Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 5501, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34535655

ABSTRACT

Fibrotic scar tissue limits central nervous system regeneration in adult mammals. The extent of fibrotic tissue generation and distribution of stromal cells across different lesions in the brain and spinal cord has not been systematically investigated in mice and humans. Furthermore, it is unknown whether scar-forming stromal cells have the same origin throughout the central nervous system and in different types of lesions. In the current study, we compared fibrotic scarring in human pathological tissue and corresponding mouse models of penetrating and non-penetrating spinal cord injury, traumatic brain injury, ischemic stroke, multiple sclerosis and glioblastoma. We show that the extent and distribution of stromal cells are specific to the type of lesion and, in most cases, similar between mice and humans. Employing in vivo lineage tracing, we report that in all mouse models that develop fibrotic tissue, the primary source of scar-forming fibroblasts is a discrete subset of perivascular cells, termed type A pericytes. Perivascular cells with a type A pericyte marker profile also exist in the human brain and spinal cord. We uncover type A pericyte-derived fibrosis as a conserved mechanism that may be explored as a therapeutic target to improve recovery after central nervous system lesions.


Subject(s)
Central Nervous System/pathology , Cicatrix/pathology , Pericytes/pathology , Aging/physiology , Animals , Astrocytes/pathology , Brain Injuries, Traumatic/pathology , Brain Ischemia/pathology , Brain Neoplasms/pathology , Cerebral Cortex/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Extracellular Matrix/metabolism , Fibroblasts/pathology , Fibrosis , Glioblastoma/pathology , Humans , Ischemic Stroke/pathology , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , Receptor, Platelet-Derived Growth Factor beta/metabolism , Spinal Cord/pathology , Spinal Cord/ultrastructure , Spinal Cord Injuries/pathology , Stromal Cells/pathology
2.
Brain ; 140(3): 692-706, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28115364

ABSTRACT

Transplanted neurons derived from stem cells have been proposed to improve function in animal models of human disease by various mechanisms such as neuronal replacement. However, whether the grafted neurons receive functional synaptic inputs from the recipient's brain and integrate into host neural circuitry is unknown. Here we studied the synaptic inputs from the host brain to grafted cortical neurons derived from human induced pluripotent stem cells after transplantation into stroke-injured rat cerebral cortex. Using the rabies virus-based trans-synaptic tracing method and immunoelectron microscopy, we demonstrate that the grafted neurons receive direct synaptic inputs from neurons in different host brain areas located in a pattern similar to that of neurons projecting to the corresponding endogenous cortical neurons in the intact brain. Electrophysiological in vivo recordings from the cortical implants show that physiological sensory stimuli, i.e. cutaneous stimulation of nose and paw, can activate or inhibit spontaneous activity in grafted neurons, indicating that at least some of the afferent inputs are functional. In agreement, we find using patch-clamp recordings that a portion of grafted neurons respond to photostimulation of virally transfected, channelrhodopsin-2-expressing thalamo-cortical axons in acute brain slices. The present study demonstrates, for the first time, that the host brain regulates the activity of grafted neurons, providing strong evidence that transplanted human induced pluripotent stem cell-derived cortical neurons can become incorporated into injured cortical circuitry. Our findings support the idea that these neurons could contribute to functional recovery in stroke and other conditions causing neuronal loss in cerebral cortex.


Subject(s)
Brain Injuries/surgery , Evoked Potentials, Somatosensory/physiology , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/transplantation , Synapses/physiology , Action Potentials , Afferent Pathways/physiology , Animals , Brain/cytology , Brain/ultrastructure , Brain Injuries/etiology , Cell Line, Transformed , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/ultrastructure , Disease Models, Animal , Humans , Lysine/analogs & derivatives , Lysine/metabolism , Male , Neurons/physiology , Neurons/ultrastructure , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Nude , Rats, Sprague-Dawley , Stroke/complications , Synapses/ultrastructure , Ventral Thalamic Nuclei/cytology
3.
J Neurosci ; 36(15): 4182-95, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-27076418

ABSTRACT

Stroke is a leading cause of disability and currently lacks effective therapy enabling long-term functional recovery. Ischemic brain injury causes local inflammation, which involves both activated resident microglia and infiltrating immune cells, including monocytes. Monocyte-derived macrophages (MDMs) exhibit a high degree of functional plasticity. Here, we determined the role of MDMs in long-term spontaneous functional recovery after middle cerebral artery occlusion in mice. Analyses by flow cytometry and immunocytochemistry revealed that monocytes home to the stroke-injured hemisphere., and that infiltration peaks 3 d after stroke. At day 7, half of the infiltrating MDMs exhibited a bias toward a proinflammatory phenotype and the other half toward an anti-inflammatory phenotype, but during the subsequent 2 weeks, MDMs with an anti-inflammatory phenotype dominated. Blocking monocyte recruitment using the anti-CCR2 antibody MC-21 during the first week after stroke abolished long-term behavioral recovery, as determined in corridor and staircase tests, and drastically decreased tissue expression of anti-inflammatory genes, including TGFß, CD163, and Ym1. Our results show that spontaneously recruited monocytes to the injured brain early after the insult contribute to long-term functional recovery after stroke. SIGNIFICANCE STATEMENT: For decades, any involvement of circulating immune cells in CNS repair was completely denied. Only over the past few years has involvement of monocyte-derived macrophages (MDMs) in CNS repair received appreciation. We show here, for the first time, that MDMs recruited to the injured brain early after ischemic stroke contribute to long-term spontaneous functional recovery through inflammation-resolving activity. Our data raise the possibility that inadequate recruitment of MDMs to the brain after stroke underlies the incomplete functional recovery seen in patients and that boosting homing of MDMs with an anti-inflammatory bias to the injured brain tissue may be a new therapeutic approach to promote long-term improvement after stroke.


Subject(s)
Macrophages , Monocytes , Recovery of Function , Stroke/physiopathology , Animals , Antibodies, Blocking/pharmacology , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/biosynthesis , Antigens, Differentiation, Myelomonocytic/genetics , Behavior, Animal/drug effects , Chimera , Functional Laterality , Infarction, Middle Cerebral Artery/physiopathology , Inflammation/pathology , Lectins/biosynthesis , Lectins/genetics , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Monocytes/pathology , Neuronal Plasticity/physiology , Psychomotor Performance/drug effects , Receptors, CCR2/antagonists & inhibitors , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Recovery of Function/drug effects , Stroke/pathology , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , beta-N-Acetylhexosaminidases/biosynthesis , beta-N-Acetylhexosaminidases/genetics
4.
Neurobiol Dis ; 83: 1-15, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26299391

ABSTRACT

Ischemic stroke triggers neurogenesis from neural stem/progenitor cells (NSPCs) in the subventricular zone (SVZ) and migration of newly formed neuroblasts toward the damaged striatum where they differentiate to mature neurons. Whether it is the injury per se or the associated inflammation that gives rise to this endogenous neurogenic response is unknown. Here we showed that inflammation without corresponding neuronal loss caused by intrastriatal lipopolysaccharide (LPS) injection leads to striatal neurogenesis in rats comparable to that after a 30 min middle cerebral artery occlusion, as characterized by striatal DCX+ neuroblast recruitment and mature NeuN+/BrdU+ neuron formation. Using global gene expression analysis, changes in several factors that could potentially regulate striatal neurogenesis were identified in microglia sorted from SVZ and striatum of LPS-injected and stroke-subjected rats. Among the upregulated factors, one chemokine, CXCL13, was found to promote neuroblast migration from neonatal mouse SVZ explants in vitro. However, neuroblast migration to the striatum was not affected in constitutive CXCL13 receptor CXCR5(-/-) mice subjected to stroke. Infarct volume and pro-inflammatory M1 microglia/macrophage density were increased in CXCR5(-/-) mice, suggesting that microglia-derived CXCL13, acting through CXCR5, might be involved in neuroprotection following stroke. Our findings raise the possibility that the inflammation accompanying an ischemic insult is the major inducer of striatal neurogenesis after stroke.


Subject(s)
Corpus Striatum/physiopathology , Encephalitis/physiopathology , Infarction, Middle Cerebral Artery/physiopathology , Neural Stem Cells/physiology , Neurogenesis , Neurons/physiology , Stroke/physiopathology , Animals , Cell Death , Cell Movement/drug effects , Chemokine CXCL13/pharmacology , Chemokine CXCL13/physiology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Doublecortin Protein , Encephalitis/chemically induced , Encephalitis/metabolism , Gene Expression , Infarction, Middle Cerebral Artery/pathology , Inflammation Mediators/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Lateral Ventricles/physiopathology , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/metabolism , Neurons/pathology , Rats , Rats, Wistar , Receptors, CXCR5/genetics , Receptors, CXCR5/physiology , Stroke/pathology
5.
Science ; 346(6206): 237-41, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25301628

ABSTRACT

Neurogenesis is restricted in the adult mammalian brain; most neurons are neither exchanged during normal life nor replaced in pathological situations. We report that stroke elicits a latent neurogenic program in striatal astrocytes in mice. Notch1 signaling is reduced in astrocytes after stroke, and attenuated Notch1 signaling is necessary for neurogenesis by striatal astrocytes. Blocking Notch signaling triggers astrocytes in the striatum and the medial cortex to enter a neurogenic program, even in the absence of stroke, resulting in 850 ± 210 (mean ± SEM) new neurons in a mouse striatum. Thus, under Notch signaling regulation, astrocytes in the adult mouse brain parenchyma carry a latent neurogenic program that may potentially be useful for neuronal replacement strategies.


Subject(s)
Astrocytes/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Receptor, Notch1/physiology , Signal Transduction , Stroke/physiopathology , Animals , Astrocytes/cytology , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Gene Deletion , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurons/cytology , Receptor, Notch1/genetics , Stroke/pathology
6.
Restor Neurol Neurosci ; 32(4): 547-58, 2014.
Article in English | MEDLINE | ID: mdl-24916776

ABSTRACT

PURPOSE: Induced pluripotent stem cells (iPSCs) improve behavior and form neurons after implantation into the stroke-injured adult rodent brain. How the aged brain responds to grafted iPSCs is unknown. We determined survival and differentiation of grafted human fibroblast-derived iPSCs and their ability to improve recovery in aged rats after stroke. METHODS: Twenty-four months old rats were subjected to 30 min distal middle cerebral artery occlusion causing neocortical damage. After 48 h, animals were transplanted intracortically with human iPSC-derived long-term neuroepithelial-like stem (hiPSC-lt-NES) cells. Controls were subjected to stroke and were vehicle-injected. RESULTS: Cell-grafted animals performed better than vehicle-injected recipients in cylinder test at 4 and 7 weeks. At 8 weeks, cell proliferation was low (0.7 %) and number of hiPSC-lt-NES cells corresponded to 49.2% of that of implanted cells. Transplanted cells expressed markers of neuroblasts and mature and GABAergic neurons. Cell-grafted rats exhibited less activated microglia/macrophages in injured cortex and neuronal loss was mitigated. CONCLUSIONS: Our study provides the first evidence that grafted human iPSCs survive, differentiate to neurons and ameliorate functional deficits in stroke-injured aged brain.


Subject(s)
Aging , Brain Injuries/surgery , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/transplantation , Infarction, Middle Cerebral Artery/complications , Recovery of Function/physiology , Analysis of Variance , Animals , Cell Differentiation , Disease Models, Animal , Doublecortin Domain Proteins , ELAV Proteins/metabolism , Exploratory Behavior/physiology , Humans , Ki-67 Antigen/metabolism , Male , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism , gamma-Aminobutyric Acid/metabolism
7.
Nat Neurosci ; 17(6): 801-3, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24747576

ABSTRACT

It has been unclear whether ischemic stroke induces neurogenesis or neuronal DNA rearrangements in the human neocortex. Using immunohistochemistry; transcriptome, genome and ploidy analyses; and determination of nuclear bomb test-derived (14)C concentration in neuronal DNA, we found neither to be the case. A large proportion of cortical neurons displayed DNA fragmentation and DNA repair a short time after stroke, whereas neurons at chronic stages after stroke showed DNA integrity, demonstrating the relevance of an intact genome for survival.


Subject(s)
Cellular Senescence/physiology , DNA Fragmentation , DNA Repair/physiology , Neocortex/pathology , Neurons/pathology , Stroke/genetics , Stroke/pathology , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Neocortex/physiology , Neurons/physiology
8.
Brain ; 136(Pt 12): 3561-77, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24148272

ABSTRACT

Stem cell-based approaches to restore function after stroke through replacement of dead neurons require the generation of specific neuronal subtypes. Loss of neurons in the cerebral cortex is a major cause of stroke-induced neurological deficits in adult humans. Reprogramming of adult human somatic cells to induced pluripotent stem cells is a novel approach to produce patient-specific cells for autologous transplantation. Whether such cells can be converted to functional cortical neurons that survive and give rise to behavioural recovery after transplantation in the stroke-injured cerebral cortex is not known. We have generated progenitors in vitro, expressing specific cortical markers and giving rise to functional neurons, from long-term self-renewing neuroepithelial-like stem cells, produced from adult human fibroblast-derived induced pluripotent stem cells. At 2 months after transplantation into the stroke-damaged rat cortex, the cortically fated cells showed less proliferation and more efficient conversion to mature neurons with morphological and immunohistochemical characteristics of a cortical phenotype and higher axonal projection density as compared with non-fated cells. Pyramidal morphology and localization of the cells expressing the cortex-specific marker TBR1 in a certain layered pattern provided further evidence supporting the cortical phenotype of the fated, grafted cells, and electrophysiological recordings demonstrated their functionality. Both fated and non-fated cell-transplanted groups showed bilateral recovery of the impaired function in the stepping test compared with vehicle-injected animals. The behavioural improvement at this early time point was most likely not due to neuronal replacement and reconstruction of circuitry. At 5 months after stroke in immunocompromised rats, there was no tumour formation and the grafted cells exhibited electrophysiological properties of mature neurons with evidence of integration in host circuitry. Our findings show, for the first time, that human skin-derived induced pluripotent stem cells can be differentiated to cortical neuronal progenitors, which survive, differentiate to functional neurons and improve neurological outcome after intracortical implantation in a rat stroke model.


Subject(s)
Cerebral Cortex/cytology , Induced Pluripotent Stem Cells/physiology , Infarction, Middle Cerebral Artery/surgery , Neurons/physiology , Recovery of Function/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Cerebral Cortex/transplantation , Disease Models, Animal , Electric Stimulation , Glutaminase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/transplantation , Infarction, Middle Cerebral Artery/pathology , Neurons/classification , Neurons/drug effects , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , Rats , Rats, Nude , Rats, Sprague-Dawley
9.
Neurobiol Dis ; 52: 191-203, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23276704

ABSTRACT

Neural stem/progenitor cells (NSPCs) in subventricular zone (SVZ) produce new striatal neurons during several months after stroke, which may contribute to recovery. Intracerebral grafts of NSPCs can exert beneficial effects after stroke through neuronal replacement, trophic actions, neuroprotection, and modulation of inflammation. Here we have explored whether human fetal striatum-derived NSPC-grafts influence striatal neurogenesis and promote recovery in stroke-damaged brain. T cell-deficient rats were subjected to 1h middle cerebral artery occlusion (MCAO). Human fetal NSPCs or vehicle were implanted into ipsilateral striatum 48 h after MCAO, animals were assessed behaviorally, and perfused at 6 or 14 weeks. Grafted human NSPCs survived in all rats, and a subpopulation had differentiated to neuroblasts or mature neurons at 6 and 14 weeks. Numbers of proliferating cells in SVZ and new migrating neuroblasts and mature neurons were higher, and numbers of activated microglia/macrophages were lower in the ischemic striatum of NSPC-grafted compared to vehicle-injected group both at 6 and 14 weeks. A fraction of grafted NSPCs projected axons from striatum to globus pallidus. The NSPC-grafted rats showed improved functional recovery in stepping and cylinder tests from 6 and 12 weeks, respectively. Our data show, for the first time, that intrastriatal implants of human fetal NSPCs exert a long-term enhancement of several steps of striatal neurogensis after stroke. The grafts also suppress striatal inflammation and ameliorate neurological deficits. Our findings support the idea that combination of NSPC transplantation and stimulation of neurogenesis from endogenous NSPCs may become a valuable strategy for functional restoration after stroke.


Subject(s)
Infarction, Middle Cerebral Artery/surgery , Motor Skills/physiology , Neural Stem Cells/transplantation , Neurogenesis/physiology , Recovery of Function/physiology , Animals , Cell Movement/physiology , Corpus Striatum/physiopathology , Humans , Infarction, Middle Cerebral Artery/physiopathology , Male , Neurons/physiology , Rats , Rats, Nude , Walking/physiology
10.
Stem Cells ; 30(6): 1120-33, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22495829

ABSTRACT

Reprogramming of adult human somatic cells to induced pluripotent stem cells (iPSCs) is a novel approach to produce patient-specific cells for autologous transplantation. Whether such cells survive long-term, differentiate to functional neurons, and induce recovery in the stroke-injured brain are unclear. We have transplanted long-term self-renewing neuroepithelial-like stem cells, generated from adult human fibroblast-derived iPSCs, into the stroke-damaged mouse and rat striatum or cortex. Recovery of forepaw movements was observed already at 1 week after transplantation. Improvement was most likely not due to neuronal replacement but was associated with increased vascular endothelial growth factor levels, probably enhancing endogenous plasticity. Transplanted cells stopped proliferating, could survive without forming tumors for at least 4 months, and differentiated to morphologically mature neurons of different subtypes. Neurons in intrastriatal grafts sent axonal projections to the globus pallidus. Grafted cells exhibited electrophysiological properties of mature neurons and received synaptic input from host neurons. Our study provides the first evidence that transplantation of human iPSC-derived cells is a safe and efficient approach to promote recovery after stroke and can be used to supply the injured brain with new neurons for replacement.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Neurons/cytology , Stem Cell Transplantation/methods , Stroke/pathology , Stroke/surgery , Aged , Animals , Brain/cytology , Brain/pathology , Cell Differentiation/physiology , Cells, Cultured , Female , Humans , Immunohistochemistry , Mice , Rats
11.
Stroke ; 40(10): 3378-83, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19661466

ABSTRACT

BACKGROUND AND PURPOSE: The functional role of arteriolo-arteriolar anastomosis (AAA) between the middle cerebral artery (MCA) and anterior cerebral artery in local hemodynamics is unknown, and was investigated here. METHODS: Blood flow in AAAs was examined using fluorescein isothiocyanate-labeled red blood cells (RBCs) as a flow indicator in 16 anesthetized C57BL/6J mice before and after MCA occlusion up to 7 experimental days. RESULTS: We observed paradoxical flow in AAAs; labeled RBCs entered from both the MCA and anterior cerebral artery sides and the opposing flows met at a branching T-junction, where the flows combined and passed into a penetrating arteriole. The dually fed T-junction was not fixed in position, but functionally jumped to adjacent T-junctions in response to changing hemodynamic conditions. On MCA occlusion, RBC flow from the MCA side immediately stopped. After a period of "hesitation," blood started to move retrogradely in one of the MCA branches toward the MCA stem. The retrograde blood flow was statistically significantly (P<0.05), serving to feed blood to other MCA branches after a lag period. In capillaries, MCA occlusion induced immediate RBC disappearance in the ischemic core and to a lesser extent in the marginal zone near AAAs. At day 3 after ischemia, we recognized the beginning of remodeling with angiogenesis centering on AAAs. CONCLUSIONS: AAAs appear to play a key role in local hemodynamic homeostasis, both in the normal state and in the development of collateral channels and revascularization during ischemia.


Subject(s)
Anterior Cerebral Artery/physiology , Arterioles/physiology , Brain Ischemia/physiopathology , Brain/blood supply , Hemodynamics/physiology , Homeostasis/physiology , Middle Cerebral Artery/physiology , Animals , Anterior Cerebral Artery/anatomy & histology , Arterioles/anatomy & histology , Brain/physiology , Capillaries/cytology , Capillaries/physiology , Cerebrovascular Circulation/physiology , Disease Models, Animal , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Mice, Inbred C57BL , Middle Cerebral Artery/anatomy & histology , Recovery of Function/physiology , Staining and Labeling/methods
12.
Clin Hemorheol Microcirc ; 35(1-2): 223-6, 2006.
Article in English | MEDLINE | ID: mdl-16899932

ABSTRACT

OBJECTIVES: The aim of the present study was the experimental analysis of blood rheological disorders in the microcirculation during endotoxic shock. METHODS: Experiments were carried out on 30 white laboratory rats of both sexes weighing about 200 g. The experimental animals were divided in two groups: the control group and the group undergoing experimental endotoxic shock. The following specific hemorheological properties were investigated in all animals during the experiments: red blood cells (RBC) aggregability, their deformability and the systemic hematocrit. RESULTS: We found that all the investigated hemorheological parameters appreciably changed underconditions of endotoxic shock. The RBC aggregability index was increased by a mean of 136%. As to the RBC deformability index was found to be decreased by 71%, and the systemic hematocrit lowered by 31%, as compared to the same parameters in the control group. CONCLUSION: The data obtained provide us with evidence that the hemorheological derangements associated with the development of the heavy microcirculatory disorders during endotoxic shock, are most significant factors.


Subject(s)
Erythrocyte Aggregation/drug effects , Erythrocyte Deformability/drug effects , Hematocrit , Microcirculation/pathology , Shock, Septic/blood , Animals , Erythrocyte Aggregation/physiology , Erythrocyte Deformability/physiology , Female , Hemorheology , Lipopolysaccharides/pharmacology , Male , Microcirculation/drug effects , Microcirculation/physiology , Rats , Shock, Septic/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...