One-Year Follow-Up After Bone Marrow Stromal Cell Treatment in Middle-Aged Female Rats With Stroke (original) (raw)
Related papers
Treatment of stroke in rat with intracarotid administration of marrow stromal cells
Neurology, 2001
Article abstract-Objective: To measure the therapeutic efficacy for the treatment of stroke with intra-arterial administration of bone marrow stromal cells (MSC). Background: MSC have characteristics of stem and progenitor cells. The hypothesis that MSC injected into the internal carotid artery after stroke enter into ischemic brain and improve neurologic recovery was tested. Methods: Twenty-five adult Wistar rats were subjected to transient (2-hour) middle cerebral artery occlusion alone (n ϭ 9), or treated with intracarotid arterial injection of 200 L phosphate-buffered saline (n ϭ 8) or 2 ϫ 10 6 MSC in 200 L phosphate-buffered saline (n ϭ 8) 1 day after ischemia. MSC were harvested and isolated from additional adult rats and then cultured and labeled with bromodeoxyuridine. Rats were subjected to neurologic functional tests (adhesive-removal, modified neurologic severity scores) before and at 1, 7, and 14 days after middle cerebral artery occlusion. Immunohistochemistry was used to identify cell-specific proteins of bromodeoxyuridine-reactive MSC. Results: Bromodeoxyuridine-reactive cells (~21% of 2 ϫ 10 6 injected MSC) distributed throughout the territory of the middle cerebral artery by 14 days after ischemia. Some bromodeoxyuridine-reactive cells expressed proteins characteristic of astrocytes and neurons. Rats with intra-arterial transplantation of MSC exhibited improvement on the adhesive-removal test (p Ͻ 0.05) and the modified neurologic severity scores (p Ͻ 0.05) at 14 days compared with controls. Conclusions: MSC injected intra-arterially are localized and directed to the territory of the middle cerebral artery, and these cells foster functional improvement after cerebral ischemia.
Therapeutic window for treatment of cortical ischemia with bone marrow-derived cells in rats
Brain Research, 2010
The beneficial effect of treatment with bone marrow mononuclear cells (BMMCs) was evaluated in different therapeutic windows in a rat model of focal ischemia induced by thermocoagulation of the blood vessels in the left motor, somestesic, and sensorimotor cortices. We also compared the therapeutic benefits between BMMCs and bone marrowderived mesenchymal stem cells (MSCs). BMMCs and MSCs were obtained from donor rats and injected into the jugular vein after ischemia. BMMCs-treated animals received approximately 3 × 10 7 cells at post-ischemic days (PIDs) 1, 7, 14, or 30. MSCs-treated animals received approximately 3 × 10 6 cells at PIDs 1 and 30. Control animals received only the vehicle. The animals were then evaluated for functional sensorimotor recovery weekly with behavioral tests (cylinder test and adhesive test). Significant recovery of sensorimotor function was only observed in the cylinder test in animals treated with BMMCs at PIDs 1 and 7. Similar effects were also observed in the animals treated with MSCs 1 day after ischemia, but not in animals treated with MSCs 30 days after ischemia. Significant decrease in glial scarring did not seem to be a mechanism of action of BMMCs, since treatment with BMMCs did not change the level of expression of GFAP, indicating no significant change in the astrocytic scar in the periphery of the ischemic lesion. These results suggest that BMMCs might be an efficient treatment protocol for stroke only in the acute/subacute phase of the disease, and its efficiency in inducing functional recovery is similar to that of MSCs.
Gliosis and brain remodeling after treatment of stroke in rats with marrow stromal cells
Glia, 2005
The long-term (4-month) responses to treatment of stroke in the older adult rat, using rat bone marrow stromal cells (MSCs), have not been investigated. Retired breeder rats were subjected to middle cerebral artery occlusion (MCAo) alone, or injected intravenously with 3 ϫ 10 6 MSCs, at 7 days after MCAo. Functional recovery was measured using an adhesive-removal patch test and a modified neurological severity score. Bromodeoxyuridine, a cell proliferation marker, was injected daily for 14 before sacrifice. Animals were sacrificed 4 months after stroke. Double immunostaining was used to identify cell proliferation and cell types for axons, astrocytes, microglia, and oligodendrocytes. MSC treatment induced significant improvement in neurological outcome after MCAo compared with control rats. MSC treatment reduced the thickness of the scar wall (P Ͻ 0.05) and reduced the numbers of microglia/macrophages within the scar wall (P Ͻ 0.01). Double staining showed increased expression of an axonal marker (GAP-43), among reactive astrocytes in the scar boundary zone and in the subventricular zone in the treated rats. Bromodeoxyuridine in cells preferentially colocalized with markers of astrocytes (GFAP) and oligodendrocytes (RIP) in the ipsilateral hemisphere, and gliogenesis was enhanced in the subventricular zone of the rats treated with MSCs. This is the first report to show that MSCs injected at 7 days after stroke improve long-term neurological outcome in older animals. Brain tissue repair is an ongoing process with reactive gliosis, which persists for at least 4 months after stroke. Reactive astrocytes responding to MSC treatment of ischemia may also promote axonal regeneration during long-term recovery.
PLoS ONE, 2014
Intranasal treatment with C57BL/6 MSCs reduces lesion volume and improves motor and cognitive behavior in the neonatal hypoxic-ischemic (HI) mouse model. In this study, we investigated the potential of human MSCs (hMSCs) to treat HI brain injury in the neonatal mouse. Assessing the regenerative capacity of hMSCs is crucial for translation of our knowledge to the clinic. We determined the neuroregenerative potential of hMSCs in vitro and in vivo by intranasal administration 10 d post-HI in neonatal mice. HI was induced in P9 mouse pups. 1610 6 or 2610 6 hMSCs were administered intranasally 10 d post-HI. Motor behavior and lesion volume were measured 28 d post-HI. The in vitro capacity of hMSCs to induce differentiation of mouse neural stem cell (mNSC) was determined using a transwell co-culture differentiation assay. To determine which chemotactic factors may play a role in mediating migration of MSCs to the lesion, we performed a PCR array on 84 chemotactic factors 10 days following sham-operation, and at 10 and 17 days post-HI. Our results show that 2610 6 hMSCs decrease lesion volume, improve motor behavior, and reduce scar formation and microglia activity. Moreover, we demonstrate that the differentiation assay reflects the neuroregenerative potential of hMSCs in vivo, as hMSCs induce mNSCs to differentiate into neurons in vitro. We also provide evidence that the chemotactic factor CXCL10 may play an important role in hMSC migration to the lesion site. This is suggested by our finding that CXCL10 is significantly upregulated at 10 days following HI, but not at 17 days after HI, a time when MSCs no longer reach the lesion when given intranasally. The results described in this work also tempt us to contemplate hMSCs not only as a potential treatment option for neonatal encephalopathy, but also for a plethora of degenerative and traumatic injuries of the nervous system.
Journal of Neuroscience Research, 2003
The present study investigates the induction of neurogenesis, reduction of apoptosis, and promotion of basic fibroblast growth factor (bFGF) expression as possible mechanisms by which treatment of stroke with bone marrow stromal cells (MSCs) improves neurological functional recovery. Additionally, for the first time, we treated cerebral ischemia in female rats with intraveneous administration of MSCs. Female rats were subjected to 2 hr of middle cerebral artery occlusion (MCAo), followed by an injection of 3 ϫ 10 6 male (for Y chromosome labeling) rat MSCs or phosphate-buffered saline (PBS) into the tail vein 24 hr after MCAo. All animals received daily injection of bromodeoxyuridine (BrdU; 50 mg/kg, i.p.) for 13 days after treatment for identification of newly synthesized DNA. Animals were sacrificed at 14 days after MCAo. Behavioral tests (rotarod and adhesive-removal tests) were performed. In situ hybridization, immunohistochemistry, and terminal deoxynucleotidyltransferase (TdT)-mediated dUTP-biotin nick-end labeling (TUNEL) were performed to identify transplanted MSCs (Y chromosome), BrdU, bFGF, and apoptotic cells in the brain. Significant recovery of behavior was found in MSC-treated rats at 7 days in the somatosensory test and at 14 days in the motor test after MCAo compared with control, PBS-treated animals (P Ͻ .05). MSCs were found to survive and preferentially localize to the ipsilateral ischemic hemisphere. Significantly more BrdU-positive cells were located in the subventricular zone (P Ͻ .05), and significantly fewer apoptotic cells and more bFGF immunoreactive cell were found in the ischemic boundary area (P Ͻ .05) of MSCtreated rats than in PBS-treated animals. Here we demonstrate that intravenously administered male MSCs increase bFGF expression, reduce apoptosis, promote endogenous cellular proliferation, and improve functional recovery after stroke in female rats.
Human marrow stromal cell therapy for stroke in rat: neurotrophins and functional recovery
2002
Objective: To test the effect of IV-injected human bone marrow stromal cells (hMSC) on neurologic functional deficits after stroke in rats. Methods: Rats were subjected to transient middle cerebral artery occlusion and IV injected with 3 ϫ 10 6 hMSC 1 day after stroke. Functional outcome was measured before and 1, 7, and 14 days after stroke. Mixed lymphocyte reaction and the development of cytotoxic T lymphocytes measured the immune rejection of hMSC. A monoclonal antibody specific to human cellular nuclei (mAb1281) was used to identify hMSC and to measure neural phenotype. ELISA analyzed neurotrophin levels in cerebral tissue from hMSC-treated or nontreated rats. Bromodeoxyuridine injections were used to identify newly formed cells. Results: Significant recovery of function was found in rats treated with hMSC at 14 days compared with control rats with ischemia. Few (1 to 5%) hMSC expressed proteins phenotypic of brain parenchymal cells. Brain-derived neurotrophic factor and nerve growth factor significantly increased, and apoptotic cells significantly decreased in the ischemic boundary zone; significantly more bromodeoxyuridine-reactive cells were detected in the subventricular zone of the ischemic hemisphere of rats treated with hMSC. hMSC induced proliferation of lymphocytes without the induction of cytotoxic T lymphocytes. Conclusion: Neurologic benefit resulting from hMSC treatment of stroke in rats may derive from the increase of growth factors in the ischemic tissue, the reduction of apoptosis in the penumbral zone of the lesion, and the proliferation of endogenous cells in the subventricular zone. NEUROLOGY 2002;59:514 -523 Bone marrow stromal cells (MSC; also referred to as mesenchymal stem and progenitor cells) are multipotent and capable of aiding the repair of tissues in vitro and in vivo. 1 MSC normally give rise to bone, cartilage, and mesenchymal cells, 2 and MSC can differentiate into myocytes, hepatocytes, glial cells, and neurons. 3-6 MSC can pass through the blood-brain barrier and migrate throughout forebrain and cere-
Journal of The Neurological Sciences, 2001
. We tested the hypothesis that bone marrow stromal cells MSCs transplanted into the ischemic boundary zone, survive, differentiate Ž . and improve functional recovery after middle cerebral artery occlusion MCAo . MSCs were harvested from adult rats and cultured with Ž . Ž . Ž . or without nerve growth factor NGF . For cellular identification, MSCs were prelabeled with bromodeoxyuridine BrdU . Rats n s 24 Ž . were subjected to 2 h of MCAo, received grafts at 24 h and were euthanized at 14 days after MCAo. Test groups consisted of: 1 Ž . Ž . Ž . Ž . control-MCAo alone n s 8 ; 2 intracerebral transplantation of MSCs n s 8 ; 3 intracerebral transplantation of MSCs cultured with Ž . Ž NGF n s 8 . Immunohistochemistry was used to identify cells from MSCs. Behavioral tests rotarod, adhesive-removal and modified w x. neurological severity score NSS were performed before and after MCAo. The data demonstrate that MSCs survive, migrate and Ž . Ž . differentiate into phenotypic neural cells. Significant recovery of somatosensory behavior p -0.05 and NSS p -0.05 were found in animals transplanted with MSCs compared with control animals. Animals that received MSCs cultured with NGF displayed significant Ž . Ž . Ž . recovery in motor p -0.05 , somatosensory p -0.05 and NSS p -0.05 behavioral tests compared with control animals. Our data suggest that intracerebral transplantation of MSCs may provide a powerful autoplastic therapy for stroke. q
2009
Aims: Previous studies on the therapeutic time window for intravascular administration of bone marrow stem cells (BMSCs) after stroke have shown that early intervention (from 3 h after onset) in the middle cerebral artery occlusion (MCAO) rat model is the most effective approach to reduce ischaemic lesion size. We have confirmed these observations but noticed that 2 weeks after transplantation, almost none of the grafted BMSCs could be detected in or around the lesion. The present experiments aimed to assess the fate and kinetics of intravascularly injected BMSCs shortly after administration in correlation to the development of the ischaemic lesion after MCAO. Methods: We administered a syngeneic suspension of complete (haematopoietic and mesenchymal) BMSCs via the carotid artery to rats at 2 h after MCAO onset. We examined the distribution and tissue location of BMSCs within the first 24 h after arterial administration by perfusion-fixating rats and performing immunohistochemical analysis at different time points. Results: The vast majority (>95%) of BMSCs appeared to become trapped in the spleen shortly after injection. Six hours after implantation, together with the appearance of activated microglia, the first BMSCs could be detected in and around the lesion; their number gradually increased during the first 12 h after implantation but started to decrease at 24 h. The implanted BMSCs were surrounded by activated and phagocytotic microglia. Conclusion: Our results show that ischaemic lesion size reduction can already be achieved by the early transient presence at the lesion site of intravascularly implanted BMSCs, possibly mediated via activated microglia.