Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Transplant ; 17(7): 1729-1741, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28035752

ABSTRACT

The emergence of skin-containing vascularized composite allografts (VCAs) has provided impetus to understand factors affecting rejection and tolerance of skin. VCA tolerance can be established in miniature swine across haploidentical MHC barriers using mixed chimerism. Because the deceased donor pool for VCAs does not permit MHC antigen matching, clinical VCAs are transplanted across varying MHC disparities. We investigated whether sharing of MHC class I or II antigens between donors and recipients influences VCA skin tolerance. Miniature swine were conditioned nonmyeloablatively and received hematopoietic stem cell transplants and VCAs across MHC class I (n = 3) or class II (n = 3) barriers. In vitro immune responsiveness was assessed, and VCA skin-resident leukocytes were characterized by flow cytometry. Stable mixed chimerism was established in all animals. MHC class II-mismatched chimeras were tolerant of VCAs. MHC class I-mismatched animals, however, rejected VCA skin, characterized by infiltration of recipient-type CD8+ lymphocytes. Systemic donor-specific nonresponsiveness was maintained, including after VCA rejection. This study shows that MHC antigen matching influences VCA skin rejection and suggests that local regulation of immune tolerance is critical in long-term acceptance of all VCA components. These results help elucidate novel mechanisms underlying skin tolerance and identify clinically relevant VCA tolerance strategies.


Subject(s)
Composite Tissue Allografts/transplantation , Graft Rejection/prevention & control , Major Histocompatibility Complex/immunology , Skin Transplantation/adverse effects , Transplantation Chimera/immunology , Transplantation Tolerance/immunology , Vascularized Composite Allotransplantation/adverse effects , Animals , Composite Tissue Allografts/immunology , Composite Tissue Allografts/pathology , Graft Rejection/etiology , Graft Survival/immunology , Isoantibodies/blood , Isoantibodies/immunology , Swine , Swine, Miniature
2.
Am J Transplant ; 14(12): 2713-22, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25278264

ABSTRACT

Mixed chimerism approaches for induction of tolerance of solid organ transplants have been applied successfully in animal models and in the clinic. However, in xenogeneic models (pig-to-primate), host macrophages participate in the rapid clearance of porcine hematopoietic progenitor cells, hindering the ability to achieve mixed chimerism. CD47 is a cell-surface molecule that interacts in a species-specific manner with SIRPα receptors on macrophages to inhibit phagocytosis and expression of human CD47 (hCD47) on porcine cells has been shown to inhibit phagocytosis by primate macrophages. We report here the generation of hCD47 transgenic GalT-KO miniature swine that express hCD47 in all blood cell lineages. The effect of hCD47 expression on xenogeneic hematopoietic engraftment was tested in an in vivo mouse model of human hematopoietic cell engraftment. High-level porcine chimerism was observed in the bone marrow of hCD47 progenitor cell recipients and smaller but readily measurable chimerism levels were observed in the peripheral blood of these recipients. In contrast, transplantation of WT progenitor cells resulted in little or no bone marrow engraftment and no detectable peripheral chimerism. These results demonstrate a substantial protective effect of hCD47 expression on engraftment and persistence of porcine cells in this model, presumably by modulation of macrophage phagocytosis.


Subject(s)
Bone Marrow/immunology , CD47 Antigen/immunology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Immune Tolerance/immunology , Transplantation Chimera/immunology , Animals , Animals, Genetically Modified , CD47 Antigen/metabolism , Chimerism , Galactosyltransferases/genetics , Gene Knockout Techniques , Graft Survival/immunology , Humans , Macrophages/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Phagocytosis/physiology , Swine , Swine, Miniature , Transplantation Conditioning , Transplantation, Heterologous
3.
Am J Transplant ; 14(2): 343-55, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24405666

ABSTRACT

Vascularized composite allograft (VCA) transplantation can restore form and function following severe craniofacial injuries, extremity amputations or massive tissue loss. The induction of transplant tolerance would eliminate the need for long-term immunosuppression, realigning the risk-benefit ratio for these life-enhancing procedures. Skin, a critical component of VCA, has consistently presented the most stringent challenge to transplant tolerance. Here, we demonstrate, in a clinically relevant miniature swine model, induction of immunologic tolerance of VCAs across MHC barriers by induction of stable hematopoietic mixed chimerism. Recipient conditioning consisted of T cell depletion with CD3-immunotoxin, and 100 cGy total body irradiation prior to hematopoietic cell transplantation (HCT) and a 45-day course of cyclosporine A. VCA transplantation was performed either simultaneously to induction of mixed chimerism or into established mixed chimeras 85-150 days later. Following withdrawal of immunosuppression both VCAs transplanted into stable chimeras (n=4), and those transplanted at the time of HCT (n=2) accepted all components, including skin, without evidence of rejection to the experimental end point 115-504 days posttransplant. These data demonstrate that tolerance across MHC mismatches can be induced in a clinically relevant VCA model, providing proof of concept for long-term immunosuppression-free survival.


Subject(s)
Composite Tissue Allografts/immunology , Graft Rejection/immunology , Graft Survival/immunology , Hematopoietic Stem Cell Transplantation , Major Histocompatibility Complex/immunology , Vascularized Composite Allotransplantation , Animals , Composite Tissue Allografts/pathology , Histocompatibility , Immunoenzyme Techniques , Immunosuppressive Agents/therapeutic use , Lymphocyte Culture Test, Mixed , Swine , Swine, Miniature , T-Lymphocytes, Regulatory/immunology , Transplantation Chimera/immunology , Transplantation Tolerance/immunology
4.
Dev Neurosci ; 35(2-3): 130-9, 2013.
Article in English | MEDLINE | ID: mdl-23548575

ABSTRACT

Toll-like receptors (TLRs) are members of the pattern recognition receptor family that detect components of foreign pathogens or endogenous molecules released in response to injury. Recent studies demonstrate that TLRs also have a functional role in regulating neuronal proliferation in the developing brain. This study investigated cellular expression of TLR3 using immunohistochemistry on human brain tissue. The tissue sections analysed contained anterior and lateral periventricular white matter from the frontal and parietal lobes in post-mortem neonatal cases with a postmenstrual age range of 23.6-31.4 weeks. In addition to preterm brains without overt pathology (control), preterm pathology cases with evidence of white matter injuries (WMI) were also examined. In order to identify TLR-positive cells, we utilized standard double-labelling immunofluorescence co-labelling techniques and confocal microscopy to compare co-expression of TLR3 with a neuronal marker (NeuN) or with glial markers (GFAP for astrocytes, Iba-1 for microglia and Olig2 for oligodendrocytes). We observed an increase in the neuronal (28 vs. 17%) and astroglial (38 vs. 21%) populations in the WMI group compared to controls in the anterior regions of the periventricular white matter in the frontal lobe. The increase in neurons and astrocytes in the WMI cases was associated with an increase in TLR3 immunoreactivity. This expression was significantly increased in the astroglia. The morphology of the TLR3 signal in the control cases was globular and restricted to the perinuclear region of the neurons and astrocytes, whilst in the cases of WMI, both neuronal, axonal and astroglial TLR3 expression was more diffuse (i.e., a different intracellular distribution) and could be detected along the extensions of the processes. This study demonstrates for the first time that neurons and glial cells in human neonatal periventricular white matter express TLR3 during development. The patterns of TLR3 expression were altered in the presence of WMI, which might influence normal developmental processes within the immature brain. Identifying changes in TLR3 expression during fetal development may be key to understanding the reduced volumes of grey matter and impaired cortical development seen in preterm infants.


Subject(s)
Brain/metabolism , Infant, Extremely Premature/metabolism , Neuroglia/metabolism , Neurons/metabolism , Toll-Like Receptor 3/biosynthesis , Brain/growth & development , Brain/pathology , Brain Injuries/metabolism , Humans , Infant, Extremely Premature/growth & development , Infant, Newborn , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Toll-Like Receptor 3/analysis
5.
Brain Behav Immun ; 24(5): 776-83, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19903519

ABSTRACT

Although the role of microglial activation in neural injury remains controversial, there is increasing evidence for a detrimental effect in the immature brain, which may occur in response to release of neurotoxic substances including pro-inflammatory cytokines. However, the signaling mechanisms involved in microglial-induced neuronal cell death are unclear. Microglia isolated from the brains of wild-type (WT) or MyD88 knockout (KO) mice were exposed to PBS or the TLR4-ligand LPS (100 ng/mL) for 2, 6, 14, or 24 h, and the microglia-conditioned medium (MCM) collected. Detection of multiple inflammatory molecules in MCM was performed using a mouse 22-plex cytokine microbead array kit. Primary neuronal cultures were supplemented with the 14 or 24 h MCM, and the degree of neuronal apoptosis examined after exposure for 24 h. Results showed a rapid and sustained elevation in multiple inflammatory mediators in the MCM of WT microglia exposed to LPS, which was largely inhibited in MyD88 KO microglia. There was a significant increase in apoptotic death measured at 24 h in cultured neurons exposed to CM from either 14 or 24 h LPS-stimulated WT microglia (p<.05 vs. WT control). By contrast, there was no increase in apoptotic death in cultured neurons exposed to CM from 14 or 24 h LPS-stimulated MyD88 KO microglia (p=.15 vs. MyD88 KO control). These data suggest that MyD88-dependent activation of microglia by LPS causes release of factors directly toxic to neurons.


Subject(s)
Cytokines/metabolism , Microglia/metabolism , Myeloid Differentiation Factor 88/metabolism , Neurons/metabolism , Analysis of Variance , Animals , Apoptosis/drug effects , Apoptosis/immunology , Brain/immunology , Brain/metabolism , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Microglia/drug effects , Microglia/immunology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Neurons/drug effects , Neurons/immunology , Signal Transduction/drug effects , Signal Transduction/immunology
6.
Burns ; 43(8): 1717-1724, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28602591

ABSTRACT

Burn is associated with a considerable burden of morbidity worldwide. Early excision of burned tissue and skin grafting of the resultant wound has been established as a mainstay of modern burn therapy. However, in large burns, donor sites for autologous skin may be limited. Numerous alternatives, from cadaver skin to synthetic substitutes have been described, each with varying benefits and limitations. We previously proposed the use of genetically modified (alpha-1,3-galactosyl transferase knockout, GalT-KO) porcine skin as a viable skin alternative. In contrast to wild type porcine skin, which has been used as a biologic dressing following glutaraldehyde fixation, GalT-KO porcine skin is a viable graft, which is not susceptible to loss by hyperacute rejection, and undergoes graft take and healing, prior to eventual rejection, comparable to cadaver allogeneic skin. In the current study we aimed to perform a detailed functional analysis of GalT-KO skin grafts in comparison to allogeneic grafts for temporary closure of full thickness wounds using our baboon dorsum wound model. Grafts were assessed by measurement of fluid loss, wound infection rate, and take, and healed appearance, of secondary autologous grafts following xenograft rejection. Comparison was also made between fresh and cryopreserved grafts. No statistically significant difference was identified between GalT-KO and allogeneic skin grafts in any of the assessed parameters, and graft take and function was not adversely effected by the freeze-thaw process. These data demonstrate that GalT-KO porcine grafts are functionally comparable to allogeneic skin grafts for temporary closure of full thickness wounds, and support their consideration as an alternative to cadaver allogeneic skin in the emergency management of large burns.


Subject(s)
Burns/surgery , Galactosyltransferases/genetics , Skin Transplantation/methods , Animals , Animals, Genetically Modified , Disease Models, Animal , Papio , Skin/pathology , Swine , Swine, Miniature , Transplantation, Heterologous , Wound Healing/physiology , Wound Infection
7.
Placenta ; 27(2-3): 322-6, 2006.
Article in English | MEDLINE | ID: mdl-16338476

ABSTRACT

Toll-like receptor 4 (TLR-4) mediates Gram-negative bacterial-induced inflammatory responses, including production of pro-inflammatory cytokines. Maternal infection and inflammation play an important role in preterm birth and neonatal brain damage. The localization of placental TLR-4 as well as changes during normal gestation are critical issues in understanding the role of toll-like receptors in defending the placento-fetal unit from maternal infection. We therefore investigated, by immunohistochemistry (IHC) and Western blot, the subcellular localization of TLR-4 in first trimester and term human placenta. In both term placenta (n=4) and first trimester placenta villous samples (n=5), immunoreactivity for TLR-4 was found in the cytoplasm of the syncytiotrophoblast, with darker staining in some areas of the maternal facing plasma membrane (MVM). In addition, TLR-4 was found to be expressed in the first trimester cytotrophoblast cells. Using Western blot analysis, TLR-4 was identified in both placental homogenates and isolated MVM and the fetal facing basal membrane (BM). TLR-4 expression in MVM was significantly higher in term (n=9) as compared to first trimester (n=2) samples. We have shown for the first time that the subcellular localization of TLR-4 in term placenta is preferentially in the MVM compared to BM. The MVM is continuously bathed in maternal blood, suggesting that from this vantage point TLR-4 can initiate a rapid response to maternal bacterial infection.


Subject(s)
Cell Membrane/chemistry , Placenta/metabolism , Pregnancy Trimester, First/metabolism , Toll-Like Receptor 4/analysis , Toll-Like Receptor 4/metabolism , Female , Humans , Placenta/chemistry , Placenta/cytology , Pregnancy
8.
Acta Physiol (Oxf) ; 214(4): 481-96, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25912364

ABSTRACT

AIM: Bestrophins are putative calcium-activated chloride channels. Recently, cell-protective functions for Bestrophin-3 (Best3) were proposed. Best3 exists in different splice variants. We have here examined expression, alternative splicing and localization of Best3 in mouse podocytes under normal conditions and during endoplasmic reticulum (ER) stress. METHODS: Best3 expression was determined on the mRNA level using quantitative PCR and on the protein level by immunohistochemistry and Western blotting. RESULTS: Staining for Best3 was pronounced in glomeruli and was detected in cultured mouse podocytes. Best3 did not co-localize with markers for endothelial cells (CD31), podocyte foot processes (synaptopodin) or microtubules (actin). However, immunogold-based electron microscopy and co-localization with nestin showed Best3 presence in podocyte primary processes and cell bodies. Only two splice variants of Best3 mRNA (both lacking exons 2 and 3, and one also lacking exon 6), but no full-length variant, were detected. ER stress induced by lipopolysaccharides in vivo transiently elevated mRNA levels of total Best3 and its two splice variants with different time courses. In cultured podocytes under ER stress induced by thapsigargin, the expression of total Best3, its splice variants and nestin transiently increased with similar time courses. The ER stress marker C/EBP homologous protein (CHOP) and nestin mRNA increased during ER stress in vivo and in vitro. CONCLUSIONS: Best3 is localized intracellularly in cell bodies and primary processes of mouse podocytes and is co-localized with nestin. Two splice variants of Best3 are expressed in glomeruli and in cultured podocytes, and their expression is differentially regulated in ER stress.


Subject(s)
Eye Proteins/biosynthesis , Podocytes/metabolism , Animals , Blotting, Western , Cells, Cultured , Endoplasmic Reticulum Stress/physiology , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Protein Isoforms , RNA, Messenger , Reverse Transcriptase Polymerase Chain Reaction
9.
Int J Dev Neurosci ; 45: 44-54, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25702527

ABSTRACT

Preterm born infants have high rates of brain injury, leading to motor and neurocognitive problems in later life. Infection and resulting inflammation of the fetus and newborn are highly associated with these disabilities. However, there are no established neuroprotective therapies. Microglial activation and expression of many cytokines play a key role in normal brain function and development, as well as being deleterious. Thus, treatment must achieve a delicate balance between possible beneficial and harmful effects. In this review, we discuss potential neuroprotective strategies targeting systemic infection or the resulting systemic and central inflammatory responses. We highlight the central importance of timing of treatment and the critical lack of studies of delayed treatment of infection/inflammation.


Subject(s)
Brain Injuries/prevention & control , Brain Injuries/physiopathology , Central Nervous System Infections/prevention & control , Central Nervous System Infections/physiopathology , Encephalitis/prevention & control , Encephalitis/physiopathology , Brain/physiopathology , Brain Injuries/diagnosis , Central Nervous System Infections/diagnosis , Encephalitis/diagnosis , Evidence-Based Medicine , Female , Humans , Infant, Newborn , Male , Neuroprotective Agents/therapeutic use , Treatment Outcome
10.
J Neuropathol Exp Neurol ; 58(3): 255-64, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10197817

ABSTRACT

The cholinergic neurotransmitter system is vital for several brainstem functions including cardiorespiratory control and central chemosensitivity. This study has examined aspects of the cholinergic neurotransmitter system in the brainstem of sudden infant death syndrome (SIDS) and control infants. The cellular localisation and the optical density of the immunoreactivity of the cholinergic enzyme choline acetyltransferase (CHAT-IR) and the muscarinic acetylcholine receptor m2 (m2-IR) in the medulla was described in 14 SIDS and 9 control cases. There was a reduction in the number of CHAT-IR neurons in the hypoglossal nucleus (control: 71.2+/-8.3% vs SIDS: 46.1+/-5.3%) and the dorsal motor nucleus of the vagus (DMV) (control: 77.2+/-5.0% vs SIDS: 52.5+/-7.4%) and reduced optical density of CHAT-IR in the hypoglossal nucleus (control: 0.20+/-0.01 vs SIDS; 0.14+/-0.02) in SIDS infants. In contrast there were no changes in the optical density of m2-IR in the hypoglossal nucleus, the DMV, or the arcuate nucleus. Hypoplasia of the arcuate nucleus was observed in one SIDS infant. These results suggest that there is a specific defect in some cholinergic motor neurons in the medulla of SIDS infants. This could lead to abnormal control of cardiovascular and respiratory function and airway patency and may be one of the contributing factors in the etiology of SIDS.


Subject(s)
Brain Stem/chemistry , Choline O-Acetyltransferase/analysis , Receptors, Muscarinic/analysis , Sudden Infant Death , Brain Stem/enzymology , Humans , Immunohistochemistry , Infant , Infant, Newborn , Postmortem Changes
11.
J Cereb Blood Flow Metab ; 13(4): 609-16, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8314914

ABSTRACT

Intraventricular injection of insulin-like growth factor 1 (IGF-1) 2 h after hypoxic-ischemic injury reduces neuronal loss. To clarify the mode of action, we compared histological outcome between treatment groups in the following three studies: 0, 0.5, 5, and 50 micrograms IGF-1 given 2 h after injury; 0 and 20 micrograms IGF-1 given 1 h before; and 20 micrograms IGF-1 and insulin or vehicle alone given 2 h after. Unilateral hypoxic-ischemic injury was induced in adult rats by ligation of the right carotid and exposure to 6% O2 for 10 min. Histological outcome was evaluated in the cortex, striatum, and hippocampus 5 days later. Five to 50 micrograms IGF-1 reduced the incidence of infarction and neuronal loss in a dose-dependent manner in all regions (p < 0.05), and 50 micrograms reduced the infarction rate from 87 to 26% (p < 0.01). Pretreatment did not alter outcome. IGF-1 improved outcome compared with equimolar doses of insulin (p < 0.05) and did not affect systemic glucose concentrations or cortical temperature. The results indicate that the neuronal protective effects of IGF-1 are specific and are not mediated via insulin receptors, hypothermia, or hypoglycemic mechanisms. Centrally administered IGF-1 appears to provide worthwhile trophic support to cells within most cerebral structures after transient hypoxic-ischemic injury.


Subject(s)
Brain Ischemia/pathology , Hypoxia, Brain/pathology , Somatomedins/pharmacology , Animals , Body Temperature/drug effects , Brain/drug effects , Brain/physiopathology , Brain Ischemia/complications , Brain Ischemia/physiopathology , Cerebral Infarction/etiology , Cerebral Infarction/prevention & control , Dose-Response Relationship, Drug , Hypoxia, Brain/complications , Hypoxia, Brain/physiopathology , Male , Rats , Rats, Wistar , Somatomedins/administration & dosage , Time Factors
12.
Neuroscience ; 100(2): 327-33, 2000.
Article in English | MEDLINE | ID: mdl-11008170

ABSTRACT

Intrauterine growth restriction is a risk factor for neurological and behavioural deficits in children although the precise underlying biological correlate for this is unclear. The present study shows that animals with intrauterine growth restriction, induced by a period of reduced placental blood flow during the second half of pregnancy, demonstrate reduced numbers of neurons in the hippocampus and the cerebellum in conjunction with retarded dendritic and axonal growth within these structures. Intrauterine growth restriction was induced at 30 days gestational age by unilateral uterine artery ligation in pregnant guinea-pigs. At one week of age, the total number of CA1 pyramidal neurons in the hippocampus and the Purkinje neurons in the cerebellum were determined using the combined fractionator/optical disector technique. The Cavalieri Principle was used to determine the volume of specific regions within the hippocampus and cerebellum. The body weight of animals that were classified as intrauterine growth-restricted was reduced by 42% (n=8) compared with control animals (n=8, P<0.001), while there was a smaller effect on brain weight (16% reduction, P<0.01). Estimates of the total number of neurons showed a reduction in CA1 pyramidal neurons in growth-restricted animals (4.19+/-0.43x10(5)) compared with control (5.20+/-0.44x10(5), P<0.01), and the volume of the stratum oriens layer above the CA1 region, which contains the apical dendrites of the CA1 pyramidal neurons, was reduced by 21% (P<0.01) in growth-restricted animals. In the cerebellum there was a reduction in the number of Purkinje neurons in growth-restricted animals (3.97+/-0.50x10(5)) compared with control (5.13+/-0.52x10(5), P<0.01), and in the volume of the molecular layer (17%, P<0.05), the internal granular layer (22%, P<0.01) and in the volume of the cerebellar white matter (23%, P<0.01). These results show that a period of placental insufficiency during the second half of pregnancy can effect brain development in a way which could lead to neurological and behavioural deficits in the postnatal animal.


Subject(s)
Cerebellum/pathology , Fetal Growth Retardation/pathology , Hippocampus/pathology , Neurons/cytology , Purkinje Cells/pathology , Pyramidal Cells/pathology , Animals , Animals, Newborn , Cell Count , Cerebellum/embryology , Cerebellum/growth & development , Female , Guinea Pigs , Hippocampus/embryology , Hippocampus/growth & development , Pregnancy
13.
Neurosci Lett ; 332(1): 57-60, 2002 Oct 25.
Article in English | MEDLINE | ID: mdl-12377384

ABSTRACT

The induction of the p75 neurotrophin receptor (p75NTR) on striatal cholinergic neurons by global hypoxic-ischemia has been reported to promote neuron survival. We have found, however, while the p75NTR-expressing neurons survive the insult for the first 5 days, subsequently they undergo shrinkage, loss of choline acetyl transferase (ChAT) expression, and more than 96% are eventually lost by 8 days. In contrast ChAT-expressing cells in the surrounding region of the infarction, do not express p75NTR and there is no evidence of neuronal loss. These results suggest the expression of p75NTR on cholinergic interneurons of the rat striatum is associated with delayed neuronal degeneration.


Subject(s)
Brain Ischemia/metabolism , Cholinergic Fibers/metabolism , Corpus Striatum/metabolism , Nerve Degeneration/metabolism , Receptors, Nerve Growth Factor/biosynthesis , Animals , Brain Ischemia/pathology , Corpus Striatum/pathology , Nerve Degeneration/pathology , Rats , Rats, Wistar , Receptor, Nerve Growth Factor
14.
Clin Perinatol ; 20(2): 305-25, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8358953

ABSTRACT

Following a severe asphyxial episode many cells can recover metabolically, and a cascade of processes are triggered in which intervention, even some hours later, can allow rescue of some cells that would otherwise die. A number of principles, however, needs to be carefully considered before extrapolating from animal to human trials. In particular, the effects on long-term outcome and on those who are compromised by intrauterine growth retardation need to be determined. It is critical to be able to identify rapidly those infants in terms of nature and severity of injury who are most likely to benefit from treatment. The dimension of time and phase of injury or recovery are key factors to effective intervention. Novel continuous cerebral function monitoring techniques such as those based on real-time spectral analysis of the EEG activity, cortical impedance monitoring, and near-infrared spectroscopy have considerable potential for determining the severity and pathophysiologic phase of injury on line.


Subject(s)
Asphyxia Neonatorum/physiopathology , Adaptation, Physiological , Asphyxia Neonatorum/complications , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/therapy , Brain Diseases/epidemiology , Brain Diseases/etiology , Brain Diseases/pathology , Free Radicals , Gestational Age , Growth Substances/therapeutic use , Humans , Infant, Newborn , Monitoring, Physiologic , Prognosis , Reactive Oxygen Species , Reperfusion Injury/etiology , Reperfusion Injury/pathology
15.
Neuroscience ; 253: 21-8, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-23994184

ABSTRACT

Experimental studies show that inflammation reduces the regenerative capacity in the adult brain. Less is known about how early postnatal inflammation affects neurogenesis, stem cell proliferation, cell survival and learning and memory in young adulthood. In this study we examined if an early-life inflammatory challenge alters cell proliferation and survival in distinct anatomical regions of the hippocampus and whether learning and memory were affected. Lipopolysaccharide (LPS, 1mg/kg) was administered to mice on postnatal day (P) 9 and proliferation and survival of hippocampal cells born either prior to (24h before LPS), or during the inflammatory insult (48 h after LPS) was evaluated. Long-term cell survival of neurons and astrocytes was determined on P 41 and P 60 in the dorsal and ventral horns of the hippocampus. On day 50 the mice were tested in the trace fear conditioning (TFC) paradigm. There was no effect on the survival of neurons and astrocytes that were born before LPS injection. In contrast, the number of neurons and astrocytes that were born after LPS injection were reduced on P 41. The LPS-induced reduction in cell numbers was specific for the dorsal hippocampus. Neither early (48 h after LPS) or late (33 days after LPS) proliferation of cells was affected by neonatal inflammation and neonatal LPS did not alter the behavior of young adult mice in the TFC test. These data highlight that neonatal inflammation specifically affects survival of dividing neurons and astrocytes, but not post-mitotic cells. The reduction in cell survival could be attributed to less cell survival in the dorsal hippocampus, but had no effect on learning and memory in the young adult.


Subject(s)
Cell Proliferation/drug effects , Hippocampus/drug effects , Lipopolysaccharides/toxicity , Neurons/drug effects , Stem Cells/drug effects , Age Factors , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Survival/drug effects , Doublecortin Domain Proteins , Doublecortin Protein , Female , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neurogenesis/drug effects , Neurons/physiology , Neuropeptides/metabolism , Stem Cells/physiology
19.
Neuroscience ; 160(3): 606-15, 2009 May 19.
Article in English | MEDLINE | ID: mdl-19285118

ABSTRACT

Injury to the cerebellum and brainstem is becoming increasingly recognized in prematurely born infants. The role of infection/inflammation in mediating damage to those structures in the preterm brain is largely unknown. Preterm fetal sheep (70% gestation) received either saline-vehicle (control group; n=11) or Escherichia coli lipopolysaccharide (100 ng intravenous [i.v.]; lipopolysaccharide [LPS] group; n=9), and were allowed to recover for 3 days before sacrifice. A diffuse pattern of cerebellar white matter damage was observed in all animals exposed to LPS, while focal cerebellar white matter lesions were observed in three out of nine animals, and an intragyral white matter hemorrhage in one animal. Cerebellar white matter injury was associated with a statistically significant loss of oligodendrocyte transcription factor-2-positive oligodendrocytes and increased terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling-positive cell counts. Ionized calcium binding adapter molecule 1 (Iba1)-positive cells which had the morphology of activated microglia were commonly observed in areas of injury. There was no obvious injury to the cerebellar cortex or to cerebellar Purkinje cells, and no obvious injury in any region of the brainstem. These data provide support for a role of infection/inflammation in selective white matter injury in the immature cerebellum, and demonstrate a differential vulnerability of the brainstem and cerebellar white matter to injury at this time.


Subject(s)
Cerebellar Diseases/etiology , Cerebellum/pathology , Endotoxemia/complications , Fetal Diseases/pathology , Lipopolysaccharides/toxicity , Nerve Fibers, Myelinated/pathology , Animals , Brain Stem/pathology , Cell Death , Cerebellar Cortex/embryology , Cerebellar Cortex/pathology , Cerebellar Diseases/embryology , Cerebellar Diseases/pathology , Cerebellum/embryology , DNA-Binding Proteins/metabolism , Endotoxemia/pathology , Endotoxins/administration & dosage , Escherichia coli , Female , Microglia/metabolism , Microglia/pathology , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Pregnancy , Sheep
20.
Semin Neonatol ; 5(1): 41-51, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10802749

ABSTRACT

Although the aetiology of antenatal brain injury is often unclear, procedures can be employed to prevent or reduce the risk of injury. Defective neuropore closure can be prevented by periconceptional administration of folic acid, and the incidence of other severe malformations and genetic disorders can be reduced by early identification and termination of pregnancy. Antenatal identification of IUGR, administration of corticosteroids to cases with pending preterm birth, and treatment of maternal/fetal infections would also reduce the incidence of injury. Mothers can decrease the risk of injury by maintaining a good diet, avoiding smoking, alcohol intake and exposure to TORCH infections during pregnancy.


Subject(s)
Brain Injuries/congenital , Brain Injuries/prevention & control , Fetal Hypoxia/etiology , Fetal Hypoxia/prevention & control , Prenatal Care/methods , Anti-Inflammatory Agents/therapeutic use , Central Nervous System/abnormalities , Female , Fetal Alcohol Spectrum Disorders/complications , Fetal Growth Retardation/complications , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/prevention & control , Folic Acid/therapeutic use , Humans , Obstetric Labor, Premature/prevention & control , Pregnancy , Pregnancy Complications, Infectious/prevention & control , Risk Factors , Smoking/adverse effects , Steroids
SELECTION OF CITATIONS
SEARCH DETAIL