Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
CNS Neurosci Ther ; 22(3): 200-11, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26775604

ABSTRACT

AIMS: Traumatic brain injury (TBI) occurs when the head is impacted by an external force causing either a closed or penetrating head injury through a direct or accelerating impact. In laboratory research, most of the TBI animal models focus on a specific region to cause brain injury, but traumatic injuries in patients do not always impact the same brain regions. The aim of this study was to examine the histopathological effects of different angles of mechanical injury by manipulating the trajectory of the controlled cortical impact injury (CCI) model in adult Sprague-Dawley rats. METHODS: The CCI model was manipulated as follows: conventional targeting of the frontal cortex, farthest right angle targeting the frontal cortex, closest right angle targeting the frontal cortex, olfactory bulb injury, and cerebellar injury. Three days after TBI, brains were harvested to analyze cortical and hippocampal cell loss, neuroinflammatory response, and neurogenesis via immunohistochemistry. RESULTS: Results revealed cell death in the M1 region of the cortex across all groups, and in the CA3 area from olfactory bulb injury group. This observed cell death involved upregulation of inflammation as evidenced by rampant MHCII overexpression in cortex, but largely spared Ki-67/nestin neurogenesis in the hippocampus during this acute phase of TBI. CONCLUSION: These results indicate a trajectory-dependent injury characterized by exacerbation of inflammation and different levels of impaired cell proliferation and neurogenesis. Such multiple brain areas showing varying levels of cell death after region-specific CCI model may closely mimic the clinical manifestations of TBI.


Subject(s)
Brain Injuries/pathology , Cerebral Cortex/pathology , Hippocampus/pathology , Analysis of Variance , Animals , Cerebral Cortex/metabolism , Disease Models, Animal , Gene Expression Regulation/physiology , Hippocampus/metabolism , Histocompatibility Antigens Class II/metabolism , Ki-67 Antigen/metabolism , Male , Microglia/pathology , Nestin/metabolism , Rats , Rats, Sprague-Dawley
2.
CNS Neurosci Ther ; 21(4): 348-56, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25399760

ABSTRACT

Stroke is a significant unmet clinical need. The current stroke treatment of tissue plasminogen activator is limited to the very acute 4.5 h after disease onset which benefits only less than 3% of ischemic stroke patients. Our overarching hypothesis advances the notion that gender, which has been established as a comorbidity factor of stroke, plays a key role in regenerative medicine, in particular stem cell therapy. We hypothesize that gender is a key factor in culture-induced stemness of adult stem cells. Our goal is to provide new evidence supporting gender effects on stroke and stem cells for the purpose of enhancing our understanding of the pathophysiology of the disease and developing novel stem cell-based therapeutics targeting gender-relevant stress hormones as manifested in a stroke-postpartum depression paradigm.


Subject(s)
Adult Stem Cells/physiology , Depression, Postpartum/physiopathology , Sex Characteristics , Stroke/physiopathology , Animals , Depression, Postpartum/therapy , Female , Hormones/metabolism , Humans , Male , Stroke/therapy
3.
Front Cell Neurosci ; 8: 227, 2014.
Article in English | MEDLINE | ID: mdl-25165432

ABSTRACT

Accumulating preclinical evidence suggests the use of amnion as a source of stem cells for investigations of basic science concepts related to developmental cell biology, but also for stem cells' therapeutic applications in treating human disorders. We previously reported isolation of viable rat amniotic fluid-derived stem (AFS) cells. Subsequently, we recently reported the therapeutic benefits of intravenous transplantation of AFS cells in a rodent model of ischemic stroke. Parallel lines of investigations have provided safety and efficacy of stem cell therapy for treating stroke and other neurological disorders. This review article highlights the need for investigations of mechanisms underlying AFS cells' therapeutic benefits and discusses lab-to-clinic translational gating items in an effort to optimize the clinical application of the cell transplantation for stroke.

4.
Front Neurol ; 5: 147, 2014.
Article in English | MEDLINE | ID: mdl-25161645

ABSTRACT

Treatments for neonatal hypoxic-ischemic encephalopathy (HIE) have been limited. The aim of this paper is to offer translational research guidance on stem cell therapy for neonatal HIE by examining clinically relevant animal models, practical stem cell sources, safety and efficacy of endpoint assays, as well as a general understanding of modes of action of this cellular therapy. In order to do so, we discuss the clinical manifestations of HIE, highlighting its overlapping pathologies with stroke and providing insights on the potential of cell therapy currently investigated in stroke, for HIE. To this end, we draw guidance from recommendations outlined in stem cell therapeutics as an emerging paradigm for stroke or STEPS, which have been recently modified to Baby STEPS to cater for the "neonatal" symptoms of HIE. These guidelines recognized that neonatal HIE exhibit distinct disease symptoms from adult stroke in need of an innovative translational approach that facilitates the entry of cell therapy in the clinic. Finally, new information about recent clinical trials and insights into combination therapy are provided with the vision that stem cell therapy may benefit from available treatments, such as hypothermia, already being tested in children diagnosed with HIE.

5.
Int J Mol Sci ; 15(9): 15225-43, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25170809

ABSTRACT

Cell therapy now constitutes an important area of regenerative medicine. The aging of the population has mandated the discovery and development of new and innovative therapeutic modalities to combat devastating disorders such as stroke. Menstrual blood and Sertoli cells represent two sources of viable transplantable cells that are gender-specific, both of which appear to have potential as donor cells for transplantation in stroke. During the subacute phase of stroke, the use of autologous cells offers effective and practical clinical application and is suggestive of the many benefits of using the aforementioned gender-specific cells. For example, in addition to being exceptionally immunosuppressive, testis-derived Sertoli cells secrete many growth and trophic factors and have been shown to aid in the functional recovery of animals transplanted with fetal dopaminergic cells. Correspondingly, menstrual blood cells are easily obtainable and exhibit angiogenic characteristics, proliferative capability, and pluripotency. Of further interest is the ability of menstrual blood cells, following transplantation in stroke models, to migrate to the infarct site, secrete neurotrophic factors, regulate the inflammatory response, and be steered towards neural differentiation. From cell isolation to transplantation, we emphasize in this review paper the practicality and relevance of the experimental and clinical use of gender-specific stem cells, such as Sertoli cells and menstrual blood cells, in the treatment of stroke.


Subject(s)
Adult Stem Cells/cytology , Cell Differentiation , Sex Characteristics , Stem Cell Transplantation/methods , Adult Stem Cells/transplantation , Animals , Female , Humans , Male
6.
Front Syst Neurosci ; 8: 116, 2014.
Article in English | MEDLINE | ID: mdl-25009475

ABSTRACT

Modified mesenchymal stromal cells (MSCs) display a unique mechanism of action during the repair phase of traumatic brain injury by exhibiting the ability to build a biobridge between the neurogenic niche and the site of injury. Immunohistochemistry and laser capture assay have visualized this biobridge in the area between the neurogenic subventricular zone and the injured cortex. This biobridge expresses high levels of extracellular matrix metalloproteinases (MMPs), which are initially co-localized with a stream of transplanted MSCs, but later this region contains only few to non-detectable grafts and becomes overgrown by newly recruited host cells. We have reported that long-distance migration of host cells from the neurogenic niche to the injured brain site can be attained via these transplanted stem cell-paved biobridges, which serve as a key regenerative process for the initiation of endogenous repair mechanisms. Thus, far the two major schools of discipline in stem cell repair mechanisms support the idea of "cell replacement" and the bystander effects of "trophic factor secretion." Our novel observation of stem cell-paved biobridges as pathways for directed migration of host cells from neurogenic niche toward the injured brain site adds another mode of action underlying stem cell therapy. More in-depth investigations on graft-host interaction will likely aid translational research focused on advancing this stem cell-paved biobridge from its current place, as an equally potent repair mechanism as cell replacement and trophic factor secretion, into a new treatment strategy for traumatic brain injury and other neurological disorders.

7.
Cell Med ; 6(3): 111-122, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24999442

ABSTRACT

Stroke is the third most common cause of death and severe disability among Western populations. Overall, the incidence of stroke is uniformly higher in men than in women. Stroke is rare in women during the reproductive years, and rapidly increases after menopause, strongly suggesting that estrogen (E2) plays an important role in the prevention of stroke. Ongoing studies are currently evaluating both the benefits and risks associated with E2 replacement therapy and hormone replacement therapy in stroke. Equally important is the role of E2 receptor (ER), as studies indicate that ER populations in several tissue sites may significantly change during stress and aging. Such changes may affect the patient's susceptibility to neurological disorders including stroke, and greatly affect the response to selective E2 receptor modulators (SERMs). Replacement therapies may be inefficient with low ER levels. The goal of this review paper is to discuss an animal model that will allow investigations of the potential therapeutic effects of E2 and its derivatives in stroke. We hypothesize that E2 neuroprotection is, in part, receptor mediated. This hypothesis is a proof of principle approach to demonstrate a role for specific ER subtypes in E2 neuroprotection. To accomplish this, we use a retroviral mediated gene transfer strategy that express subtypes of the ER gene in regions of the rat brain most susceptible to neuronal damage, namely the striatum and cortex. The animal model is exposed to experimental stroke conditions involving middle cerebral artery occlusion (MCAo) method, and eventually the extent of neuronal damage will be evaluated. A reduction in neuronal damage is expected when E2 is administered with specific ER subtypes. From this animal model, an optimal E2 dose and treatment regimen can be determined. The animal model can help identify potential E2-like therapeutics in stroke, and screen for beneficial or toxic additives present in commercial E2 preparations that are currently available. Such studies will be informative in designing drug therapies for stroke.

8.
Cell Med ; 6(3): 123-127, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24999443

ABSTRACT

Multiple sclerosis (MS) is characterized by widespread immunomodulatory demyelination of the CNS resulting in nerve cell dysfunction. Accordingly, treatment strategies have been centered on immunodulation and remyelination, with the former primarily focused on reducing the pathology rather than enhancing myelin repair which the latter targets. While conceding to the emerging view of heterogeneity in the pathology of MS, which precludes variations in degree of immune response (i.e., inflammation) and demyelination, the concept of enhancing myelin repair is appealing since it is likely to provide both disease-reducing and disease-inhibiting therapeutic approach to MS. In this regard, we and several others, have proposed that cell replacement therapy is an effective strategy to repair the myelin in MS. Here, we hypothesize that transplantation of mouse bone marrow-derived oligodendrocytes (BMDOs) and BMDOs transfected with Ephrin proteins (BMDO+Ephrin), which are known to enhance cell and axonal migratory capacity, may produce therapeutic benefits in animal models of MS.

9.
J Neurosci ; 34(1): 313-26, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24381292

ABSTRACT

Traumatic brain injury (TBI) survivors exhibit motor and cognitive symptoms from the primary injury that can become aggravated over time because of secondary cell death. In the present in vivo study, we examined the beneficial effects of human adipose-derived stem cells (hADSCs) in a controlled cortical impact model of mild TBI using young (6 months) and aged (20 months) F344 rats. Animals were transplanted intravenously with 4 × 10(6) hADSCs (Tx), conditioned media (CM), or vehicle (unconditioned media) at 3 h after TBI. Significant amelioration of motor and cognitive functions was revealed in young, but not aged, Tx and CM groups. Fluorescent imaging in vivo and ex vivo revealed 1,1' dioactadecyl-3-3-3',3'-tetramethylindotricarbocyanine iodide-labeled hADSCs in peripheral organs and brain after TBI. Spatiotemporal deposition of hADSCs differed between young and aged rats, most notably reduced migration to the aged spleen. Significant reduction in cortical damage and hippocampal cell loss was observed in both Tx and CM groups in young rats, whereas less neuroprotection was detected in the aged rats and mainly in the Tx group but not the CM group. CM harvested from hADSCs with silencing of either NEAT1 (nuclear enriched abundant transcript 1) or MALAT1 (metastasis associated lung adenocarcinoma transcript 1), long noncoding RNAs (lncRNAs) known to play a role in gene expression, lost the efficacy in our model. Altogether, hADSCs are promising therapeutic cells for TBI, and lncRNAs in the secretome is an important mechanism of cell therapy. Furthermore, hADSCs showed reduced efficacy in aged rats, which may in part result from decreased homing of the cells to the spleen.


Subject(s)
Adipose Tissue/transplantation , Brain Injuries/surgery , Cognition Disorders/surgery , Motor Skills Disorders/surgery , Nerve Degeneration/surgery , Stem Cell Transplantation/methods , Adipose Tissue/cytology , Age Factors , Animals , Brain/metabolism , Brain/pathology , Brain/surgery , Brain Injuries/metabolism , Brain Injuries/pathology , Cells, Cultured , Cognition Disorders/metabolism , Cognition Disorders/pathology , Humans , Infusions, Intravenous , Male , Motor Skills Disorders/metabolism , Motor Skills Disorders/pathology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Rats , Rats, Inbred F344 , Tissue Distribution/physiology
10.
J Stem Cell Res Ther ; 4(4): 12912, 2013 May 01.
Article in English | MEDLINE | ID: mdl-24077523

ABSTRACT

Stem cells exert therapeutic effects against ischemic stroke via transplantation of exogenous stem cells or stimulation of endogenous stem cells within the neurogenic niches of subventricular zone and subgranular zone, or recruited from the bone marrow through peripheral circulation. In this paper, we review the different sources of stem cells that have been tested in animal models of stroke. In addition, we discuss specific mechanisms of action, in particular neurovascular repair by endothelial progenitor cells, as key translational research for advancing the clinical applications of stem cells for ischemic stroke.

11.
Neuroreport ; 24(18): 1067-71, 2013 Dec 18.
Article in English | MEDLINE | ID: mdl-24169604

ABSTRACT

With limited clinical trials on stem cell therapy for adult stroke underway, the assessment of efficacy also needs to be considered for neonatal hypoxic-ischemic brain injury, considering its distinct symptoms. The critical nature of this condition leads to establishment of deficits that last a lifetime. Here, we will highlight the progress of current translational research, commenting on the critical nature of the disease, stem cell sources, the use of hypothermia, safety and efficacy of each treatment, modes of action, and the possibility of combination therapy. With this in mind, we reference translational guidelines established by a consortium of research partners called Stem cell Therapeutics as an Emerging Paradigm for Stroke (STEPS). The guidelines of STEPS are directed toward evaluating outcomes of cell therapy in adult stroke; however, we identify the overlapping pathology, as we believe that these guidelines will serve well in the investigation of neonatal hypoxic-ischemic therapy. Finally, we discuss emerging treatments and a case report, altogether suggesting that the potential for these treatments to be used in synergy has arrived and the time for advancing stem cell use in combination with hypothermia for cerebral palsy is now.


Subject(s)
Cerebral Palsy/therapy , Critical Care , Hypothermia, Induced , Hypoxia-Ischemia, Brain/therapy , Stem Cell Transplantation , Cerebral Palsy/surgery , Combined Modality Therapy , Humans , Hypoxia-Ischemia, Brain/surgery , Infant, Newborn , Translational Research, Biomedical
12.
Int J Mol Sci ; 14(9): 17410-9, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23979422

ABSTRACT

In pursuit of neurological therapies, the opioid system, specifically delta opioid receptors and delta opioid peptides, demonstrates promising therapeutic potential for stroke, Parkinson's disease, and other degenerative neurological conditions. Recent studies offer strong evidence in support of the therapeutic use of delta opioid receptors, and provide insights into the underlying mechanisms of action. Delta opioid receptors have been shown to confer protective effects by mediating ionic homeostasis and activating endogenous neuroprotective pathways. Additionally, delta opioid agonists such as (D-Ala 2, D-Leu 5) enkephalin (DADLE) have been shown to decrease apoptosis and promote neuronal survival. In its entirety, the delta opioid system represents a promising target for neural therapies.


Subject(s)
Neuroprotective Agents/therapeutic use , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Animals , Enkephalin, Leucine-2-Alanine/therapeutic use , Humans , Stroke/drug therapy
13.
PLoS One ; 8(5): e63553, 2013.
Article in English | MEDLINE | ID: mdl-23675488

ABSTRACT

BACKGROUND: Comprehensive stroke studies reveal diaschisis, a loss of function due to pathological deficits in brain areas remote from initial ischemic lesion. However, blood-brain barrier (BBB) competence in subacute diaschisis is uncertain. The present study investigated subacute diaschisis in a focal ischemic stroke rat model. Specific focuses were BBB integrity and related pathogenic processes in contralateral brain areas. METHODOLOGY/PRINCIPAL FINDINGS: In ipsilateral hemisphere 7 days after transient middle cerebral artery occlusion (tMCAO), significant BBB alterations characterized by large Evans Blue (EB) parenchymal extravasation, autophagosome accumulation, increased reactive astrocytes and activated microglia, demyelinization, and neuronal damage were detected in the striatum, motor and somatosensory cortices. Vascular damage identified by ultrastuctural and immunohistochemical analyses also occurred in the contralateral hemisphere. In contralateral striatum and motor cortex, major ultrastructural BBB changes included: swollen and vacuolated endothelial cells containing numerous autophagosomes, pericyte degeneration, and perivascular edema. Additionally, prominent EB extravasation, increased endothelial autophagosome formation, rampant astrogliosis, activated microglia, widespread neuronal pyknosis and decreased myelin were observed in contralateral striatum, and motor and somatosensory cortices. CONCLUSIONS/SIGNIFICANCE: These results demonstrate focal ischemic stroke-induced pathological disturbances in ipsilateral, as well as in contralateral brain areas, which were shown to be closely associated with BBB breakdown in remote brain microvessels and endothelial autophagosome accumulation. This microvascular damage in subacute phase likely revealed ischemic diaschisis and should be considered in development of treatment strategies for stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Ischemia/complications , Stroke/etiology , Stroke/metabolism , Animals , Astrocytes/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/ultrastructure , Corpus Striatum/blood supply , Corpus Striatum/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Infarction, Middle Cerebral Artery/complications , Male , Microglia/metabolism , Microvessels/metabolism , Microvessels/pathology , Microvessels/ultrastructure , Motor Cortex/blood supply , Motor Cortex/metabolism , Motor Cortex/pathology , Myelin Sheath/metabolism , Neurons/metabolism , Permeability , Phagosomes , Rats , Stroke/pathology
14.
Future Neurol ; 8(2): 193-203, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23565051

ABSTRACT

Stem cell therapy for adult stroke has reached limited clinical trials. Here, we provide translational research guidance on stem cell therapy for neonatal hypoxic-ischemic brain injury requiring a careful consideration of clinically relevant animal models, feasible stem cell sources, and validated safety and efficacy endpoint assays, as well as a general understanding of modes of action of this cellular therapy. To this end, we refer to existing translational guidelines, in particular the recommendations outlined in the consortium of academicians, industry partners and regulators called Stem cell Therapeutics as an Emerging Paradigm for Stroke or STEPS. Although the STEPS guidelines are directed at enhancing the successful outcome of cell therapy in adult stroke, we highlight overlapping pathologies between adult stroke and neonatal hypoxic-ischemic brain injury. We are, however, cognizant that the neonatal hypoxic-ischemic brain injury displays disease symptoms distinct from adult stroke in need of an innovative translational approach that facilitates the entry of cell therapy in the clinic. Finally, insights into combination therapy are provided with the vision that stem cell therapy may benefit from available treatments, such as hypothermia, already being tested in children diagnosed with hypoxic-ischemic brain injury.

15.
J Clin Med ; 2(4): 220-241, 2013 Dec.
Article in English | MEDLINE | ID: mdl-25177494

ABSTRACT

With a constellation of stem cell sources available, researchers hope to utilize their potential for cellular repair as a therapeutic target for disease. However, many lab-to-clinic translational considerations must be given in determining their efficacy, variables such as the host response, effects on native tissue, and potential for generating tumors. This review will discuss the current knowledge of stem cell research in neurological disease, mainly stroke, with a focus on the benefits, limitations, and clinical potential.

16.
PLoS One ; 7(9): e45256, 2012.
Article in English | MEDLINE | ID: mdl-23028885

ABSTRACT

Inflammation in the brain plays a major role in neurodegenerative diseases. In particular, microglial cell activation is believed to be associated with the pathogenesis of neurodegenerative diseases, including Parkinson's disease (PD). An increase in microglia activation has been shown in the substantia nigra pars compacta (SNpc) of PD models when there has been a decrease in tyrosine hydroxylase (TH) positive cells. This may be a sign of neurotoxicity due to prolonged activation of microglia in both early and late stages of disease progression. Natural products, such as spirulina, derived from blue green algae, are believed to help reverse this effect due to its anti-inflammatory/anti-oxidant properties. An adeno-associated virus vector (AAV9) for α-synuclein was injected in the substantia nigra of rats to model Parkinson's disease and to study the effects of spirulina on the inflammatory response. One month prior to surgeries, rats were fed either a diet enhanced with spirulina or a control diet. Immunohistochemistry was analyzed with unbiased stereological methods to quantify lesion size and microglial activation. As hypothesized, spirulina was neuroprotective in this α-synuclein model of PD as more TH+ and NeuN+ cells were observed; spirulina concomitantly decreased the numbers of activated microglial cells as determined by MHCII expression. This decrease in microglia activation may have been due, in part, to the effect of spirulina to increase expression of the fractalkine receptor (CX3CR1) on microglia. With this study we hypothesize that α-synuclein neurotoxicity is mediated, at least in part, via an interaction with microglia. We observed a decrease in activated microglia in the rats that received a spirulina- enhanced diet concomitant to neuroprotection. The increase in CX3CR1 in the groups that received spirulina, suggests a potential mechanism of action.


Subject(s)
Diet , Microglia/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/diet therapy , Spirulina/chemistry , Substantia Nigra/drug effects , alpha-Synuclein/adverse effects , Animals , CX3C Chemokine Receptor 1 , Dependovirus/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Injections, Intraventricular , Male , Microglia/metabolism , Microglia/pathology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Rats , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
17.
J Neuroinflammation ; 8: 9, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21266082

ABSTRACT

BACKGROUND: Parkinson's disease is characterized by a progressive loss of dopaminergic neurons in the substantia nigra. The cause of the neurodegeneration is unknown. Neuroinflammation has been clearly shown in Parkinson's disease and may be involved in the progressive nature of the disease. Microglia are capable of producing neuronal damage through the production of bioactive molecules such as cytokines, as well as reactive oxygen species (ROS), and nitric oxide (NO). The inflammatory response in the brain is tightly regulated at multiple levels. One form of immune regulation occurs via neurons. Fractalkine (CX3CL1), produced by neurons, suppresses the activation of microglia. CX3CL1 is constitutively expressed. It is not known if addition of exogenous CX3CL1 beyond otherwise physiologically normal levels could decrease microglia activation and thereby minimize the secondary neurodegeneration following a neurotoxic insult. METHODS: The intrastriatal 6-hydroxydopamine (6-OHDA) rat model of Parkinson disease, was used to test the hypothesis that exogenous CX3CL1 could be neuroprotective. Treatment with recombinant CX3CL1 was delivered to the striatum by an osmotic minipump for 28 days beginning 7 days after the initial insult. Unbiased stereological methods were used to quantify the lesion size in the striatum, the amount of neuronal loss in the substantia nigra, and the amount of microglia activation. RESULTS: As hypothesized, CX3CL1 was able to suppress this microglia activation. The reduced microglia activation was found to be neuroprotective as the CX3CL1 treated rats had a smaller lesion volume in the striatum and importantly significantly fewer neurons were lost in the CX3CL1 treated rats. CONCLUSION: These findings demonstrated that CX3CL1 plays a neuroprotective role in 6-OHDA-induced dopaminergic lesion and it might be an effective therapeutic target for many neurodegenerative diseases, including Parkinson disease and Alzheimer disease, where inflammation plays an important role.


Subject(s)
Chemokine CX3CL1/pharmacology , Chemokine CX3CL1/toxicity , Microglia/drug effects , Microglia/physiology , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Animals , Chemokine CX3CL1/genetics , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Humans , Male , Microglia/cytology , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Neurons/cytology , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Oxidopamine/pharmacology , Parkinson Disease/physiopathology , Rats , Rats, Inbred F344 , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology
18.
BMC Neurosci ; 9: 22, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-18275610

ABSTRACT

BACKGROUND: Neurogenesis continues to occur throughout life but dramatically decreases with increasing age. This decrease is mostly related to a decline in proliferative activity as a result of an impoverishment of the microenvironment of the aged brain, including a reduction in trophic factors and increased inflammation. RESULTS: We determined that human umbilical cord blood mononuclear cells (UCBMC) given peripherally, by an intravenous injection, could rejuvenate the proliferative activity of the aged neural stem/progenitor cells. This increase in proliferation lasted for at least 15 days after the delivery of the UCBMC. Along with the increase in proliferation following UCBMC treatment, an increase in neurogenesis was also found in the aged animals. The increase in neurogenesis as a result of UCBMC treatment seemed to be due to a decrease in inflammation, as a decrease in the number of activated microglia was found and this decrease correlated with the increase in neurogenesis. CONCLUSION: The results demonstrate that a single intravenous injection of UCBMC in aged rats can significantly improve the microenvironment of the aged hippocampus and rejuvenate the aged neural stem/progenitor cells. Our results raise the possibility of a peripherally administered cell therapy as an effective approach to improve the microenvironment of the aged brain.


Subject(s)
Aging/pathology , Fetal Blood/physiology , Hippocampus/cytology , Neurons/cytology , Regeneration/physiology , Stem Cells/cytology , Animals , Cell Cycle/physiology , Cell Proliferation , Cellular Senescence/physiology , Humans , Injections, Intravenous , Leukocytes, Mononuclear/cytology , Male , Microglia/cytology , Phenotype , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...