibition of neurogenesis in chronic epilepsy. Epileptic seizure-induced neurodegeneration activates the mobilization of neural stem cells during neurogenesis to substitute for neural loss in temporal lobe epilepsy (TLE), which is the most refractory type of epilepsy. Moreover, recurrent epileptic seizures in TLE trigger neurogenesis in certain brain regions. However, AHN is a transient acute epileptic seizure that terminated with 1-4 weeks following status epilepticus (SE). Nevertheless, adult AHN is dramatically reduced in chronic epilepsy and associated with the development of cognitive impairment in TLE. These findings indicate that impairment of AHN is linked with the severity of epileptic seizures. Hence, neurogenesis activators may attenuate the pathogenesis of TLE. Therefore, this review aims to discuss and explain the beneficial role of AHN in TLE and how neurogenesis activators could be effective in the management of epilepsy.
Collapse
Affiliation(s)
- Ahmed Salem Al-Dhahi
- Consultant Neurology, Department of Neuroscience, King Fahad Specialist Hospital, Tabuk, Saudi Arabia.
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq; Jabir ibn Hayyan Medical University Al-Ameer Qu./ Najaf - Iraq Po. Box (13) Kufa, Iraq.
| | - Ahmed M Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University, Arish Branch, Arish 45511, Egypt.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Mohali, India; Department of Research & Development, Funogen, Athens, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten, Herdecke, Heusnerstrasse 40, Wuppertal 42283, Germany.
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhur University, Damanhur 22511, AlBeheira, Egypt.
| |
Collapse
|
2
|
Kodali M, Madhu LN, Somayaji Y, Attaluri S, Huard C, Panda PK, Shankar G, Rao S, Shuai B, Gonzalez JJ, Oake C, Hering C, Babu RS, Kotian S, Shetty AK. Residual microglia following short-term PLX5622 treatment in 5xFAD mice exhibit diminished NLRP3 inflammasome and mTOR signaling, and enhanced autophagy. Aging Cell 2025; 24:e14398. [PMID: 39571180 PMCID: PMC11822669 DOI: 10.1111/acel.14398] [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: 06/19/2024] [Revised: 10/09/2024] [Accepted: 10/17/2024] [Indexed: 11/27/2024] Open
Abstract
While moderately activated microglia in Alzheimer's disease (AD) are pivotal in clearing amyloid beta (Aβ), hyperactivated microglia perpetuate neuroinflammation. Prior investigations reported that the elimination of ~80% of microglia through inhibition of the colony-stimulating factor 1 receptor (CSF1R) during the advanced stage of neuroinflammation in 5xFamilial AD (5xFAD) mice mitigates synapse loss and neurodegeneration. Furthermore, prolonged CSF1R inhibition diminished the development of parenchymal plaques. Nonetheless, the effects of short-term CSF1R inhibition during the early stages of neuroinflammation on residual microglia are unknown. Therefore, we investigated the effects of 10-day CSF1R inhibition using PLX5622 in three-month-old female 5xFAD mice, a stage characterized by the onset of neuroinflammation and minimal Aβ plaques. We observed ~65% microglia depletion in the hippocampus and cerebral cortex. The leftover microglia displayed a noninflammatory phenotype with reduced NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome complexes. Moreover, plaque-associated microglia were reduced with diminished Clec7a expression. Additionally, phosphorylated S6 ribosomal protein and the protein sequestosome 1 analysis suggested reduced mechanistic targets of rapamycin (mTOR) signaling and autophagy in microglia and neurons within the hippocampus and cerebral cortex. Biochemical assays validated the inhibition of NLRP3 inflammasome activation, decreased mTOR signaling in the hippocampus and cerebral cortex, and enhanced autophagy in the hippocampus. However, short-term CSF1R inhibition did not influence Aβ plaques, soluble Aβ-42 levels, astrocyte hypertrophy, or hippocampal neurogenesis. Thus, short-term CSF1R inhibition during the early stages of neuroinflammation in 5xFAD mice promotes the retention of homeostatic microglia with diminished inflammasome activation and mTOR signaling, alongside increased autophagy.
Collapse
Affiliation(s)
- Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Leelavathi N. Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Charles Huard
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Prashanta Kumar Panda
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Goutham Shankar
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Shama Rao
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Jenny J. Gonzalez
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Chris Oake
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Catherine Hering
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Roshni Sara Babu
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Sanya Kotian
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| |
Collapse
|
3
|
Madhu LN, Kodali M, Upadhya R, Rao S, Somayaji Y, Attaluri S, Shuai B, Kirmani M, Gupta S, Maness N, Rao X, Cai JJ, Shetty AK. Extracellular vesicles from human-induced pluripotent stem cell-derived neural stem cells alleviate proinflammatory cascades within disease-associated microglia in Alzheimer's disease. J Extracell Vesicles 2024; 13:e12519. [PMID: 39499013 PMCID: PMC11536387 DOI: 10.1002/jev2.12519] [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: 03/26/2024] [Revised: 08/17/2024] [Accepted: 08/28/2024] [Indexed: 11/07/2024] Open
Abstract
As current treatments for Alzheimer's disease (AD) lack disease-modifying interventions, novel therapies capable of restraining AD progression and maintaining better brain function have great significance. Anti-inflammatory extracellular vesicles (EVs) derived from human induced pluripotent stem cell (hiPSC)-derived neural stem cells (NSCs) hold promise as a disease-modifying biologic for AD. This study directly addressed this issue by examining the effects of intranasal (IN) administrations of hiPSC-NSC-EVs in 3-month-old 5xFAD mice. IN administered hiPSC-NSC-EVs incorporated into microglia, including plaque-associated microglia, and encountered astrocyte soma and processes in the brain. Single-cell RNA sequencing revealed transcriptomic changes indicative of diminished activation of microglia and astrocytes. Multiple genes linked to disease-associated microglia, NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3)-inflammasome and interferon-1 (IFN-1) signalling displayed reduced expression in microglia. Adding hiPSC-NSC-EVs to cultured human microglia challenged with amyloid-beta oligomers resulted in similar effects. Astrocytes also displayed reduced expression of genes linked to IFN-1 and interleukin-6 signalling. Furthermore, the modulatory effects of hiPSC-NSC-EVs on microglia in the hippocampus persisted 2 months post-EV treatment without impacting their phagocytosis function. Such effects were evidenced by reductions in microglial clusters and inflammasome complexes, concentrations of mediators, and end products of NLRP3 inflammasome activation, the expression of genes and/or proteins involved in the activation of p38/mitogen-activated protein kinase and IFN-1 signalling, and unaltered phagocytosis function. The extent of astrocyte hypertrophy, amyloid-beta plaques, and p-tau were also reduced in the hippocampus. Such modulatory effects of hiPSC-NSC-EVs also led to better cognitive and mood function. Thus, early hiPSC-NSC-EV intervention in AD can maintain better brain function by reducing adverse neuroinflammatory signalling cascades, amyloid-beta plaque load, and p-tau. These results reflect the first demonstration of the efficacy of hiPSC-NSC-EVs to restrain neuroinflammatory signalling cascades in an AD model by inducing transcriptomic changes in activated microglia and reactive astrocytes.
Collapse
Affiliation(s)
- Leelavathi N. Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Shama Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Maha Kirmani
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Shreyan Gupta
- Department of Veterinary Integrative BiosciencesTexas A&M College of Veterinary Medicine, College StationTexasUSA
| | - Nathaniel Maness
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Xiaolan Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - James J. Cai
- Department of Veterinary Integrative BiosciencesTexas A&M College of Veterinary Medicine, College StationTexasUSA
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| |
Collapse
|
4
|
Kodali M, Madhu LN, Kolla VSV, Attaluri S, Huard C, Somayaji Y, Shuai B, Jordan C, Rao X, Shetty S, Shetty AK. FDA-approved cannabidiol [Epidiolex ®] alleviates Gulf War Illness-linked cognitive and mood dysfunction, hyperalgesia, neuroinflammatory signaling, and declined neurogenesis. Mil Med Res 2024; 11:61. [PMID: 39169440 PMCID: PMC11340098 DOI: 10.1186/s40779-024-00563-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Chronic Gulf War Illness (GWI) is characterized by cognitive and mood impairments, as well as persistent neuroinflammation and oxidative stress. This study aimed to investigate the efficacy of Epidiolex®, a Food and Drug Administration (FDA)-approved cannabidiol (CBD), in improving brain function in a rat model of chronic GWI. METHODS Six months after exposure to low doses of GWI-related chemicals [pyridostigmine bromide, N,N-diethyl-meta-toluamide (DEET), and permethrin (PER)] along with moderate stress, rats with chronic GWI were administered either vehicle (VEH) or CBD (20 mg/kg, oral) for 16 weeks. Neurobehavioral tests were conducted on 11 weeks after treatment initiation to evaluate the performance of rats in tasks related to associative recognition memory, object location memory, pattern separation, and sucrose preference. The effect of CBD on hyperalgesia was also examined. The brain tissues were processed for immunohistochemical and molecular studies following behavioral tests. RESULTS GWI rats treated with VEH exhibited impairments in all cognitive tasks and anhedonia, whereas CBD-treated GWI rats showed improvements in all cognitive tasks and no anhedonia. Additionally, CBD treatment alleviated hyperalgesia in GWI rats. Analysis of hippocampal tissues from VEH-treated rats revealed astrocyte hypertrophy and increased percentages of activated microglia presenting NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) complexes as well as elevated levels of proteins involved in NLRP3 inflammasome activation and Janus kinase/signal transducers and activators of the transcription (JAK/STAT) signaling. Furthermore, there were increased concentrations of proinflammatory and oxidative stress markers along with decreased neurogenesis. In contrast, the hippocampus from CBD-treated GWI rats displayed reduced levels of proteins mediating the activation of NLRP3 inflammasomes and JAK/STAT signaling, normalized concentrations of proinflammatory cytokines and oxidative stress markers, and improved neurogenesis. Notably, CBD treatment did not alter the concentration of endogenous cannabinoid anandamide in the hippocampus. CONCLUSIONS The use of an FDA-approved CBD (Epidiolex®) has been shown to effectively alleviate cognitive and mood impairments as well as hyperalgesia associated with chronic GWI. Importantly, the improvements observed in rats with chronic GWI in this study were attributed to the ability of CBD to significantly suppress signaling pathways that perpetuate chronic neuroinflammation.
Collapse
Affiliation(s)
- Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Venkata Sai Vashishta Kolla
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Charles Huard
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Chase Jordan
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Xiaolan Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Sanath Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA.
| |
Collapse
|
5
|
Kodali M, Madhu LN, Somayaji Y, Attaluri S, Huard C, Panda PK, Shankar G, Rao S, Shuai B, Gonzalez JJ, Oake C, Hering C, Babu RS, Kotian S, Shetty AK. Residual Microglia Following Short-term PLX5622 Treatment in 5xFAD Mice Exhibit Diminished NLRP3 Inflammasome and mTOR Signaling, and Enhanced Autophagy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.603157. [PMID: 39071343 PMCID: PMC11275929 DOI: 10.1101/2024.07.11.603157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Chronic neuroinflammation represents a prominent hallmark of Alzheimer's disease (AD). While moderately activated microglia are pivotal in clearing amyloid beta (Aβ), hyperactivated microglia perpetuate neuroinflammation. Prior investigations have indicated that the elimination of ∼80% of microglia through a month-long inhibition of the colony-stimulating factor 1 receptor (CSF1R) during the advanced stage of neuroinflammation in 5xFamilial AD (5xFAD) mice mitigates synapse loss and neurodegeneration without impacting Aβ levels. Furthermore, prolonged CSF1R inhibition diminished the development of parenchymal plaques. Nonetheless, the immediate effects of short-term CSF1R inhibition during the early stages of neuroinflammation on residual microglial phenotype or metabolic fitness are unknown. Therefore, we investigated the effects of 10-day CSF1R inhibition in three-month-old female 5xFAD mice, a stage characterized by the onset of neuroinflammation and minimal Aβ plaques. We observed ∼65% microglia depletion in the hippocampus and cerebral cortex. The leftover microglia demonstrated a noninflammatory phenotype, with highly branched and ramified processes and reduced NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome complexes. Moreover, plaque-associated microglia were reduced in number with diminished Clec7a (dectin-1) expression. Additionally, both microglia and neurons displayed reduced mechanistic target of rapamycin (mTOR) signaling and autophagy. Biochemical assays validated the inhibition of NLRP3 inflammasome activation, decreased mTOR signaling, and enhanced autophagy. However, short-term CSF1R inhibition did not influence Aβ plaques, soluble Aβ-42 levels, or hippocampal neurogenesis. Thus, short-term CSF1R inhibition during the early stages of neuroinflammation in 5xFAD mice promotes the retention of homeostatic microglia with diminished inflammasome activation and mTOR signaling, alongside increased autophagy.
Collapse
|
6
|
Madhu LN, Kodali M, Upadhya R, Rao S, Shuai B, Somayaji Y, Attaluri S, Kirmani M, Gupta S, Maness N, Rao X, Cai J, Shetty AK. Intranasally Administered EVs from hiPSC-derived NSCs Alter the Transcriptomic Profile of Activated Microglia and Conserve Brain Function in an Alzheimer's Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576313. [PMID: 38293018 PMCID: PMC10827207 DOI: 10.1101/2024.01.18.576313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Antiinflammatory extracellular vesicles (EVs) derived from human induced pluripotent stem cell (hiPSC)-derived neural stem cells (NSCs) hold promise as a disease-modifying biologic for Alzheimer's disease (AD). This study directly addressed this issue by examining the effects of intranasal administrations of hiPSC-NSC-EVs to 3-month-old 5xFAD mice. The EVs were internalized by all microglia, which led to reduced expression of multiple genes associated with disease-associated microglia, inflammasome, and interferon-1 signaling. Furthermore, the effects of hiPSC-NSC-EVs persisted for two months post-treatment in the hippocampus, evident from reduced microglial clusters, inflammasome complexes, and expression of proteins and/or genes linked to the activation of inflammasomes, p38/mitogen-activated protein kinase, and interferon-1 signaling. The amyloid-beta (Aβ) plaques, Aβ-42, and phosphorylated-tau concentrations were also diminished, leading to better cognitive and mood function in 5xFAD mice. Thus, early intervention with hiPSC-NSC-EVs in AD may help maintain better brain function by restraining the progression of adverse neuroinflammatory signaling cascades.
Collapse
|
7
|
Ayyubova G, Kodali M, Upadhya R, Madhu LN, Attaluri S, Somayaji Y, Shuai B, Rao S, Shankar G, Shetty AK. Extracellular vesicles from hiPSC-NSCs can prevent peripheral inflammation-induced cognitive dysfunction with inflammasome inhibition and improved neurogenesis in the hippocampus. J Neuroinflammation 2023; 20:297. [PMID: 38087314 PMCID: PMC10717852 DOI: 10.1186/s12974-023-02971-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Extracellular vesicles (EVs) released by human induced pluripotent stem cell-derived neural stem cells (hiPSC-NSCs) are enriched with miRNAs and proteins capable of mediating robust antiinflammatory activity. The lack of tumorigenic and immunogenic properties and ability to permeate the entire brain to incorporate into microglia following intranasal (IN) administrations makes them an attractive biologic for curtailing chronic neuroinflammation in neurodegenerative disorders. We tested the hypothesis that IN administrations of hiPSC-NSC-EVs can alleviate chronic neuroinflammation and cognitive impairments induced by the peripheral lipopolysaccharide (LPS) challenge. Adult male, C57BL/6J mice received intraperitoneal injections of LPS (0.75 mg/kg) for seven consecutive days. Then, the mice received either vehicle (VEH) or hiPSC-NSC-EVs (~ 10 × 109 EVs/administration, thrice over 6 days). A month later, mice in all groups were investigated for cognitive function with behavioral tests and euthanized for histological and biochemical studies. Mice receiving VEH after LPS displayed deficits in associative recognition memory, temporal pattern processing, and pattern separation. Such impairments were associated with an increased incidence of activated microglia presenting NOD-, LRR-, and pyrin domain containing 3 (NLRP3) inflammasomes, elevated levels of NLRP3 inflammasome mediators and end products, and decreased neurogenesis in the hippocampus. In contrast, the various cognitive measures in mice receiving hiPSC-NSC-EVs after LPS were closer to naive mice. Significantly, these mice displayed diminished microglial activation, NLRP3 inflammasomes, proinflammatory cytokines, and a level of neurogenesis matching age-matched naïve controls. Thus, IN administrations of hiPSC-NSC-EVs are an efficacious approach to reducing chronic neuroinflammation-induced cognitive impairments.
Collapse
Affiliation(s)
- Gunel Ayyubova
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Shama Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Goutham Shankar
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA.
| |
Collapse
|
8
|
Wei ZYD, Liang K, Shetty AK. Role of Microglia, Decreased Neurogenesis and Oligodendrocyte Depletion in Long COVID-Mediated Brain Impairments. Aging Dis 2023; 14:1958-1966. [PMID: 37815903 PMCID: PMC10676788 DOI: 10.14336/ad.2023.10918] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 10/12/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of a recent worldwide coronavirus disease-2019 (COVID-19) pandemic. SARS-CoV-2 primarily causes an acute respiratory infection but can progress into significant neurological complications in some. Moreover, patients with severe acute COVID-19 could develop debilitating long-term sequela. Long-COVID is characterized by chronic symptoms that persist months after the initial infection. Common complaints are fatigue, myalgias, depression, anxiety, and "brain fog," or cognitive and memory impairments. A recent study demonstrated that a mild COVID-19 respiratory infection could generate elevated proinflammatory cytokines and chemokines in the cerebral spinal fluid. This commentary discusses findings from this study, demonstrating that even a mild respiratory SARS-CoV-2 infection can cause considerable neuroinflammation with microglial and macrophage reactivity. Such changes could also be gleaned by measuring chemokines and cytokines in the circulating blood. Moreover, neuroinflammation caused by mild SARS-CoV-2 infection can also impair hippocampal neurogenesis, deplete oligodendrocytes, and decrease myelinated axons. All these changes likely contribute to cognitive deficits in long-COVID syndrome. Therefore, strategies capable of restraining neuroinflammation, maintaining better hippocampal neurogenesis, and preserving oligodendrocyte lineage differentiation and maturation may prevent or reduce the incidence of long-COVID after SARS-CoV-2 respiratory infection.
Collapse
Affiliation(s)
- Zhuang-Yao D. Wei
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, USA.
| | - Ketty Liang
- Sam Houston State University College of Osteopathic Medicine, Conroe, TX, USA.
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, USA.
| |
Collapse
|
9
|
Kodali M, Madhu LN, Reger RL, Milutinovic B, Upadhya R, Gonzalez JJ, Attaluri S, Shuai B, Gitai DLG, Rao S, Choi JM, Jung SY, Shetty AK. Intranasally administered human MSC-derived extracellular vesicles inhibit NLRP3-p38/MAPK signaling after TBI and prevent chronic brain dysfunction. Brain Behav Immun 2023; 108:118-134. [PMID: 36427808 PMCID: PMC9974012 DOI: 10.1016/j.bbi.2022.11.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/21/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
Traumatic brain injury (TBI) leads to lasting brain dysfunction with chronic neuroinflammation typified by nucleotide-binding domain leucine-rich repeat and pyrin domain-containing receptor 3 (NLRP3) inflammasome activation in microglia. This study probed whether a single intranasal (IN) administration of human mesenchymal stem cell-derived extracellular vesicles (hMSC-EVs) naturally enriched with activated microglia-modulating miRNAs can avert chronic adverse outcomes of TBI. Small RNA sequencing confirmed the enrichment of miRNAs capable of modulating activated microglia in hMSC-EV cargo. IN administration of hMSC-EVs into adult mice ninety minutes after the induction of a unilateral controlled cortical impact injury resulted in their incorporation into neurons and microglia in both injured and contralateral hemispheres. A single higher dose hMSC-EV treatment also inhibited NLRP3 inflammasome activation after TBI, evidenced by reduced NLRP3, apoptosis-associated speck-like protein containing a CARD, activated caspase-1, interleukin-1 beta, and IL-18 levels in the injured brain. Such inhibition in the acute phase of TBI endured in the chronic phase, which could also be gleaned from diminished NLRP3 inflammasome activation in microglia of TBI mice receiving hMSC-EVs. Proteomic analysis and validation revealed that higher dose hMSC-EV treatment thwarted the chronic activation of the p38 mitogen-activated protein kinase (MAPK) signaling pathway by IL-18, which decreased the release of proinflammatory cytokines. Inhibition of the chronic activation of NLRP3-p38/MAPK signaling after TBI also prevented long-term cognitive and mood impairments. Notably, the animals receiving higher doses of hMSC-EVs after TBI displayed better cognitive and mood function in all behavioral tests than animals receiving the vehicle after TBI. A lower dose of hMSC-EV treatment also partially improved cognitive and mood function. Thus, an optimal IN dose of hMSC-EVs naturally enriched with activated microglia-modulating miRNAs can inhibit the chronic activation of NLRP3-p38/MAPK signaling after TBI and prevent lasting brain dysfunction.
Collapse
Affiliation(s)
- Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Roxanne L Reger
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Bojana Milutinovic
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Jenny J Gonzalez
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Daniel L G Gitai
- Institute of Biological Sciences and Health, Federal University of Alagoas, Brazil
| | - Shama Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Jong M Choi
- Advanced Technology Core, Mass Spectrometry and Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Sung Y Jung
- The Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA.
| |
Collapse
|
10
|
Promise of irisin to attenuate cognitive dysfunction in aging and Alzheimer's disease. Ageing Res Rev 2022; 78:101637. [PMID: 35504553 PMCID: PMC9844023 DOI: 10.1016/j.arr.2022.101637] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 01/19/2023]
Abstract
Strategies proficient for relieving cognitive impairments in aging and Alzheimer's disease (AD) have an enormous impact. Regular physical exercise (PE) can prevent age-related dementia and slow down AD progression. However, such a lifestyle change is likely not achievable for individuals displaying age-related frailty. Hence, drugs or biologics that could simulate the benefits of PE have received much attention. Previous studies suggested that the fibronectin-domain III containing 5 (FNDC5) underlies the PE-mediated improved cognitive function. A recent study reports that PE-related cognitive benefits in aging and AD are mediated by irisin, the cleaved form of FNDC5 released into the blood after PE. Such a conclusion was apparent from the deletion of irisin through a global knockout of FNDC5, leading to the loss of PE-induced cognitive benefits or inducing memory impairments in adult or aged models. Furthermore, in AD models, peripherally administered irisin mimicked the cognitive benefits of PE by modulating neuroinflammation. This short review discusses the promise of irisin to simulate the cognitive benefits of PE in age- and AD-related dementia. In addition, critical issues such as how blood-borne irisin acts on neural cells, the role of the brain-derived neurotrophic factor in irisin-mediated cognitive benefits, and irisin's ability to inhibit neuroinflammatory cascades in aging and AD are discussed.
Collapse
|
11
|
Attaluri S, Upadhya R, Kodali M, Madhu LN, Upadhya D, Shuai B, Shetty AK. Brain-Specific Increase in Leukotriene Signaling Accompanies Chronic Neuroinflammation and Cognitive Impairment in a Model of Gulf War Illness. Front Immunol 2022; 13:853000. [PMID: 35572589 PMCID: PMC9099214 DOI: 10.3389/fimmu.2022.853000] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Persistent cognitive impairment is a primary central nervous system-related symptom in veterans afflicted with chronic Gulf War Illness (GWI). Previous studies in a rat model have revealed that cognitive dysfunction in chronic GWI is associated with neuroinflammation, typified by astrocyte hypertrophy, activated microglia, and enhanced proinflammatory cytokine levels. Studies in a mouse model of GWI have also shown upregulation of several phospholipids that serve as reservoirs of arachidonic acid, a precursor of leukotrienes (LTs). However, it is unknown whether altered LT signaling is a component of chronic neuroinflammatory conditions in GWI. Therefore, this study investigated changes in LT signaling in the brain of rats displaying significant cognitive impairments six months after exposure to GWI-related chemicals and moderate stress. The concentration of cysteinyl LTs (CysLTs), LTB4, and 5-Lipoxygenase (5-LOX), the synthesizing enzyme of LTs, were evaluated. CysLT and LTB4 concentrations were elevated in the hippocampus and the cerebral cortex, along with enhanced 5-LOX expression in neurons and microglia. Such changes were also associated with increased proinflammatory cytokine levels in the hippocampus and the cerebral cortex. Enhanced CysLT and LTB4 levels in the brain could also be gleaned from their concentrations in brain-derived extracellular vesicles in the circulating blood. The circulating blood in GWI rats displayed elevated proinflammatory cytokines with no alterations in CysLT and LTB4 concentrations. The results provide new evidence that a brain-specific increase in LT signaling is another adverse alteration that potentially contributes to the maintenance of chronic neuroinflammation in GWI. Therefore, drugs capable of modulating LT signaling may reduce neuroinflammation and improve cognitive function in GWI. Additional findings demonstrate that altered LT levels in the brain could be tracked efficiently by analyzing brain-derived EVs in the circulating blood.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX, United States
| |
Collapse
|
12
|
Attaluri S, Arora M, Madhu LN, Kodali M, Shuai B, Melissari L, Upadhya R, Rao X, Bates A, Mitra E, Ghahfarouki KR, Ravikumar MNV, Shetty AK. Oral Nano-Curcumin in a Model of Chronic Gulf War Illness Alleviates Brain Dysfunction with Modulation of Oxidative Stress, Mitochondrial Function, Neuroinflammation, Neurogenesis, and Gene Expression. Aging Dis 2022; 13:583-613. [PMID: 35371600 PMCID: PMC8947830 DOI: 10.14336/ad.2021.0829] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/29/2021] [Indexed: 12/14/2022] Open
Abstract
Unrelenting cognitive and mood impairments concomitant with incessant oxidative stress and neuroinflammation are among the significant symptoms of chronic Gulf War Illness (GWI). Curcumin (CUR), an antiinflammatory compound, has shown promise to alleviate brain dysfunction in a model of GWI following intraperitoneal administrations at a high dose. However, low bioavailability after oral treatment has hampered its clinical translation. Therefore, this study investigated the efficacy of low-dose, intermittent, oral polymer nanoparticle encapsulated CUR (nCUR) for improving brain function in a rat model of chronic GWI. Intermittent administration of 10 or 20 mg/Kg nCUR for 8 weeks in the early phase of GWI improved brain function and reduced oxidative stress (OS) and neuroinflammation. We next examined the efficacy of 12-weeks of intermittent nCUR at 10 mg/Kg in GWI animals, with treatment commencing 8 months after exposure to GWI-related chemicals and stress, mimicking treatment for the persistent cognitive and mood dysfunction displayed by veterans with GWI. GWI rats receiving nCUR exhibited better cognitive and mood function associated with improved mitochondrial function and diminished neuroinflammation in the hippocampus. Improved mitochondrial function was evident from normalized expression of OS markers, antioxidants, and mitochondrial electron transport genes, and complex proteins. Lessened neuroinflammation was noticeable from reductions in astrocyte hypertrophy, NF-kB, activated microglia with NLRP3 inflammasomes, and multiple proinflammatory cytokines. Moreover, nCUR treated animals displayed enhanced neurogenesis with a normalized expression of synaptophysin puncta, and multiple genes linked to cognitive dysfunction. Thus, low-dose, intermittent, oral nCUR therapy has promise for improving brain function in veterans with GWI.
Collapse
Affiliation(s)
- Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA.
| | - Meenakshi Arora
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, Texas, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA.
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA.
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA.
| | - Laila Melissari
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA.
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA.
| | - Xiaolan Rao
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA.
| | - Adrian Bates
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA.
| | - Eeshika Mitra
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA.
| | - Keyhan R Ghahfarouki
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA.
| | - M. N. V Ravikumar
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, Texas, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA.
| |
Collapse
|
13
|
Kodali M, Mishra V, Hattiangady B, Attaluri S, Gonzalez JJ, Shuai B, Shetty AK. Moderate, intermittent voluntary exercise in a model of Gulf War Illness improves cognitive and mood function with alleviation of activated microglia and astrocytes, and enhanced neurogenesis in the hippocampus. Brain Behav Immun 2021; 97:135-149. [PMID: 34245811 PMCID: PMC9885810 DOI: 10.1016/j.bbi.2021.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/28/2021] [Accepted: 07/04/2021] [Indexed: 02/01/2023] Open
Abstract
Persistent cognitive and mood impairments in Gulf War Illness (GWI) are associated with chronic neuroinflammation, typified by hypertrophied astrocytes, activated microglia, and increased proinflammatory mediators in the brain. Using a rat model, we investigated whether a simple lifestyle change such as moderate voluntary physical exercise would improve cognitive and mood function in GWI. Because veterans with GWI exhibit fatigue and post-exertional malaise, we employed an intermittent voluntary running exercise (RE) regimen, which prevented exercise-induced stress. The GWI rats were provided access to running wheels three days per week for 13 weeks, commencing ten weeks after the exposure to GWI-related chemicals and stress (GWI-RE group). Groups of age-matched sedentary GWI rats (GWI-SED group) and naïve rats were maintained parallelly. Interrogation of rats with behavioral tests after the 13-week RE regimen revealed improved hippocampus-dependent object location memory and pattern separation function and reduced anxiety-like behavior in the GWI-RE group compared to the GWI-SED group. Moreover, 13 weeks of RE in GWI rats significantly reversed activated microglia with short and less ramified processes into non-inflammatory/antiinflammatory microglia with highly ramified processes and reduced the hypertrophy of astrocytes. Moreover, the production of new neurons in the hippocampus was enhanced when examined eight weeks after the commencement of RE. Notably, increased neurogenesis continued even after the cessation of RE. Collectively, the results suggest that even a moderate, intermittent physical exercise has the promise to improve brain function in veterans with GWI in association with suppression of neuroinflammation and enhancement of hippocampal neurogenesis.
Collapse
Affiliation(s)
- Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, United States,Research Service, Olin E. Teague Veterans Affairs Medical Center, Central Texas Veterans Health Care System, Temple, TX, United States
| | - Vikas Mishra
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, United States,Research Service, Olin E. Teague Veterans Affairs Medical Center, Central Texas Veterans Health Care System, Temple, TX, United States
| | - Bharathi Hattiangady
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, United States,Research Service, Olin E. Teague Veterans Affairs Medical Center, Central Texas Veterans Health Care System, Temple, TX, United States
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, United States
| | - Jenny Jaimes Gonzalez
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, United States
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, United States,Research Service, Olin E. Teague Veterans Affairs Medical Center, Central Texas Veterans Health Care System, Temple, TX, United States
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, United States,Research Service, Olin E. Teague Veterans Affairs Medical Center, Central Texas Veterans Health Care System, Temple, TX, United States,Corresponding author at: Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine, 1114 TAMU, 206 Olsen Boulevard, College Station, TX 77843, United States. (A.K. Shetty)
| |
Collapse
|
14
|
Madhu LN, Kodali M, Attaluri S, Shuai B, Melissari L, Rao X, Shetty AK. Melatonin improves brain function in a model of chronic Gulf War Illness with modulation of oxidative stress, NLRP3 inflammasomes, and BDNF-ERK-CREB pathway in the hippocampus. Redox Biol 2021; 43:101973. [PMID: 33933884 PMCID: PMC8105671 DOI: 10.1016/j.redox.2021.101973] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/05/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023] Open
Abstract
Persistent cognitive and mood dysfunction is the primary CNS symptom in veterans afflicted with Gulf War Illness (GWI). This study investigated the efficacy of melatonin (MEL) for improving cognitive and mood function with antioxidant, antiinflammatory, and pro-cognitive effects in a rat model of chronic GWI. Six months after exposure to GWI-related chemicals and stress, rats were treated with vehicle or MEL (5, 10, 20, 40, and 80 mg/kg) for eight weeks. Behavioral tests revealed cognitive and mood dysfunction in GWI rats receiving vehicle, which were associated with elevated oxidative stress, reduced NRF2, catalase and mitochondrial complex proteins, astrocyte hypertrophy, activated microglia with NLRP3 inflammasomes, elevated proinflammatory cytokines, waned neurogenesis, and synapse loss in the hippocampus. MEL at 10 mg/kg alleviated simple and associative recognition memory dysfunction and anhedonia, along with reduced oxidative stress, enhanced glutathione and complex III, and reduced NLRP3 inflammasomes, IL-18, TNF-α, and IFN-γ. MEL at 20 mg/kg also normalized NRF2 and catalase and increased microglial ramification. MEL at 40 mg/kg, in addition, reduced astrocyte hypertrophy, activated microglia, NF-kB-NLRP3-caspase-1 signaling, IL-1β, MCP-1, and MIP-1α. Moreover, MEL at 80 mg/kg activated the BDNF-ERK-CREB signaling pathway, enhanced neurogenesis and diminished synapse loss in the hippocampus, and improved a more complex hippocampus-dependent cognitive function. Thus, MEL therapy is efficacious for improving cognitive and mood function in a rat model of chronic GWI, and MEL's effect was dose-dependent. The study provides the first evidence of MEL's promise for alleviating neuroinflammation and cognitive and mood impairments in veterans with chronic GWI. A low dose of Melatonin alleviated recognition memory dysfunction and anhedonia in a model of chronic GWI. A moderate dose of Melatonin improved more complex cognitive function in a model of chronic GWI. Melatonin treatment reduced oxidative stress and enhanced mitochondrial complex proteins in the GWI brain. Melatonin inhibited NLRP3 inflammasomes and proinflammatory cytokines in the GWI brain. Melatonin activated the BDNF-ERK-CREB signaling pathway and enhanced neurogenesis in the GWI brain.
Collapse
Affiliation(s)
- Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Laila Melissari
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Xiaolan Rao
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA.
| |
Collapse
|
15
|
Hattiangady B, Kuruba R, Shuai B, Grier R, Shetty AK. Hippocampal Neural Stem Cell Grafting after Status Epilepticus Alleviates Chronic Epilepsy and Abnormal Plasticity, and Maintains Better Memory and Mood Function. Aging Dis 2020; 11:1374-1394. [PMID: 33269095 PMCID: PMC7673840 DOI: 10.14336/ad.2020.1020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022] Open
Abstract
Hippocampal damage after status epilepticus (SE) leads to multiple epileptogenic changes, which lead to chronic temporal lobe epilepsy (TLE). Morbidities such as spontaneous recurrent seizures (SRS) and memory and mood impairments are seen in a significant fraction of SE survivors despite the administration of antiepileptic drugs after SE. We examined the efficacy of bilateral intra-hippocampal grafting of neural stem/progenitor cells (NSCs) derived from the embryonic day 19 rat hippocampi, six days after SE for restraining SE-induced SRS, memory, and mood impairments in the chronic phase. Grafting of NSCs curtailed the progression of SRS at 3-5 months post-SE and reduced the frequency and severity of SRS activity when examined at eight months post-SE. Reduced SRS activity was also associated with improved memory function. Graft-derived cells migrated into different hippocampal cell layers, differentiated into GABA-ergic interneurons, astrocytes, and oligodendrocytes. Significant percentages of graft-derived cells also expressed beneficial neurotrophic factors such as the fibroblast growth factor-2, brain-derived neurotrophic factor, insulin-like growth factor-1 and glial cell line-derived neurotrophic factor. NSC grafting protected neuropeptide Y- and parvalbumin-positive host interneurons, diminished the abnormal migration of newly born neurons, and rescued the reelin+ interneurons in the dentate gyrus. Besides, grafting led to the maintenance of a higher level of normal neurogenesis in the chronic phase after SE and diminished aberrant mossy fiber sprouting in the dentate gyrus. Thus, intrahippocampal grafting of hippocampal NSCs shortly after SE considerably curbed the progression of epileptogenic processes and SRS, which eventually resulted in less severe chronic epilepsy devoid of significant cognitive and mood impairments.
Collapse
Affiliation(s)
- Bharathi Hattiangady
- 1Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA.,2Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA.,3Department of Surgery (Neurosurgery) Duke University Medical Center, Durham, NC, USA.,4Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Ramkumar Kuruba
- 3Department of Surgery (Neurosurgery) Duke University Medical Center, Durham, NC, USA.,4Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Bing Shuai
- 1Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA.,2Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA.,3Department of Surgery (Neurosurgery) Duke University Medical Center, Durham, NC, USA.,4Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Remedios Grier
- 3Department of Surgery (Neurosurgery) Duke University Medical Center, Durham, NC, USA.,4Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Ashok K Shetty
- 1Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA.,2Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA.,3Department of Surgery (Neurosurgery) Duke University Medical Center, Durham, NC, USA.,4Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, NC, USA
| |
Collapse
|
16
|
Intranasally Administered Human MSC-Derived Extracellular Vesicles Pervasively Incorporate into Neurons and Microglia in both Intact and Status Epilepticus Injured Forebrain. Int J Mol Sci 2019; 21:ijms21010181. [PMID: 31888012 PMCID: PMC6981466 DOI: 10.3390/ijms21010181] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles (EVs) derived from human bone marrow mesenchymal stem cells (hMSCs) have great promise as biologics to treat neurological and neurodegenerative conditions due to their robust antiinflammatory and neuroprotective properties. Besides, intranasal (IN) administration of EVs has caught much attention because the procedure is noninvasive, amenable for repetitive dispensation, and leads to a quick penetration of EVs into multiple regions of the forebrain. Nonetheless, it is unknown whether brain injury-induced signals are essential for the entry of IN-administered EVs into different brain regions. Therefore, in this study, we investigated the distribution of IN-administered hMSC-derived EVs into neurons and microglia in the intact and status epilepticus (SE) injured rat forebrain. Ten billion EVs labeled with PKH26 were dispensed unilaterally into the left nostril of naïve rats, and rats that experienced two hours of kainate-induced SE. Six hours later, PKH26 + EVs were quantified from multiple forebrain regions using serial brain sections processed for different neural cell markers and confocal microscopy. Remarkably, EVs were seen bilaterally in virtually all regions of intact and SE-injured forebrain. The percentage of neurons incorporating EVs were comparable for most forebrain regions. However, in animals that underwent SE, a higher percentage of neurons incorporated EVs in the hippocampal CA1 subfield and the entorhinal cortex, the regions that typically display neurodegeneration after SE. In contrast, the incorporation of EVs by microglia was highly comparable in every region of the forebrain measured. Thus, unilateral IN administration of EVs is efficient for delivering EVs bilaterally into neurons and microglia in multiple regions in the intact or injured forebrain. Furthermore, incorporation of EVs by neurons is higher in areas of brain injury, implying that injury-related signals likely play a role in targeting of EVs into neurons, which may be beneficial for EV therapy in various neurodegenerative conditions including traumatic brain injury, stroke, multiple sclerosis, and Alzheimer's disease.
Collapse
|
17
|
Extracellular Vesicles in the Forebrain Display Reduced miR-346 and miR-331-3p in a Rat Model of Chronic Temporal Lobe Epilepsy. Mol Neurobiol 2019; 57:1674-1687. [PMID: 31813125 DOI: 10.1007/s12035-019-01797-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/22/2019] [Indexed: 12/20/2022]
Abstract
An initial precipitating injury in the brain, such as after status epilepticus (SE), evolves into chronic temporal lobe epilepsy (TLE). We investigated changes in the miRNA composition of extracellular vesicles (EVs) in the forebrain after the establishment of SE-induced chronic TLE. We induced SE in young Fischer 344 rats through graded intraperitoneal injections of kainic acid, which resulted in consistent spontaneous recurrent seizures at ~ 3 months post-SE. We isolated EVs from the entire forebrain of chronically epileptic rats and age-matched naïve control animals through an ultracentrifugation method and performed miRNA-sequencing studies to discern changes in the miRNA composition of forebrain-derived EVs in chronic epilepsy. EVs from both naïve and epileptic forebrains displayed spherical or cup-shaped morphology, a comparable size range, and CD63 expression but lacked the expression of a deep cellular marker GM130. However, miRNA-sequencing studies suggested downregulation of 3 miRNAs (miR-187-5p, miR-346, and miR-331-3p) and upregulation of 4 miRNAs (miR-490-5p, miR-376b-3p, miR-493-5p, and miR-124-5p) in EVs from epileptic forebrains with fold changes ranging from 1.5 to 2.4 (p < 0.0006; FDR < 0.05). By using geNorm and Normfinder software, we identified miR-487 and miR-221 as the best combination of reference genes for measurement of altered miRNAs found in the epileptic forebrain through qRT-PCR studies. The validation revealed that only miR-346 and miR-331-3p were significantly downregulated in EVs from the epileptic forebrain. The enrichment pathway analysis of these miRNAs showed an overrepresentation of signaling pathways that are linked to molecular mechanisms underlying chronic epilepsy, including GABA-ergic (miR-346 targets) and mTOR (miR-331-3p targets) systems. Thus, the packaging of two miRNAs into EVs in neural cells is considerably altered in chronic epilepsy. Functional studies on these two miRNAs may uncover their role in the pathophysiology and treatment of TLE.
Collapse
|
18
|
Shetty AK, Attaluri S, Kodali M, Shuai B, Shetty GA, Upadhya D, Hattiangady B, Madhu LN, Upadhya R, Bates A, Rao X. Monosodium luminol reinstates redox homeostasis, improves cognition, mood and neurogenesis, and alleviates neuro- and systemic inflammation in a model of Gulf War Illness. Redox Biol 2019; 28:101389. [PMID: 31778892 PMCID: PMC6888767 DOI: 10.1016/j.redox.2019.101389] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/12/2019] [Accepted: 11/15/2019] [Indexed: 12/15/2022] Open
Abstract
Enduring brain dysfunction is amid the highly manifested symptoms in veterans with Gulf War Illness (GWI). Animal studies have established that lasting brain dysfunction in GWI is concomitant with augmented oxidative stress, inflammation, and declined neurogenesis in the brain, and systemic inflammation. We hypothesize that drugs capable of restoring redox homeostasis in GWI will improve cognitive and mood function with modulation of neuroinflammation and neurogenesis. We examined the efficacy of monosodium luminol-GVT (MSL), a drug that promotes redox homeostasis, for improving cognitive and mood function in GWI rats. Young rats were exposed to GWI-related chemicals and moderate restraint stress for four weeks. Four months later, GWI rats received different doses of MSL or vehicle for eight weeks. Behavioral analyses in the last three weeks of treatment revealed that GWI rats receiving higher doses of MSL displayed better cognitive and mood function associated with reinstatement of redox homeostasis. Such restoration was evident from the normalized expression of multiple genes encoding proteins involved in combating oxidative stress in the brain and the return of several oxidative stress markers to control levels in the brain and the circulating blood. Sustained redox homeostasis by MSL also resulted in antiinflammatory and pro-neurogenic effects, which were apparent from reduced densities of hypertrophied astrocytes and activated microglia, and increased neurogenesis with augmented neural stem cell proliferation. Moreover, MSL treatment normalized the concentration of multiple proinflammatory markers in the circulating blood. Thus, MSL treatment reinstated redox homeostasis in an animal model of GWI, which resulted in alleviation of both brain and systemic inflammation, improved neurogenesis, and better cognitive and mood function. Brain dysfunction in an animal model of Gulf War Illness is linked with persistently elevated oxidative stress. Monosodium Luminol treatment reinstated redox homeostasis in a model of Gulf War Illness. Reinstatement of redox balance improved cognitive and mood function. Restoration of redox balance modulated reactive astrocytes and activated microglia in the brain. Return of redox homeostasis enhanced neurogenesis and suppressed systemic inflammation.
Collapse
Affiliation(s)
- Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA.
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Geetha A Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Dinesh Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Bharathi Hattiangady
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Adrian Bates
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Xiaolan Rao
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| |
Collapse
|
19
|
Seong KJ, Kim HJ, Cai B, Kook MS, Jung JY, Kim WJ. Toll-like receptor 2 promotes neurogenesis from the dentate gyrus after photothrombotic cerebral ischemia in mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:145-153. [PMID: 29520167 PMCID: PMC5840073 DOI: 10.4196/kjpp.2018.22.2.145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/03/2017] [Accepted: 12/07/2017] [Indexed: 01/27/2023]
Abstract
The subgranular zone (SGZ) of hippocampal dentate gyrus (HDG) is a primary site of adult neurogenesis. Toll-like receptors (TLRs), are involved in neural system development of Drosophila and innate immune response of mammals. TLR2 is expressed abundantly in neurogenic niches such as adult mammalian hippocampus. It regulates adult hippocampal neurogenesis. However, the role of TLR2 in adult neurogenesis is not well studied in global or focal cerebral ischemia. Therefore, this study aimed to investigate the role of TLR2 in adult neurogenesis after photochemically induced cerebral ischemia. At 7 days after photothrombotic ischemic injury, the number of bromodeoxyuridine (BrdU)-positive cells was increased in both TLR2 knock-out (KO) mice and wild-type (WT) mice. However, the increment rate of BrdU-positive cells was lower in TLR2 KO mice compared to that in WT mice. The number of doublecortin (DCX) and neuronal nuclei (NeuN)-positive cells in HDG was decreased after photothrombotic ischemia in TLR2 KO mice compared to that in WT mice. The survival rate of cells in HDG was decreased in TLR2 KO mice compared to that in WT mice. In contrast, the number of cleaved-caspase 3 (apoptotic marker) and the number of GFAP (glia marker)/BrdU double-positive cells in TLR2 KO mice were higher than that in WT mice. These results suggest that TLR2 can promote adult neurogenesis from neural stem cell of hippocampal dentate gyrus through increasing proliferation, differentiation, and survival from neural stem cells after ischemic injury of the brain.
Collapse
Affiliation(s)
- Kyung-Joo Seong
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Hyeong-Jun Kim
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Bangrong Cai
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Min-Suk Kook
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Ji-Yeon Jung
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Won-Jae Kim
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| |
Collapse
|
20
|
Kodali M, Hattiangady B, Shetty G, Bates A, Shuai B, Shetty A. Curcumin treatment leads to better cognitive and mood function in a model of Gulf War Illness with enhanced neurogenesis, and alleviation of inflammation and mitochondrial dysfunction in the hippocampus. Brain Behav Immun 2018; 69:499-514. [PMID: 29454881 PMCID: PMC7023905 DOI: 10.1016/j.bbi.2018.01.009] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/11/2018] [Accepted: 01/15/2018] [Indexed: 01/10/2023] Open
Abstract
Diminished cognitive and mood function are among the most conspicuous symptoms of Gulf War Illness (GWI). Our previous studies in a rat model of GWI have demonstrated that persistent cognitive and mood impairments are associated with substantially declined neurogenesis, chronic low-grade inflammation, increased oxidative stress and mitochondrial dysfunction in the hippocampus. We tested the efficacy of curcumin (CUR) to maintain better cognitive and mood function in a rat model of GWI because of its neurogenic, antiinflammatory, antioxidant, and memory and mood enhancing properties. Male rats were exposed daily to low doses of GWI-related chemicals, pyridostigmine bromide, N,N-diethyl-m-toluamide (DEET) and permethrin, and 5-minutes of restraint stress for 28 days. Animals were next randomly assigned to two groups, which received daily CUR or vehicle treatment for 30 days. Animals also received 5'-bromodeoxyuridine during the last seven days of treatment for analysis of neurogenesis. Behavioral studies through object location, novel object recognition and novelty suppressed feeding tests performed sixty days after treatment revealed better cognitive and mood function in CUR treated GWI rats. These rats also displayed enhanced neurogenesis and diminished inflammation typified by reduced astrocyte hypertrophy and activated microglia in the hippocampus. Additional studies showed that CUR treatment to GWI rats enhanced the expression of antioxidant genes and normalized the expression of multiple genes related to mitochondrial respiration. Thus, CUR therapy is efficacious for maintaining better memory and mood function in a model of GWI. Enhanced neurogenesis, restrained inflammation and oxidative stress with normalized mitochondrial respiration may underlie better memory and mood function mediated by CUR treatment.
Collapse
Affiliation(s)
- M. Kodali
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College Station, TX, USA,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - B. Hattiangady
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College Station, TX, USA,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - G.A. Shetty
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College Station, TX, USA,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - A. Bates
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College Station, TX, USA,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - B. Shuai
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College Station, TX, USA,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - A.K. Shetty
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College Station, TX, USA,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA,Corresponding author at: Institute for Regenerative Medicine, Texas A&M Health Science Center, College of Medicine, 1114 TAMU, 206 Olsen Boulevard, College Station, TX 77843, USA. (A.K. Shetty)
| |
Collapse
|
21
|
Prospects of Cannabidiol for Easing Status Epilepticus-Induced Epileptogenesis and Related Comorbidities. Mol Neurobiol 2018; 55:6956-6964. [PMID: 29372545 DOI: 10.1007/s12035-018-0898-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/09/2018] [Indexed: 12/21/2022]
Abstract
The hippocampus is one of the most susceptible regions in the brain to be distraught with status epilepticus (SE) induced injury. SE can occur from numerous causes and is more frequent in children and the elderly population. Administration of a combination of antiepileptic drugs can abolish acute seizures in most instances of SE but cannot prevent the morbidity typically seen in survivors of SE such as cognitive and mood impairments and spontaneous recurrent seizures. This is primarily due to the inefficiency of antiepileptic drugs to modify the evolution of SE-induced initial precipitating injury into a series of epileptogenic changes followed by a state of chronic epilepsy. Chronic epilepsy is typified by spontaneous recurrent seizures, cognitive dysfunction, and depression, which are associated with persistent inflammation, significantly waned neurogenesis, and abnormal synaptic reorganization. Thus, alternative approaches that are efficient not only for curtailing SE-induced initial brain injury, neuroinflammation, aberrant neurogenesis, and abnormal synaptic reorganization but also for thwarting or restraining the progression of SE into a chronic epileptic state are needed. In this review, we confer the promise of cannabidiol, an active ingredient of Cannabis sativa, for preventing or easing SE-induced neurodegeneration, neuroinflammation, cognitive and mood impairments, and the spontaneous recurrent seizures.
Collapse
|
22
|
Castro OW, Upadhya D, Kodali M, Shetty AK. Resveratrol for Easing Status Epilepticus Induced Brain Injury, Inflammation, Epileptogenesis, and Cognitive and Memory Dysfunction-Are We There Yet? Front Neurol 2017; 8:603. [PMID: 29180982 PMCID: PMC5694141 DOI: 10.3389/fneur.2017.00603] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 10/30/2017] [Indexed: 12/29/2022] Open
Abstract
Status epilepticus (SE) is a medical emergency exemplified by self-sustaining, unceasing seizures or swiftly recurring seizure events with no recovery between seizures. The early phase after SE event is associated with neurodegeneration, neuroinflammation, and abnormal neurogenesis in the hippocampus though the extent of these changes depends on the severity and duration of seizures. In many instances, over a period, the initial precipitating injury caused by SE leads to temporal lobe epilepsy (TLE), typified by spontaneous recurrent seizures, cognitive, memory and mood impairments associated with chronic inflammation, reduced neurogenesis, abnormal synaptic reorganization, and multiple molecular changes in the hippocampus. While antiepileptic drugs are efficacious for terminating or greatly reducing seizures in most cases of SE, they have proved ineffective for easing SE-induced epileptogenesis and TLE. Despite considerable advances in elucidating SE-induced multiple cellular, electrophysiological, and molecular changes in the brain, efficient strategies that prevent SE-induced TLE development are yet to be discovered. This review critically confers the efficacy and promise of resveratrol, a phytoalexin found in the skin of red grapes, for easing SE-induced neurodegeneration, neuroinflammation, aberrant neurogenesis, and for restraining the evolution of SE-induced brain injury into a chronic epileptic state typified by spontaneous recurrent seizures, and learning, memory, and mood impairments.
Collapse
Affiliation(s)
- Olagide W Castro
- Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas, United States.,Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, United States.,Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceio, Brazil
| | - Dinesh Upadhya
- Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas, United States.,Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, United States.,Department of Anatomy, Kasturba Medical College, Manipal University, Manipal, India
| | - Maheedhar Kodali
- Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas, United States.,Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, United States
| | - Ashok K Shetty
- Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas, United States.,Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, United States
| |
Collapse
|
23
|
Cipriani R, Chara JC, Rodríguez-Antigüedad A, Matute C. Effects of FTY720 on brain neurogenic niches in vitro and after kainic acid-induced injury. J Neuroinflammation 2017; 14:147. [PMID: 28738875 PMCID: PMC5525223 DOI: 10.1186/s12974-017-0922-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 07/14/2017] [Indexed: 01/28/2023] Open
Abstract
Background FTY720 (fingolimod, Gilenya™) is an oral, blood-brain barrier (BBB)-passing drug approved as immunomodulatory treatment for relapsing-remitting form of the multiple sclerosis (MS). In addition, FTY720 exerts several effects in the central nervous system (CNS), ranging from neuroprotection to reduction of neuroinflammation. However, the neurogenic and oligodendrogenic potential of FTY720 has been poorly investigated. In this study, we assessed the effect of FTY720 on the production of new neurons and oligodendrocytes from neural stem/precursor cells both in vitro and in vivo. Methods Neural stem cells (NSCs) derived from the young rat subventricular zone (SVZ) were exposed to FTY720 (10, 100 nM), and their differentiation into neurons and oligodendrocytes was measured using immunofluorescence for anti-β-III tubulin or CNPase (2′,3′-cyclic nucleotide 3′-phosphodiesterase) as markers of mature neurons or oligodendrocytes, respectively. In addition, intracerebroventricular (icv) administration of kainic acid (KA; 0.5 μg/2 μl) in Sprague-Dawley rats was used as an in vivo model of neuronal death and inflammation. FTY720 was applied icv (1 μg/2 μl), together with KA, plus intraperitoneally (ip; 1 mg/kg) 24 h before, and daily, until sacrifice 8 days after KA injection. To visualize cell proliferation in the hippocampus and in white matter regions, rats were administered 5-bromo-2-deoxyuridine (BrdU) 100 mg/kg, ip injected every 2 days. Immunohistochemical analyses were performed on rat brain slices to measure the production of new neuronal precursors (doublecortin/DCX+ cells) and new oligodendrocytes precursors (proteoglycan/NG2+ cells). Results In this study, we observed that FTY720 increased postnatal NSCs differentiation into both neurons and oligodendrocytes in vitro. In turn, in adult animals, FTY720 enhanced the percentage of BrdU+ cells coexpressing DCX marker, both in basal (FTY720 alone) and in neurodegenerative (FTY720 + KA) conditions. However, FTY720 had only a partial effect on proliferation and differentiation of oligodendrocyte progenitor cell (OPC) population in vivo. Conclusions FTY720 promotes neurogenesis and oligodendrogenesis in vitro under basal conditions. In addition, it increases the generation of neuroblasts and oligodendrocytes after excitotoxic brain injury. This suggests that FTY720 has the potential to activate the neurogenic niche and thus favour tissue repair after lesion. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0922-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raffaela Cipriani
- Centro de Investigaciones Biomédicas en Red (CIBERNED), Achucarro Basque Center for Neuroscience and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), E-48940, Leioa, Spain.
| | - Juan Carlos Chara
- Centro de Investigaciones Biomédicas en Red (CIBERNED), Achucarro Basque Center for Neuroscience and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), E-48940, Leioa, Spain
| | | | - Carlos Matute
- Centro de Investigaciones Biomédicas en Red (CIBERNED), Achucarro Basque Center for Neuroscience and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), E-48940, Leioa, Spain
| |
Collapse
|
24
|
Ihunwo AO, Tembo LH, Dzamalala C. The dynamics of adult neurogenesis in human hippocampus. Neural Regen Res 2016; 11:1869-1883. [PMID: 28197172 PMCID: PMC5270414 DOI: 10.4103/1673-5374.195278] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2016] [Indexed: 02/06/2023] Open
Abstract
The phenomenon of adult neurogenesis is now an accepted occurrence in mammals and also in humans. At least two discrete places house stem cells for generation of neurons in adult brain. These are olfactory system and the hippocampus. In animals, newly generated neurons have been directly or indirectly demonstrated to generate a significant amount of new neurons to have a functional role. However, the data in humans on the extent of this process is still scanty and such as difficult to comprehend its functional role in humans. This paper explores the available data on as extent of adult hippocampal neurogenesis in humans and makes comparison to animal data.
Collapse
Affiliation(s)
- Amadi O. Ihunwo
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Lackson H. Tembo
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Charles Dzamalala
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
25
|
Upadhya D, Hattiangady B, Shetty GA, Zanirati G, Kodali M, Shetty AK. Neural Stem Cell or Human Induced Pluripotent Stem Cell-Derived GABA-ergic Progenitor Cell Grafting in an Animal Model of Chronic Temporal Lobe Epilepsy. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2016; 38:2D.7.1-2D.7.47. [PMID: 27532817 PMCID: PMC5313261 DOI: 10.1002/cpsc.9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Grafting of neural stem cells (NSCs) or GABA-ergic progenitor cells (GPCs) into the hippocampus could offer an alternative therapy to hippocampal resection in patients with drug-resistant chronic epilepsy, which afflicts >30% of temporal lobe epilepsy (TLE) cases. Multipotent, self-renewing NSCs could be expanded from multiple regions of the developing and adult brain, human embryonic stem cells (hESCs), and human induced pluripotent stem cells (hiPSCs). On the other hand, GPCs could be generated from the medial and lateral ganglionic eminences of the embryonic brain and from hESCs and hiPSCs. To provide comprehensive methodologies involved in testing the efficacy of transplantation of NSCs and GPCs in a rat model of chronic TLE, NSCs derived from the rat medial ganglionic eminence (MGE) and MGE-like GPCs derived from hiPSCs are taken as examples in this unit. The topics comprise description of the required materials, reagents and equipment, methods for obtaining rat MGE-NSCs and hiPSC-derived MGE-like GPCs in culture, generation of chronically epileptic rats, intrahippocampal grafting procedure, post-grafting evaluation of the effects of grafts on spontaneous recurrent seizures and cognitive and mood impairments, analyses of the yield and the fate of graft-derived cells, and the effects of grafts on the host hippocampus. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Dinesh Upadhya
- Institute for Regenerative Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas
| | - Bharathi Hattiangady
- Institute for Regenerative Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas
| | - Geetha A Shetty
- Institute for Regenerative Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas
| | - Gabriele Zanirati
- Institute for Regenerative Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas
| |
Collapse
|
26
|
Shetty AK, Hattiangady B. Grafted Subventricular Zone Neural Stem Cells Display Robust Engraftment and Similar Differentiation Properties and Form New Neurogenic Niches in the Young and Aged Hippocampus. Stem Cells Transl Med 2016; 5:1204-15. [PMID: 27194744 PMCID: PMC4996439 DOI: 10.5966/sctm.2015-0270] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 04/01/2016] [Indexed: 12/30/2022] Open
Abstract
The engraftment and differentiation of alkaline phosphatase-positive neural stem cells (NSCs) expanded from the postnatal subventricular zone (SVZ), 3 months after grafting into the intact young or aged rat hippocampus, were examined. Both young and aged hippocampi supported robust engraftment and similar differentiation of SVZ-NSC graft-derived cells. As clinical application of neural stem cell (NSC) grafting into the brain would also encompass aged people, critical evaluation of engraftment of NSC graft-derived cells in the aged hippocampus has significance. We examined the engraftment and differentiation of alkaline phosphatase-positive NSCs expanded from the postnatal subventricular zone (SVZ), 3 months after grafting into the intact young or aged rat hippocampus. Graft-derived cells engrafted robustly into both young and aged hippocampi. Although most graft-derived cells pervasively migrated into different hippocampal layers, the graft cores endured and contained graft-derived neurons expressing neuron-specific nuclear antigen (NeuN) and γ-amino butyric acid in both groups. A fraction of migrated graft-derived cells in the neurogenic subgranular zone-granule cell layer also expressed NeuN. Neuronal differentiation was, however, occasionally seen amid graft-derived cells that had migrated into non-neurogenic regions, where substantial fractions differentiated into S-100β+ astrocytes, NG2+ oligodendrocyte progenitors, or Olig2+ putative oligodendrocytes. In both age groups, graft cores located in non-neurogenic regions displayed many doublecortin-positive (DCX+) immature neurons at 3 months after grafting. Analyses of cells within graft cores using birth dating and putative NSC markers revealed that DCX+ neurons were newly born neurons derived from engrafted cells and that putative NSCs persisted within the graft cores. Thus, both young and aged hippocampi support robust engraftment and similar differentiation of SVZ-NSC graft-derived cells. Furthermore, some grafted NSCs retain the “stemness” feature and produce new neurons even at 3 months after grafting, implying that grafting of SVZ-NSCs into the young or aged hippocampus leads to establishment of new neurogenic niches in non-neurogenic regions. Significance The results demonstrate that advanced age of the host at the time of grafting has no major adverse effects on engraftment, migration, and differentiation of grafted subventricular zone-neural stem cells (SVZ-NSCs) in the intact hippocampus, as both young and aged hippocampi promoted excellent engraftment, migration, and differentiation of SVZ-NSC graft-derived cells in the present study. Furthermore, SVZ-NSC grafts showed ability for establishing neurogenic niches in non-neurogenic regions, generating new neurons for extended periods after grafting. This phenomenon will be beneficial if these niches can continuously generate new neurons and glia in the grafted hippocampus, as newly generated neurons and glia are expected to improve, not only the microenvironment, but also the plasticity and function of the aged hippocampus. Overall, these results have significance because the potential application of NSC grafting for treatment of neurodegenerative disorders at early stages of disease progression and age-related impairments would mostly involve aged persons as recipients.
Collapse
Affiliation(s)
- Ashok K Shetty
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas, USAResearch Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USADepartment of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, USADivision of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USAResearch and Surgery Services, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Bharathi Hattiangady
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas, USAResearch Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USADepartment of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, USADivision of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USAResearch and Surgery Services, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| |
Collapse
|
27
|
Gao F, Song X, Zhu D, Wang X, Hao A, Nadler JV, Zhan RZ. Dendritic morphology, synaptic transmission, and activity of mature granule cells born following pilocarpine-induced status epilepticus in the rat. Front Cell Neurosci 2015; 9:384. [PMID: 26500490 PMCID: PMC4596052 DOI: 10.3389/fncel.2015.00384] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 09/14/2015] [Indexed: 01/23/2023] Open
Abstract
To understand the potential role of enhanced hippocampal neurogenesis after pilocarpine-induced status epilepticus (SE) in the development of epilepsy, we quantitatively analyzed the geometry of apical dendrites, synaptic transmission, and activation levels of normotopically distributed mature newborn granule cells in the rat. SE in male Sprague-Dawley rats (between 6 and 7 weeks old) lasting for more than 2 h was induced by an intraperitoneal injection of pilocarpine. The complexity, spine density, miniature post-synaptic currents, and activity-regulated cytoskeleton-associated protein (Arc) expression of granule cells born 5 days after SE were studied between 10 and 17 weeks after CAG-GFP retroviral vector-mediated labeling. Mature granule cells born after SE had dendritic complexity similar to that of granule cells born naturally, but with denser mushroom-like spines in dendritic segments located in the outer molecular layer. Miniature inhibitory post-synaptic currents (mIPSCs) were similar between the controls and rats subjected to SE; however, smaller miniature excitatory post-synaptic current (mEPSC) amplitude with a trend toward less frequent was found in mature granule cells born after SE. After maturation, granule cells born after SE did not show denser Arc expression in the resting condition or 2 h after being activated by pentylenetetrazol-induced transient seizure activity than vicinal GFP-unlabeled granule cells. Thus our results suggest that normotopic granule cells born after pilocarpine-induced SE are no more active when mature than age-matched, naturally born granule cells.
Collapse
Affiliation(s)
- Fei Gao
- Department of Physiology, Shandong University School of Medicine Jinan, China
| | - Xueying Song
- Department of Physiology, Shandong University School of Medicine Jinan, China
| | - Dexiao Zhu
- Department of Physiology, Shandong University School of Medicine Jinan, China
| | - Xiaochen Wang
- Department of Physiology, Shandong University School of Medicine Jinan, China
| | - Aijun Hao
- Department of Histology and Embryology, Shandong University School of Medicine Jinan, China
| | - J Victor Nadler
- Departments of Pharmacology and Neurobiology, Duke University Medical Center Durham, NC, USA
| | - Ren-Zhi Zhan
- Department of Physiology, Shandong University School of Medicine Jinan, China
| |
Collapse
|
28
|
FGF Signaling Is Necessary for Neurogenesis in Young Mice and Sufficient to Reverse Its Decline in Old Mice. J Neurosci 2015; 35:10217-23. [PMID: 26180198 DOI: 10.1523/jneurosci.1469-15.2015] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED The mechanisms regulating hippocampal neurogenesis remain poorly understood. Particularly unclear is the extent to which age-related declines in hippocampal neurogenesis are due to an innate decrease in precursor cell performance or to changes in the environment of these cells. Several extracellular signaling factors that regulate hippocampal neurogenesis have been identified. However, the role of one important family, FGFs, remains uncertain. Although a body of literature suggests that FGFs can promote the proliferation of cultured adult hippocampal precursor cells, their requirement for adult hippocampal neurogenesis in vivo and the cell types within the neurogenic lineage that might depend on FGFs remain unclear. Here, specifically targeting adult neural precursor cells, we conditionally express an activated form of an FGF receptor or delete the FGF receptors that are expressed in these cells. We find that FGF receptors are required for neural stem-cell maintenance and that an activated receptor expressed in all precursors can increase the number of neurons produced. Moreover, in older mice, an activated FGF receptor can rescue the age-related decline in neurogenesis to a level found in young adults. These results suggest that the decrease in neurogenesis with age is not simply due to fewer stem cells, but also to declining signals in their niche. Thus, enhancing FGF signaling in precursors can be used to reverse age-related declines in hippocampal neurogenesis. SIGNIFICANCE STATEMENT Hippocampal deficits can result from trauma, neurodegeneration, or aging and can lead to loss of memory and mood control. The addition of new neurons to the hippocampus facilitates memory formation, but how this process is regulated and how we might manipulate it to reverse hippocampal dysfunction remains unclear. The FGF signaling pathway has been hypothesized to be important, but its role in generating new neurons had been poorly defined. Our study indicates that FGF signaling maintains and expands subsets of neural precursor cells. Moreover, in older mice, increasing FGF signaling is sufficient to reverse the decline in neuron production to levels found in young adults, providing a potential means of reversing age-related hippocampal deficits.
Collapse
|
29
|
Dentate gyrus expression of nestin-immunoreactivity in patients with drug-resistant temporal lobe epilepsy and hippocampal sclerosis. Seizure 2015; 27:75-9. [DOI: 10.1016/j.seizure.2015.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 02/03/2015] [Accepted: 02/12/2015] [Indexed: 01/02/2023] Open
|
30
|
Effect of the N-methyl-D-aspartate NR2B subunit antagonist ifenprodil on precursor cell proliferation in the hippocampus. J Neurosci Res 2014; 92:679-91. [DOI: 10.1002/jnr.23347] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 11/12/2013] [Accepted: 11/14/2013] [Indexed: 12/27/2022]
|
31
|
Cavarsan CF, Queiroz CM, dos Santos JG, Xavier GF, Mello LE, Covolan L. Reduced hippocampal dentate cell proliferation and impaired spatial memory performance in aged-epileptic rats. Front Neurol 2013; 4:106. [PMID: 23898322 PMCID: PMC3724058 DOI: 10.3389/fneur.2013.00106] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 07/12/2013] [Indexed: 11/13/2022] Open
Abstract
Increased adult neurogenesis is observed after training in hippocampal-dependent tasks and also after acutely induced status epilepticus (SE) although the specific roles of these cells are still a matter of debate. In this study, we investigated hippocampal cell proliferation and differentiation and the spatial learning performance in young or aged chronically epileptic rats. Status was induced by pilocarpine in 3 or 20-month old rats. Either 2 or 20 months later, rats were treated with bromodeoxyuridine (BrdU) and subsequently underwent to 8-day schedule of water maze (WM) tests. As expected, learning curves were faster in young than in aged animals (P < 0.001). Chronically epileptic animals exhibited impaired learning curves compared to age-matched controls. Interestingly, the duration of epilepsy (2 or 20 months) did not correlate with the memory impairment of aged-epileptic animals. The number of BrdU-positive cells was greater in young-epileptic subjects than in age-matched controls. In contrast, cell proliferation was not increased in aged-epileptic animals, irrespective of the time of SE induction. Finally, dentate cell proliferation was not related to performance in the WM. Based on the present results we conclude that even though aging and epilepsy lead to impairments in spatial learning, their effects are not additive.
Collapse
Affiliation(s)
- Clarissa F. Cavarsan
- Department of Physiology, Universidade Federal de São Paulo – UNIFESP, São Paulo, Brazil
| | - Claudio M. Queiroz
- Brain Institute, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | | | - Gilberto F. Xavier
- Department of Physiology, Biosciences Institute, Universidade de São Paulo – USP, São Paulo, Brazil
| | - Luiz Eugênio Mello
- Department of Physiology, Universidade Federal de São Paulo – UNIFESP, São Paulo, Brazil
| | - Luciene Covolan
- Department of Physiology, Universidade Federal de São Paulo – UNIFESP, São Paulo, Brazil
| |
Collapse
|
32
|
Fuentealba LC, Obernier K, Alvarez-Buylla A. Adult neural stem cells bridge their niche. Cell Stem Cell 2012; 10:698-708. [PMID: 22704510 DOI: 10.1016/j.stem.2012.05.012] [Citation(s) in RCA: 269] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Major developments in the neural stem cell (NSC) field in recent years provide new insights into the nature of the NSC niche. In this perspective, we integrate recent anatomical data on the organization of the two main neurogenic niches in the adult brain, the ventricular-subventricular zone (V-SVZ) and the subgranular zone (SGZ), with signaling pathways that control the behavior of NSCs. NSCs in the adult brain stretch into physiologically distinct compartments of their niche. We propose how adult NSCs' morphology may allow these cells to integrate multiple signaling pathways arising from unique locations of their niche.
Collapse
Affiliation(s)
- Luis C Fuentealba
- Department of Neurological Surgery and Institute for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kirsten Obernier
- Department of Neurological Surgery and Institute for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery and Institute for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
33
|
Shetty AK, Hattiangady B, Rao MS, Shuai B. Neurogenesis response of middle-aged hippocampus to acute seizure activity. PLoS One 2012; 7:e43286. [PMID: 22912847 PMCID: PMC3422269 DOI: 10.1371/journal.pone.0043286] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 07/23/2012] [Indexed: 01/19/2023] Open
Abstract
Acute Seizure (AS) activity in young adult age conspicuously modifies hippocampal neurogenesis. This is epitomized by both increased addition of new neurons to the granule cell layer (GCL) by neural stem/progenitor cells (NSCs) in the dentate subgranular zone (SGZ), and greatly enhanced numbers of newly born neurons located abnormally in the dentate hilus (DH). Interestingly, AS activity in old age does not induce such changes in hippocampal neurogenesis. However, the effect of AS activity on neurogenesis in the middle-aged hippocampus is yet to be elucidated. We examined hippocampal neurogenesis in middle-aged F344 rats after a continuous AS activity for >4 hrs, induced through graded intraperitoneal injections of the kainic acid. We labeled newly born cells via daily intraperitoneal injections of the 5'-bromodeoxyuridine (BrdU) for 12 days, commencing from the day of induction of AS activity. AS activity enhanced the addition of newly born BrdU+ cells by 5.6 fold and newly born neurons (expressing both BrdU and doublecortin [DCX]) by 2.2 fold to the SGZ-GCL. Measurement of the total number of DCX+ newly born neurons also revealed a similar trend. Furthermore, AS activity increased DCX+ newly born neurons located ectopically in the DH (2.7 fold increase and 17% of total newly born neurons). This rate of ectopic migration is however considerably less than what was observed earlier for the young adult hippocampus after similar AS activity. Thus, the plasticity of hippocampal neurogenesis to AS activity in middle age is closer to its response observed in the young adult age. However, the extent of abnormal migration of newly born neurons into the DH is less than that of the young adult hippocampus after similar AS activity. These results also point out a highly divergent response of neurogenesis to AS activity between middle age and old age.
Collapse
Affiliation(s)
- Ashok K Shetty
- Research Service, Veterans Affairs Medical Centers of Durham, North Carolina, and Temple, Texas, United States of America.
| | | | | | | |
Collapse
|
34
|
Abstract
The importance of adult neurogenesis has only recently been accepted, resulting in a completely new field of investigation within stem cell biology. The regulation and functional significance of adult neurogenesis is currently an area of highly active research. G-protein-coupled receptors (GPCRs) have emerged as potential modulators of adult neurogenesis. GPCRs represent a class of proteins with significant clinical importance, because approximately 30% of all modern therapeutic treatments target these receptors. GPCRs bind to a large class of neurotransmitters and neuromodulators such as norepinephrine, dopamine, and serotonin. Besides their typical role in cellular communication, GPCRs are expressed on adult neural stem cells and their progenitors that relay specific signals to regulate the neurogenic process. This review summarizes the field of adult neurogenesis and its methods and specifies the roles of various GPCRs and their signal transduction pathways that are involved in the regulation of adult neural stem cells and their progenitors. Current evidence supporting adult neurogenesis as a model for self-repair in neuropathologic conditions, adult neural stem cell therapeutic strategies, and potential avenues for GPCR-based therapeutics are also discussed.
Collapse
Affiliation(s)
- Van A Doze
- Department of Molecular Cardiology, NB50, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | | |
Collapse
|
35
|
Artegiani B, Calegari F. Age-related cognitive decline: can neural stem cells help us? Aging (Albany NY) 2012; 4:176-86. [PMID: 22466406 PMCID: PMC3348478 DOI: 10.18632/aging.100446] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 03/29/2012] [Indexed: 02/07/2023]
Abstract
Several studies suggest that an increase in adult neurogenesis has beneficial effects on emotional behavior and cognitive performance including learning and memory. The observation that aging has a negative effect on the proliferation of neural stem cells has prompted several laboratories to investigate new systems to artificially increase neurogenesis in senescent animals as a means to compensate for age-related cognitive decline. In this review we will discuss the systemic, cellular, and molecular changes induced by aging and affecting the neurogenic niche at the level of neural stem cell proliferation, their fate change, neuronal survival, and subsequent integration in the neuronal circuitry. Particular attention will be given to those manipulations that increase neurogenesis in the aged brain as a potential avenue towards therapy.
Collapse
Affiliation(s)
- Benedetta Artegiani
- DFG-Research Center and Cluster of Excellence for Regenerative Therapies Dresden, Technische Universität Dresden, Germany
| | | |
Collapse
|
36
|
Shapiro LA, Wang L, Upadhyaya P, Ribak CE. Seizure-induced Increased Neurogenesis Occurs in the Dentate Gyrus of Aged Sprague-Dawley Rats. Aging Dis 2011; 2:286-293. [PMID: 22396880 PMCID: PMC3295072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 08/18/2011] [Accepted: 08/18/2011] [Indexed: 05/31/2023] Open
Abstract
Neurogenesis in the hippocampal dentate gyrus persists throughout the lifespan of mammals, however, the rate of neurogenesis decreases as the animal ages. Although seizures increase neurogenesis in young adult brains, this relationship has not been shown in aged animals. Using doublecortin (DCX) immunocytochemistry, the number of DCX-labeled cells in the dentate gyrus from aged rats (23 months of age) was assessed 30 days following pilocarpine-induced seizures and was compared to the number obtained from age-matched control rats. DCX-labeled cells were located in the subgranular zone, at the border between the hilus and the granule cell layer, and within the granule cell layer in both epileptic and control aged brains. When comparing the aged epileptic rats to age-matched controls, there was a significant increase in the number of DCX-labeled cells that was almost four and a half-fold. Therefore, aged rats also display an increase in adult neurogenesis following seizures.
Collapse
Affiliation(s)
- Lee A. Shapiro
- Dept. of Surgery and Neurosurgery, College of Medicine, Texas A&M Health Science Center, Scott & White Hospital, Central Texas Veterans Health Care System, Temple, TX 76504, USA
| | - Lulu Wang
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Pooja Upadhyaya
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Charles E. Ribak
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, USA
| |
Collapse
|
37
|
Arisi GM, Ruch M, Foresti ML, Mukherjee S, Ribak CE, Shapiro LA. Astrocyte Alterations in the Hippocampus Following Pilocarpine-induced Seizures in Aged Rats. Aging Dis 2011; 2:294-300. [PMID: 22396881 PMCID: PMC3295071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Revised: 08/25/2011] [Accepted: 08/26/2011] [Indexed: 05/31/2023] Open
Abstract
It is known that the incidence of epilepsy increases with age, but only a few studies have investigated the consequences and mechanisms of seizure and epilepsy in aged animals. Astrocytic changes are known to directly influence neuronal excitability and seizure susceptibility. However, information regarding alterations to astrocytes after seizures in aged animals is lacking in the literature. In the present study, the density and morphology of astrocytes expressing GFAP were investigated in the hippocampus of aged rats that experienced status epilepticus induced by pilocarpine. One month after seizures, astrocytes in aged rats have increased volume and present activated morphology. Despite these morphological changes, the density of astrocytes was not altered in the hippocampus of aged rats after seizures.
Collapse
Affiliation(s)
- Gabriel M Arisi
- Scott & White Hospital, Neuroscience Research Institute, Temple, TX, USA
- Central Texas Veterans Health System, Temple, TX, USA
| | - Megan Ruch
- Scott & White Hospital, Neuroscience Research Institute, Temple, TX, USA
- Central Texas Veterans Health System, Temple, TX, USA
| | - Maira L Foresti
- Scott & White Hospital, Neuroscience Research Institute, Temple, TX, USA
- Central Texas Veterans Health System, Temple, TX, USA
| | - Sanjib Mukherjee
- Scott & White Hospital, Neuroscience Research Institute, Temple, TX, USA
- Central Texas Veterans Health System, Temple, TX, USA
| | - Charles E Ribak
- Department of Anatomy and Neurobiology, University of California at Irvine, School of Medicine, Irvine, CA 92697-1275, USA
| | - Lee A Shapiro
- Scott & White Hospital, Neuroscience Research Institute, Temple, TX, USA
- Central Texas Veterans Health System, Temple, TX, USA
- Department of Surgery, Neurosurgery and Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA
| |
Collapse
|
38
|
Hattiangady B, Kuruba R, Shetty AK. Acute Seizures in Old Age Leads to a Greater Loss of CA1 Pyramidal Neurons, an Increased Propensity for Developing Chronic TLE and a Severe Cognitive Dysfunction. Aging Dis 2011; 2:1-17. [PMID: 21339903 PMCID: PMC3041587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Revised: 01/26/2010] [Accepted: 01/26/2010] [Indexed: 05/30/2023] Open
Abstract
The aged population displays an enhanced risk for developing acute seizure (AS) activity. However, it is unclear whether AS activity in old age would result in a greater magnitude of hippocampal neurodegeneration and inflammation, and an increased predilection for developing chronic temporal lobe epilepsy (TLE) and cognitive dysfunction. Therefore, we addressed these issues in young-adult (5-months old) and aged (22-months old) F344 rats after three-hours of AS activity, induced through graded intraperitoneal injections of kainic acid (KA), and terminated through a diazepam injection. During the three-hours of AS activity, both young adult and aged groups exhibited similar numbers of stage-V motor seizures but the numbers of stage-IV motor seizures were greater in the aged group. In both age groups, three-hour AS activity induced degeneration of 50-55% of neurons in the dentate hilus, 22-32% of neurons in the granule cell layer and 49-52% neurons in the CA3 pyramidal cell layer without showing any interaction between the age and AS activity. However, degeneration of neurons in the CA1 pyramidal cell layer showed a clear interaction between the age and AS activity (12% in the young adult group and 56% in the aged group), suggesting that an advanced age makes the CA1 pyramidal neurons more susceptible to die with AS activity. The extent of inflammation measured through the numbers of activated microglial cells was similar between the two age groups. Interestingly, the predisposition for developing chronic TLE at 2-3 months after AS activity was 60% for young adult rats but 100% for aged rats. Moreover, both frequency & intensity of spontaneous recurrent seizures in the chronic phase after AS activity were 6-12 folds greater in aged rats than in young adult rats. Furthermore, aged rats lost their ability for spatial learning even in a scrupulous eleven-session water maze learning paradigm after AS activity, in divergence from young adult rats which retained the ability for spatial learning but had memory retrieval dysfunction after AS activity. Thus, AS activity in old age results in a greater loss of hippocampal CA1 pyramidal neurons, an increased propensity for developing robust chronic TLE, and a severe cognitive dysfunction.
Collapse
Affiliation(s)
| | | | - Ashok K. Shetty
- Correspondence should be addressed to: Ashok K. Shetty, M.Sc., Ph.D., Division of Neurosurgery, DUMC Box 3807, Duke University Medical Center, Durham NC 27710. E-mail:
| |
Collapse
|
39
|
Avanzi R, Cavarsan C, Santos J, Hamani C, Mello L, Covolan L. Basal dendrites are present in newly born dentate granule cells of young but not aged pilocarpine-treated chronic epileptic rats. Neuroscience 2010; 170:687-91. [DOI: 10.1016/j.neuroscience.2010.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 07/29/2010] [Accepted: 08/01/2010] [Indexed: 01/23/2023]
|
40
|
Lugert S, Basak O, Knuckles P, Haussler U, Fabel K, Götz M, Haas CA, Kempermann G, Taylor V, Giachino C. Quiescent and active hippocampal neural stem cells with distinct morphologies respond selectively to physiological and pathological stimuli and aging. Cell Stem Cell 2010; 6:445-56. [PMID: 20452319 DOI: 10.1016/j.stem.2010.03.017] [Citation(s) in RCA: 539] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 02/15/2010] [Accepted: 03/18/2010] [Indexed: 12/16/2022]
Abstract
New neurons are generated in the adult hippocampus throughout life by neural stem/progenitor cells (NSCs), and neurogenesis is a plastic process responsive to external stimuli. We show that canonical Notch signaling through RBP-J is required for hippocampal neurogenesis. Notch signaling distinguishes morphologically distinct Sox2(+) NSCs, and within these pools subpopulations can shuttle between mitotically active or quiescent. Radial and horizontal NSCs respond selectively to neurogenic stimuli. Physical exercise activates the quiescent radial population whereas epileptic seizures induce expansion of the horizontal NSC pool. Surprisingly, reduced neurogenesis correlates with a loss of active horizontal NSCs in aged mice rather than a total loss of stem cells, and the transition to a quiescent state is reversible to rejuvenate neurogenesis in the brain. The discovery of multiple NSC populations with Notch dependence but selective responses to stimuli and reversible quiescence has important implications for the mechanisms of adaptive learning and also for regenerative therapy.
Collapse
Affiliation(s)
- Sebastian Lugert
- Department of Molecular Embryology, Max Planck Institute of Immunobiology, Stubeweg 51, D-79108 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Tan YF, Preston E, Wojtowicz JM. Enhanced post-ischemic neurogenesis in aging rats. Front Neurosci 2010; 4. [PMID: 20877422 PMCID: PMC2944628 DOI: 10.3389/fnins.2010.00163] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 08/16/2010] [Indexed: 01/11/2023] Open
Abstract
Hippocampal neurogenesis persists in adult mammals, but its rate declines dramatically with age. Evidence indicates that experimentally-reduced levels of neurogenesis (e.g., by irradiation) in young rats has profound influence on cognition as determined by learning and memory tests. In the present study we asked whether in middle-aged, 10- to 13-months-old rats, cell production can be restored toward the level present in young rats. To manipulate neurogenesis we induced bilateral carotid occlusion with hypotension. This procedure is known to increase neurogenesis in young rats, presumably in a compensatory manner, but until now, has never been tested in aging rats. Cell production was measured at 10, 35, and 90 days after ischemia. The results indicate that neuronal proliferation and differentiation can be transiently restored in middle-aged rats. Furthermore, the effects are more pronounced in the dorsal as opposed to ventral hippocampus thus restoring the dorso-ventral gradient seen in younger rats. Our results support previous findings showing that some of the essential features of the age-dependent decline in neurogenesis are reversible. Thus, it may be possible to manipulate neurogenesis and improve learning and memory in old age.
Collapse
Affiliation(s)
- Yao-Fang Tan
- Department of Physiology, University of Toronto Toronto, ON, Canada
| | | | | |
Collapse
|
42
|
Shetty AK, Hattiangady B, Rao MS. Vulnerability of hippocampal GABA-ergic interneurons to kainate-induced excitotoxic injury during old age. J Cell Mol Med 2010. [PMID: 20141618 DOI: 10.1111/j.1582-4934.2008.00675.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Hippocampal inhibitory interneurons expressing glutamate decarboxylase-67 (GAD-67) considerably decline in number during old age. Studies in young adult animals further suggest that hippocampal GAD-67+ interneuron population is highly vulnerable to excitotoxic injury. However, the relative susceptibility of residual GAD-67+ interneurons in the aged hippocampus to excitotoxic injury is unknown. To elucidate this, using both adult and aged F344 rats, we performed stereological counting of GAD-67+ interneurons in different layers of the dentate gyrus and CA1 & CA3 sub-fields, at 3 months post-excitotoxic hippocampal injury inflicted through an intracerebroventricular administration of kainic acid (KA). Substantial reductions of GAD-67+ interneurons were found in all hippocampal layers and sub-fields after KA-induced injury in adult animals. Contrastingly, there was no significant change in GAD-67+ interneuron population in any of the hippocampal layers and sub-fields following similar injury in aged animals. Furthermore, the stability of GAD-67+ interneurons in aged rats after KA was not attributable to milder injury, as the overall extent of KA-induced hippocampal principal neuron loss was comparable between adult and aged rats. Interestingly, because of the age-related disparity in vulnerability of interneurons to injury, the surviving GAD-67+ interneuron population in the injured aged hippocampus remained comparable to that observed in the injured adult hippocampus despite enduring significant reductions in interneuron number with aging. Thus, unlike in the adult hippocampus, an excitotoxic injury to the aged hippocampus does not result in significantly decreased numbers of GAD-67+ interneurons. Persistence of GAD-67+ interneuron population in the injured aged hippocampus likely reflects an age-related change in the response of GAD-67+ interneurons to excitotoxic hippocampal injury. These results have implications towards understanding mechanisms underlying the evolution of initial precipitating injury into temporal lobe epilepsy in the elderly population.
Collapse
Affiliation(s)
- Ashok K Shetty
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, NC, USA.
| | | | | |
Collapse
|
43
|
Shetty AK, Hattiangady B, Rao MS. Vulnerability of hippocampal GABA-ergic interneurons to kainate-induced excitotoxic injury during old age. J Cell Mol Med 2010; 13:2408-23. [PMID: 20141618 DOI: 10.1111/j.1582-4934.2009.00675.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Hippocampal inhibitory interneurons expressing glutamate decarboxylase-67 (GAD-67) considerably decline in number during old age. Studies in young adult animals further suggest that hippocampal GAD-67+ interneuron population is highly vulnerable to excitotoxic injury. However, the relative susceptibility of residual GAD-67+ interneurons in the aged hippocampus to excitotoxic injury is unknown. To elucidate this, using both adult and aged F344 rats, we performed stereological counting of GAD-67+ interneurons in different layers of the dentate gyrus and CA1 & CA3 sub-fields, at 3 months post-excitotoxic hippocampal injury inflicted through an intracerebroventricular administration of kainic acid (KA). Substantial reductions of GAD-67+ interneurons were found in all hippocampal layers and sub-fields after KA-induced injury in adult animals. Contrastingly, there was no significant change in GAD-67+ interneuron population in any of the hippocampal layers and sub-fields following similar injury in aged animals. Furthermore, the stability of GAD-67+ interneurons in aged rats after KA was not attributable to milder injury, as the overall extent of KA-induced hippocampal principal neuron loss was comparable between adult and aged rats. Interestingly, because of the age-related disparity in vulnerability of interneurons to injury, the surviving GAD-67+ interneuron population in the injured aged hippocampus remained comparable to that observed in the injured adult hippocampus despite enduring significant reductions in interneuron number with aging. Thus, unlike in the adult hippocampus, an excitotoxic injury to the aged hippocampus does not result in significantly decreased numbers of GAD-67+ interneurons. Persistence of GAD-67+ interneuron population in the injured aged hippocampus likely reflects an age-related change in the response of GAD-67+ interneurons to excitotoxic hippocampal injury. These results have implications towards understanding mechanisms underlying the evolution of initial precipitating injury into temporal lobe epilepsy in the elderly population.
Collapse
Affiliation(s)
- Ashok K Shetty
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, NC, USA.
| | | | | |
Collapse
|
44
|
Abstract
Aged animals have been used by researchers to better understand the differences between the young and the aged brain and how these differences may provide insight into the mechanisms of acute seizures and epilepsy in the elderly. To date, there have been relatively few studies dedicated to the modeling of acute seizures and epilepsy in aged, healthy animals. Inherent challenges to this area of research include the costs associated with the purchase and maintenance of older animals and, at times, the unexpected and potentially confounding comorbidities associated with aging. However, recent studies using a variety of in vivo and in vitro models of acute seizures and epilepsy in mice and rats have built upon early investigations in the field, all of which has provided an expanded vision of seizure generation and epileptogenesis in the aged brain. Results of these studies could potentially translate to new and tailored interventional approaches that limit or prevent the development of epilepsy in the elderly.
Collapse
Affiliation(s)
- Kevin M Kelly
- Drexel University College of Medicine, Center for Neuroscience Research, Allegheny-Singer Research Institute, Allegheny General Hospital Pittsburgh, Pittsburgh, PA 15212-4772, USA.
| |
Collapse
|
45
|
Shetty AK, Hattiangady B, Rao MS, Shuai B. Deafferentation enhances neurogenesis in the young and middle aged hippocampus but not in the aged hippocampus. Hippocampus 2010; 21:631-46. [PMID: 20333732 DOI: 10.1002/hipo.20776] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2010] [Indexed: 12/13/2022]
Abstract
Increased neurogenesis in the dentate gyrus (DG) after brain insults such as excitotoxic lesions, seizures, or stroke is a well known phenomenon in the young hippocampus. This plasticity reflects an innate compensatory response of neural stem cells (NSCs) in the young hippocampus to preserve function or minimize damage after injury. However, injuries to the middle-aged and aged hippocampi elicit either no or dampened neurogenesis response, which could be due to an altered plasticity of NSCs and/or the hippocampus with age. We examined whether the plasticity of NSCs to increase neurogenesis in response to a milder injury such as partial deafferentation is preserved during aging. We quantified DG neurogenesis in the hippocampus of young, middle-aged, and aged F344 rats after partial deafferentation. A partial deafferentation of the left hippocampus without any apparent cell loss was induced via administration of Kainic acid (0.5 μg in 1.0 μl) into the right lateral ventricle of the brain. In this model, degeneration of CA3 pyramidal neurons and dentate hilar neurons in the right hippocampus results in loss of commissural axons which leads to partial deafferentation of the dendrites of dentate granule cells and CA1-CA3 pyramidal neurons in the left hippocampus. Quantification of newly born cells that are added to the dentate granule cell layer at postdeafferentation days 4-15 using 5'-bromodeoxyuridine (BrdU) labeling revealed greatly increased addition of newly born cells (∼three fold increase) in the deafferented young and middle-aged hippocampi but not in the deafferented aged hippocampus. Measurement of newly born neurons using doublecortin (DCX) immunostaining also revealed similar findings. Analyses using BrdU-DCX dual immunofluorescence demonstrated no changes in neuronal fate-choice decision of newly born cells after deafferentation, in comparison to the age-matched naive hippocampus in all age groups. Thus, the plasticity of hippocampal NSCs to increase DG neurogenesis in response to a milder injury such as partial hippocampal deafferentation is preserved until middle age but lost at old age.
Collapse
Affiliation(s)
- Ashok K Shetty
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, North Carolina, USA.
| | | | | | | |
Collapse
|
46
|
Ndode-Ekane X, Hayward N, Gröhn O, Pitkänen A. Vascular changes in epilepsy: functional consequences and association with network plasticity in pilocarpine-induced experimental epilepsy. Neuroscience 2010; 166:312-32. [DOI: 10.1016/j.neuroscience.2009.12.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 11/25/2009] [Accepted: 12/01/2009] [Indexed: 01/07/2023]
|
47
|
Ahlenius H, Visan V, Kokaia M, Lindvall O, Kokaia Z. Neural stem and progenitor cells retain their potential for proliferation and differentiation into functional neurons despite lower number in aged brain. J Neurosci 2009; 29:4408-19. [PMID: 19357268 PMCID: PMC6665731 DOI: 10.1523/jneurosci.6003-08.2009] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 02/05/2009] [Accepted: 03/02/2009] [Indexed: 12/18/2022] Open
Abstract
Neurogenesis in the subventricular zone (SVZ), which gives rise to new neurons in the olfactory bulb, continues throughout life but declines with increasing age. Little is known about how aging affects the intrinsic properties of the neural stem and progenitor cells (NSCs) in SVZ and the functional characteristics of their neuronal progeny. Here, we have compared the properties of NSCs isolated from embryonic lateral ganglionic eminence and adult and aged SVZ in mice using in vivo and in vitro systems, analyzed their gene expression profile, and studied their electrophysiological characteristics before and after differentiation into neurons. We show a loss of NSCs in SVZ from aged mice accompanied by reduced expression of genes for NSC markers, developmentally important transcription factors, and neurogenic factors. However, when isolated in vitro, the NSCs from SVZ of aged animals have capacity for proliferation and multilineage differentiation, including production of functional neurons, similar to that of NSCs in adult mice, albeit with lower efficacy. These properties are of major importance when considering therapeutic applications of neuronal replacement from endogenous NSCs in the injured, aged brain.
Collapse
Affiliation(s)
- Henrik Ahlenius
- Laboratory of Neural Stem Cell Biology
- Lund Stem Cell Center, SE-221 84 Lund, Sweden
| | - Violeta Visan
- Laboratory of Neurogenesis and Cell Therapy, and
- Lund Stem Cell Center, SE-221 84 Lund, Sweden
| | - Merab Kokaia
- Experimental Epilepsy Group, Section of Restorative Neurology, Wallenberg Neuroscience Center, University Hospital, SE-221 84 Lund, Sweden, and
| | - Olle Lindvall
- Laboratory of Neurogenesis and Cell Therapy, and
- Lund Stem Cell Center, SE-221 84 Lund, Sweden
| | - Zaal Kokaia
- Laboratory of Neural Stem Cell Biology
- Lund Stem Cell Center, SE-221 84 Lund, Sweden
| |
Collapse
|
48
|
Abstract
Psychiatric and neurologic disorders take an enormous toll on society. Alleviating the devastating symptoms and consequences of neuropsychiatric disorders such as addiction, depression, epilepsy, and schizophrenia is a main force driving clinical and basic researchers alike. By elucidating these disease neuromechanisms, researchers hope to better define treatments and preventive therapies. Research suggests that regulation of adult hippocampal neurogenesis represents a promising approach to treating and perhaps preventing mental illness. Here we appraise the role of adult hippocampal neurogenesis in major psychiatric and neurologic disorders within the essential framework of recent progress made in understanding "normal" adult neurogenesis. Topics addressed include the following: the life cycle of an adult hippocampal stem cell and the implications for aging; links between learning and hippocampal neurogenesis; the reciprocal relationship between cocaine self-administration and adult hippocampal neurogenesis; the role of adult neurogenesis in an animal model of depression and response to antidepressant exposure; the impact of neonatal seizures on dentate gyrus neurogenesis; and the contribution of a schizophrenia-susceptibility gene to adult hippocampal neurogenesis. These topics are discussed in light of the regulation of adult neurogenesis, the relationship to normal neurogenesis in adulthood and aging, and, importantly, the manipulation of neurogenesis to promote mental health and treat mental illness.
Collapse
|
49
|
Molecular and cellular basis of epileptogenesis in symptomatic epilepsy. Epilepsy Behav 2009; 14 Suppl 1:16-25. [PMID: 18835369 DOI: 10.1016/j.yebeh.2008.09.023] [Citation(s) in RCA: 216] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 09/18/2008] [Accepted: 09/18/2008] [Indexed: 12/21/2022]
Abstract
Epileptogenesis refers to a process in which an initial brain-damaging insult triggers a cascade of molecular and cellular changes that eventually lead to the occurrence of spontaneous seizures. Cellular alterations include neurodegeneration, neurogenesis, axonal sprouting, axonal injury, dendritic remodeling, gliosis, invasion of inflammatory cells, angiogenesis, alterations in extracellular matrix, and acquired channelopathies. Large-scale molecular profiling of epileptogenic tissue has provided information about the molecular pathways that can initiate and maintain cellular alterations. Currently we are learning how these pathways contribute to postinjury epileptogenesis and recovery process and whether they could be used as treatment targets.
Collapse
|
50
|
Kuruba R, Hattiangady B, Shetty AK. Hippocampal neurogenesis and neural stem cells in temporal lobe epilepsy. Epilepsy Behav 2009; 14 Suppl 1:65-73. [PMID: 18796338 PMCID: PMC2654382 DOI: 10.1016/j.yebeh.2008.08.020] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 08/28/2008] [Accepted: 08/29/2008] [Indexed: 12/12/2022]
Abstract
Virtually all mammals, including humans, exhibit neurogenesis throughout life in the hippocampus, a learning and memory center in the brain. Numerous studies in animal models imply that hippocampal neurogenesis is important for functions such as learning, memory, and mood. Interestingly, hippocampal neurogenesis is very sensitive to physiological and pathological stimuli. Certain pathological stimuli such as seizures alter both the amount and the pattern of neurogenesis, though the overall effect depends on the type of seizures. Acute seizures are classically associated with augmentation of neurogenesis and migration of newly born neurons into ectopic regions such as the hilus and the molecular layer of the dentate gyrus. Additional studies suggest that abnormally migrated newly born neurons play a role in the occurrence of epileptogenic hippocampal circuitry characteristically seen after acute seizures, status epilepticus, or head injury. Recurrent spontaneous seizures such as those typically observed in chronic temporal lobe epilepsy are associated with substantially reduced neurogenesis, which, interestingly, coexists with learning and memory impairments and depression. In this review, we discuss both the extent and the potential implications of abnormal hippocampal neurogenesis induced by acute seizures as well as recurrent spontaneous seizures. We also discuss the consequences of chronic spontaneous seizures on differentiation of neural stem cell progeny in the hippocampus and strategies that are potentially useful for normalizing neurogenesis in chronic temporal lobe epilepsy.
Collapse
Affiliation(s)
- Ramkumar Kuruba
- Department of Surgery (Neurosurgery) Duke University Medical Center, Durham, NC 27710,Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, NC 27705
| | - Bharathi Hattiangady
- Department of Surgery (Neurosurgery) Duke University Medical Center, Durham, NC 27710,Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, NC 27705
| | - Ashok K. Shetty
- Department of Surgery (Neurosurgery) Duke University Medical Center, Durham, NC 27710,Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, NC 27705,Correspondence should be addressed to: Ashok K. Shetty, M.Sc., Ph.D., Professor, Division of Neurosurgery, Box 3807, Duke University Medical Center, Durham NC 27710, Phone: 919-286-0411, Ext. 7096, E-mail:
| |
Collapse
|