Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
iScience ; 26(6): 106923, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37283810

ABSTRACT

While insulin resistance (IR) is associated with inflammation in white adipose tissue, we report a non-inflammatory adipose mechanism of high fat-induced IR mediated by loss of Pref-1. Pref-1, released from adipose Pref-1+ cells with characteristics of M2 macrophages, endothelial cells or progenitors, inhibits MIF release from both Pref-1+ cells and adipocytes by binding with integrin ß1 and inhibiting the mobilization of p115. High palmitic acid induces PAR2 expression in Pref-1+ cells, downregulating Pref-1 expression and release in an AMPK-dependent manner. The loss of Pref-1 increases adipose MIF secretion contributing to non-inflammatory IR in obesity. Treatment with Pref-1 blunts the increase in circulating plasma MIF levels and subsequent IR induced by a high palmitic acid diet. Thus, high levels of fatty acids suppress Pref-1 expression and secretion, through increased activation of PAR2, resulting in an increase in MIF secretion and a non-inflammatory adipose mechanism of IR.

2.
Front Cell Dev Biol ; 9: 659531, 2021.
Article in English | MEDLINE | ID: mdl-33959612

ABSTRACT

Myeloid cell leukemia-1 (Mcl-1), an anti-apoptotic Bcl-2 protein, regulates neural precursor cell (NPC) survival in both the developing and adult mammalian nervous system. It is unclear when during the neurogenic period Mcl-1 becomes necessary for NPC survival and whether Bax is the sole pro-apoptotic target of Mcl-1. To address these questions, we used the nervous system-specific Nestin-Cre Mcl-1 conditional knockout mouse line (Mcl-1 CKO) to assess the anti-apoptotic role of Mcl-1 in developmental neurogenesis. Loss of Mcl-1 resulted in a wave of apoptosis beginning in the brainstem and cervical spinal cord at embryonic day 9.5 (E9.5) and in the forebrain at E10.5. Apoptosis was first observed ventrally in each region and spread dorsally over time. Within the spinal cord, apoptosis also spread in a rostral to caudal direction following the path of differentiation. Breeding the Mcl-1 CKO mouse with the Bax null mouse rescued the majority of NPC from apoptosis except in the dorsomedial brainstem and ventral thoracic spinal cord where only 50% were rescued. This demonstrates that Mcl-1 promotes NPC survival primarily by inhibiting the activation of Bax, but that Bax is not the sole pro-apoptotic target of Mcl-1 during embryonic neurogenesis. Interestingly, although co-deletion of Bax rescued the majority of NPC apoptosis, it resulted in embryonic lethality at E13, whereas conditional deletion of both Mcl-1 and Bax rescued embryonic lethality. In summary, this study demonstrates the widespread dependency on Mcl-1 during nervous system development.

3.
Eur J Neurosci ; 52(4): 3196-3214, 2020 08.
Article in English | MEDLINE | ID: mdl-31945213

ABSTRACT

Ischemic stroke causes immediate cell death within the infarct core, whereas cells in the surrounding penumbra region are damaged but can be salvaged. Rapid restoration of blood flow can rescue these cells in the first few hours post-stroke. It remains unclear how long cells within the penumbra region can survive. To address this, we compared the acute cellular response within the ischemic core versus the penumbra region following an Endothelin-1-induced focal ischemic stroke in mice. We used vascular labelling to distinguish the three regions of blood perfusion: the non-perfused core; the hypo-perfused penumbra; and the perfused region. Within 4 hr post-stroke ~80% of neurons died in the non-perfused core, while 40% of neurons died in the hypo-perfused region. From 4 to 24 hr post-stroke, surviving neurons within the hypo-perfused region underwent extensive dendritic and axonal degeneration. Breakdown of the blood brain barrier, microglia activation, monocyte/neutrophil infiltration and astrogliosis, however, was not observed until 24 hr post-stroke. The cellular response within the hypo-perfused region was distinct from the non-perfused core. The core was comprised primarily of infiltrating peripheral monocytes and leukocytes, whereas the hypo-perfused region contained activated microglia and astrocytes. This study shows that small, localized ischemic strokes exhibit altered breakdown of the BBB in comparison with larger strokes. Furthermore, the massive degeneration of neuronal processes within the penumbra region suggests that the timeline to salvage surviving neurons is limited. In summary, the findings from this study reinforce the urgent need for therapeutic strategies targeting the acute hours post-stroke.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Animals , Astrocytes , Mice , Neurons
4.
Neuroscience ; 406: 637-652, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30731155

ABSTRACT

Obesity is associated with worse neurological outcomes following overt ischemic strokes. The majority of strokes however, are covert, small strokes that often evade detection. How obesity impacts the cellular response to covert strokes is unclear. Here, we used a diet-induced obesity model by feeding mice a high fat diet (HFD) and examining its impact on the behavioral and cellular responses to either an Endothelin-1-induced focal ischemic stroke or a saline injection (control). Specifically, we examined cells in regions with different levels of blood perfusion: the non-perfused core, the hypo-perfused surround and the perfused region around the infarct. We show that HFD selectively exacerbated the response to stroke but not to saline injections. Stroke affected the composition of microglia/macrophages, astrocytes and neurons within each region of perfusion. In the non-perfused core, the majority of cells were Iba-1+ microglia and macrophages. HFD resulted in a greater infiltration of CD68+ macrophages into the infarct core while CD68+ /TMEM119+ microglia were reduced. Furthermore, there was a trend towards an increased spread of the astrogliosis scar from the infarct border in the HFD condition. Within the hypo-perfused region, significantly fewer neurons survived in HFD-fed mice than Chow-fed mice, suggesting that neurons in the HFD condition have an increased vulnerability. In summary, diet-induced obesity exacerbates covert-like stroke injuries by worsening the cellular responses in the varying levels of perfusion across the infarct.


Subject(s)
Brain Ischemia/physiopathology , Diet, High-Fat , Neurons/physiology , Stroke/physiopathology , Animals , Astrocytes/physiology , Brain Ischemia/complications , Inflammation/complications , Inflammation/physiopathology , Macrophages/physiology , Male , Mice, Inbred C57BL , Microglia/physiology , Obesity/complications , Stroke/complications
5.
Cell Death Differ ; 26(8): 1501-1515, 2019 08.
Article in English | MEDLINE | ID: mdl-30361616

ABSTRACT

During neurogenesis, proliferating neural precursor cells (NPC) exit the cell cycle and differentiate into postmitotic neurons. The proteins that regulate cell survival through the stages of differentiation, however, are still poorly understood. Here, we examined the roles of the anti-apoptotic Bcl-2 proteins, Mcl-1 and Bcl-xL, in promoting survival as cells progress through the stages of neurogenesis in the mouse embryonic central nervous system. We used Nestin-mediated, nervous system-specific conditional deletion of mcl-1, bcl-x or both to identify their distinct and overlapping roles. Individual conditional deletion of mcl-1 (MKO) and bcl-x (BKO) suggested sequential roles in promoting cell survival during developmental neurogenesis. In the MKO embryo, apoptosis begins at embryonic day 10 (E10) in the proliferating NPC population throughout the entire developing nervous system. In the BKO embryo, apoptosis begins later at E11 within the postmitotic neuron populations. In the double (mcl-1 and bcl-x) conditional knockout (DKO), cell death extended throughout both proliferating and non-proliferating cell populations resulting in embryonic lethality at E12, earlier than in either the MKO or BKO. Apoptotic cell death of the entire central nervous system in the DKO demonstrates that both genes are necessary for cell survival during developmental neurogenesis. To determine whether Mcl-1 and Bcl-xL have overlapping anti-apoptotic roles during neurogenesis, we examined the impact of gene dosage. Loss of a single bcl-x allele in the MKO embryo exasperated apoptotic cell death within the NPC population revealing a novel anti-apoptotic role for Bcl-xL in proliferating NPCs. Cells were rescued from apoptosis in both the MKO and BKO embryos by breeding with the Bax null mouse line indicating that Mcl-1 and Bcl-xL have a common pro-apoptotic target during developmental neurogenesis. Taken together, these findings demonstrate that Mcl-1 and Bcl-xL are the two essential anti-apoptotic Bcl-2 proteins required for the survival of the developing mammalian nervous system.


Subject(s)
Central Nervous System/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , bcl-X Protein/metabolism , Animals , Cell Differentiation , Cell Survival , Mice , Mice, Inbred C57BL , Neurons/metabolism
6.
Mol Cell Neurosci ; 77: 34-46, 2016 12.
Article in English | MEDLINE | ID: mdl-27665712

ABSTRACT

The bcl-2 family of survival and death promoting proteins play a key role in regulating cell numbers during nervous system development. Bcl-xL, an anti-apoptotic bcl-2 family member is highly expressed in the developing nervous system. However; the early embryonic lethality of the bcl-x germline null mouse precluded an investigation into its role in nervous system development. To identify the role of bcl-x in spinal cord neurogenesis, we generated a central nervous system-specific bcl-x conditional knockout (BKO) mouse. Apoptotic cell death in the BKO embryo was initially detected at embryonic day 11 (E11) in the ventrolateral aspect of the spinal cord corresponding to the location of motor neurons. Apoptosis reached its peak at E13 having spread across the ventral and into the dorsal spinal cord. By E18, the wave of apoptosis had passed and only a few apoptotic cells were observed. The duration and direction of spread of apoptosis across the spinal cord is consistent with the spatial and temporal sequence of neuronal differentiation. Motor neurons, the first neurons to become post mitotic in the spinal cord, were also the first apoptotic cells. As neurogenesis spread across the spinal cord, later born neuronal populations such as Lim2+ interneurons were also affected. The onset of apoptosis occurred in cells that had exited the cell cycle within the previous 24h and initiated neural differentiation as demonstrated by BrdU birthdating and ßIII tubulin immunohistochemistry. This data demonstrates that spinal cord neurons become Bcl-xL dependent at an early post mitotic stage in developmental neurogenesis.


Subject(s)
Neurogenesis , Spinal Cord/metabolism , bcl-X Protein/metabolism , Animals , Apoptosis , Cell Cycle , Mice , Mice, Inbred C57BL , Motor Neurons/cytology , Motor Neurons/metabolism , Spinal Cord/cytology , Spinal Cord/embryology , bcl-X Protein/genetics
7.
J Vis Exp ; (98): e52701, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25993447

ABSTRACT

The cylinder test is routinely used to predict focal ischemic damage to the forelimb motor cortex in rodents. When placed in the cylinder, rodents explore by rearing and touching the walls of the cylinder with their forelimb paws for postural support. Following ischemic injury to the forelimb sensorimotor cortex, rats rely more heavily on their unaffected forelimb paw for postural support resulting in fewer touches with their affected paw which is termed forelimb asymmetry. In contrast, focal ischemic damage in the mouse brain fails to result in comparable consistent deficits in forelimb asymmetry. While forelimb asymmetry deficits are infrequently observed, mice do demonstrate a novel behaviour post stroke termed "paw-dragging". Paw-dragging is the tendency for a mouse to drag its affected paw along the cylinder wall rather than directly push off from the wall when dismounting from a rear to a four-legged stance. We have previously demonstrated that paw-dragging behaviour is highly sensitive to small cortical ischemic injuries to the forelimb motor cortex. Here we provide a detailed protocol for paw-dragging analysis. We define what a paw-drag is and demonstrate how to quantify paw-dragging behaviour. The cylinder test is a simple and inexpensive test to administer and does not require pre-training or food deprivation strategies. In using paw-dragging analysis with the cylinder test, it fills a niche for predicting cortical ischemic injuries such as photothrombosis and Endothelin-1 (ET-1)-induced ischemia--two models that are ever-increasing in popularity and produce smaller focal injuries than middle cerebral artery occlusion. Finally, measuring paw-dragging behaviour in the cylinder test will allow studies of functional recovery after cortical injury using a wide cohort of transgenic mouse strains where previous forelimb asymmetry analysis has failed to detect consistent deficits.


Subject(s)
Brain Injuries/diagnosis , Brain Injuries/physiopathology , Animals , Brain Injuries/chemically induced , Brain Ischemia/chemically induced , Brain Ischemia/diagnosis , Brain Ischemia/physiopathology , Disease Models, Animal , Endothelin-1 , Forelimb/physiopathology , Male , Mice , Motor Cortex/physiopathology , Rats , Recovery of Function , Stroke/chemically induced , Stroke/diagnosis , Stroke/physiopathology
8.
J Neurosci Methods ; 233: 34-44, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24915635

ABSTRACT

BACKGROUND: Despite the availability of numerous transgenic mouse lines to study the role of individual genes in promoting neural repair following stroke, few studies have availed of this technology, primarily due to the lack of a reproducible ischemic injury model in the mouse. Intracortical injections of Endothelin-1 (ET1) a potent vasoconstrictive agent, reliably produces focal infarcts with concomitant behavioral deficits in rats. In contrast, ET1 infarcts in mice are significantly smaller and do not generate consistent behavioral deficits. NEW METHOD: We have modified the ET1 ischemia model to target the anterior forelimb motor cortex (aFMC) and show that this generates a reproducible focal ischemic injury in mice with consistent behavioral deficits. Furthermore, we have developed a novel analysis of the cylinder test by quantifying paw-dragging behavior. RESULTS: ET1 injections which damage deep layer neurons in the aFMC generate reproducible deficits on the staircase test. Cylinder test analysis showed no forelimb asymmetry post-injection; however, we observed a novel paw-dragging behavior in mice which is a positive sign of damage to the FMC. COMPARISON WITH EXISTING METHODS: Previous ET1 studies have demonstrated inconsistent behavioral deficits; however, targeting ET1 injections to the aFMC reliably results in staircase deficits. We show that analysis of paw-dragging behavior in the cylinder test is a more sensitive measure of damage to the FMC than the classical forelimb asymmetry analysis. CONCLUSIONS: We have developed a focal ischemic injury model in the mouse that results in reproducible behavioral deficits and can be used to test future regenerative therapies.


Subject(s)
Disease Models, Animal , Forelimb/physiopathology , Motor Cortex/physiopathology , Stroke/physiopathology , Animals , Brain Ischemia/complications , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Count , Endothelin-1 , Immunohistochemistry , Male , Mice , Motor Cortex/pathology , Movement Disorders/etiology , Movement Disorders/pathology , Movement Disorders/physiopathology , Neurons/pathology , Neurons/physiology , Random Allocation , Reproducibility of Results , Severity of Illness Index , Stroke/complications , Stroke/pathology
9.
Development ; 140(15): 3118-27, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23824576

ABSTRACT

Cortical development requires the precise timing of neural precursor cell (NPC) terminal mitosis. Although cell cycle proteins regulate terminal mitosis, the factors that influence the cell cycle machinery are incompletely understood. Here we show in mice that myeloid cell leukemia 1 (Mcl1), an anti-apoptotic Bcl-2 protein required for the survival of NPCs, also regulates their terminal differentiation through the cell cycle regulator p27(Kip1). A BrdU-Ki67 cell profiling assay revealed that in utero electroporation of Mcl1 into NPCs in the embryonic neocortex increased NPC cell cycle exit (the leaving fraction). This was further supported by a decrease in proliferating NPCs (Pax6(+) radial glial cells and Tbr2(+) neural progenitors) and an increase in differentiating cells (Dcx(+) neuroblasts and Tbr1(+) neurons). Similarly, BrdU birth dating demonstrated that Mcl1 promotes premature NPC terminal mitosis giving rise to neurons of the deeper cortical layers, confirming their earlier birthdate. Changes in Mcl1 expression within NPCs caused concomitant changes in the levels of p27(Kip1) protein, a key regulator of NPC differentiation. Furthermore, in the absence of p27(Kip1), Mcl1 failed to induce NPC cell cycle exit, demonstrating that p27(Kip1) is required for Mcl1-mediated NPC terminal mitosis. In summary, we have identified a novel physiological role for anti-apoptotic Mcl1 in regulating NPC terminal differentiation.


Subject(s)
Brain/embryology , Brain/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Neural Stem Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Brain/cytology , Cell Cycle Checkpoints , Cell Differentiation , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Doublecortin Protein , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitosis , Myeloid Cell Leukemia Sequence 1 Protein , Neural Stem Cells/cytology , Neurogenesis , Pregnancy , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/genetics
10.
J Neurosci ; 33(13): 5773-84, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23536090

ABSTRACT

Ubiquitous classical (typical) calpains, calpain-1 and calpain-2, are Ca(+2)-dependent cysteine proteases, which have been associated with numerous physiological and pathological cellular functions. However, a clear understanding of the role of calpains in the CNS has been hampered by the lack of appropriate deletion paradigms in the brain. In this study, we describe a unique model of conditional deletion of both calpain-1 and calpain-2 activities in mouse brain, which more definitively assesses the role of these ubiquitous proteases in brain development/function and pathology. Surprisingly, we show that these calpains are not critical for gross CNS development. However, calpain-1/calpain-2 loss leads to reduced dendritic branching complexity and spine density deficits associated with major deterioration in hippocampal long-term potentiation and spatial memory. Moreover, calpain-1/calpain-2-deficient neurons were significantly resistant to injury induced by excitotoxic stress or mitochondrial toxicity. Examination of downstream target showed that the conversion of the Cdk5 activator, p35, to pathogenic p25 form, occurred only in the presence of calpain and that it played a major role in calpain-mediated neuronal death. These findings unequivocally establish two central roles of calpain-1/calpain-2 in CNS function in plasticity and neuronal death.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/pathology , Brain , Calpain/deficiency , Long-Term Potentiation/physiology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Biophysics , Brain/embryology , Brain/growth & development , Brain/pathology , Brain Injuries/chemically induced , Brain Injuries/physiopathology , Bromodeoxyuridine/metabolism , Cell Death/drug effects , Cell Death/genetics , Dendrites/metabolism , Dendrites/pathology , Dendrites/ultrastructure , Disease Models, Animal , Electric Stimulation , Embryo, Mammalian , Evoked Potentials/drug effects , Evoked Potentials/physiology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Female , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Hippocampus/cytology , In Vitro Techniques , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Long-Term Potentiation/genetics , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , N-Methylaspartate/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Phosphotransferases , Psychomotor Performance , RNA, Messenger/metabolism , Silver Staining , Transfection
11.
Mol Cell Neurosci ; 49(4): 439-47, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22357134

ABSTRACT

Since the discovery of neural precursor cells (NPCs) in the adult mammalian brain, there has been a lot of excitement surrounding the potential for regeneration in the adult brain. For instance, many studies have shown that a significant number of NPCs will migrate to a site of injury and differentiate into all of the neural lineages. However, one of the main challenges affecting endogenous neural regeneration is that many of the NPCs that migrate to the injury site ultimately undergo apoptosis. Therefore, we sought to determine whether myeloid cell leukemia-1 (Mcl-1), an anti-apoptotic Bcl-2 protein, would promote the survival of adult NPCs by impeding apoptosis. To do this, we first confirmed that Mcl-1 is endogenously expressed within the adult NPC population using BrdU labeling assays. Next, we conditionally deleted Mcl-1 in adult NPCs using cre/lox technology and expressed Cre from the NPC-specific promoter Nestin. In vitro, cells that had Mcl-1 conditionally deleted had a 2-fold increase in apoptosis when compared to controls. In vivo, we used electroporation to conditionally delete Mcl-1 in adult NPCs and assessed apoptosis at 72h. after electroporation. As in our in vitro results, there was a 2-fold increase in apoptosis when Mcl-1 was conditionally deleted. Finally, we found that Mcl-1 over-expression reduced the endogenous rate of adult NPC apoptosis 2-fold in vitro. Collectively, these results demonstrate that Mcl-1 is crucial for the survival of adult NPCs and may be a promising target for future neural regeneration therapies.


Subject(s)
Adult Stem Cells/metabolism , Apoptosis/physiology , Brain/metabolism , Neural Stem Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Adult Stem Cells/cytology , Animals , Blotting, Western , Brain/cytology , Cell Survival/physiology , Immunohistochemistry , Mice , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein , Neural Stem Cells/cytology , Transfection
12.
EMBO J ; 30(2): 395-407, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21139567

ABSTRACT

Apoptosis has an important role during development to regulate cell number. In differentiated cells, however, activation of autophagy has a critical role by enabling cells to remain functional following stress. In this study, we show that the antiapoptotic BCL-2 homologue MCL-1 has a key role in controlling both processes in a developmentally regulated manner. Specifically, MCL-1 degradation is an early event not only following induction of apoptosis, but also under nutrient deprivation conditions where MCL-1 levels regulate activation of autophagy. Furthermore, deletion of MCL-1 in cortical neurons of transgenic mice activates a robust autophagic response. This autophagic response can, however, be converted to apoptosis by either reducing the levels of the autophagy regulator Beclin-1, or by a concomitant activation of BAX. Our results define a pathway whereby MCL-1 has a key role in determining cell fate, by coordinately regulating apoptosis and autophagy.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Analysis of Variance , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Mice, Knockout , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-2-Associated X Protein/metabolism
13.
Mol Cell Biol ; 29(17): 4701-13, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19564414

ABSTRACT

We have previously shown that p107, a member of the retinoblastoma (Rb) cell cycle regulatory family, has a unique function in regulating the pool of neural precursor cells. As the pool of progenitors is regulated by a limiting supply of trophic factors, we asked if the Rb/E2F pathway may control the size of the progenitor population by regulating the levels of growth factors or their receptors. Here, we demonstrate that fibroblast growth factor 2 (FGF2) is aberrantly upregulated in the brains of animals lacking Rb family proteins and that the gene encoding the FGF2 ligand is directly regulated by p107 and E2F3. Chromatin immunoprecipitation assays demonstrated that E2F3 and p107 occupy E2F consensus sites on the FGF2 promoter in the context of native chromatin. To evaluate the physiological consequence of FGF2 deregulation in both p107 and E2F3 mutants, we measured neural progenitor responsiveness to growth factors. Our results demonstrate that E2F3 and p107 are each mediators of FGF2 growth factor responsiveness in neural progenitor cells. These results support a model whereby p107 regulates the pool of FGF-responsive progenitors by directly regulating FGF2 gene expression in vivo. By identifying novel roles for p107/E2F in regulating genes outside of the classical cell cycle machinery targets, we uncover a new mechanism whereby Rb/E2F mediates proliferation through regulating growth factor responsiveness.


Subject(s)
E2F3 Transcription Factor/metabolism , Fibroblast Growth Factor 2/metabolism , Neurons/physiology , Signal Transduction/physiology , Stem Cells/physiology , Animals , Base Sequence , Cell Proliferation , Cells, Cultured , E2F3 Transcription Factor/genetics , Female , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Neurons/cytology , Pregnancy , Promoter Regions, Genetic , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Sequence Alignment , Stem Cells/cytology
14.
J Neurosci ; 28(24): 6068-78, 2008 Jun 11.
Article in English | MEDLINE | ID: mdl-18550749

ABSTRACT

Despite the importance of Mcl-1, an anti-apoptotic Bcl-2 family member, in the regulation of apoptosis, little is known regarding its role in nervous system development and injury-induced neuronal cell death. Because germline deletion of Mcl-1 results in peri-implantation lethality, we address the function of Mcl-1 in the nervous system using two different conditional Mcl-1 mouse mutants in the developing nervous system. Here, we show for the first time that Mcl-1 is required for neuronal development. Neural precursors within the ventricular zone and newly committed neurons in the cortical plate express high levels of Mcl-1 throughout cortical neurogenesis. Loss of Mcl-1 in neuronal progenitors results in widespread apoptosis. Double labeling with active caspase 3 and Tuj1 reveals that newly committed Mcl1 deficient neurons undergo apoptosis as they commence migration away from the ventricular zone. Examination of neural progenitor differentiation in vitro demonstrated that cell death in the absence of Mcl1 is cell autonomous. Although conditional deletion of Mcl-1 in cultured neurons does not trigger apoptosis, loss of Mcl-1 sensitizes neurons to an acute DNA damaging insult. Indeed, the rapid reduction of Mcl-1 mRNA and protein levels are early events after DNA damage in neurons, and maintaining high Mcl-1 levels can protect neurons against death. Together, our results are the first to demonstrate the requirement of Mcl-1, an anti-apoptotic Bcl-2 family protein, for cortical neurogenesis and the survival of neurons after DNA damage.


Subject(s)
Apoptosis/physiology , Central Nervous System/embryology , DNA Damage/physiology , Gene Expression Regulation, Developmental/physiology , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Age Factors , Animals , Apoptosis/genetics , Caspase 3/metabolism , Cell Differentiation , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Central Nervous System/cytology , DNA Damage/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Mutation/genetics , Myeloid Cell Leukemia Sequence 1 Protein , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Stem Cells/physiology , Transfection/methods , Tubulin/metabolism
15.
Mol Cell Neurosci ; 38(3): 349-58, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18499474

ABSTRACT

Mammalian HES1 and HES5 are abundant in developing CNS and inhibit neurogenesis, while HES6 promotes neurogenesis. An early serotonergic differentiation marker, the 5-HT1A receptor, is repressed by HES5 and DEAF1 which recognize the C(-1019), but not G(-1019) allele of a human 5-HT1A promoter polymorphism associated with mood disorders. We tested whether HES1 and HES6 regulate transcriptional activity at this element. HES1 strongly repressed 5-HT1A transcription in neuronal and non-neuronal cells, while HES6 reversed HES1- and HES5-mediated repression. Mutation of a putative HES consensus site blocked HES1 and HES5, but, unlike HES5, HES1 repressed at the G(-1019) allele. To address its role in vivo, the temporal expression of 5-HT1A receptor RNA and protein was examined in HES1-/- mice, and elevated levels in E12.5 hindbrain and midbrain were observed. Thus, HES1 and HES6 oppositely regulate 5-HT1A receptor transcription and HES1 is required for its correct developmental expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/physiology , Polymorphism, Genetic/physiology , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT1A/metabolism , Transcription, Genetic/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Female , Homeodomain Proteins/genetics , Humans , Mice , Mice, Knockout , Pregnancy , Rats , Receptor, Serotonin, 5-HT1A/biosynthesis , Transcription Factor HES-1
16.
J Neurosci ; 27(22): 5926-35, 2007 May 30.
Article in English | MEDLINE | ID: mdl-17537963

ABSTRACT

Early forebrain development is characterized by extensive proliferation of neural precursors coupled with complex structural transformations; however, little is known regarding the mechanisms by which these processes are integrated. Here, we show that deficiency of the cell cycle regulatory protein, E2F4, results in the loss of ventral telencephalic structures and impaired self-renewal of neural precursor cells. The mechanism underlying aberrant ventral patterning lies in a dramatic loss of Sonic hedgehog (Shh) expression specifically in this region. The E2F4-deficient phenotype can be recapitulated by interbreeding mice heterozygous for E2F4 with those lacking one allele of Shh, suggesting a genetic interaction between these pathways. Treatment of E2F4-deficient cells with a Hh agonist rescues stem cell self-renewal and cells expressing the homeodomain proteins that specify the ventral telencephalic structures. Finally, we show that E2F4 deficiency results in impaired activity of Shh forebrain-specific enhancers. In conclusion, these studies establish a novel requirement for the cell cycle regulatory protein, E2F4, in the development of the ventral telencephalon.


Subject(s)
Cell Cycle/physiology , E2F4 Transcription Factor/physiology , Telencephalon/embryology , Telencephalon/metabolism , Animals , Cells, Cultured , E2F4 Transcription Factor/deficiency , Female , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Telencephalon/cytology , Telencephalon/growth & development
17.
J Cell Biol ; 178(1): 129-39, 2007 Jul 02.
Article in English | MEDLINE | ID: mdl-17591923

ABSTRACT

The Retinoblastoma protein p107 regulates the neural precursor pool in both the developing and adult brain. As p107-deficient mice exhibit enhanced levels of Hes1, we questioned whether p107 regulates neural precursor self-renewal through the repression of Hes1. p107 represses transcription at the Hes1 promoter. Despite an expanded neural precursor population, p107-null mice exhibit a striking reduction in the number of cortical neurons. Hes1 deficiency rescues neurosphere numbers in p107-null embryos. We find that the loss of a single Hes1 allele in vivo restores the number of neural precursor cells at the ventricular zone. Neuronal birthdating analysis reveals a dramatic reduction in the rate of neurogenesis, demonstrating impairment in p107(-/-) progenitors to commit to a neuronal fate. The loss of a single Hes1 allele restores the number of newly generated neurons in p107-deficient brains. Together, we identify a novel function for p107 in promoting neural progenitor commitment to a neuronal fate.


Subject(s)
Gene Expression Regulation, Developmental , Neurons/metabolism , Retinoblastoma-Like Protein p107/deficiency , Stem Cells/metabolism , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cerebral Cortex/cytology , Embryo, Mammalian , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Kinetics , Mice , Mice, Knockout , Models, Biological , Proliferating Cell Nuclear Antigen/analysis , Promoter Regions, Genetic , Retinoblastoma-Like Protein p107/genetics , Transcription Factor HES-1 , Transcription, Genetic
18.
Mol Cell Biol ; 27(13): 4825-43, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17452454

ABSTRACT

The cell cycle regulatory retinoblastoma (Rb) protein is a key regulator of neural precursor proliferation; however, its role has been expanded to include a novel cell-autonomous role in mediating neuronal migration. We sought to determine the Rb-interacting factors that mediate both the cell cycle and migration defects. E2F1 and E2F3 are likely Rb-interacting candidates that we have shown to be deregulated in the absence of Rb. Using mice with compound null mutations of Rb and E2F1 or E2F3, we asked to what extent either E2F1 or E2F3 interacts with Rb in neurogenesis. Here, we report that E2F1 and E2F3 are both functionally relevant targets in neural precursor proliferation, cell cycle exit, and laminar patterning. Each also partially mediates the Rb requirement for neuronal survival. Neuronal migration, however, is specifically mediated through E2F3, beyond its role in cell cycle regulation. This study not only outlines overlapping and distinct functions for E2Fs in neurogenesis but also is the first to establish a physiologically relevant role for the Rb/E2F pathway beyond cell cycle regulation in vivo.


Subject(s)
Cell Cycle , Cell Movement , E2F3 Transcription Factor/metabolism , Neurons/cytology , Retinoblastoma Protein/metabolism , Animals , Cell Proliferation , Cell Survival , E2F1 Transcription Factor/metabolism , Female , Gene Expression Regulation , Interneurons/cytology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Protein Binding , Stem Cells/cytology , Stem Cells/metabolism , Telencephalon/embryology , Telencephalon/metabolism
19.
Am J Physiol Renal Physiol ; 292(2): F868-75, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16885154

ABSTRACT

We investigated the role of the prostaglandin E(2) (PGE(2)) EP(1) receptor in modulating urine concentration as it is expressed along the renal collecting duct where arginine-vasopressin (AVP) exerts its anti-diuretic activity, and in the paraventricular and supraoptic nuclei of the hypothalamus where AVP is synthesized. The urine osmolality of EP(1)-null mice (EP(1)(-/-)) failed to match levels achieved by wild-type (WT) counterparts upon water deprivation (WD) for 24 h. This difference was reflected by higher plasma osmolality in WD EP(1)(-/-) mice. Along the collecting duct, the induction and subapical to plasma membrane translocation of the aquaporin-2 water channel in WD EP(1)(-/-) mice appeared equivalent to that of WD WT mice as determined by quantitative RT-PCR and immunohistochemistry. However, medullary interstitial osmolalities dropped significantly in EP(1)(-/-) mice following WD. Furthermore, urinary AVP levels of WD EP(1)(-/-) mice were significantly lower than those of WD WT mice. This deficit could be traced back to a blunted induction of hypothalamic AVP mRNA expression in WD EP(1)(-/-) mice as determined by quantitative RT-PCR. Administration of the AVP mimetic [deamino-Cys(1),D-Arg(8)]-vasopressin restored a significant proportion of the urine concentrating ability of WD EP(1)(-/-) mice. When mice were water loaded to suppress endogenous AVP production, urine osmolalities increased equally for WT and EP(1)(-/-) mice. These data suggest that PGE(2) modulates urine concentration by acting at EP(1) receptors, not in the collecting duct, but within the hypothalamus to promote AVP synthesis in response to acute WD.


Subject(s)
Kidney Concentrating Ability/genetics , Receptors, Prostaglandin E/deficiency , Water Deprivation/physiology , Animals , Aquaporin 2/biosynthesis , Arginine Vasopressin/biosynthesis , Deamino Arginine Vasopressin/pharmacology , Mice , Receptors, Prostaglandin E, EP1 Subtype
20.
EMBO J ; 24(24): 4381-91, 2005 Dec 21.
Article in English | MEDLINE | ID: mdl-16308563

ABSTRACT

Precise cell cycle regulation is critical for nervous system development. To assess the role of the cell cycle regulator, retinoblastoma (Rb) protein, in forebrain development, we studied mice with telencephalon-specific Rb deletions. We examined the role of Rb in neuronal specification and migration of diverse neuronal populations. Although layer specification occurred at the appropriate time in Rb mutants, migration of early-born cortical neurons was perturbed. Consistent with defects in radial migration, neuronal cell death in Rb mutants specifically affected Cajal-Retzius neurons. In the ventral telencephalon, although calbindin- and Lhx6-expressing cortical neurons were generated at embryonic day 12.5, their tangential migration into the neocortex was dramatically and specifically reduced in the mutant marginal zone. Cell transplantation assays revealed that defects in tangential migration arose owing to a cell-autonomous loss of Rb in migrating interneurons and not because of a defective cortical environment. These results revealed a cell-autonomous role for Rb in regulating the tangential migration of cortical interneurons. Taken together, we reveal a novel requirement for the cell cycle protein, Rb, in the regulation of neuronal migration.


Subject(s)
Gene Expression Regulation, Developmental , Neurons/metabolism , Retinoblastoma Protein/metabolism , Animals , Apoptosis , Body Patterning , Calbindins , Cell Cycle , Cell Differentiation , Cell Lineage , Cell Movement , Cell Survival , Coculture Techniques , Embryo, Mammalian/metabolism , Genotype , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , LIM-Homeodomain Proteins , Mice , Models, Anatomic , Mutation , Nerve Tissue Proteins/metabolism , Retinoblastoma/metabolism , S100 Calcium Binding Protein G/metabolism , Stem Cells/metabolism , Telencephalon/metabolism , Time Factors , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...