1
|
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: 2.0] [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
|
2
|
Alayli A, Lockard G, Gordon J, Connolly J, Monsour M, Schimmel S, Dela Peña I, Borlongan CV. Stem Cells: Recent Developments Redefining Epilepsy Therapy. Cell Transplant 2023; 32:9636897231158967. [PMID: 36919673 PMCID: PMC10021095 DOI: 10.1177/09636897231158967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/31/2023] [Accepted: 02/06/2023] [Indexed: 03/16/2023] Open
Abstract
The field of stem cell therapy is growing rapidly and hopes to offer an alternative solution to diseases that are historically treated medically or surgically. One such focus of research is the treatment of medically refractory epilepsy, which is traditionally approached from a surgical or interventional standpoint. Research shows that stem cell transplantation has potential to offer significant benefits to the epilepsy patient by reducing seizure frequency, intensity, and neurological deficits that often result from the condition. This review explores the basic science progress made on the topic of stem cells and epilepsy by focusing on experiments using animal models and highlighting the most recent developments from the last 4 years.
Collapse
Affiliation(s)
- Adam Alayli
- University of South Florida Morsani
College of Medicine, Tampa, FL, USA
| | - Gavin Lockard
- University of South Florida Morsani
College of Medicine, Tampa, FL, USA
| | - Jonah Gordon
- University of South Florida Morsani
College of Medicine, Tampa, FL, USA
| | - Jacob Connolly
- University of South Florida Morsani
College of Medicine, Tampa, FL, USA
| | - Molly Monsour
- University of South Florida Morsani
College of Medicine, Tampa, FL, USA
| | - Samantha Schimmel
- University of South Florida Morsani
College of Medicine, Tampa, FL, USA
| | - Ike Dela Peña
- Department of Pharmaceutical and
Administrative Sciences, Loma Linda University, Loma Linda, CA, USA
| | - Cesar V. Borlongan
- Center of Excellence for Aging and
Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL,
USA
| |
Collapse
|
3
|
Zayed MA, Sultan S, Alsaab HO, Yousof SM, Alrefaei GI, Alsubhi NH, Alkarim S, Al Ghamdi KS, Bagabir SA, Jana A, Alghamdi BS, Atta HM, Ashraf GM. Stem-Cell-Based Therapy: The Celestial Weapon against Neurological Disorders. Cells 2022; 11:3476. [PMID: 36359871 PMCID: PMC9655836 DOI: 10.3390/cells11213476] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/15/2022] [Accepted: 10/24/2022] [Indexed: 09/01/2023] Open
Abstract
Stem cells are a versatile source for cell therapy. Their use is particularly significant for the treatment of neurological disorders for which no definitive conventional medical treatment is available. Neurological disorders are of diverse etiology and pathogenesis. Alzheimer's disease (AD) is caused by abnormal protein deposits, leading to progressive dementia. Parkinson's disease (PD) is due to the specific degeneration of the dopaminergic neurons causing motor and sensory impairment. Huntington's disease (HD) includes a transmittable gene mutation, and any treatment should involve gene modulation of the transplanted cells. Multiple sclerosis (MS) is an autoimmune disorder affecting multiple neurons sporadically but induces progressive neuronal dysfunction. Amyotrophic lateral sclerosis (ALS) impacts upper and lower motor neurons, leading to progressive muscle degeneration. This shows the need to try to tailor different types of cells to repair the specific defect characteristic of each disease. In recent years, several types of stem cells were used in different animal models, including transgenic animals of various neurologic disorders. Based on some of the successful animal studies, some clinical trials were designed and approved. Some studies were successful, others were terminated and, still, a few are ongoing. In this manuscript, we aim to review the current information on both the experimental and clinical trials of stem cell therapy in neurological disorders of various disease mechanisms. The different types of cells used, their mode of transplantation and the molecular and physiologic effects are discussed. Recommendations for future use and hopes are highlighted.
Collapse
Affiliation(s)
- Mohamed A. Zayed
- Physiology Department, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Physiology Department, Faculty of Medicine, Menoufia University, Menoufia 32511, Egypt
| | - Samar Sultan
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Regenerative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Shimaa Mohammad Yousof
- Physiology Department, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Medical Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ghadeer I. Alrefaei
- Department of Biology, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Nouf H. Alsubhi
- Department of Biological Sciences, College of Science & Arts, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| | - Saleh Alkarim
- Embryonic and Cancer Stem Cell Research Group, King Fahad Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Biology Department, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Embryonic Stem Cells Research Unit, Biology Department, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Kholoud S. Al Ghamdi
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Sali Abubaker Bagabir
- Genetic Unit, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Ankit Jana
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Campus-11, Patia, Bhubaneswar 751024, Odisha, India
| | - Badrah S. Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hazem M. Atta
- Clinical Biochemistry Department, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Ghulam Md Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, University City, Sharjah 27272, United Arab Emirates
| |
Collapse
|
4
|
Upadhya D, Attaluri S, Liu Y, Hattiangady B, Castro OW, Shuai B, Dong Y, Zhang SC, Shetty AK. Grafted hPSC-derived GABA-ergic interneurons regulate seizures and specific cognitive function in temporal lobe epilepsy. NPJ Regen Med 2022; 7:38. [PMID: 35915118 PMCID: PMC9343458 DOI: 10.1038/s41536-022-00234-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 06/24/2022] [Indexed: 11/25/2022] Open
Abstract
Interneuron loss/dysfunction contributes to spontaneous recurrent seizures (SRS) in chronic temporal lobe epilepsy (TLE), and interneuron grafting into the epileptic hippocampus reduces SRS and improves cognitive function. This study investigated whether graft-derived gamma-aminobutyric acid positive (GABA-ergic) interneurons directly regulate SRS and cognitive function in a rat model of chronic TLE. Human pluripotent stem cell-derived medial ganglionic eminence-like GABA-ergic progenitors, engineered to express hM4D(Gi), a designer receptor exclusively activated by designer drugs (DREADDs) through CRISPR/Cas9 technology, were grafted into hippocampi of chronically epileptic rats to facilitate the subsequent silencing of graft-derived interneurons. Such grafting substantially reduced SRS and improved hippocampus-dependent cognitive function. Remarkably, silencing of graft-derived interneurons with a designer drug increased SRS and induced location memory impairment but did not affect pattern separation function. Deactivation of DREADDs restored both SRS control and object location memory function. Thus, transplanted GABA-ergic interneurons could directly regulate SRS and specific cognitive functions in TLE.
Collapse
Affiliation(s)
- Dinesh Upadhya
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA.,Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA.,Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA.,Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA
| | - Yan Liu
- Waisman Center, Departments of Neuroscience and Neurology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Bharathi Hattiangady
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA.,Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA
| | - Olagide W Castro
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA.,Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA.,Institute of Biological Sciences and Health, Federal Univ of Alagoas (UFAL), Maceio, AL, Brazil
| | - Bing Shuai
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA.,Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA
| | - Yi Dong
- Waisman Center, Departments of Neuroscience and Neurology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Su-Chun Zhang
- Waisman Center, Departments of Neuroscience and Neurology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA. .,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA. .,Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA.
| |
Collapse
|
5
|
Waloschková E, Gonzalez-Ramos A, Mikroulis A, Kudláček J, Andersson M, Ledri M, Kokaia M. Human Stem Cell-Derived GABAergic Interneurons Establish Efferent Synapses onto Host Neurons in Rat Epileptic Hippocampus and Inhibit Spontaneous Recurrent Seizures. Int J Mol Sci 2021; 22:ijms222413243. [PMID: 34948040 PMCID: PMC8705828 DOI: 10.3390/ijms222413243] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 11/17/2022] Open
Abstract
Epilepsy is a complex disorder affecting the central nervous system and is characterised by spontaneously recurring seizures (SRSs). Epileptic patients undergo symptomatic pharmacological treatments, however, in 30% of cases, they are ineffective, mostly in patients with temporal lobe epilepsy. Therefore, there is a need for developing novel treatment strategies. Transplantation of cells releasing γ-aminobutyric acid (GABA) could be used to counteract the imbalance between excitation and inhibition within epileptic neuronal networks. We generated GABAergic interneuron precursors from human embryonic stem cells (hESCs) and grafted them in the hippocampi of rats developing chronic SRSs after kainic acid-induced status epilepticus. Using whole-cell patch-clamp recordings, we characterised the maturation of the grafted cells into functional GABAergic interneurons in the host brain, and we confirmed the presence of functional inhibitory synaptic connections from grafted cells onto the host neurons. Moreover, optogenetic stimulation of grafted hESC-derived interneurons reduced the rate of epileptiform discharges in vitro. We also observed decreased SRS frequency and total time spent in SRSs in these animals in vivo as compared to non-grafted controls. These data represent a proof-of-concept that hESC-derived GABAergic neurons can exert a therapeutic effect on epileptic animals presumably through establishing inhibitory synapses with host neurons.
Collapse
Affiliation(s)
- Eliška Waloschková
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, 221 84 Lund, Sweden; (A.G.-R.); (A.M.); (J.K.); (M.A.); (M.L.)
- Correspondence: (E.W.); (M.K.)
| | - Ana Gonzalez-Ramos
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, 221 84 Lund, Sweden; (A.G.-R.); (A.M.); (J.K.); (M.A.); (M.L.)
| | - Apostolos Mikroulis
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, 221 84 Lund, Sweden; (A.G.-R.); (A.M.); (J.K.); (M.A.); (M.L.)
| | - Jan Kudláček
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, 221 84 Lund, Sweden; (A.G.-R.); (A.M.); (J.K.); (M.A.); (M.L.)
- Department of Physiology, Second Faculty of Medicine, Charles University, 150 06 Prague, Czech Republic
| | - My Andersson
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, 221 84 Lund, Sweden; (A.G.-R.); (A.M.); (J.K.); (M.A.); (M.L.)
| | - Marco Ledri
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, 221 84 Lund, Sweden; (A.G.-R.); (A.M.); (J.K.); (M.A.); (M.L.)
| | - Merab Kokaia
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, 221 84 Lund, Sweden; (A.G.-R.); (A.M.); (J.K.); (M.A.); (M.L.)
- Correspondence: (E.W.); (M.K.)
| |
Collapse
|
6
|
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: 8] [Impact Index Per Article: 2.7] [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
|
7
|
GABAergic deficits in absence of LPA 1 receptor, associated anxiety-like and coping behaviors, and amelioration by interneuron precursor transplants into the dorsal hippocampus. Brain Struct Funct 2021; 226:1479-1495. [PMID: 33792787 DOI: 10.1007/s00429-021-02261-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 03/17/2021] [Indexed: 02/05/2023]
Abstract
Defects in GABAergic function can cause anxiety- and depression-like behaviors among other neuropsychiatric disorders. Therapeutic strategies using the transplantation of GABAergic interneuron progenitors derived from the medial ganglionic eminence (MGE) into the adult hippocampus reversed the symptomatology in multiple rodent models of interneuron-related pathologies. In turn, the lysophosphatidic acid receptor LPA1 has been reported to be essential for hippocampal function. Converging evidence suggests that deficits in LPA1 receptor signaling represent a core feature underlying comparable hippocampal dysfunction and behaviors manifested in common neuropsychiatric conditions. Here, we first analyzed the GABAergic interneurons in the hippocampus of wild-type and maLPA1-null mice, lacking the LPA1 receptor. Our data revealed a reduction in the number of neurons expressing GABA, calcium-binding proteins, and neuropeptides such as somatostatin and neuropeptide Y in the hippocampus of maLPA1-null mice. Then, we used interneuron precursor transplants to test links between hippocampal GABAergic interneuron deficit, cell-based therapy, and LPA1 receptor-dependent psychiatric disease-like phenotypes. For this purpose, we transplanted MGE-derived interneuron precursors into the adult hippocampus of maLPA1-null mice, to test their effects on GABAergic deficit and behavioral symptoms associated with the absence of the LPA1 receptor. Transplant studies in maLPA1-null mice showed that grafted cells were able to restore the hippocampal host environment, decrease the anxiety-like behaviors and neutralize passive coping, with no abnormal effects on motor activity. Furthermore, grafted MGE-derived cells maintained their normal differentiation program. These findings reinforce the use of cell-based strategies for brain disorders and suggest that the LPA1 receptor represents a potential target for interneuron-related neuropsychiatric disorders.
Collapse
|
8
|
Sadanandan N, Saft M, Gonzales-Portillo B, Borlongan CV. Multipronged Attack of Stem Cell Therapy in Treating the Neurological and Neuropsychiatric Symptoms of Epilepsy. Front Pharmacol 2021; 12:596287. [PMID: 33815100 PMCID: PMC8010689 DOI: 10.3389/fphar.2021.596287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/15/2021] [Indexed: 11/13/2022] Open
Abstract
Epilepsy stands as a life-threatening disease that is characterized by unprovoked seizures. However, an important characteristic of epilepsy that needs to be examined is the neuropsychiatric aspect. Epileptic patients endure aggression, depression, and other psychiatric illnesses. Therapies for epilepsy can be divided into two categories: antiepileptic medications and surgical resection. Antiepileptic drugs are used to attenuate heightened neuronal firing and to lessen seizure frequency. Alternatively, surgery can also be conducted to physically cut out the area of the brain that is assumed to be the root cause for the anomalous firing that triggers seizures. While both treatments serve as viable approaches that aim to regulate seizures and ameliorate the neurological detriments spurred by epilepsy, they do not serve to directly counteract epilepsy's neuropsychiatric traits. To address this concern, a potential new treatment involves the use of stem cells. Stem cell therapy has been employed in experimental models of neurological maladies, such as Parkinson's disease, and neuropsychiatric illnesses like depression. Cell-based treatments for epilepsy utilizing stem cells such as neural stem cells (NSCs), mesenchymal stem cells (MSCs), and interneuron grafts have been explored in preclinical and clinical settings, highlighting both the acute and chronic stages of epilepsy. However, it is difficult to create an animal model to capitalize on all the components of epilepsy due to the challenges in delineating the neuropsychiatric aspect. Therefore, further preclinical investigation into the safety and efficacy of stem cell therapy in addressing both the neurological and the neuropsychiatric components of epilepsy is warranted in order to optimize cell dosage, delivery, and timing of cell transplantation.
Collapse
Affiliation(s)
| | | | | | - Cesar V. Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| |
Collapse
|
9
|
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: 2.3] [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
|
10
|
D'Alessio L, Mesarosova L, Anink JJ, Kochen S, Solís P, Oddo S, Konopka H, Iyer AM, Mühlebner A, Lucassen PJ, Aronica E, van Vliet EA. Reduced expression of the glucocorticoid receptor in the hippocampus of patients with drug-resistant temporal lobe epilepsy and comorbid depression. Epilepsia 2020; 61:1595-1605. [PMID: 32652588 PMCID: PMC7496961 DOI: 10.1111/epi.16598] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 05/16/2020] [Accepted: 06/11/2020] [Indexed: 12/23/2022]
Abstract
Objective Depressive disorders are common among about 50% of the patients with drug‐resistant temporal lobe epilepsy (TLE). The underlying etiology remains elusive, but hypothalamus‐pituitary‐adrenal (HPA) axis activation due to changes in glucocorticoid receptor (GR) protein expression could play an important role. Therefore, we set out to investigate expression of the GR in the hippocampus, an important brain region for HPA axis feedback, of patients with drug‐resistant TLE, with and without comorbid depression. Methods GR expression was studied using immunohistochemistry on hippocampal sections from well‐characterized TLE patients with depression (TLE + D, n = 14) and without depression (TLE − D, n = 12) who underwent surgery for drug‐resistant epilepsy, as well as on hippocampal sections from autopsy control cases (n = 9). Video–electroencephalography (EEG), magnetic resonance imaging (MRI), and psychiatric and memory assessments were performed prior to surgery. Results Abundant GR immunoreactivity was present in dentate gyrus granule cells and CA1 pyramidal cells of controls. In contrast, neuronal GR expression was lower in patients with TLE, particularly in the TLE + D group. Quantitative analysis showed a smaller GR+ area in TLE + D as compared to TLE − D patients and controls. Furthermore, the ratio between the number of GR+/NeuN+ cells was lower in patients with TLE + D as compared to TLE − D and correlated negatively with the depression severity based on psychiatric history. The expression of the GR was also lower in glial cells of TLE + D compared to TLE − D patients and correlated negatively to the severity of depression. Significance Reduced hippocampal GR expression may be involved in the etiology of depression in patients with TLE and could constitute a biological marker of depression in these patients.
Collapse
Affiliation(s)
- Luciana D'Alessio
- Universidad de Buenos Aires, IBCN-CONICET, Centro de Epilepsia Hospital Ramos Mejía y Hospital El Cruce, ENyS-CONICET, Buenos Aires, Argentina
| | - Lucia Mesarosova
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Jasper J Anink
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Silvia Kochen
- Universidad de Buenos Aires, IBCN-CONICET, Centro de Epilepsia Hospital Ramos Mejía y Hospital El Cruce, ENyS-CONICET, Buenos Aires, Argentina
| | - Patricia Solís
- Universidad de Buenos Aires, IBCN-CONICET, Centro de Epilepsia Hospital Ramos Mejía y Hospital El Cruce, ENyS-CONICET, Buenos Aires, Argentina
| | - Silvia Oddo
- Universidad de Buenos Aires, IBCN-CONICET, Centro de Epilepsia Hospital Ramos Mejía y Hospital El Cruce, ENyS-CONICET, Buenos Aires, Argentina
| | - Hector Konopka
- Universidad de Buenos Aires, IBCN-CONICET, Centro de Epilepsia Hospital Ramos Mejía y Hospital El Cruce, ENyS-CONICET, Buenos Aires, Argentina
| | - Anand M Iyer
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Angelika Mühlebner
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| | - Erwin A van Vliet
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands.,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
11
|
Curcumin alleviates neuroinflammation, enhances hippocampal neurogenesis, and improves spatial memory after traumatic brain injury. Brain Res Bull 2020; 162:84-93. [PMID: 32502596 DOI: 10.1016/j.brainresbull.2020.05.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/24/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
Cognitive decline is one of the most obvious symptoms of traumatic brain injury (TBI). Previous studies have demonstrated that cognitive decline is related to substantially increased neuroinflammation and decreased neurogenesis in the hippocampus in a rat model of TBI. Using this model, we explored the role of curcumin (Cur) in ameliorating TBI-impaired spatial memory because Cur has been shown to exhibit anti-chronic-neuroinflammatory, neurogenesis-promoting, and memory-improving properties. Animals received daily Cur or vehicle treatment for 28 days after TBI and also received 50-bromodeoxyuridine(BrdU) for the first 7 days of the treatment for assaying neurogenesis. An optimal Cur dose of 30 mg/kg, selected from a range of 10-50 mg/kg, was used for the present study. Neuroinflammation was evaluated by astrocyte hypertrophy, activated microglia, and inflammatory factors in the hippocampus. Behavioral water-maze studies were conducted for 5 days, starting at 35-day post-TBI. The tropomyosin receptor kinase B (Trkb) inhibitor, ANA-12, was used to test the role of the brain-derived neurotrophic factor (BDNF)/ TrkB/Phosphoinositide 3-kinase (PI3K)/Akt signaling pathway in regulating inflammation and neurogenesis in the hippocampus. Treatment with Cur ameliorated the spatial memory of TBI rats, reduced TBI-induced chronic inflammation, typified by diminished astrocyte hypertrophy, reduction in activated microglia, declined inflammatory factors, and increased neurogenesis in the hippocampus. We also found that BDNF/Trkb/PI3K/Akt signaling was involved in the effects of Cur in TBI rats. Thus, Cur treatment can ameliorate the spatial memory in a murine model of TBI, which may be attributable to decreased chronic neuroinflammation, increased hippocampal neurogenesis, and/or BDNF/Trkb/PI3K/Akt signaling.
Collapse
|
12
|
Gebril HM, Rose RM, Gesese R, Emond MP, Huo Y, Aronica E, Boison D. Adenosine kinase inhibition promotes proliferation of neural stem cells after traumatic brain injury. Brain Commun 2020; 2:fcaa017. [PMID: 32322821 PMCID: PMC7158236 DOI: 10.1093/braincomms/fcaa017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/26/2019] [Accepted: 01/01/2020] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a major public health concern and remains a leading cause of disability and socio-economic burden. To date, there is no proven therapy that promotes brain repair following an injury to the brain. In this study, we explored the role of an isoform of adenosine kinase expressed in the cell nucleus (ADK-L) as a potential regulator of neural stem cell proliferation in the brain. The rationale for this hypothesis is based on coordinated expression changes of ADK-L during foetal and postnatal murine and human brain development indicating a role in the regulation of cell proliferation and plasticity in the brain. We first tested whether the genetic disruption of ADK-L would increase neural stem cell proliferation after TBI. Three days after TBI, modelled by a controlled cortical impact, transgenic mice, which lack ADK-L (ADKΔneuron) in the dentate gyrus (DG) showed a significant increase in neural stem cell proliferation as evidenced by significant increases in doublecortin and Ki67-positive cells, whereas animals with transgenic overexpression of ADK-L in dorsal forebrain neurons (ADK-Ltg) showed an opposite effect of attenuated neural stem cell proliferation. Next, we translated those findings into a pharmacological approach to augment neural stem cell proliferation in the injured brain. Wild-type C57BL/6 mice were treated with the small molecule adenosine kinase inhibitor 5-iodotubercidin for 3 days after the induction of TBI. We demonstrate significantly enhanced neural stem cell proliferation in the DG of 5-iodotubercidin-treated mice compared to vehicle-treated injured animals. To rule out the possibility that blockade of ADK-L has any effects in non-injured animals, we quantified baseline neural stem cell proliferation in ADKΔneuron mice, which was not altered, whereas baseline neural stem cell proliferation in ADK-Ltg mice was enhanced. Together these findings demonstrate a novel function of ADK-L involved in the regulation of neural stem cell proliferation after TBI.
Collapse
Affiliation(s)
- Hoda M Gebril
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA.,Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR 97232, USA
| | - Rizelle Mae Rose
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR 97232, USA
| | - Raey Gesese
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR 97232, USA
| | - Martine P Emond
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR 97232, USA
| | - Yuqing Huo
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center and Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen (SEIN) Nederland, Heemstede, The Netherlands
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
13
|
Andrejew R, Glaser T, Oliveira-Giacomelli Á, Ribeiro D, Godoy M, Granato A, Ulrich H. Targeting Purinergic Signaling and Cell Therapy in Cardiovascular and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:275-353. [PMID: 31898792 DOI: 10.1007/978-3-030-31206-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular purines exert several functions in physiological and pathophysiological mechanisms. ATP acts through P2 receptors as a neurotransmitter and neuromodulator and modulates heart contractility, while adenosine participates in neurotransmission, blood pressure, and many other mechanisms. Because of their capability to differentiate into mature cell types, they provide a unique therapeutic strategy for regenerating damaged tissue, such as in cardiovascular and neurodegenerative diseases. Purinergic signaling is pivotal for controlling stem cell differentiation and phenotype determination. Proliferation, differentiation, and apoptosis of stem cells of various origins are regulated by purinergic receptors. In this chapter, we selected neurodegenerative and cardiovascular diseases with clinical trials using cell therapy and purinergic receptor targeting. We discuss these approaches as therapeutic alternatives to neurodegenerative and cardiovascular diseases. For instance, promising results were demonstrated in the utilization of mesenchymal stem cells and bone marrow mononuclear cells in vascular regeneration. Regarding neurodegenerative diseases, in general, P2X7 and A2A receptors mostly worsen the degenerative state. Stem cell-based therapy, mainly through mesenchymal and hematopoietic stem cells, showed promising results in improving symptoms caused by neurodegeneration. We propose that purinergic receptor activity regulation combined with stem cells could enhance proliferative and differentiation rates as well as cell engraftment.
Collapse
Affiliation(s)
- Roberta Andrejew
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Talita Glaser
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Ágatha Oliveira-Giacomelli
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Deidiane Ribeiro
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Mariana Godoy
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.,Laboratory of Neurodegenerative Diseases, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro Granato
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
14
|
Upadhya D, Kodali M, Gitai D, Castro OW, Zanirati G, Upadhya R, Attaluri S, Mitra E, Shuai B, Hattiangady B, Shetty AK. A Model of Chronic Temporal Lobe Epilepsy Presenting Constantly Rhythmic and Robust Spontaneous Seizures, Co-morbidities and Hippocampal Neuropathology. Aging Dis 2019; 10:915-936. [PMID: 31595192 PMCID: PMC6764729 DOI: 10.14336/ad.2019.0720] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/20/2019] [Indexed: 12/18/2022] Open
Abstract
Many animal prototypes illustrating the various attributes of human temporal lobe epilepsy (TLE) are available. These models have been invaluable for comprehending multiple epileptogenic processes, modifications in electrophysiological properties, neuronal hyperexcitability, neurodegeneration, neural plasticity, and chronic neuroinflammation in TLE. Some models have also uncovered the efficacy of new antiepileptic drugs or biologics for alleviating epileptogenesis, cognitive impairments, or spontaneous recurrent seizures (SRS). Nonetheless, the suitability of these models for testing candidate therapeutics in conditions such as chronic TLE is debatable because of a lower frequency of SRS and an inconsistent pattern of SRS activity over days, weeks or months. An ideal prototype of chronic TLE for investigating novel therapeutics would need to display a large number of SRS with a dependable frequency and severity and related co-morbidities. This study presents a new kainic acid (KA) model of chronic TLE generated through induction of status epilepticus (SE) in 6-8 weeks old male F344 rats. A rigorous characterization in the chronic epilepsy period validated that the animal prototype mimicked the most salient features of robust chronic TLE. Animals displayed a constant frequency and intensity of SRS across weeks and months in the 5th and 6th month after SE, as well as cognitive and mood impairments. Moreover, SRS frequency displayed a rhythmic pattern with 24-hour periodicity and a consistently higher number of SRS in the daylight period. Besides, the model showed many neuropathological features of chronic TLE, which include a partial loss of inhibitory interneurons, reduced neurogenesis with persistent aberrant migration of newly born neurons, chronic neuroinflammation typified by hypertrophied astrocytes and rod-shaped microglia, and a significant aberrant mossy fiber sprouting in the hippocampus. This consistent chronic seizure model is ideal for investigating the efficacy of various antiepileptic drugs and biologics as well as understanding multiple pathophysiological mechanisms underlying chronic epilepsy.
Collapse
Affiliation(s)
- Dinesh Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - Daniel Gitai
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - Olagide W Castro
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - Gabriele Zanirati
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - Eeshika Mitra
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - Bharathi Hattiangady
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| |
Collapse
|
15
|
Lybrand ZR, Goswami S, Hsieh J. Stem cells: A path towards improved epilepsy therapies. Neuropharmacology 2019; 168:107781. [PMID: 31539537 DOI: 10.1016/j.neuropharm.2019.107781] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
Abstract
Despite the immense growth of new anti-seizure drugs (ASDs), approximately one-third of epilepsy patients remain resistant to current treatment options. Advancements in whole genome sequencing technology continues to identify an increasing number of epilepsy-associated genes at a rate that is outpacing the development of in vivo animal models. Patient-derived induced pluripotent stem cells (iPSCs) show promise in providing a platform for modeling genetic epilepsies, high throughput drug screening, and personalized medicine. This is largely due to the ease of collecting donor cells for iPSC reprogramming, and their ability to be maintained in vitro, while preserving the patient's genetic background. In this review, we summarize the current state of iPSC research in epilepsy and closely related syndromes, discuss the growing need for high-throughput drug screening (HTS), and review the use of stem cell technology for the purpose of autologous transplantation for epilepsy stem cell therapy. Although the use of iPSC technology, as it applies to ASD discovery, is in its infancy, we highlight the significant progress that has been made in phenotype and assay development to facilitate systematic HTS for personalized medicine. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Zane R Lybrand
- Department of Biology and Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Sonal Goswami
- Department of Biology and Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Jenny Hsieh
- Department of Biology and Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
16
|
Alia C, Terrigno M, Busti I, Cremisi F, Caleo M. Pluripotent Stem Cells for Brain Repair: Protocols and Preclinical Applications in Cortical and Hippocampal Pathologies. Front Neurosci 2019; 13:684. [PMID: 31447623 PMCID: PMC6691396 DOI: 10.3389/fnins.2019.00684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/14/2019] [Indexed: 12/13/2022] Open
Abstract
Brain injuries causing chronic sensory or motor deficit, such as stroke, are among the leading causes of disability worldwide, according to the World Health Organization; furthermore, they carry heavy social and economic burdens due to decreased quality of life and need of assistance. Given the limited effectiveness of rehabilitation, novel therapeutic strategies are required to enhance functional recovery. Since cell-based approaches have emerged as an intriguing and promising strategy to promote brain repair, many efforts have been made to study the functional integration of neurons derived from pluripotent stem cells (PSCs), or fetal neurons, after grafting into the damaged host tissue. PSCs hold great promises for their clinical applications, such as cellular replacement of damaged neural tissues with autologous neurons. They also offer the possibility to create in vitro models to assess the efficacy of drugs and therapies. Notwithstanding these potential applications, PSC-derived transplanted neurons have to match the precise sub-type, positional and functional identity of the lesioned neural tissue. Thus, the requirement of highly specific and efficient differentiation protocols of PSCs in neurons with appropriate neural identity constitutes the main challenge limiting the clinical use of stem cells in the near future. In this Review, we discuss the recent advances in the derivation of telencephalic (cortical and hippocampal) neurons from PSCs, assessing specificity and efficiency of the differentiation protocols, with particular emphasis on the genetic and molecular characterization of PSC-derived neurons. Second, we address the remaining challenges for cellular replacement therapies in cortical brain injuries, focusing on electrophysiological properties, functional integration and therapeutic effects of the transplanted neurons.
Collapse
Affiliation(s)
- Claudia Alia
- CNR Neuroscience Institute, National Research Council (CNR), Pisa, Italy
| | - Marco Terrigno
- Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Irene Busti
- CNR Neuroscience Institute, National Research Council (CNR), Pisa, Italy.,Department of Neuroscience, Psychology, Drugs and Child Health Area, School of Psychology, University of Florence, Florence, Italy
| | - Federico Cremisi
- Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy.,Biophysics Institute (IBF), National Research Council (CNR), Pisa, Italy
| | - Matteo Caleo
- CNR Neuroscience Institute, National Research Council (CNR), Pisa, Italy.,Department of Biomedical Sciences, University of Padua, Padua, Italy.,Padua Neuroscience Center, University of Padua, Padua, Italy
| |
Collapse
|
17
|
Depression and Temporal Lobe Epilepsy: Expression Pattern of Calbindin Immunoreactivity in Hippocampal Dentate Gyrus of Patients Who Underwent Epilepsy Surgery with and without Comorbid Depression. Behav Neurol 2019; 2019:7396793. [PMID: 31191739 PMCID: PMC6525951 DOI: 10.1155/2019/7396793] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 12/05/2022] Open
Abstract
Purpose Changes in calbindin (CB) expression have been reported in patients with temporal lobe epilepsy (TLE) with controversial implications on hippocampal functions. The aim of this study was to determine the CB immunoreactivity in hippocampal dentate gyrus of patients who underwent epilepsy surgery for drug-resistant TLE with and without comorbid depression and/or memory deficits. Methods Selected hippocampal samples from patients with TLE who underwent epilepsy surgery were included. Clinical and complementary assessment: EEG, video-EEG, MRI, psychiatric assessment (structured clinical interview, DSM-IV), and memory assessment (Rey auditory verbal learning test, RAVLT; Rey-Osterrieth complex figure test, RCFT), were determined before surgery. Hippocampal sections were processed using immunoperoxidase with the anti-calbindin antibody. The semiquantitative analysis of CB immunoreactivity was determined in dentate gyrus by computerized image analysis (ImageJ). Results Hippocampal sections of patients with TLE and HS (n = 24) and postmortem controls (n = 5) were included. A significant reduction of CB+ cells was found in patients with TLE (p < 0.05, Student's t-test). Among TLE cases (n = 24), depression (n = 12) and memory deficit (n = 17) were determined. Depression was associated with a higher % of cells with the CB dendritic expression (CB-sprouted cells) (F(1, 20) = 11.81, p = 0.003, hp2 = 0.37), a higher CB+ area (μm2) (F(1, 20) = 5.33, p = 0.032, hp2 = 0.21), and a higher optical density (F(1, 20) = 15.09, p = 0.001, hp2 = 0.43) (two-way ANOVA). The GAF scale (general assessment of functioning) of DSM-IV inversely correlated with the % of CB-sprouted cells (r = −0.52, p = 0.008) and with the CB+ area (r = −0.46, p = 0.022). Conclusions In this exploratory study, comorbid depression was associated with a differential pattern of CB cell loss in dentate gyrus combined with a higher CB sprouting. These changes may indicate granular cell dysmaturation associated to the epileptic hyperexcitability phenomena. Further investigations should be carried out to confirm these preliminary findings.
Collapse
|
18
|
Leite Góes Gitai D, de Andrade TG, Dos Santos YDR, Attaluri S, Shetty AK. Chronobiology of limbic seizures: Potential mechanisms and prospects of chronotherapy for mesial temporal lobe epilepsy. Neurosci Biobehav Rev 2019; 98:122-134. [PMID: 30629979 PMCID: PMC7023906 DOI: 10.1016/j.neubiorev.2019.01.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/11/2022]
Abstract
Mesial Temporal Lobe Epilepsy (mTLE) characterized by progressive development of complex partial seizures originating from the hippocampus is the most prevalent and refractory type of epilepsy. One of the remarkable features of mTLE is the rhythmic pattern of occurrence of spontaneous seizures, implying a dependence on the endogenous clock system for seizure threshold. Conversely, circadian rhythms are affected by epilepsy too. Comprehending how the circadian system and seizures interact with each other is essential for understanding the pathophysiology of epilepsy as well as for developing innovative therapies that are efficacious for better seizure control. In this review, we confer how the temporal dysregulation of the circadian clock in the hippocampus combined with multiple uncoupled oscillators could lead to periodic seizure occurrences and comorbidities. Unraveling these associations with additional research would help in developing chronotherapy for mTLE, based on the chronobiology of spontaneous seizures. Notably, differential dosing of antiepileptic drugs over the circadian period and/or strategies that resynchronize biological rhythms may substantially improve the management of seizures in mTLE patients.
Collapse
Affiliation(s)
- Daniel Leite Góes Gitai
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas, USA; Institute of Biological Sciences and Health, Federal University of Alagoas, Maceio, Alagoas, Brazil
| | | | | | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, 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 Station, Texas, USA; Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USA.
| |
Collapse
|
19
|
Xu K, Liu F, Xu W, Liu J, Chen S, Wu G. Transplanting GABAergic Neurons Differentiated from Neural Stem Cells into Hippocampus Inhibits Seizures and Epileptiform Discharges in Pilocarpine-Induced Temporal Lobe Epilepsy Model. World Neurosurg 2019; 128:e1-e11. [PMID: 30790741 DOI: 10.1016/j.wneu.2019.01.245] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVE This study aimed to explore whether intrahippocampal transplantation of GABAergic neurons generated in vitro ameliorated seizures and epileptiform discharges via increasing γ-aminobutyric acid (GABA)-associated inhibition mediated by the addition of new GABAergic neurons. METHODS Neural stem cells (NSCs) isolated from newborn rats were induced and differentiated into GABAergic neurons. A total of 36 Pilocarpine-induced pharmacoresistant epileptic rats were divided into 3 groups: PBS (phosphate-buffered saline) group, NSCs group, and GABAergic neurons group (GABA group), with an additional 10 normal rats used (normal rat control group). The effects of grafting on spontaneous recurrent seizures (SRS) were examined and hippocampal GABA content was measured after grafting. RESULTS In the GABA group, the frequency of electroencephalography decreased significantly compared with the PBS group (P < 0.001), but there was no significant difference between the GABA group and NSCs group. Compared with the PBS group, the overall frequency and duration of SRS significantly decreased in the transplantation group, especially in the GABA group (P < 0.01). The number of GABAergic neurons was highest in the GABA group compared with the other groups (P < 0.001). Furthermore, hippocampal GABA concentrations significantly increased in the GABA group. CONCLUSIONS We show that GABAergic neurons generated in vitro from NSCs and grafted into the hippocampi of chronically epileptic rats can significantly reduce the frequency of electroencephalography and frequency and duration of SRS via increasing GABA-associated inhibition mediated by the addition of new GABAergic neurons.
Collapse
Affiliation(s)
- Kaya Xu
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang City, China
| | - Feng Liu
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang City, China
| | - Wei Xu
- Public Health School, Guizhou Medical University, Guizhou, Guiyang City, China
| | - Jian Liu
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang City, China
| | - Shuxuan Chen
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang City, China
| | - Guofeng Wu
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang City, China.
| |
Collapse
|
20
|
Human induced pluripotent stem cell-derived MGE cell grafting after status epilepticus attenuates chronic epilepsy and comorbidities via synaptic integration. Proc Natl Acad Sci U S A 2018; 116:287-296. [PMID: 30559206 PMCID: PMC6320542 DOI: 10.1073/pnas.1814185115] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This study provides evidence that human induced pluripotent stem cell (hiPSC)-derived medial ganglionic eminence (MGE) cell grafting into the hippocampus after status epilepticus can greatly reduce the frequency of spontaneous seizures in the chronic phase through both antiepileptogenic and antiepileptic effects. The antiepileptogenic changes comprised reductions in host interneuron loss, abnormal neurogenesis, and aberrant mossy fiber sprouting, whereas the antiepileptic effects were evident from an increased occurrence of seizures after silencing of graft-derived interneurons. Additional curative impacts of grafting comprised improved cognitive and mood function. The results support the application of autologous human MGE cell therapy for temporal lobe epilepsy. Autologous cell therapy is advantageous as such a paradigm can avoid immune suppression and promote enduring graft–host integration. Medial ganglionic eminence (MGE)-like interneuron precursors derived from human induced pluripotent stem cells (hiPSCs) are ideal for developing patient-specific cell therapy in temporal lobe epilepsy (TLE). However, their efficacy for alleviating spontaneous recurrent seizures (SRS) or cognitive, memory, and mood impairments has never been tested in models of TLE. Through comprehensive video- electroencephalographic recordings and a battery of behavioral tests in a rat model, we demonstrate that grafting of hiPSC-derived MGE-like interneuron precursors into the hippocampus after status epilepticus (SE) greatly restrained SRS and alleviated cognitive, memory, and mood dysfunction in the chronic phase of TLE. Graft-derived cells survived well, extensively migrated into different subfields of the hippocampus, and differentiated into distinct subclasses of inhibitory interneurons expressing various calcium-binding proteins and neuropeptides. Moreover, grafting of hiPSC-MGE cells after SE mediated several neuroprotective and antiepileptogenic effects in the host hippocampus, as evidenced by reductions in host interneuron loss, abnormal neurogenesis, and aberrant mossy fiber sprouting in the dentate gyrus (DG). Furthermore, axons from graft-derived interneurons made synapses on the dendrites of host excitatory neurons in the DG and the CA1 subfield of the hippocampus, implying an excellent graft–host synaptic integration. Remarkably, seizure-suppressing effects of grafts were significantly reduced when the activity of graft-derived interneurons was silenced by a designer drug while using donor hiPSC-MGE cells expressing designer receptors exclusively activated by designer drugs (DREADDs). These results implied the direct involvement of graft-derived interneurons in seizure control likely through enhanced inhibitory synaptic transmission. Collectively, the results support a patient-specific MGE cell grafting approach for treating TLE.
Collapse
|
21
|
Backofen-Wehrhahn B, Gey L, Bröer S, Petersen B, Schiff M, Handreck A, Stanslowsky N, Scharrenbroich J, Weißing M, Staege S, Wegner F, Niemann H, Löscher W, Gernert M. Anticonvulsant effects after grafting of rat, porcine, and human mesencephalic neural progenitor cells into the rat subthalamic nucleus. Exp Neurol 2018; 310:70-83. [PMID: 30205107 DOI: 10.1016/j.expneurol.2018.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/20/2018] [Accepted: 09/07/2018] [Indexed: 12/11/2022]
Abstract
Cell transplantation based therapy is a promising strategy for treating intractable epilepsies. Inhibition of the subthalamic nucleus (STN) or substantia nigra pars reticulata (SNr) is a powerful experimental approach for remote control of different partial seizure types, when targeting the seizure focus is not amenable. Here, we tested the hypothesis that grafting of embryonic/fetal neural precursor cells (NPCs) from various species (rat, human, pig) into STN or SNr of adult rats induces anticonvulsant effects. To rationally refine this approach, we included NPCs derived from the medial ganglionic eminence (MGE) and ventral mesencephalon (VM), both of which are able to develop a GABAergic phenotype. All VM- and MGE-derived cells showed intense migration behavior after grafting into adult rats, developed characteristics of inhibitory interneurons, and survived at least up to 4 months after transplantation. By using the intravenous pentylenetetrazole (PTZ) seizure threshold test in adult rats, transient anticonvulsant effects were observed after bilateral grafting of NPCs derived from human and porcine VM into STN, but not after SNr injection (site-specificity). In contrast, MGE-derived NPCs did not cause anticonvulsant effects after grafting into STN or SNr (cell-specificity). Neither induction of status epilepticus by lithium-pilocarpine to induce neuronal damage prior to the PTZ test nor pretreatment of MGE cells with retinoic acid and potassium chloride to increase differentiation into GABAergic neurons could enhance anticonvulsant effectiveness of MGE cells. This is the first proof-of-principle study showing anticonvulsant effects by bilateral xenotransplantation of NPCs into the STN. Our study highlights the value of VM-derived NPCs for interneuron-based cell grafting targeting the STN.
Collapse
Affiliation(s)
- Bianca Backofen-Wehrhahn
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Laura Gey
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Sonja Bröer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Björn Petersen
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Mariensee, Germany
| | - Miriam Schiff
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Annelie Handreck
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | | | - Jessica Scharrenbroich
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Michael Weißing
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Selma Staege
- Center for Systems Neuroscience, Hannover, Germany; Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Florian Wegner
- Center for Systems Neuroscience, Hannover, Germany; Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Heiner Niemann
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Mariensee, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Manuela Gernert
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
22
|
Zhu Q, Naegele JR, Chung S. Cortical GABAergic Interneuron/Progenitor Transplantation as a Novel Therapy for Intractable Epilepsy. Front Cell Neurosci 2018; 12:167. [PMID: 29997478 PMCID: PMC6028694 DOI: 10.3389/fncel.2018.00167] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/30/2018] [Indexed: 11/24/2022] Open
Abstract
Epilepsy is a severe neurological disease affecting more than 70 million people worldwide that is characterized by unpredictable and abnormal electrical discharges resulting in recurrent seizures. Although antiepileptic drugs (AEDs) are the mainstay of epilepsy treatment for seizure control, about one third of patients with epilepsy suffer from intractable seizures that are unresponsive to AEDs. Furthermore, the patients that respond to AEDs typically experience adverse systemic side effects, underscoring the urgent need to develop new therapies that target epileptic foci rather than more systemic interventions. Neurosurgical removal of affected brain tissues or implanting neurostimulator devices are effective options only for a fraction of patients with drug-refractory seizures, so it is imperative to develop treatments that are more generally applicable and restorative in nature. Considering the abnormalities of GABAergic inhibitory interneurons in epileptic brain tissues, one strategy with considerable promise is to restore normal circuit function by transplanting GABAergic interneurons/progenitors into the seizure focus. In this review, we focus on recent studies of cortical GABAergic interneuron transplantation to treat epilepsy and discuss critical issues in moving this promising experimental therapeutic treatment into clinic.
Collapse
Affiliation(s)
- Qian Zhu
- Translational Stem Cell Neurobiology Laboratory, Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, United States
| | - Janice R. Naegele
- Hall-Atwater Laboratory, Department of Biology, Program in Neuroscience and Behavior, Wesleyan University, Middletown, CT, United States
| | - Sangmi Chung
- Translational Stem Cell Neurobiology Laboratory, Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
23
|
Falcicchia C, Simonato M, Verlengia G. New Tools for Epilepsy Therapy. Front Cell Neurosci 2018; 12:147. [PMID: 29896092 PMCID: PMC5986878 DOI: 10.3389/fncel.2018.00147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/14/2018] [Indexed: 12/19/2022] Open
Abstract
One third of the epilepsies are refractory to conventional antiepileptic drugs (AEDs) and, therefore, identification of new therapies is highly needed. Here, we briefly describe two approaches, direct cell grafting and gene therapy, that may represent alternatives to conventional drugs for the treatment of focal epilepsies. In addition, we discuss more in detail some new tools, cell based-biodelivery systems (encapsulated cell biodelivery (ECB) devices) and new generation gene therapy vectors, which may help in the progress toward clinical translation. The field is advancing rapidly, and there is optimism that cell and/or gene therapy strategies will soon be ready for testing in drug-resistant epileptic patients.
Collapse
Affiliation(s)
- Chiara Falcicchia
- Department of Medical Sciences, Section of Pharmacology, and Neuroscience Center, University of Ferrara and National Institute of Neuroscience, Ferrara, Italy
| | - Michele Simonato
- Department of Medical Sciences, Section of Pharmacology, and Neuroscience Center, University of Ferrara and National Institute of Neuroscience, Ferrara, Italy.,School of Medicine, University Vita-Salute San Raffaele, Milan, Italy
| | - Gianluca Verlengia
- Department of Medical Sciences, Section of Pharmacology, and Neuroscience Center, University of Ferrara and National Institute of Neuroscience, Ferrara, Italy.,School of Medicine, University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
24
|
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: 59] [Impact Index Per Article: 9.8] [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
|
25
|
Fukumura S, Sasaki M, Kataoka-Sasaki Y, Oka S, Nakazaki M, Nagahama H, Morita T, Sakai T, Tsutsumi H, Kocsis JD, Honmou O. Intravenous infusion of mesenchymal stem cells reduces epileptogenesis in a rat model of status epilepticus. Epilepsy Res 2018; 141:56-63. [PMID: 29475054 DOI: 10.1016/j.eplepsyres.2018.02.008] [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: 09/11/2017] [Revised: 01/22/2018] [Accepted: 02/13/2018] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Status epilepticus (SE) causes neuronal cell death, aberrant mossy fiber sprouting (MFS), and cognitive deteriorations. The present study tested the hypothesis that systemically infused mesenchymal stem cells (MSCs) reduce epileptogenesis by inhibiting neuronal cell death and suppressing aberrant MFS, leading to cognitive function preservation in a rat model of epilepsy. METHODS SE was induced using the lithium-pilocarpine injection model. The seizure frequency was scored using a video-monitoring system and the Morris water maze test was carried out to evaluate cognitive function. Comparisons were made between MSCs- and vehicle-infused rats. Immunohistochemical staining was performed to detect Green fluorescent protein (GFP)+ MSCs and to quantify the number of GAD67+ and NeuN+ neurons in the hippocampus. Manganese-enhanced magnetic resonance imaging (MEMRI) and Timm staining were also performed to assess the MFS. RESULTS MSC infusion inhibited epileptogenesis and preserved cognitive function after SE. The infused GFP+ MSCs were accumulated in the hippocampus and were associated with the preservation of GAD67+ and NeuN+ hippocampal neurons. Furthermore, the MSC infusion suppressed the aberrant MFS in the hippocampus as evidenced by MEMRI and Timm staining. CONCLUSIONS This study demonstrated that the intravenous infusion of MSCs mitigated epileptogenesis, thus advancing MSCs as an effective approach for epilepsy in clinical practice.
Collapse
Affiliation(s)
- Shinobu Fukumura
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan; Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan; Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA; Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT, 06516, USA.
| | - Yuko Kataoka-Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Shinichi Oka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Masahito Nakazaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Hiroshi Nagahama
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Tomonori Morita
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Takuro Sakai
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan; Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Hiroyuki Tsutsumi
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Jeffery D Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA; Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan; Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA; Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| |
Collapse
|
26
|
Lippert T, Gelineau L, Napoli E, Borlongan CV. Harnessing neural stem cells for treating psychiatric symptoms associated with fetal alcohol spectrum disorder and epilepsy. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:10-22. [PMID: 28365374 DOI: 10.1016/j.pnpbp.2017.03.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/09/2017] [Indexed: 12/20/2022]
Abstract
Brain insults with progressive neurodegeneration are inherent in pathological symptoms that represent many psychiatric illnesses. Neural network disruptions characterized by impaired neurogenesis have been recognized to precede, accompany, and possibly even exacerbate the evolution and progression of symptoms of psychiatric disorders. Here, we focus on the neurodegeneration and the resulting psychiatric symptoms observed in fetal alcohol spectrum disorder and epilepsy, in an effort to show that these two diseases are candidate targets for stem cell therapy. In particular, we provide preclinical evidence in the transplantation of neural stem cells (NSCs) in both conditions, highlighting the potential of this cell-based treatment for correcting the psychiatric symptoms that plague these two disorders. Additionally, we discuss the challenges of NSC transplantation and offer insights into the mechanisms that may mediate the therapeutic benefits and can be exploited to overcome the hurdles of translating this therapy from the laboratory to the clinic. Our ultimate goal is to advance stem cell therapy for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Trenton Lippert
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd. MDC 78, Tampa, FL 33612, USA
| | - Lindsey Gelineau
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd. MDC 78, Tampa, FL 33612, USA
| | - Eleonora Napoli
- Department of Molecular Biosciences, 3011 VM3B 1089 Veterinary Medicine Drive, University of California Davis, Davis, CA 95616, USA..
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd. MDC 78, Tampa, FL 33612, USA.
| |
Collapse
|
27
|
Milczarek O, Jarocha D, Starowicz-Filip A, Kwiatkowski S, Badyra B, Majka M. Multiple Autologous Bone Marrow-Derived CD271 + Mesenchymal Stem Cell Transplantation Overcomes Drug-Resistant Epilepsy in Children. Stem Cells Transl Med 2017; 7:20-33. [PMID: 29224250 PMCID: PMC5746144 DOI: 10.1002/sctm.17-0041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022] Open
Abstract
There is a need among patients suffering from drug‐resistant epilepsy (DRE) for more efficient and less toxic treatments. The objective of the present study was to assess the safety, feasibility, and potential efficacy of autologous bone marrow cell transplantation in pediatric patients with DRE. Two females and two males (11 months to 6 years) were enrolled and underwent a combined therapy consisting of autologous bone marrow nucleated cells (BMNCs) transplantation (intrathecal: 0.5 × 109; intravenous: 0.38 × 109–1.72 × 109) followed by four rounds of intrathecal bone marrow mesenchymal stem cells (BMMSCs) transplantation (18.5 × 106–40 × 106) every 3 months. The BMMSCs used were a unique population derived from CD271‐positive cells. The neurological evaluation included magnetic resonance imaging, electroencephalography (EEG), and cognitive development assessment. The characteristics of BMMSCs were evaluated. Four intravenous and 20 intrathecal transplantations into the cerebrospinal fluid were performed. There were no adverse events, and the therapy was safe and feasible over 2 years of follow‐up. The therapy resulted in neurological and cognitive improvement in all patients, including a reduction in the number of epileptic seizures (from 10 per day to 1 per week) and an absence of status epilepticus episodes (from 4 per week to 0 per week). The number of discharges on the EEG evaluation was decreased, and cognitive improvement was noted with respect to reactions to light and sound, emotions, and motor function. An analysis of the BMMSCs' characteristics revealed the expression of neurotrophic, proangiogenic, and tissue remodeling factors, and the immunomodulatory potential. Our results demonstrate the safety and feasibility of BMNCs and BMMSCs transplantations and the considerable neurological and cognitive improvement in children with DRE. stemcellstranslationalmedicine2018;7:20–33
Collapse
Affiliation(s)
- Olga Milczarek
- Departments of Children Surgery, Jagiellonian University School of Medicine, Cracow, Poland
| | - Danuta Jarocha
- Transplantation, Institute of Pediatrics, Jagiellonian University School of Medicine, Cracow, Poland
| | - Anna Starowicz-Filip
- Department of Medical Psychology, Jagiellonian University School of Medicine, Cracow, Poland
| | - Stanislaw Kwiatkowski
- Departments of Children Surgery, Jagiellonian University School of Medicine, Cracow, Poland
| | - Bogna Badyra
- Transplantation, Institute of Pediatrics, Jagiellonian University School of Medicine, Cracow, Poland
| | - Marcin Majka
- Transplantation, Institute of Pediatrics, Jagiellonian University School of Medicine, Cracow, Poland
| |
Collapse
|
28
|
Maljevic S, Reid CA, Petrou S. Models for discovery of targeted therapy in genetic epileptic encephalopathies. J Neurochem 2017; 143:30-48. [PMID: 28742937 DOI: 10.1111/jnc.14134] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/20/2022]
Abstract
Epileptic encephalopathies are severe disorders emerging in the first days to years of life that commonly include refractory seizures, various types of movement disorders, and different levels of developmental delay. In recent years, many de novo occurring variants have been identified in individuals with these devastating disorders. To unravel disease mechanisms, the functional impact of detected variants associated with epileptic encephalopathies is investigated in a range of cellular and animal models. This review addresses efforts to advance and use such models to identify specific molecular and cellular targets for the development of novel therapies. We focus on ion channels as the best-studied group of epilepsy genes. Given the clinical and genetic heterogeneity of epileptic encephalopathy disorders, experimental models that can reflect this complexity are critical for the development of disease mechanisms-based targeted therapy. The convergence of technological advances in gene sequencing, stem cell biology, genome editing, and high throughput functional screening together with massive unmet clinical needs provides unprecedented opportunities and imperatives for precision medicine in epileptic encephalopathies.
Collapse
Affiliation(s)
- Snezana Maljevic
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Christopher A Reid
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Steven Petrou
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia.,University of Melbourne, Melbourne, Australia
| |
Collapse
|
29
|
Rao G, Mashkouri S, Aum D, Marcet P, Borlongan CV. Contemplating stem cell therapy for epilepsy-induced neuropsychiatric symptoms. Neuropsychiatr Dis Treat 2017; 13:585-596. [PMID: 28260906 PMCID: PMC5328607 DOI: 10.2147/ndt.s114786] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Epilepsy is a debilitating disease that impacts millions of people worldwide. While unprovoked seizures characterize its cardinal symptom, an important aspect of epilepsy that remains to be addressed is the neuropsychiatric component. It has been documented for millennia in paintings and literature that those with epilepsy can suffer from bouts of aggression, depression, and other psychiatric ailments. Current treatments for epilepsy include the use of antiepileptic drugs and surgical resection. Antiepileptic drugs reduce the overall firing of the brain to mitigate the rate of seizure occurrence. Surgery aims to remove a portion of the brain that is suspected to be the source of aberrant firing that leads to seizures. Both options treat the seizure-generating neurological aspect of epilepsy, but fail to directly address the neuropsychiatric components. A promising new treatment for epilepsy is the use of stem cells to treat both the biological and psychiatric components. Stem cell therapy has been shown efficacious in treating experimental models of neurological disorders, including Parkinson's disease, and neuropsychiatric diseases, such as depression. Additional research is necessary to see if stem cells can treat both neurological and neuropsychiatric aspects of epilepsy. Currently, there is no animal model that recapitulates all the clinical hallmarks of epilepsy. This could be due to difficulty in characterizing the neuropsychiatric component of the disease. In advancing stem cell therapy for treating epilepsy, experimental testing of the safety and efficacy of allogeneic and autologous transplantation will require the optimization of cell dosage, delivery, and timing of transplantation in a clinically relevant model of epilepsy with both neurological and neuropsychiatric symptoms of the disease as the primary outcome measures.
Collapse
Affiliation(s)
- Gautam Rao
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sherwin Mashkouri
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - David Aum
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Paul Marcet
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
30
|
Romariz SAA, Paiva DS, Galindo LT, Barnabé GF, Guedes VA, Borlongan CV, Longo BM. Medial Ganglionic Eminence Cells Freshly Obtained or Expanded as Neurospheres Show Distinct Cellular and Molecular Properties in Reducing Epileptic Seizures. CNS Neurosci Ther 2016; 23:127-134. [PMID: 27770487 DOI: 10.1111/cns.12650] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 09/11/2016] [Accepted: 09/13/2016] [Indexed: 01/20/2023] Open
Abstract
AIMS Medial ganglionic eminence (MGE) progenitors give rise to inhibitory interneurons and may serve as an alternative cell source for large-scale cell transplantation for epilepsy after in vitro expansion. We investigated whether modifications in the culture medium of MGE neurospheres affect neuronal differentiation and expression of MGE-specific genes. In vivo, we compared anticonvulsant effects and cell differentiation pattern among neurospheres grown in different culture media and compared them with freshly harvested MGE cells. METHODS We used four variations of cell culture: standard, containing growth factors (EGF/FGF-2) (GF); addition of retinoic acid (GF-RA); withdrawal of EGF/FGF-2 (WD); and addition of retinoic acid and withdrawal of EGF/FGF-2 (WD-RA). Based on in vitro results neurosphere-grown (WD-RA or GF conditions) or fresh MGE cells were transplanted into the hippocampus. RESULTS In vitro WD-RA showed increased neuronal population and higher expression of Dlx1, Nkx2.1, and Lhx6 genes in comparison with GF culture condition. After transplantation, fresh MGE cells and neurospheres (GF) showed anticonvulsant effects. However, fresh MGE cells differentiated preferentially into inhibitory neurons, while GF gave rise to glial cells. CONCLUSION We conclude that freshly isolated and neurosphere-grown MGE cells reduced seizures by different mechanisms (inhibitory interneurons vs. astrocytes). Fresh MGE cells appear more appropriate for cell therapies targeting inhibitory interneurons for conferring anticonvulsant outcomes.
Collapse
Affiliation(s)
- Simone A A Romariz
- Departamento de Fisiologia, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Daisyléa S Paiva
- Departamento de Fisiologia, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Layla T Galindo
- Departamento de Bioquímica, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Gabriela F Barnabé
- Ludwig Institute for Cancer Research at Instituto Sírio-Libanês de Ensino e Pesquisa, São Paulo, SP, Brazil
| | - Vivian A Guedes
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Cesario V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Beatriz M Longo
- Departamento de Fisiologia, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| |
Collapse
|
31
|
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: 2.8] [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
|
32
|
GABA-ergic cell therapy for epilepsy: Advances, limitations and challenges. Neurosci Biobehav Rev 2015; 62:35-47. [PMID: 26748379 DOI: 10.1016/j.neubiorev.2015.12.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 11/06/2015] [Accepted: 12/28/2015] [Indexed: 01/04/2023]
Abstract
Diminution in the number of gamma-amino butyric acid positive (GABA-ergic) interneurons and their axon terminals, and/or alterations in functional inhibition are conspicuous brain alterations believed to contribute to the persistence of seizures in acquired epilepsies such as temporal lobe epilepsy. This has steered a perception that replacement of lost GABA-ergic interneurons would improve inhibitory synaptic neurotransmission in the epileptic brain region and thereby reduce the occurrence of seizures. Indeed, studies using animal prototypes have reported that grafting of GABA-ergic progenitors derived from multiple sources into epileptic regions can reduce seizures. This review deliberates recent advances, limitations and challenges concerning the development of GABA-ergic cell therapy for epilepsy. The efficacy and limitations of grafts of primary GABA-ergic progenitors from the embryonic lateral ganglionic eminence and medial ganglionic eminence (MGE), neural stem/progenitor cells expanded from MGE, and MGE-like progenitors generated from human pluripotent stem cells for alleviating seizures and co-morbidities of epilepsy are conferred. Additional studies required for possible clinical application of GABA-ergic cell therapy for epilepsy are also summarized.
Collapse
|
33
|
Mishra V, Shuai B, Kodali M, Shetty GA, Hattiangady B, Rao X, Shetty AK. Resveratrol Treatment after Status Epilepticus Restrains Neurodegeneration and Abnormal Neurogenesis with Suppression of Oxidative Stress and Inflammation. Sci Rep 2015; 5:17807. [PMID: 26639668 PMCID: PMC4671086 DOI: 10.1038/srep17807] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/06/2015] [Indexed: 12/18/2022] Open
Abstract
Antiepileptic drug therapy, though beneficial for restraining seizures, cannot thwart status epilepticus (SE) induced neurodegeneration or down-stream detrimental changes. We investigated the efficacy of resveratrol (RESV) for preventing SE-induced neurodegeneration, abnormal neurogenesis, oxidative stress and inflammation in the hippocampus. We induced SE in young rats and treated with either vehicle or RESV, commencing an hour after SE induction and continuing every hour for three-hours on SE day and twice daily thereafter for 3 days. Seizures were terminated in both groups two-hours after SE with a diazepam injection. In contrast to the vehicle-treated group, the hippocampus of animals receiving RESV during and after SE presented no loss of glutamatergic neurons in hippocampal cell layers, diminished loss of inhibitory interneurons expressing parvalbumin, somatostatin and neuropeptide Y in the dentate gyrus, reduced aberrant neurogenesis with preservation of reelin + interneurons, lowered concentration of oxidative stress byproduct malondialdehyde and pro-inflammatory cytokine tumor necrosis factor-alpha, normalized expression of oxidative stress responsive genes and diminished numbers of activated microglia. Thus, 4 days of RESV treatment after SE is efficacious for thwarting glutamatergic neuron degeneration, alleviating interneuron loss and abnormal neurogenesis, and suppressing oxidative stress and inflammation. These results have implications for restraining SE-induced chronic temporal lobe epilepsy.
Collapse
Affiliation(s)
- Vikas Mishra
- Institute for Regenerative Medicine, Texas A & M Health Science Center College of Medicine at Scott & White, Temple, Texas, USA
- Research Service, Olin E. Teague Veterans’ Affairs Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USA
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Texas A & M Health Science Center College of Medicine at Scott & White, Temple, Texas, USA
- Research Service, Olin E. Teague Veterans’ Affairs Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USA
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Texas A & M Health Science Center College of Medicine at Scott & White, Temple, Texas, USA
- Research Service, Olin E. Teague Veterans’ Affairs Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USA
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA
| | - Geetha A. Shetty
- Institute for Regenerative Medicine, Texas A & M Health Science Center College of Medicine at Scott & White, Temple, Texas, USA
- Research Service, Olin E. Teague Veterans’ Affairs Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USA
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA
| | - Bharathi Hattiangady
- Institute for Regenerative Medicine, Texas A & M Health Science Center College of Medicine at Scott & White, Temple, Texas, USA
- Research Service, Olin E. Teague Veterans’ Affairs Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USA
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA
| | - Xiaolan Rao
- Institute for Regenerative Medicine, Texas A & M Health Science Center College of Medicine at Scott & White, Temple, Texas, USA
- Research Service, Olin E. Teague Veterans’ Affairs Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USA
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Texas A & M Health Science Center College of Medicine at Scott & White, Temple, Texas, USA
- Research Service, Olin E. Teague Veterans’ Affairs Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USA
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA
| |
Collapse
|
34
|
Oliveira Á, Illes P, Ulrich H. Purinergic receptors in embryonic and adult neurogenesis. Neuropharmacology 2015; 104:272-81. [PMID: 26456352 DOI: 10.1016/j.neuropharm.2015.10.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 10/01/2015] [Accepted: 10/04/2015] [Indexed: 01/14/2023]
Abstract
ATP (adenosine 5'-triphosphate), one of the most ancient neurotransmitters, exerts essential functions in the brain, including neurotransmission and modulation of synaptic activity. Moreover, this nucleotide has been attributed with trophic properties and experimental evidence points to the participation of ATP-activated P2X and P2Y purinergic receptors in embryonic brain development as well as in adult neurogenesis for maintenance of normal brain functions and neuroregeneration upon brain injury. We discuss here the available data on purinergic P2 receptor expression and function during brain development and in the neurogenic zones of the adult brain, as well as the insights based on the use of in vitro stem cell cultures. While several P2 receptor subtypes were shown to be expressed during in vitro and in vivo neurogenesis, specific functions have been proposed for P2Y1, P2Y2 metabotropic as well as P2X2 ionotropic receptors to promote neurogenesis. Further, the P2X7 receptor is suggested to function in the maintenance of pools of neural stem and progenitor cells through induction of proliferation or cell death, depending on the microenvironment. Pathophysiological actions have been proposed for this receptor in worsening damage in brain disease. The P2X7 receptor and possibly additional P2 receptor subtypes have been implicated in pathophysiology of neurological diseases including Parkinson's disease, Alzheimer's disease and epilepsy. New strategies in cell therapy could involve modulation of purinergic signaling, either in the achievement of more effective protocols to obtain viable and homogeneous cell populations or in the process of functional engraftment of transplanted cells into the damaged brain. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Ágatha Oliveira
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-900, Av. Prof. Lineu Prestes, 748, Brazil
| | - Peter Illes
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie der Universität Leipzig, Haertelstrasse 16-18, 04107 Leipzig, Germany.
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-900, Av. Prof. Lineu Prestes, 748, Brazil.
| |
Collapse
|
35
|
Potential of GABA-ergic cell therapy for schizophrenia, neuropathic pain, and Alzheimer's and Parkinson's diseases. Brain Res 2015; 1638:74-87. [PMID: 26423935 DOI: 10.1016/j.brainres.2015.09.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 08/26/2015] [Accepted: 09/08/2015] [Indexed: 12/17/2022]
Abstract
Several neurological and psychiatric disorders present hyperexcitability of neurons in specific regions of the brain or spinal cord, partly because of some loss and/or dysfunction of gamma-amino butyric acid positive (GABA-ergic) inhibitory interneurons. Strategies that enhance inhibitory neurotransmission in the affected brain regions may therefore ease several or most deficits linked to these disorders. This perception has incited a huge interest in testing the efficacy of GABA-ergic interneuron cell grafting into regions of the brain or spinal cord exhibiting hyperexcitability, dearth of GABA-ergic interneurons or impaired inhibitory neurotransmission, using preclinical models of neurological and psychiatric disorders. Interneuron progenitors from the embryonic ventral telencephalon capable of differentiating into diverse subclasses of interneurons have particularly received much consideration because of their ability for dispersion, migration and integration with the host neural circuitry after grafting. The goal of this review is to discuss the premise, scope and advancement of GABA-ergic cell therapy for easing neurological deficits in preclinical models of schizophrenia, chronic neuropathic pain, Alzheimer's disease and Parkinson's disease. As grafting studies in these prototypes have so far utilized either primary cells from the embryonic medial and lateral ganglionic eminences or neural progenitor cells expanded from these eminences as donor material, the proficiency of these cell types is highlighted. Moreover, future studies that are essential prior to considering the possible clinical application of these cells for the above neurological conditions are proposed. Particularly, the need for grafting studies utilizing medial ganglionic eminence-like progenitors generated from human pluripotent stem cells via directed differentiation approaches or somatic cells through direct reprogramming methods are emphasized. This article is part of a Special Issue entitled SI: PSC and the brain.
Collapse
|
36
|
Park GY, Lee EM, Seo MS, Seo YJ, Oh JS, Son WC, Kim KS, Kim JS, Kang JK, Kang KS. Preserved Hippocampal Glucose Metabolism on 18F-FDG PET after Transplantation of Human Umbilical Cord Blood-derived Mesenchymal Stem Cells in Chronic Epileptic Rats. J Korean Med Sci 2015; 30:1232-40. [PMID: 26339161 PMCID: PMC4553668 DOI: 10.3346/jkms.2015.30.9.1232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 05/18/2015] [Indexed: 11/20/2022] Open
Abstract
Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) may be a promising modality for treating medial temporal lobe epilepsy. (18)F-fluorodeoxyglucose positron emission tomography (FDG-PET) is a noninvasive method for monitoring in vivo glucose metabolism. We evaluated the efficacy of hUCB-MSCs transplantation in chronic epileptic rats using FDG-PET. Rats with recurrent seizures were randomly assigned into three groups: the stem cell treatment (SCT) group received hUCB-MSCs transplantation into the right hippocampus, the sham control (ShC) group received same procedure with saline, and the positive control (PC) group consisted of treatment-negative epileptic rats. Normal rats received hUCB-MSCs transplantation acted as the negative control (NC). FDG-PET was performed at pre-treatment baseline and 1- and 8-week posttreatment. Hippocampal volume was evaluated and histological examination was done. In the SCT group, bilateral hippocampi at 8-week after transplantation showed significantly higher glucose metabolism (0.990 ± 0.032) than the ShC (0.873 ± 0.087; P < 0.001) and PC groups (0.858 ± 0.093; P < 0.001). Histological examination resulted that the transplanted hUCB-MSCs survived in the ipsilateral hippocampus and migrated to the contralateral hippocampus but did not differentiate. In spite of successful engraftment, seizure frequency among the groups was not significantly different. Transplanted hUCB-MSCs can engraft and migrate, thereby partially restoring bilateral hippocampal glucose metabolism. The results suggest encouraging effect of hUCB-MSCs on restoring epileptic networks.
Collapse
Affiliation(s)
- Ga Young Park
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- The Asan Institute for Life Science, Seoul, Korea
| | - Eun Mi Lee
- Department of Neurology, Ulsan University Hospital, Ulsan, Korea
| | - Min-Soo Seo
- Adult Stem Cell Research, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Yoo-Jin Seo
- Adult Stem Cell Research, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Jungsu S. Oh
- Department of Nuclear Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Woo-Chan Son
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ki Soo Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jae Seung Kim
- Department of Nuclear Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Joong Koo Kang
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
37
|
Venugopal C, Chandanala S, Prasad HC, Nayeem D, Bhonde RR, Dhanushkodi A. Regenerative therapy for hippocampal degenerative diseases: lessons from preclinical studies. J Tissue Eng Regen Med 2015; 11:321-333. [PMID: 26118731 DOI: 10.1002/term.2052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 04/08/2015] [Accepted: 04/29/2015] [Indexed: 12/30/2022]
Abstract
Increase in life expectancy has put neurodegenerative diseases on the rise. Amongst these, degenerative diseases involving hippocampus like Alzheimer's disease (AD) and temporal lobe epilepsy (TLE) are ranked higher as it is vulnerable to excitotoxicity induced neuronal dysfunction and death resulting in cognitive impairment. Modern medicines have not succeeded in halting the progression of these diseases rendering them incurable and often fatal. Under such scenario, regenerative studies employing stem cells or their by-products in animal models of AD and TLE have yielded encourageing results. This review focuses on the distinct cell types, such as hippocampal cell lines, neural precursor cells, embryonic stem cells derived neural precursor cells, induced pluripotent stem cells, induced neurons and mesenchymal stem cells, which can be employed to rescue hippocampal functions in neurodegenerative diseases like AD and TLE. Besides, the divergent mechanisms through which cell based therapy confer neuroprotection, current impediments and possible improvements in stem cell transplantation strategies are discussed. Authors are aware of the voluminous literature available on this issue and have made a sincere attempt to put forth the current status of research in the field of cell based therapy concurrently discussing the promise it holds for combating neurodegenerative diseases like AD and TLE in the near future. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Chaitra Venugopal
- School of Regenerative Medicine, Manipal University, Bangalore, India
| | | | | | - Danish Nayeem
- School of Regenerative Medicine, Manipal University, Bangalore, India
| | - Ramesh R Bhonde
- School of Regenerative Medicine, Manipal University, Bangalore, India
| | | |
Collapse
|
38
|
Agadi S, Shetty AK. Concise Review: Prospects of Bone Marrow Mononuclear Cells and Mesenchymal Stem Cells for Treating Status Epilepticus and Chronic Epilepsy. Stem Cells 2015; 33:2093-103. [PMID: 25851047 DOI: 10.1002/stem.2029] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 03/16/2015] [Indexed: 12/22/2022]
Abstract
Mononuclear cells (MNCs) and mesenchymal stem cells (MSCs) derived from the bone marrow and other sources have received significant attention as donor cells for treating various neurological disorders due to their robust neuroprotective and anti-inflammatory effects. Moreover, it is relatively easy to procure these cells from both autogenic and allogenic sources. Currently, there is considerable interest in examining the usefulness of these cells for conditions such as status epilepticus (SE) and chronic epilepsy. A prolonged seizure activity in SE triggers neurodegeneration in the limbic brain areas, which elicits epileptogenesis and evolves into a chronic epileptic state. Because of their potential for providing neuroprotection, diminishing inflammation and curbing epileptogenesis, early intervention with MNCs or MSCs appears attractive for treating SE as such effects may restrain the development of chronic epilepsy typified by spontaneous seizures and learning and memory impairments. Delayed administration of these cells after SE may also be useful for easing spontaneous seizures and cognitive dysfunction in chronic epilepsy. This concise review evaluates the current knowledge and outlook pertaining to MNC and MSC therapies for SE and chronic epilepsy. In the first section, the behavior of these cells in animal models of SE and their efficacy to restrain neurodegeneration, inflammation, and epileptogenesis are discussed. The competence of these cells for suppressing seizures and improving cognitive function in chronic epilepsy are conferred in the next section. The final segment ponders issues that need to be addressed to pave the way for clinical application of these cells for SE and chronic epilepsy.
Collapse
Affiliation(s)
- Satish Agadi
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas, USA.,Department of Pediatrics, McLane's Children's Hospital, Baylor Scott & White Health, Temple, Texas, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas, USA.,Research Service, Olin E. Teague Veterans Affairs Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USA.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA
| |
Collapse
|
39
|
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: 1.0] [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
|
40
|
Leung A, Ahn S, Savvidis G, Kim Y, Iskandar D, Luna MJ, Kim KS, Cunningham M, Chung S. Optimization of pilocarpine-mediated seizure induction in immunodeficient NodScid mice. Epilepsy Res 2014; 109:114-8. [PMID: 25524850 DOI: 10.1016/j.eplepsyres.2014.10.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/07/2014] [Accepted: 10/28/2014] [Indexed: 11/25/2022]
Abstract
Temporal lobe epilepsy (TLE) has been modeled in mice using pilocarpine induction, with variable results depending on specific strains. To allow efficient xenotransplantation for the purpose of optimizing potential cell-based therapy of human TLE, we have determined the optimal dosing strategy to produce spontaneous recurring seizures in immunodeficient NodScid mice. Multiple 100mg/kg injections of pilocarpine have been shown to be more effective than single 300-400mg/kg injections for inducing spontaneous seizures in NodScid mice. Under our optimal conditions, 88.1 ± 2.9% of the mice experienced status epilepticus (SE) with a survival rate of 61.8 ± 5.9%. Surviving SE mice displayed spontaneous recurrent seizures at a frequency of 2.8 ± 0.9 seizures/day for a duration of 41.1 ± 3.5s. The widely used method of a single injection of pilocarpine was significantly less efficient in inducing seizures in NodScid mice. Therefore, we have determined that a multiple injection "ramping up" of 100mg/kg of pilocarpine is optimal for inducing TLE-like spontaneous seizures in NodScid mice. Using this method, mice with SE efficiently developed SRS and expressed mossy fiber sprouting, a signature histopathological feature of TLE.
Collapse
Affiliation(s)
- Amanda Leung
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Sandra Ahn
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - George Savvidis
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Yeachan Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Danielle Iskandar
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Maria Jose Luna
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Kwang-Soo Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Miles Cunningham
- Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Sangmi Chung
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
41
|
Cunningham M, Cho JH, Leung A, Savvidis G, Ahn S, Moon M, Lee PKJ, Han JJ, Azimi N, Kim KS, Bolshakov VY, Chung S. hPSC-derived maturing GABAergic interneurons ameliorate seizures and abnormal behavior in epileptic mice. Cell Stem Cell 2014; 15:559-73. [PMID: 25517465 DOI: 10.1016/j.stem.2014.10.006] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 08/25/2014] [Accepted: 10/16/2014] [Indexed: 01/04/2023]
Abstract
Seizure disorders debilitate more than 65,000,000 people worldwide, with temporal lobe epilepsy (TLE) being the most common form. Previous studies have shown that transplantation of GABA-releasing cells results in suppression of seizures in epileptic mice. Derivation of interneurons from human pluripotent stem cells (hPSCs) has been reported, pointing to clinical translation of quality-controlled human cell sources that can enhance inhibitory drive and restore host circuitry. In this study, we demonstrate that hPSC-derived maturing GABAergic interneurons (mGINs) migrate extensively and integrate into dysfunctional circuitry of the epileptic mouse brain. Using optogenetic approaches, we find that grafted mGINs generate inhibitory postsynaptic responses in host hippocampal neurons. Importantly, even before acquiring full electrophysiological maturation, grafted neurons were capable of suppressing seizures and ameliorating behavioral abnormalities such as cognitive deficits, aggressiveness, and hyperactivity. These results provide support for the potential of hPSC-derived mGIN for restorative cell therapy for epilepsy.
Collapse
Affiliation(s)
- Miles Cunningham
- Laboratory for Neural Reconstruction, Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Jun-Hyeong Cho
- Cellular Neurobiology Laboratory, Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Amanda Leung
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - George Savvidis
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Sandra Ahn
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Minho Moon
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Paula K J Lee
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Jason J Han
- MRC Electron and Light Microscopy Core Facility, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Nima Azimi
- Laboratory for Neural Reconstruction, Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Kwang-Soo Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Vadim Y Bolshakov
- Cellular Neurobiology Laboratory, Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Sangmi Chung
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA.
| |
Collapse
|
42
|
Amini E, Rezaei M, Mohamed Ibrahim N, Golpich M, Ghasemi R, Mohamed Z, Raymond AA, Dargahi L, Ahmadiani A. A Molecular Approach to Epilepsy Management: from Current Therapeutic Methods to Preconditioning Efforts. Mol Neurobiol 2014; 52:492-513. [PMID: 25195699 DOI: 10.1007/s12035-014-8876-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 08/25/2014] [Indexed: 01/16/2023]
Abstract
Epilepsy is the most common and chronic neurological disorder characterized by recurrent unprovoked seizures. The key aim in treating patients with epilepsy is the suppression of seizures. An understanding of focal changes that are involved in epileptogenesis may therefore provide novel approaches for optimal treatment of the seizure. Although the actual pathogenesis of epilepsy is still uncertain, recently growing lines of evidence declare that microglia and astrocyte activation, oxidative stress and reactive oxygen species (ROS) production, mitochondria dysfunction, and damage of blood-brain barrier (BBB) are involved in its pathogenesis. Impaired GABAergic function in the brain is probably the most accepted hypothesis regarding the pathogenesis of epilepsy. Clinical neuroimaging of patients and experimental modeling have demonstrated that seizures may induce neuronal apoptosis. Apoptosis signaling pathways are involved in the pathogenesis of several types of epilepsy such as temporal lobe epilepsy (TLE). The quality of life of patients is seriously affected by treatment-related problems and also by unpredictability of epileptic seizures. Moreover, the available antiepileptic drugs (AED) are not significantly effective to prevent epileptogenesis. Thus, novel therapies that are proficient to control seizure in people who are suffering from epilepsy are needed. The preconditioning method promises to serve as an alternative therapeutic approach because this strategy has demonstrated the capability to curtail epileptogenesis. For this reason, understanding of molecular mechanisms underlying brain tolerance induced by preconditioning is crucial to delineate new neuroprotective ways against seizure damage and epileptogenesis. In this review, we summarize the work to date on the pathogenesis of epilepsy and discuss recent therapeutic strategies in the treatment of epilepsy. We will highlight that novel therapy targeting such as preconditioning process holds great promise. In addition, we will also highlight the role of gene reprogramming and mitochondrial biogenesis in the preconditioning-mediated neuroprotective events.
Collapse
Affiliation(s)
- Elham Amini
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Human fetal brain-derived neural stem/progenitor cells grafted into the adult epileptic brain restrain seizures in rat models of temporal lobe epilepsy. PLoS One 2014; 9:e104092. [PMID: 25105891 PMCID: PMC4126719 DOI: 10.1371/journal.pone.0104092] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 07/10/2014] [Indexed: 01/10/2023] Open
Abstract
Cell transplantation has been suggested as an alternative therapy for temporal lobe epilepsy (TLE) because this can suppress spontaneous recurrent seizures in animal models. To evaluate the therapeutic potential of human neural stem/progenitor cells (huNSPCs) for treating TLE, we transplanted huNSPCs, derived from an aborted fetal telencephalon at 13 weeks of gestation and expanded in culture as neurospheres over a long time period, into the epileptic hippocampus of fully kindled and pilocarpine-treated adult rats exhibiting TLE. In vitro, huNSPCs not only produced all three central nervous system neural cell types, but also differentiated into ganglionic eminences-derived γ-aminobutyric acid (GABA)-ergic interneurons and released GABA in response to the depolarization induced by a high K+ medium. NSPC grafting reduced behavioral seizure duration, afterdischarge duration on electroencephalograms, and seizure stage in the kindling model, as well as the frequency and the duration of spontaneous recurrent motor seizures in pilocarpine-induced animals. However, NSPC grafting neither improved spatial learning or memory function in pilocarpine-treated animals. Following transplantation, grafted cells showed extensive migration around the injection site, robust engraftment, and long-term survival, along with differentiation into β-tubulin III+ neurons (∼34%), APC-CC1+ oligodendrocytes (∼28%), and GFAP+ astrocytes (∼8%). Furthermore, among donor-derived cells, ∼24% produced GABA. Additionally, to explain the effect of seizure suppression after NSPC grafting, we examined the anticonvulsant glial cell-derived neurotrophic factor (GDNF) levels in host hippocampal astrocytes and mossy fiber sprouting into the supragranular layer of the dentate gyrus in the epileptic brain. Grafted cells restored the expression of GDNF in host astrocytes but did not reverse the mossy fiber sprouting, eliminating the latter as potential mechanism. These results suggest that human fetal brain-derived NSPCs possess some therapeutic effect for TLE treatments although further studies to both increase the yield of NSPC grafts-derived functionally integrated GABAergic neurons and improve cognitive deficits are still needed.
Collapse
|
44
|
Abdanipour A, Tiraihi T, Mirnajafi-Zadeh J. Improvement of the pilocarpine epilepsy model in rat using bone marrow stromal cell therapy. Neurol Res 2013; 33:625-32. [DOI: 10.1179/1743132810y.0000000018] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
45
|
Pluchino S, Cossetti C. How stem cells speak with host immune cells in inflammatory brain diseases. Glia 2013; 61:1379-401. [PMID: 23633288 DOI: 10.1002/glia.22500] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/01/2013] [Indexed: 12/14/2022]
Abstract
Advances in stem cell biology have raised great expectations that diseases and injuries of the central nervous system (CNS) may be ameliorated by the development of non-hematopoietic stem cell medicines. Yet, the application of adult stem cells as CNS therapeutics is challenging and the interpretation of some of the outcomes ambiguous. In fact, the initial idea that stem cell transplants work only via structural cell replacement has been challenged by the observation of consistent cellular signaling between the graft and the host. Cellular signaling is the foundation of coordinated actions and flexible responses, and arises via networks of exchanging and interacting molecules that transmit patterns of information between cells. Sustained stem cell graft-to-host communication leads to remarkable trophic effects on endogenous brain cells and beneficial modulatory actions on innate and adaptive immune responses in vivo, ultimately promoting the healing of the injured CNS. Among a number of adult stem cell types, mesenchymal stem cells (MSCs) and neural stem/precursor cells (NPCs) are being extensively investigated for their ability to signal to the immune system upon transplantation in experimental CNS diseases. Here, we focus on the main cellular signaling pathways that grafted MSCs and NPCs use to establish a therapeutically relevant cross talk with host immune cells, while examining the role of inflammation in regulating some of the bidirectionality of these communications. We propose that the identification of the players involved in stem cell signaling might contribute to the development of innovative, high clinical impact therapeutics for inflammatory CNS diseases.
Collapse
Affiliation(s)
- Stefano Pluchino
- Department of Clinical Neurosciences, John van Geest Cambridge Centre for Brain Repair and Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, United Kingdom.
| | | |
Collapse
|
46
|
Miltiadous P, Kouroupi G, Stamatakis A, Koutsoudaki PN, Matsas R, Stylianopoulou F. Subventricular zone-derived neural stem cell grafts protect against hippocampal degeneration and restore cognitive function in the mouse following intrahippocampal kainic acid administration. Stem Cells Transl Med 2013; 2:185-98. [PMID: 23417642 DOI: 10.5966/sctm.2012-0074] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Temporal lobe epilepsy (TLE) is a major neurological disease, often associated with cognitive decline. Since approximately 30% of patients are resistant to antiepileptic drugs, TLE is being considered as a possible clinical target for alternative stem cell-based therapies. Given that insulin-like growth factor I (IGF-I) is neuroprotective following a number of experimental insults to the nervous system, we investigated the therapeutic potential of neural stem/precursor cells (NSCs) transduced, or not, with a lentiviral vector for overexpression of IGF-I after transplantation in a mouse model of kainic acid (KA)-induced hippocampal degeneration, which represents an animal model of TLE. Exposure of mice to the Morris water maze task revealed that unilateral intrahippocampal NSC transplantation significantly prevented the KA-induced cognitive decline. Moreover, NSC grafting protected against neurodegeneration at the cellular level, reduced astrogliosis, and maintained endogenous granule cell proliferation at normal levels. In some cases, as in the reduction of hippocampal cell loss and the reversal of the characteristic KA-induced granule cell dispersal, the beneficial effects of transplanted NSCs were manifested earlier and were more pronounced when these were transduced to express IGF-I. However, differences became less pronounced by 2 months postgrafting, since similar amounts of IGF-I were detected in the hippocampi of both groups of mice that received cell transplants. Grafted NSCs survived, migrated, and differentiated into neurons-including glutamatergic cells-and not glia, in the host hippocampus. Our results demonstrate that transplantation of IGF-I producing NSCs is neuroprotective and restores cognitive function following KA-induced hippocampal degeneration.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Behavior, Animal
- Cell Movement
- Cell Proliferation
- Cell Survival
- Cognition
- Disease Models, Animal
- Epilepsy, Temporal Lobe/chemically induced
- Epilepsy, Temporal Lobe/genetics
- Epilepsy, Temporal Lobe/metabolism
- Epilepsy, Temporal Lobe/pathology
- Epilepsy, Temporal Lobe/physiopathology
- Epilepsy, Temporal Lobe/psychology
- Epilepsy, Temporal Lobe/therapy
- Genetic Therapy/methods
- Genetic Vectors
- Glutamic Acid/metabolism
- Green Fluorescent Proteins/biosynthesis
- Green Fluorescent Proteins/genetics
- Hippocampus/metabolism
- Hippocampus/pathology
- Hippocampus/physiopathology
- Hippocampus/surgery
- Insulin-Like Growth Factor I/biosynthesis
- Insulin-Like Growth Factor I/genetics
- Kainic Acid
- Lentivirus/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Nerve Degeneration
- Neural Stem Cells/metabolism
- Neural Stem Cells/transplantation
- Neurogenesis
- Neurons/metabolism
- Neurons/pathology
- Spheroids, Cellular
- Time Factors
- Transduction, Genetic
Collapse
|
47
|
Innovative treatments for epilepsy: radiosurgery and local delivery. HANDBOOK OF CLINICAL NEUROLOGY 2012. [PMID: 22939079 DOI: 10.1016/b978-0-444-52899-5.00041-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
48
|
Roper SN, Steindler DA. Stem cells as a potential therapy for epilepsy. Exp Neurol 2012; 244:59-66. [PMID: 22265818 DOI: 10.1016/j.expneurol.2012.01.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 12/29/2011] [Accepted: 01/04/2012] [Indexed: 12/16/2022]
Abstract
Neural stem cells and neural progenitors (NSC/NPs) hold great promise in neuro-restorative therapy due to their remarkable capacity for self-renewal, plasticity, and ability to integrate into host brain circuitry. Some types of epilepsy would appear to be excellent targets for this type of therapy due to known alterations in local circuitry based on loss or malfunction of specific types of neurons in specific brain structures. Potential sources for NSC/NPs include the embryonic blastocyst, the fetal brain, and adult brain and non-neural tissues. Each of these cell types has potential strengths and weaknesses as candidates for clinical therapeutic agents. This article reviews some of the major types of NSC/NPs and how they have been studied with regard to synaptic integration into host brain circuits. It also reviews how these transplanted cells develop and interact with host brain cells in animal models of epilepsy. The field is still wide open with a number of very promising results but there are also some major challenges that will need to be addressed prior to considering clinical applications for epilepsy.
Collapse
Affiliation(s)
- Steven N Roper
- Department of Neurosurgery and the McKnight Brain Institute, University of Florida, USA.
| | | |
Collapse
|
49
|
Hattiangady B, Shetty AK. Neural stem cell grafting in an animal model of chronic temporal lobe epilepsy. ACTA ACUST UNITED AC 2012; Chapter 2:Unit2D.7. [PMID: 21913169 DOI: 10.1002/9780470151808.sc02d07s18] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neural stem cell (NSC) transplantation into the hippocampus could offer an alternative therapy to hippocampal resection in patients with drug-resistant chronic epilepsy, which afflicts ∼30% of mesial 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 induced pluripotent stem cells (iPSCs). However, to provide a comprehensive methodology involved in testing the efficacy of transplantation of NSCs in a rat model of chronic TLE, NSCs derived from the embryonic medial ganglionic eminence (MGE) are taken as an example in this unit. The topics comprise description of the required materials, reagents and equipment, and protocols for expanding MGE-NSCs in culture, generating chronically epileptic rats, the intrahippocampal grafting, post-grafting evaluation of the effects of NSC grafts on spontaneous recurrent seizures and cognitive impairments, analyses of the yield and the fate of graft-derived cells, and the effects of NSC grafts on the host hippocampus.
Collapse
Affiliation(s)
- Bharathi Hattiangady
- Institute for Regenerative Medicine, Texas A&M Health Science Center Temple, TX, USA
| | | |
Collapse
|
50
|
Sebe JY, Baraban SC. The promise of an interneuron-based cell therapy for epilepsy. Dev Neurobiol 2011; 71:107-17. [PMID: 21154914 DOI: 10.1002/dneu.20813] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Of the nearly 3 million Americans diagnosed with epilepsy, approximately 30% are unresponsive to current medications. Recent data has shown that early postnatal transplantation of interneuronal precursor cells increases GABAergic inhibition in the host brain and dramatically suppresses seizure activity in epileptic mice. In this review, we will highlight findings from seizure-prone mice and humans that demonstrate the link between dysfunctional GABAergic inhibition and hyperexcitability. In particular, we will focus on rodent models of temporal lobe epilepsy, the most common and difficult to treat form of the disease, and interneuronopathies, an emerging classification. A wealth of literature showing a causal link between reduced GABA-mediated inhibition and seizures has directed our efforts to recover the loss of inhibition via transplantation of interneuronal precursors. Numerous related studies have explored the anticonvulsant potential of cell grafts derived from a variety of brain regions, yet the mechanism underlying the effect of such heterogeneous cell transplants is unknown. In discussing our recent findings and placing them in context with what is known about epilepsy, and how related transplant approaches have progressed, we hope to initiate a frank discussion of the best path toward the translation of this approach to patients with intractable forms of epilepsy.
Collapse
Affiliation(s)
- Joy Y Sebe
- Epilepsy Research Laboratory, Department of Neurological Surgery, University of California, San Francisco, California, USA.
| | | |
Collapse
|