Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Epilepsia Open ; 8(2): 320-333, 2023 06.
Article in English | MEDLINE | ID: mdl-36625631

ABSTRACT

OBJECTIVE: STXBP1-related disorders are rare genetic epilepsies and neurodevelopmental disorders, but the impact of symptoms across clinical domains is poorly understood. Disease concept models are formal frameworks to assess the lived experience of individuals and their families and provide a basis for generating outcome measures. METHODS: We conducted semistructured, qualitative interviews with 19 caregivers of 16 individuals with STXBP1-related disorders and 7 healthcare professionals. We systematically coded themes using NVivo software and grouped concepts into the domains of symptoms, symptom impact, and caregiver impact. We quantified the frequency of concepts throughout the lifespan and across clinical subgroups stratified by seizure history and developmental trajectories. RESULTS: Over 25 hours of interviews, we coded a total of 3626 references to 38 distinct concepts. In addition to well-recognized clinical features such as developmental delay (n = 240 references), behavior (n = 201), and seizures (n = 147), we identified previously underrepresented symptoms including gastrointestinal (n = 68) and respiratory symptoms (n = 24) and pain (n = 30). The most frequently referenced symptom impacts were autonomy (n = 96), socialization (n = 64), and schooling (n = 61). Emotional impact (n = 354), support (n = 200), and daily life & activities (n = 108) were highly cited caregiver impacts. We found that seizures were more commonly referenced in infancy than in other age groups, while behavior and socialization were more likely to be referred to in childhood. We found that caregivers of individuals with ongoing seizures were less likely to reference developmental delay, possibly due to the relatively high impact of seizures. SIGNIFICANCE: STXBP1-related disorders are complex conditions affecting a wide range of clinical and social domains. We comprehensively mapped symptoms and their impact on families to generate a comprehensive disease model as a foundation for clinical endpoints in future trials.


Subject(s)
Epilepsy , Neurodevelopmental Disorders , Humans , Epilepsy/genetics , Seizures/genetics , Neurodevelopmental Disorders/genetics , Caregivers , Socialization , Munc18 Proteins/genetics
2.
Curr HIV/AIDS Rep ; 19(6): 580-591, 2022 12.
Article in English | MEDLINE | ID: mdl-36181625

ABSTRACT

PURPOSE OF REVIEW: Progressive multifocal leukoencephalopathy (PML) is a severe opportunistic infection that remains an important cause of morbidity and mortality in people living with HIV (PLWH). Immune checkpoint molecules are negative regulators of the immune response that have been targeted as a strategy to bolster anti-viral immunity in PML, with varied outcomes reported. While initiation and optimization of antiretroviral therapy remains the standard of care in HIV-related PML, the specific opportunities and risks for checkpoint blockade in these cases should be explored. RECENT FINDINGS: As of April 15, 2022, only 5 of the 53 total published cases of PML treated with checkpoint blockade had underlying HIV infection; four of these had a favorable outcome. The risk of promoting immune reconstitution inflammatory syndrome is a major concern and underscores the importance of patient selection and monitoring. Checkpoint blockade warrants further exploration as a potentially promising option for treatment escalation in HIV-related PML.


Subject(s)
HIV Infections , Immune Reconstitution Inflammatory Syndrome , JC Virus , Leukoencephalopathy, Progressive Multifocal , Humans , Leukoencephalopathy, Progressive Multifocal/drug therapy , Leukoencephalopathy, Progressive Multifocal/etiology , HIV Infections/complications , HIV Infections/drug therapy , Immune Reconstitution Inflammatory Syndrome/drug therapy , Immune Reconstitution Inflammatory Syndrome/complications , Antiviral Agents/therapeutic use
4.
Viruses ; 14(6)2022 06 08.
Article in English | MEDLINE | ID: mdl-35746716

ABSTRACT

Background: Lytic infection of oligodendrocytes by the human JC polyomavirus (JCPyV) results in the demyelinating disease called progressive multifocal leukoencephalopathy (PML). The detection of viral DNA in the cerebrospinal fluid (CSF) by PCR is an important diagnostic tool and, in conjunction with defined radiological and clinical features, can provide diagnosis of definite PML, avoiding the need for brain biopsy. The main aim of this study is to compare the droplet digital PCR (ddPCR) assay with the gold standard quantitative PCR (qPCR) for the quantification of JC viral loads in clinical samples. Methods: A total of 62 CSF samples from 31 patients with PML were analyzed to compare the qPCR gold standard technique with ddPCR to detect conserved viral DNA sequences in the JCPyV genome. As part of the validation process, ddPCR results were compared to qPCR data obtained in 42 different laboratories around the world. In addition, the characterization of a novel triplex ddPCR to detect viral DNA sequence from both prototype and archetype variants and a cellular housekeeping reference gene is described. Triplex ddPCR was used to analyze the serum from six PML patients and from three additional cohorts, including 20 healthy controls (HC), 20 patients with multiple sclerosis (MS) who had never been treated with natalizumab (no-NTZ-treated), and 14 patients with MS who were being treated with natalizumab (NTZ-treated); three from this last group seroconverted during the course of treatment with natalizumab. Results: JCPyV DNA was detected only by ddPCR for 5 of the 62 CSF samples (8%), while remaining undetected by qPCR. For nine CSF samples (15%), JCPyV DNA was at the lower limit of quantification for qPCR, set at <250 copies/mL, and therefore no relative quantitation could be determined. By contrast, exact copies of JCPyV for each of these samples were quantified by ddPCR. No differences were observed between qPCR and ddPCR when five standardized plasma samples were analyzed for JCPyV in 42 laboratories in the United States and Europe. JCPyV-DNA was undetected in all the sera from HC and MS cohorts tested by triplex ddPCR, while serum samples from six patients with PML tested positive for JCPyV. Conclusion: This study shows strong correlation between ddPCR and qPCR with increased sensitivity of the ddPCR assay. Further work will be needed to determine whether multiplex ddPCR can be useful to determine PML risk in natalizumab-treated MS patients.


Subject(s)
JC Virus , Leukoencephalopathy, Progressive Multifocal , Multiple Sclerosis , DNA, Viral/genetics , Humans , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/diagnosis , Leukoencephalopathy, Progressive Multifocal/drug therapy , Multiple Sclerosis/drug therapy , Natalizumab/therapeutic use , Real-Time Polymerase Chain Reaction , Viral Load
5.
Neurosci Biobehav Rev ; 122: 38-65, 2021 03.
Article in English | MEDLINE | ID: mdl-33359391

ABSTRACT

Hormone therapy, primarily progesterone and progestins, for central nervous system (CNS) disorders represents an emerging field of regenerative medicine. Following a failed clinical trial of progesterone for traumatic brain injury treatment, attention has shifted to the progestin Nestorone for its ability to potently and selectively transactivate progesterone receptors at relatively low doses, resulting in robust neurogenetic, remyelinating, and anti-inflammatory effects. That CNS disorders, including multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), spinal cord injury (SCI), and stroke, develop via demyelinating, cell death, and/or inflammatory pathological pathways advances Nestorone as an auspicious candidate for these disorders. Here, we assess the scientific and clinical progress over decades of research into progesterone, progestins, and Nestorone as neuroprotective agents in MS, ALS, SCI, and stroke. We also offer recommendations for optimizing timing, dosage, and route of the drug regimen, and identifying candidate patient populations, in advancing Nestorone to the clinic.


Subject(s)
Nervous System Diseases , Neuroprotective Agents , Progestins , Humans , Nervous System Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Progesterone , Progestins/therapeutic use , Receptors, Progesterone , Spinal Cord Injuries
6.
CNS Neurosci Ther ; 26(6): 603-615, 2020 06.
Article in English | MEDLINE | ID: mdl-32356605

ABSTRACT

Ischemic stroke and traumatic brain injury (TBI) comprise two particularly prevalent and costly examples of acquired brain injury (ABI). Following stroke or TBI, primary cell death and secondary cell death closely model disease progression and worsen outcomes. Mounting evidence indicates that long-term neuroinflammation extensively exacerbates the secondary deterioration of brain structure and function. Due to their immunomodulatory and regenerative properties, mesenchymal stem cell transplants have emerged as a promising approach to treating this facet of stroke and TBI pathology. In this review, we summarize the classification of cell death in ABI and discuss the prominent role of inflammation. We then consider the efficacy of bone marrow-derived mesenchymal stem/stromal cell (BM-MSC) transplantation as a therapy for these injuries. Finally, we examine recent laboratory and clinical studies utilizing transplanted BM-MSCs as antiinflammatory and neurorestorative treatments for stroke and TBI. Clinical trials of BM-MSC transplants for stroke and TBI support their promising protective and regenerative properties. Future research is needed to allow for better comparison among trials and to elaborate on the emerging area of cell-based combination treatments.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Mesenchymal Stem Cell Transplantation/methods , Stroke/metabolism , Stroke/therapy , Animals , Brain Injuries, Traumatic/pathology , Cell Death/physiology , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Mesenchymal Stem Cell Transplantation/trends , Stroke/pathology
7.
Brain Hemorrhages ; 1(1): 24-33, 2020 Mar.
Article in English | MEDLINE | ID: mdl-34056567

ABSTRACT

Hemorrhagic stroke is a global health crisis plagued by neuroinflammation in the acute and chronic phases. Neuroinflammation approximates secondary cell death, which in turn robustly contributes to stroke pathology. Both the physiological and behavioral symptoms of stroke correlate with various inflammatory responses in animal and human studies. That slowing the secondary cell death mediated by this inflammation may attenuate stroke pathology presents a novel treatment strategy. To this end, experimental therapies employing stem cell transplants support their potential for neuroprotection and neuroregeneration after hemorrhagic stroke. In this review, we evaluate experiments using different types of stem cell transplants as treatments for stroke-induced neuroinflammation. We also update this emerging area by examining recent preclinical and clinical trials that have deployed these therapies. While further investigations are warranted to solidify their therapeutic profile, the reviewed studies largely posit stem cells as safe and potent biologics for stroke, specifically owing to their mode of action for sequestering neuroinflammation and promoting neuroregenerative processes.

8.
J Cereb Blood Flow Metab ; 40(6): 1182-1192, 2020 06.
Article in English | MEDLINE | ID: mdl-31366299

ABSTRACT

Rodents display "empathy" defined as perceived physical pain or psychological stress by cagemates when co-experiencing socially distinct traumatic events. The present study tested the hypothesis that empathy occurs in adult rats subjected to an experimental neurological disorder, by allowing co-experience of stroke with cagemates. Psychological stress was measured by general locomotor activity, Rat Grimace Scale (RGS), and plasma corticosterone. Physiological correlates were measured by Western blot analysis of advanced glycation endproducts (AGE)-related proteins in the thymus. General locomotor activity was impaired in stroke animals and in non-stroke rats housed with stroke rats suggesting transfer of behavioral manifestation of psychological stress from an injured animal to a non-injured animal leading to social inhibition. RGS was higher in stroke rats regardless of social settings. Plasma corticosterone levels at day 3 after stroke were significantly higher in stroke animals housed with stroke rats, but not with non-stroke rats, indicating that empathy upregulated physiological stress level. The expression of five proteins related to AGE in the thymus reflected the observed pattern of general locomotor activity, RGS, and plasma corticosterone levels. These results indicate that stroke-induced psychological stress manifested on both the behavioral and physiological levels and appeared to be affected by empathy-associated social settings.


Subject(s)
Empathy , Infarction, Middle Cerebral Artery/psychology , Rats/psychology , Social Environment , Animals , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Male , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products/metabolism , Stress, Psychological/etiology , Stress, Psychological/metabolism , Thymus Gland/metabolism
9.
Stem Cells Transl Med ; 9(2): 203-220, 2020 02.
Article in English | MEDLINE | ID: mdl-31738023

ABSTRACT

The present study used in vitro and in vivo stroke models to demonstrate the safety, efficacy, and mechanism of action of adult human bone marrow-derived NCS-01 cells. Coculture with NCS-01 cells protected primary rat cortical cells or human neural progenitor cells from oxygen glucose deprivation. Adult rats that were subjected to middle cerebral artery occlusion, transiently or permanently, and subsequently received intracarotid artery or intravenous transplants of NCS-01 cells displayed dose-dependent improvements in motor and neurological behaviors, and reductions in infarct area and peri-infarct cell loss, much better than intravenous administration. The optimal dose was 7.5 × 106 cells/mL when delivered via the intracarotid artery within 3 days poststroke, although therapeutic effects persisted even when administered at 1 week after stroke. Compared with other mesenchymal stem cells, NCS-01 cells ameliorated both the structural and functional deficits after stroke through a broad therapeutic window. NCS-01 cells secreted therapeutic molecules, such as basic fibroblast growth factor and interleukin-6, but equally importantly we observed for the first time the formation of filopodia by NCS-01 cells under stroke conditions, characterized by cadherin-positive processes extending from the stem cells toward the ischemic cells. Collectively, the present efficacy readouts and the novel filopodia-mediated mechanism of action provide solid lab-to-clinic evidence supporting the use of NCS-01 cells for treatment of stroke in the clinical setting.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Ischemic Stroke/therapy , Stem Cell Transplantation/methods , Animals , Bone Marrow , Humans , Ischemic Stroke/pathology , Male , Rats
10.
Brain Circ ; 5(3): 97-100, 2019.
Article in English | MEDLINE | ID: mdl-31620654

ABSTRACT

This special issue of Brain Circulation presents cutting-edge research discoveries in stem cell-based regenerative medicine. Each article highlights recent advances in the fields of neurodegeneration and regenerative medicine. The selected contributions offer the groundwork for translating stem cell therapy to the clinic for treating central nervous system disorders. This issue is dedicated to Dr. Teng Ma, who passed away on May 18, 2019. Dr. Ma devoted a significant portion of his life in advancing biomedical engineering, including the utility of 3-dimensional bioreactor and magnetic resonance imaging, as a key element of the biological and therapeutic applications of stem cells for neurological disorders. Dr. Ma's research vision is celebrated in this compilation of ten articles on stem cell-based regenerative medicine.

11.
Brain Circ ; 5(3): 150-155, 2019.
Article in English | MEDLINE | ID: mdl-31620664

ABSTRACT

Moyamoya disease (MMD) represents a rare steno-occlusive disorder affecting the terminal ends of the internal carotid artery and promoting the development of a poor, abnormal vascular network at the brain's base. Primarily affecting East Asian countries over Western populations, MMD can be further divided into symptomatic and asymptomatic subtypes. The current knowledge of the underlying mechanisms and potential management strategies for asymptomatic cases of MMD are largely lacking and thus warrant investigation to elucidate the pathology of this rare disorder. Here, we assess research examining the expression profile of circular RNAs (circRNAs) of neutrophil transcriptome in asymptomatic MMD patients. These findings conclude that 123 differentially expressed circRNAs significantly contributed to metabolism, angiogenesis, and immune response. The hypoxia-inducing factor-1α signaling pathway was also revealed to be crucial in angiogenesis. We also evaluate current therapeutic options demonstrating the potential for MMD patients, such as EC-IC bypass and ischemic pre- and post-conditioning. These approaches combined with recent findings on the circRNA expression profile suggest a crucial role of anti-inflammatory and angiogenic-related mechanisms underlying MMD. Investigating the role of circRNAs and neutrophils in the asymptomatic MMD subtype may provide insight into its elusive pathology and direct future approaches to combat the progression of this rare disease.

12.
CNS Neurol Disord Drug Targets ; 18(9): 687-694, 2019.
Article in English | MEDLINE | ID: mdl-31642796

ABSTRACT

Here, we summarized recent advances in laboratory and clinical research on gut microbiome. The goal is to highlight recent discoveries on the biology and behavioral manifestations of gut microbiomes under normal and pathologic conditions. With this new scientific knowledge, we wish to cultivate cross-fertilization of science across multi-disciplines in the hopes of exploiting the gut microbiome as a key component of human development and its dysbiosis may signal pathological alterations that can be therapeutically targeted for regenerative medicine. In the end, we identify innovative research avenues that will merit from collaborations across biomedical disciplines that may facilitate the development of gut microbiome-based biomarkers and therapeutics. Gut microbiome stands as a core research area that transcends pediatric and nursing care, cancer biology, neurodegenerative disorders, cardiac function and diseases, among many other basic science and clinical arenas.


Subject(s)
Central Nervous System Diseases/metabolism , Dysbiosis/metabolism , Gastrointestinal Microbiome/physiology , Inflammation/metabolism , Animals , Humans , Models, Animal , Neurodegenerative Diseases/metabolism
13.
Brain Res ; 1725: 146432, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31491422

ABSTRACT

Therapeutic transplantation of autologous bone marrow mesenchymal stem cells (BMSCs) holds great promise for ischemic stroke, yet the efficacy is negatively impacted by aging. Here, we examined whether hypoxia conditioning could enhance aged human BMSCs-induced neuroprotection via secretome action. Primary cultured mouse neurons were exposed to oxygen glucose deprivation (OGD) to mimic ischemic stroke in vitro, then randomized into a hypoxia conditioned aged human BMSCs-conditioned medium (BMSC-hypoCM) versus normoxia conditioned (BMSC-norCM). After 22 h of reperfusion, cell viability was significantly increased in neurons treated with BMSC-hypoCM rather than BMSC-norCM. ELISA revealed that hypoxia conditioning enhanced vascular endothelial growth factor (VEGF) release into BMSC-derived CM. Blocking the VEGF receptor negated BMSC-hypoCM-induced protection for neurons against OGD insult. Altogether, our data indicates that hypoxia conditioning improves aged human BMSCs' therapeutic efficacy for neurons with ischemic challenge, in part via promoting secretion of VEGF.


Subject(s)
Brain Ischemia/metabolism , Mesenchymal Stem Cells/metabolism , Neurons/metabolism , Aged , Animals , Cell Hypoxia , Cell Survival , Culture Media, Conditioned , Disease Models, Animal , Humans , Mesenchymal Stem Cells/cytology , Primary Cell Culture , Vascular Endothelial Growth Factor A/metabolism
14.
Brain Res ; 1722: 146380, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31415765

ABSTRACT

Stroke poses a serious health and economic burden, and the lack of treatment options necessitates a viable therapy. Hypothermia represents a promising stroke therapy, yet side effects of full-body cooling, such as pneumonia, limit its clinical application. Selective endovascular cooling (SEC), via infusion of cold saline through the intraarterial artery, represents an attractive alternative by locally cooling the brain while preserving body temperature. However, the mechanisms underlying SEC are poorly understood. Brain-derived neurotrophic factor (BDNF) is a widely recognized promotor of neuroplasticity and biomarker of stroke outcomes, as well as its association with inflammation, such as IL-10. Stroke-induced neuroinflammation exacerbates damage and stems from peripheral organs, namely the spleen. The spleen has emerged as a therapeutic target for stroke, yet the effect of SEC on the splenic inflammatory response is unknown. Here, we aimed to elucidate the local and peripheral mechanisms driving SEC as a neuroprotective stroke therapy by examining brain BDNF and splenic IL-10 expression. Animals that received SEC prior to stroke displayed elevated brain BDNF expression ipsilaterally and contralaterally across the cortex, striatum, and hippocampus. SEC also upregulated splenic IL-10, suggesting alteration of the peripheral inflammatory response. The oxygen-glucose deprivation in vitro model of stroke further demonstrated that "cold" rat splenocytes protected rat primary neurons by upregulating BDNF and IL-10. Altogether these data support BDNF- and IL-10-based mechanisms underlying the neuroprotective potential of SEC therapy for stroke, and further advance the concept of exploiting the pathological link between brain and spleen as therapeutic targets.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Hypothermia, Induced , Interleukin-10/metabolism , Spleen/metabolism , Stroke/immunology , Stroke/metabolism , Animals , Brain/immunology , Carotid Artery, Internal/physiopathology , Hypothermia, Induced/methods , Male , Rats, Sprague-Dawley , Spleen/immunology
15.
Stem Cell Rev Rep ; 15(5): 690-702, 2019 10.
Article in English | MEDLINE | ID: mdl-31317505

ABSTRACT

Pharmaceuticals and cell-based regenerative medicine for Parkinson's disease (PD) offer palliative relief but do not arrest the disease progression. Cell therapy has emerged as an experimental treatment, but current cell sources such as human umbilical cord blood (hUCB) stem cells display only partial recapitulation of mature dopaminergic neuron phenotype and function. Nonetheless, stem cell grafts ameliorate PD-associated histological and behavioral deficits likely through stem cell graft-secreted therapeutic substances. We recently demonstrated the potential of hUCB-derived plasma in enhancing motor capabilities and gastrointestinal function, as well as preventing dopaminergic neuronal cell loss, in an 1-methyl-4-phenyl-1,2,3,6-tetrahydro-pyridine (MPTP) rodent model of PD. Recognizing the translational need to test in another PD model, we now examined here the effects of an intravenously transplanted combination of hUCB and plasma into the 6-hydroxydopamine (6-OHDA) lesioned adult rats. Animals received three separate doses of 4 × 106 hUCB cells with plasma beginning at 7 days after stereotaxic 6-OHDA lesion, then behaviorally and immunohistochemically evaluated over 56 days post-lesion. Whereas vehicle-treated lesioned animals exhibited the typical 6-OHDA neurobehavioral symptoms, hUCB and plasma-treated lesioned animals showed significant attenuation of motor function, gut motility, and nigral dopaminergic neuronal survival, combined with diminished pro-inflammatory microbiomes not only in the nigra, but also in the gut. Altogether these data support a regenerative medicine approach for PD by sequestering inflammation and neurotoxicity through correction of gut dysbiosis.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Gastrointestinal Microbiome , Inflammation/prevention & control , MPTP Poisoning/therapy , Neuroprotective Agents/administration & dosage , Regenerative Medicine , Umbilical Cord/cytology , Animals , Disease Models, Animal , Dopaminergic Neurons/cytology , Inflammation/etiology , Inflammation/pathology , MPTP Poisoning/etiology , MPTP Poisoning/pathology , Male , Motor Disorders/etiology , Motor Disorders/pathology , Motor Disorders/prevention & control , Rats , Rats, Sprague-Dawley , Substantia Nigra/cytology
16.
Neural Regen Res ; 14(4): 597-604, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30632499

ABSTRACT

Transplantation of human bone marrow mesenchymal stem cells (hMSCs) stands as a potent stroke therapy, but its exact mechanism remains unknown. This study investigated the anti-apoptotic mechanisms by which hMSCs exert neuroprotective effects on cerebral ischemia. Primary mixed cultures of rat neurons and astrocytes were cultured and exposed to oxygen-glucose deprivation. A two-hour period of "reperfusion" in standard medium and normoxic conditions was allowed and immediately followed by hMSCs and/or Bcl-2 antibody treatment. Cell viability of primary rat neurons and astrocytes was determined by 3-(4,5-dimethylthianol-2-yl)-2,5 diphenyl tetrazolium bromide and trypan blue exclusion methods. hMSC survival and differentiation were characterized by immunocytochemistry, while the concentration of Bcl-2 in the supernatant was measured by enzyme-linked immunosorbent assay to reveal the secretory anti-apoptotic function of hMSCs. Cultured hMSCs expressed embryonic-like stem cell phenotypic markers CXCR4, Oct4, SSEA4, and Nanog, as well as immature neural phenotypic marker Nestin. Primary rat neurons and astrocytes were protected from oxygen-glucose deprivation by hMSCs, which was antagonized by the Bcl-2 antibody. However, Bcl-2 levels in the supernatants did not differ between hMSC- and non-treated cells exposed to oxygen-glucose deprivation. Neuroprotective effects of hMSCs against cerebral ischemia were partially mediated by the anti-apoptotic mechanisms. However, further studies are warranted to fully elucidate this pathway.

17.
Prog Neurobiol ; 172: 23-39, 2019 01.
Article in English | MEDLINE | ID: mdl-30447256

ABSTRACT

Soluble epoxide hydrolase (sEH) degrades epoxides of fatty acids including epoxyeicosatrienoic acid isomers (EETs), which are produced as metabolites of the cytochrome P450 branch of the arachidonic acid pathway. EETs exert a variety of largely beneficial effects in the context of inflammation and vascular regulation. sEH inhibition is shown to be therapeutic in several cardiovascular and renal disorders, as well as in peripheral analgesia, via the increased availability of anti-inflammatory EETs. The success of sEH inhibitors in peripheral systems suggests their potential in targeting inflammation in the central nervous system (CNS) disorders. Here, we describe the current roles of sEH in the pathology and treatment of CNS disorders such as stroke, traumatic brain injury, Parkinson's disease, epilepsy, cognitive impairment, dementia and depression. In view of the robust anti-inflammatory effects of stem cells, we also outlined the potency of stem cell treatment and sEH inhibitors as a combination therapy for these CNS disorders. This review highlights the gaps in current knowledge about the pathologic and therapeutic roles of sEH in CNS disorders, which should guide future basic science research towards translational and clinical applications of sEH inhibitors for treatment of neurological diseases.


Subject(s)
Central Nervous System Agents/pharmacology , Central Nervous System Diseases/drug therapy , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Animals , Central Nervous System Agents/therapeutic use , Central Nervous System Diseases/enzymology , Enzyme Inhibitors/therapeutic use , Epoxide Hydrolases/metabolism , Humans
18.
Handb Exp Pharmacol ; 247: 277-299, 2018.
Article in English | MEDLINE | ID: mdl-28315071

ABSTRACT

Research of the opioid system and its composite receptors and ligands has revealed its promise as a potential therapy for neurodegenerative diseases such as stroke and Parkinson's Disease. In particular, delta opioid receptors (DORs) have been elucidated as a therapeutically distinguished subset of opioid receptors and a compelling target for novel intervention techniques. Research is progressively shedding light on the underlying mechanism of DORs and has revealed two mechanisms of DOR neuroprotection; DORs function to maintain ionic homeostasis and also to trigger endogenous neuroprotective pathways. Delta opioid agonists such as (D-Ala2, D-Leu5) enkephalin (DADLE) have been shown to promote neuronal survival and decrease apoptosis, resulting in a substantial amount of research for its application as a neurological therapeutic. Most notably, DADLE has demonstrated significant potential to reduce cell death following ischemic events. Current research is working to reveal the complex mechanisms of DADLE's neuroprotective properties. Ultimately, our knowledge of the DOR receptors and agonists has made the opioid system a promising target for therapeutic intervention in many neurological disorders.


Subject(s)
Enkephalin, Leucine-2-Alanine/pharmacology , Nervous System Diseases/drug therapy , Opioid Peptides/pharmacology , Receptors, Opioid, delta/drug effects , Stroke/drug therapy , Animals , Enkephalin, Leucine-2-Alanine/therapeutic use , Humans , Nervous System Diseases/physiopathology , Opioid Peptides/therapeutic use , Stroke/physiopathology
19.
Prog Neurobiol ; 158: 94-131, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28743464

ABSTRACT

Ischemic stroke is a leading cause of death worldwide. A key secondary cell death mechanism mediating neurological damage following the initial episode of ischemic stroke is the upregulation of endogenous neuroinflammatory processes to levels that destroy hypoxic tissue local to the area of insult, induce apoptosis, and initiate a feedback loop of inflammatory cascades that can expand the region of damage. Stem cell therapy has emerged as an experimental treatment for stroke, and accumulating evidence supports the therapeutic efficacy of stem cells to abrogate stroke-induced inflammation. In this review, we investigate clinically relevant stem cell types, such as hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs), very small embryonic-like stem cells (VSELs), neural stem cells (NSCs), extraembryonic stem cells, adipose tissue-derived stem cells, breast milk-derived stem cells, menstrual blood-derived stem cells, dental tissue-derived stem cells, induced pluripotent stem cells (iPSCs), teratocarcinoma-derived Ntera2/D1 neuron-like cells (NT2N), c-mycER(TAM) modified NSCs (CTX0E03), and notch-transfected mesenchymal stromal cells (SB623), comparing their potential efficacy to sequester stroke-induced neuroinflammation and their feasibility as translational clinical cell sources. To this end, we highlight that MSCs, with a proven track record of safety and efficacy as a transplantable cell for hematologic diseases, stand as an attractive cell type that confers superior anti-inflammatory effects in stroke both in vitro and in vivo. That stem cells can mount a robust anti-inflammatory action against stroke complements the regenerative processes of cell replacement and neurotrophic factor secretion conventionally ascribed to cell-based therapy in neurological disorders.


Subject(s)
Cell Death/physiology , Inflammation/therapy , Stem Cell Transplantation , Stroke/immunology , Stroke/therapy , Animals , Humans , Inflammation/etiology
20.
CNS Neurol Disord Drug Targets ; 16(4): 414-424, 2017.
Article in English | MEDLINE | ID: mdl-28322170

ABSTRACT

The opioid system has been elucidated as a potential target for therapy in a variety of neurological disorders including stroke. Delta opioid receptors have been revealed to pose an especially compelling biological function for new neuroprotective therapies. Two distinct therapeutic mechanisms have been characterized for delta opioid receptors, namely, these receptors aid in maintaining ionic homeostasis and initiate endogenous neuroprotective pathways. Specific agonists of delta opioid receptors, such as (D-Ala2, D-Leu5) enkephalin (DADLE), have displayed the ability to promote neuronal survival and mitigate apoptotic pathways. These findings have led to a significant amount of research on this molecule's potential as a neurotherapeutic. At the forefront of these efforts has been investigation into DADLE's ability to protect neurons and glial cells following ischemia. Additionally, current research is attempting to reveal the dynamic neuroprotective mechanisms that mediate DADLE's therapeutic benefits. This review article discusses the scientific evidence supporting the use of delta opioid family of receptors and ligands as a promising target for therapeutic intervention in neurological disorders, with emphasis on stroke.


Subject(s)
Brain Diseases/drug therapy , Brain Diseases/metabolism , Neuroprotective Agents/pharmacology , Receptors, Opioid, delta/metabolism , Animals , Humans , Neuroprotection/drug effects , Neuroprotection/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...