Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 15(5): 964-976, 2022 05.
Article in English | MEDLINE | ID: mdl-35869148

ABSTRACT

Intestinal epithelial barrier function is compromised in inflammatory bowel disease and barrier dysfunction contributes to disease progression. Extracellular nucleotides/nucleosides generated in gut inflammation may regulate barrier function through actions on diverse cell types. Enteric glia modulate extracellular purinergic signaling and exert pathophysiological effects on mucosal permeability. These glia may regulate inflammation with paracrine responses, theoretically mediated via adenosine 2B receptor (A2BR) signaling. As the cell-specific roles of A2BRs in models of colitis and barrier dysfunction are unclear, we studied glial A2BRs in acute dextran sodium sulfate (DSS) colitis. We performed and validated conditional ablation of glial A2BRs in Sox10CreERT2+/-;Adora2bf/f mice. Overt intestinal disease activity indices in DSS-colitis were comparable between Sox10CreERT2+/-;Adora2bf/f mice and littermate controls. However, ablating glial A2BRs protected against barrier dysfunction following acute DSS-colitis. These benefits were associated with the normalization of tight junction protein expression and localization including claudin-1, claudin-8, and occludin. Glial A2BR signaling increased levels of proinflammatory mediators in the colon and cell-intrinsic regulation of genes including Csf3, Cxcl1, Cxcl10, and Il6. Our studies show that glial A2BR signaling exacerbates immune responses during DSS-colitis and that this adenosinergic cell-specific mechanism contributes to persistent gut epithelial barrier dysfunction.


Subject(s)
Colitis , Intestinal Mucosa , Adenosine/metabolism , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Colon/metabolism , Dextran Sulfate , Disease Models, Animal , Inflammation/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Neuroglia/metabolism
2.
ASN Neuro ; 14: 17590914221083203, 2022.
Article in English | MEDLINE | ID: mdl-35593118

ABSTRACT

Enteric glia regulate gut functions in health and disease through diverse interactions with neurons and immune cells. Intracellular localization of traditional markers of enteric glia such as GFAP, s100b, and Sox10 makes them incompatible for studies that require antigen localization at the cell surface. Thus, new tools are needed for probing the heterogeneous roles of enteric glia at the protein, cell, and functional levels. Here we selected several cell surface antigens including Astrocyte Cell Surface Marker 2 (ACSA2), Cluster of differentiation 9 (CD9), lysophosphatidic acid receptor 1 (LPAR1), and Proteolipid protein 1 (PLP1) as potential markers of enteric glia. We tested their specificity for enteric glia using published single-cell/-nuclei and glia-specific translating mRNA enriched transcriptome datasets, immunolabeling, and flow cytometry. The data show that ACSA2 is a specific marker of mucosal and myenteric glia while other markers are suitable for identifying all subpopulations of enteric glia (LPAR1), glia and immune cells (CD9), or are not suitable for cell-surface labeling (PLP1). These new tools will be useful for future work focused on understanding specific glial functions in health and disease.Summary StatementThis study identifies astrocyte cell surface antigen 2 as a novel marker of myenteric glia in the intestine. This, in combination with other markers identified in this study, could be used for selective targeting of enteric glia.


Subject(s)
Antigens, Surface , Astrocytes , Animals , Antigens, Surface/metabolism , Colon , Mice , Neuroglia/metabolism , Neurons/metabolism
3.
Cell Mol Gastroenterol Hepatol ; 12(4): 1215-1237, 2021.
Article in English | MEDLINE | ID: mdl-34166814

ABSTRACT

BACKGROUND & AIMS: Enteric glial cells express type II major histocompatibility complex (MHC-II) molecules in Crohn's disease and Chagas disease, but it is unclear whether the expressed molecules are functional. We examined the capabilities of enteric glia to act as an antigen-presenting cell in vivo and whether glial MHC-II has immunomodulatory effects. METHODS: We generated Sox10CreERT2;IABfl/fl mice to ablate MHC-II in enteric glia after exposure to tamoxifen. We measured phagocytic activity and autophagy activation to assess potential peptide sources loaded onto glial MHC-II and measured T- and B-lymphocyte activation and serum and colonic tissue cytokine levels to study enteric glial immunomodulatory capabilities. RESULTS: Enteric glia express MHC-II molecules in response to a subclinical dose of interferon-γ and lipopolysaccharide in vivo. Glial MHC-II expression contributes to effective B-lymphocyte and T-lymphocyte activation with marked effects on T-helper cell (Th)17 and regulatory T cell subtypes. No effect on Th1 or Th2 subtypes was observed. Enteric glial MHC-II does not have a major effect on serum or colonic tissue cytokine levels but may influence local cytokine levels. Glial MHC-II expression requires the activation of autophagy pathways, but activating autophagy alone is not sufficient to drive glial MHC-II expression. CONCLUSIONS: Enteric glia express MHC-II as a mechanism to tune intestinal immune responses. Glial autophagy is triggered in response to proinflammatory stimuli and induces glial antigen presentation, which functions to modulate the activation of T-lymphocyte subsets involved in tolerance. These observations suggest that enteric glia may express MHC-II to maintain immune homeostasis during inflammatory conditions such as Crohn's disease.


Subject(s)
Autophagy , Enteric Nervous System/cytology , Histocompatibility Antigens Class II/genetics , Lymphocyte Activation/immunology , Neuroglia/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Cell Communication , Fluorescent Antibody Technique , Gene Expression , Gene Knockdown Techniques , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Immunophenotyping , Interferon-gamma/metabolism , Lipopolysaccharides/immunology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Knockout , Models, Biological , Phagocytosis
4.
Cell Rep ; 32(10): 108100, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32905782

ABSTRACT

Mechanisms resulting in abdominal pain include altered neuro-immune interactions in the gastrointestinal tract, but the signaling processes that link immune activation with visceral hypersensitivity are unresolved. We hypothesized that enteric glia link the neural and immune systems of the gut and that communication between enteric glia and immune cells modulates the development of visceral hypersensitivity. To this end, we manipulated a major mechanism of glial intercellular communication that requires connexin-43 and assessed the effects on acute and chronic inflammation, visceral hypersensitivity, and immune responses. Deleting connexin-43 in glia protected against the development of visceral hypersensitivity following chronic colitis. Mechanistically, the protective effects of glial manipulation were mediated by disrupting the glial-mediated activation of macrophages through the macrophage colony-stimulating factor. Collectively, our data identified enteric glia as a critical link between gastrointestinal neural and immune systems that could be harnessed by therapies to ameliorate abdominal pain.


Subject(s)
Inflammation/metabolism , Macrophages/metabolism , Neuroglia/metabolism , Animals , Humans , Mice , Phenotype
5.
Neuropharmacology ; 179: 108264, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32758565

ABSTRACT

Gulf War Illness (GWI) is a chronic multisymptom illness that includes gastrointestinal disorders. Although the exact etiology of GWI is unknown, exposure to the drug pyridostigmine bromide (PB) is considered a major factor. Exposure to PB drives enteric neuroinflammation, promotes immunosuppression, and alters physiological functions of the colon in the short term but whether exposure to PB is sufficient to promote long term dysfunction is not known. Here, we tested whether exposure to PB is sufficient to drive long term changes that reflect GWI, and whether the endogenous anti-inflammatory mediator palmitoylethanolamide (PEA) is sufficient to reduce the detrimental effects of PB in the gut and brain of mice. Exposure to PB alone was not sufficient to cause major changes in neuromuscular transmission but did drive major changes by altering the effects of PEA. Calcium imaging data show that the mechanisms responsible include a shift in receptor signaling mediated by TRPV1, endocannabinoids, and peroxisome proliferator-activated receptors alpha (PPARα). Additional mechanisms include the development of glial reactivity and changes in enteric neurochemical coding and survival. PB and PEA caused major shifts in pro-inflammatory cytokines/chemokines in the brain and colon that persisted up to 5 months following exposure. Many of the effects of PB and PEA exhibit significant sex differences. Together, these results highlight novel mechanisms whereby PB promotes long-lasting changes in nervous system and immune function by inducing occult neuroplasticity that is revealed by subsequent exposure to unrelated drugs in a sex dependent manner.


Subject(s)
Amides/pharmacology , Brain/drug effects , Ethanolamines/pharmacology , Gastrointestinal Tract/drug effects , Neuroimmunomodulation/drug effects , Palmitic Acids/pharmacology , Persian Gulf Syndrome/chemically induced , Pyridostigmine Bromide/toxicity , Amides/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain/immunology , Cholinesterase Inhibitors/toxicity , Chronic Disease , Disease Models, Animal , Ethanolamines/therapeutic use , Female , Gastrointestinal Tract/immunology , Male , Mice , Mice, Inbred C57BL , Neuroimmunomodulation/physiology , Palmitic Acids/therapeutic use , Persian Gulf Syndrome/drug therapy , Persian Gulf Syndrome/immunology
6.
Am J Physiol Gastrointest Liver Physiol ; 317(3): G314-G332, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31188623

ABSTRACT

ATP is both an important mediator of physiological gut functions such as motility and epithelial function, and a key danger signal that mediates cell death and tissue damage. The actions of extracellular ATP are regulated through the catalytic functions extracellular nucleoside triphosphate diphosphohydrolase-1 (NTPDase1), -2, -3, and -8, which ultimately generate nucleosides. Ectonucleotidases have distinct cellular associations, but the specific locations and functional roles of individual NTPDases in the intestine are still poorly understood. Here, we tested the hypothesis that differential and cell-selective regulation of purine hydrolysis by NTPDase1 and -2 plays important roles in gut physiology and disease. We studied Entpd1 and Entpd2 null mice in health and following colitis driven by 2% dextran sulfate sodium (DSS) administration using functional readouts of gut motility, epithelial barrier function, and neuromuscular communication. NTPDase1 is expressed by immune cells, and the ablation of Entpd1 altered glial numbers in the myenteric plexus. NTPDase2 is expressed by enteric glia, and the ablation of Entpd2 altered myenteric neuron numbers. Mice lacking either NTPDase1 or -2 exhibited decreased inhibitory neuromuscular transmission and altered components of inhibitory junction potentials. Ablation of Entpd2 increased gut permeability following inflammation. In conclusion, the location- and context-dependent extracellular nucleotide phosphohydrolysis by NTPDase1 and -2 substantially impacts gut function in health and disease.NEW & NOTEWORTHY Purines are important mediators of gastrointestinal physiology and pathophysiology. Nucleoside triphosphate diphosphohydrolases (NTPDases) regulate extracellular purines, but the roles of specific NTPDases in gut functions are poorly understood. Here, we used Entpd1- and Entpd2-deficient mice to show that the differential and cell-selective regulation of purine hydrolysis by NTPDase1 and -2 plays important roles in barrier function, gut motility, and neuromuscular communication in health and disease.


Subject(s)
Adenosine Triphosphatases/metabolism , Antigens, CD/metabolism , Apyrase/metabolism , Colitis/drug therapy , Colon/metabolism , Dextran Sulfate/pharmacology , Animals , Colitis/metabolism , Colon/drug effects , Female , Male , Mice , Mice, Knockout
7.
FASEB J ; 33(5): 6168-6184, 2019 05.
Article in English | MEDLINE | ID: mdl-30789759

ABSTRACT

Gulf War illness (GWI) is a chronic multisymptom disorder that is prominent in Gulf War veterans. Major unexplained symptoms of GWI include functional gastrointestinal disorders and undiagnosed illnesses, including neurologic disorders. Exposure to the antinerve gas drug pyridostigmine bromide (PB) is linked to the development of GWI, but the exact mechanisms remain unclear. Here, we tested the hypothesis that PB alters gut function by disrupting the neural and immune systems of the intestine. We exposed male and female mice to physiologically comparable amounts of PB that match the dose, route, and time frame of exposure experienced by Gulf War veterans and assessed the acute and chronic impacts on gastrointestinal functions, the functional architecture of the enteric nervous system, and immune responses in the gut and brain. Exposure to PB drove acute alterations to colonic motility and structure in both male and female mice that transitioned to chronic changes in gut functions. PB drove acute alterations to enteric neural and glial activity, glial reactivity, and neuron survival with glial reactivity persisting into the chronic phase in male mice. Despite having no effect on colonic permeability, exposure to PB caused major shifts in the expression of proinflammatory cytokines and chemokines in the colon and brain that suggest immunosuppressive effects. Interestingly, immune disruption was still evident in the colon and brain in female animals at 1 mo following exposure to PB. Together, our results show that the paradigm of PB exposure experienced by veterans of the Persian Gulf War contributes to long-lasting pathophysiology by driving enteric neuroinflammation, promoting immunosuppression, and altering functional anatomy of the colon in a sex-dependent manner.-Hernandez, S., Fried, D. E., Grubisic, V., McClain, J. L., Gulbransen, B. D. Gastrointestinal neuroimmune disruption in a mouse model of Gulf War illness.


Subject(s)
Cholinesterase Inhibitors/toxicity , Colon/drug effects , Enteric Nervous System/drug effects , Persian Gulf Syndrome/immunology , Pyridostigmine Bromide/toxicity , Animals , Brain/drug effects , Brain/immunology , Colon/immunology , Colon/physiopathology , Cytokines/metabolism , Enteric Nervous System/immunology , Enteric Nervous System/physiopathology , Female , Gastrointestinal Motility , Male , Mice , Mice, Inbred C57BL , Neuroglia/immunology , Persian Gulf Syndrome/etiology , Persian Gulf Syndrome/physiopathology
8.
Brain Res Bull ; 136: 109-117, 2018 01.
Article in English | MEDLINE | ID: mdl-28363846

ABSTRACT

The enteric nervous system, often referred to as the second brain, is the largest assembly of neurons and glia outside the central nervous system. The enteric nervous system resides within the wall of the digestive tract and regulates local gut reflexes involved in gastrointestinal motility and fluid transport; these functions can be accomplished in the absence of the extrinsic innervation from the central nervous system. It is neurons and their circuitry within the enteric nervous system that govern the gut reflexes. However, it is becoming clear that enteric glial cells are also actively involved in this process through the bidirectional signaling with neurons and other cells in the gut wall. We synthesize the recently discovered modulatory roles of enteric gliotransmission in gut motility and provide our perspective for future lines of research.


Subject(s)
Enteric Nervous System/physiology , Enteric Nervous System/physiopathology , Gastrointestinal Motility/physiology , Neuroglia/physiology , Animals , Humans
9.
Glia ; 65(5): 699-711, 2017 05.
Article in English | MEDLINE | ID: mdl-28168732

ABSTRACT

Enteric glia (EG) in the enteric nervous system can modulate neuronally regulated gut functions. Using molecular genetics, we assessed the effects that molecular entities expressed in EG and otherwise mediating two distinct mechanisms of gliotransmitter release, connexin 43 (Cx43) hemichannel vs. Ca2+ -dependent exocytosis, have on gut function. The expression of mutated Cx43G138R (which favors hemichannel, as opposed to gap-junctional activity) in EG increased gut motility in vivo, while a knock-down of Cx43 in EG resulted in the reduction of gut motility. However, inhibition of Ca2+ -dependent exocytosis in EG did not affect gut motility in vivo; rather, it increased the fecal pellet fluid content. Hampering either Cx43 expression or Ca2+ -dependent exocytosis in EG had an effect on colonic migrating motor complexes, mainly decreasing frequency and velocity of contractions ex vivo. Thus, EG can differentially modulate gut reflexes using the above two distinct mechanisms of gliotransmission.


Subject(s)
Connexin 43/metabolism , Enteric Nervous System/metabolism , Gap Junctions/metabolism , Intestinal Mucosa/metabolism , Neuroglia/metabolism , Animals , Calcium/metabolism , Mice, Inbred C57BL , Mice, Transgenic
10.
J Physiol ; 595(11): 3409-3424, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28066889

ABSTRACT

KEY POINTS: The role of enteric glial cell activity in the acute regulation of epithelial barrier and secretomotor functions of the intestines under physiological conditions is not clear. We used transgenic mice to modify glial activity and found that enteric glia significantly contribute to the neurogenic ion transport while glial activity does not appear to play a major role in the acute regulation of barrier function. The selective activation of glial activity evoked electrogenic ion transport primarily through neural pathways and was sufficient to drive electrogenic ion transport to an extent equal to the direct activation of neurogenic ion transport. These findings provide novel insight into the cellular mechanisms that control fluid transport homeostasis in the intestine and might provide novel therapeutic avenues for functional diarrheal diseases. ABSTRACT: Enteric glial cells are often implicated in the regulation of epithelial barrier and secretomotor functions of the intestines. But whether glial cell activity regulates these functions acutely under physiological conditions is not clear. We addressed this issue by using transgenic animal models to modify the activity of enteric glia, either reducing glial expression of connexin 43 in Sox10::CreERT2+/- /Cx43f/f mice or activating glial calcium responses in GFAP::hM3Dq mice, and tested the effects on colonic barrier function and electrogenic ion transport in Ussing chambers. We assessed neuronal-dependent and -independent contributions by activating or inhibiting neurogenic activity with veratridine and tetrodotoxin, respectively. Our results show that the reduction of glial Cx43 expression in Sox10::CreERT2+/- /Cx43f/f mice significantly reduced neurogenic ion transport. The selective glial activation in tissues from GFAP::hM3Dq mice evoked electrogenic ion transport to an extent equal to the direct activation of neurogenic ion transport with veratridine and glial driven responses consisted of both tetrodotoxin-sensitive and -insensitive components. The selective glial stimulation did not affect transmural ion conductance or cell-impermeant dye flux but the baseline ion conductance was more variable in Sox10::CreERT2+/- /Cx43f/f tissues. Together, our findings show that glial activity contributes to the regulation of electrogenic ion transport in the intestine through effects on neurons and possibly direct effects on epithelial cells. However, glial activity does not appear to play a major role in the acute regulation of barrier function. These findings provide novel insight into the cellular mechanisms that control fluid transport homeostasis in the intestine.


Subject(s)
Colon/metabolism , Gastrointestinal Motility , Intestinal Absorption , Intestinal Mucosa/metabolism , Neuroglia/metabolism , Animals , Colon/cytology , Colon/physiology , Connexin 43/genetics , Connexin 43/metabolism , Female , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Intestinal Mucosa/innervation , Ion Transport , Male , Mice
11.
Cereb Cortex ; 27(3): 2365-2384, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27075036

ABSTRACT

In astrocytes, the intracellular calcium (Ca2+) signaling mediated by activation of metabotropic glutamate receptor 5 (mGlu5) is crucially involved in the modulation of many aspects of brain physiology, including gliotransmission. Here, we find that the mGlu5-mediated Ca2+ signaling leading to release of glutamate is governed by mGlu5 interaction with Homer1 scaffolding proteins. We show that the long splice variants Homer1b/c are expressed in astrocytic processes, where they cluster with mGlu5 at sites displaying intense local Ca2+ activity. We show that the structural and functional significance of the Homer1b/c-mGlu5 interaction is to relocate endoplasmic reticulum (ER) to the proximity of the plasma membrane and to optimize Ca2+ signaling and glutamate release. We also show that in reactive astrocytes the short dominant-negative splice variant Homer1a is upregulated. Homer1a, by precluding the mGlu5-ER interaction decreases the intensity of Ca2+ signaling thus limiting the intensity and the duration of glutamate release by astrocytes. Hindering upregulation of Homer1a with a local injection of short interfering RNA in vivo restores mGlu5-mediated Ca2+ signaling and glutamate release and sensitizes astrocytes to apoptosis. We propose that Homer1a may represent one of the cellular mechanisms by which inflammatory astrocytic reactions are beneficial for limiting brain injury.


Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Homer Scaffolding Proteins/metabolism , Animals , Brain Ischemia/metabolism , Cations, Divalent/metabolism , Cells, Cultured , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Endoplasmic Reticulum/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homer Scaffolding Proteins/antagonists & inhibitors , Homer Scaffolding Proteins/genetics , Humans , Infant, Newborn , Male , Mice, Transgenic , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Metabotropic Glutamate/metabolism , Tissue Culture Techniques
12.
J Physiol ; 595(2): 557-570, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27106597

ABSTRACT

Glia (from Greek γλοία meaning 'glue') pertains to non-neuronal cells in the central (CNS) and peripheral nervous system (PNS) that nourish neurons and maintain homeostasis. In addition, glia are now increasingly appreciated as active regulators of numerous physiological processes initially considered exclusively under neuronal regulation. For instance, enteric glia, a collection of glial cells residing within the walls of the intestinal tract, regulate intestinal motility, a well-characterized reflex controlled by enteric neurons. Enteric glia also interact with various non-neuronal cell types in the gut wall such as enterocytes, enteroendocrine and immune cells and are therefore emerging as important local regulators of diverse gut functions. The intricate molecular mechanisms that govern glia-mediated regulation are beginning to be discovered, but much remains unknown about the functions of enteric glia in health and disease. Here we present a current view of the enteric glia and their regulatory roles in gastrointestinal (GI) (patho)physiology; from GI motility and epithelial barrier function to enteric neuroinflammation.


Subject(s)
Intestines/cytology , Intestines/physiology , Neuroglia/physiology , Animals , Gastrointestinal Motility , Humans , Neurons/physiology
13.
Mol Neurobiol ; 53(10): 6882-6896, 2016 12.
Article in English | MEDLINE | ID: mdl-26660497

ABSTRACT

In the brain, astrocytes signal to neighboring cells via regulated exocytotic release of gliosignaling molecules, such as brain-derived neurotrophic factor (BDNF). Recent studies uncovered a role of ketamine, an anesthetic and antidepressant, in the regulation of BDNF expression and in the disruption of astrocytic Ca2+ signaling, but it is unclear whether it affects astroglial BDNF release. We investigated whether ketamine affects ATP-evoked Ca2+ signaling and exocytotic release of BDNF at the single-vesicle level in cultured rat astrocytes. Cells were transfected with a plasmid encoding preproBDNF tagged with the pH-sensitive fluorescent protein superecliptic pHluorin, (BDNF-pHse) to load vesicles and measure the release of BDNF-pHse when the exocytotic fusion pore opens and alkalinizes the luminal pH. In addition, cell-attached membrane capacitance changes were recorded to monitor unitary vesicle interaction with the plasma membrane. Intracellular Ca2+ activity was monitored with Fluo-4 and confocal microscopy, which was also used to immunocytochemically characterize BDNF-pHse-laden vesicles. As revealed by double-fluorescent micrographs, BDNF-pHse localized to vesicles positive for the soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor (SNARE) proteins, vesicle-associated membrane protein 2 (VAMP2), VAMP3, and synaptotagmin IV. Ketamine treatment decreased the number of ATP-evoked BDNF-pHse fusion/secretion events (P < 0.05), the frequency of ATP-evoked transient (P < 0.001) and full-fusion exocytotic (P < 0.05) events, along with a reduction in the ATP-evoked increase in intracellular Ca2+ activity in astrocytes by ~70 % (P < 0.001). The results show that ketamine treatment suppresses ATP-triggered vesicle fusion and BDNF secretion by increasing the probability of a narrow fusion pore open state and/or by reducing astrocytic Ca2+ excitability.


Subject(s)
Adenosine Triphosphate/pharmacology , Astrocytes/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cytoplasmic Vesicles/metabolism , Exocytosis/drug effects , Ketamine/pharmacology , Animals , Astrocytes/drug effects , Biomarkers/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Hydrogen-Ion Concentration , Membrane Fusion/drug effects , Purines/metabolism , Rats, Wistar , SNARE Proteins/metabolism , Subcellular Fractions/metabolism
15.
Autism Res ; 8(5): 629-33, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25728630

ABSTRACT

Pitt-Hopkins syndrome (PTHS) is a neurodevelopmental disorder, classified as an autism spectrum disorder that is caused by the haploinsufficiency of Transcription Factor 4 (TCF4). The most common non-neurological symptoms in PTHS patients are gastrointestinal (GI) disturbances, mainly gastroesophageal reflux and severe constipation (in about 30 and 75% of PTHS patients, respectively). We hypothesized that the recently recognized mouse model of PTHS will exhibit problems with their gut function. We conducted series of in vivo tests on 15- to 19- week old male mice, heterozygous for the TCF4 functional deletion, mimicking the TCF4 haploinsufficiency in PTHS patients, and their wild type littermates. Data collection and initial analysis were performed blindly, that is, the genotyping key was received after the mean values were calculated for each individual animal, and then mean/median of each group was subsequently calculated. Body weight, fecal pellet output, and fluid content were similar between the groups, indicating normal gross growth of PTHS mice and their overall physiological GI motility and intestinal secretion/absorption. There were no significant differences in gut length and gross appearance pointing out that PTHS mice have normal gut in gross anatomical terms. However, the assessment of gut transit indicates that, while whole-gut transit velocity was similar between the groups, the upper GI and distal colon transit velocities were significantly reduced in the PTHS mice. This is the first evidence of specific gut related problems in the PTHS mice. Our study also validates the TCF4 functional knockout mice as an animal model to study PTHS-associated GI disturbances.


Subject(s)
Gastrointestinal Transit/physiology , Hyperventilation/physiopathology , Intellectual Disability/physiopathology , Animals , Disease Models, Animal , Facies , Male , Mice , Mice, Knockout
16.
Biomaterials ; 43: 50-60, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25591961

ABSTRACT

We previously used the expression of various combinations and configurations of MyoD and E12, two basic helix-loop-helix transcription factors (TF), to produce populations of myotubes assuming distinct morphology, myofibrillar development and Ca2+ dynamics, from mammalian C2C12 myoblasts in non-differentiation growth conditions. Here, we assessed the synthetically generated myotubes in terms of energetics, otherwise necessary to sustain their mechanical output as bio-actuators. We found that the myotubes exhibit changed expression of key regulators for the uptake and utilization of two major cellular fuels, glucose and lactate. Furthermore, while lactate transport was uniformly slowed in all the populations of myotubes, glucose uptake and utilization were modified by particular TF configuration. Our approach allows the production of a class of biomaterials with predetermined energetics that could be applied in biorobotics, where fuel of choice could be used, and also in reparative medicine where, for example, particular population of myotubes could be additionally employed as glucose sinks to mitigate effects of secondary metabolic syndrome.


Subject(s)
Glucose/metabolism , Lactic Acid/metabolism , Muscle Fibers, Skeletal/metabolism , MyoD Protein/metabolism , Transcription Factor 3/metabolism , Animals , Biocompatible Materials/chemistry , Biological Transport , Calcium/metabolism , Glucose Transporter Type 4/metabolism , Immunohistochemistry , Metabolic Syndrome/metabolism , Mice , Muscles/metabolism , Myofibrils/metabolism , Plasmids/metabolism , Stem Cells/metabolism
17.
ACS Appl Mater Interfaces ; 7(1): 184-92, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25511125

ABSTRACT

Botulinum neurotoxins (BoNTs) are the most lethal toxin known to human. Biodefense requires early and rapid detection of BoNTs. Traditionally, BoNTs can be detected by looking for signs of botulism in mice that receive an injection of human material, serum or stool. While the living animal assay remains the most sensitive approach, it is costly, slow and associated with legal and ethical constrains. Various biochemical, optical and mechanical methods have been developed for BoNTs detection with improved speed, but with lesser sensitivity. Here, we report a novel nanopore-based BoNT type B (BoNT-B) sensor that monitors the toxin's enzymatic activity on its substrate, a recombinant synaptic protein synaptobrevin 2 derivative. By analyzing the modulation of the pore current caused by the specific BoNT-B-digested peptide as a marker, the presence of BoNT-B at a subnanomolar concentration was identified within minutes. The nanopore detector would fill the niche for a much needed rapid and highly sensitive detection of neurotoxins, and provide an excellent system to explore biophysical mechanisms for biopolymer transportation.


Subject(s)
Biosensing Techniques/methods , Botulinum Toxins, Type A/chemistry , Peptides/chemistry , Vesicle-Associated Membrane Protein 2/chemistry , Animals , Biocatalysis , Biosensing Techniques/instrumentation , Digestion , Isomerism , Nanopores , Rats
18.
Nat Commun ; 5: 3780, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24807050

ABSTRACT

Exocytic transmitter release is regulated by the SNARE complex, which contains a vesicular protein, synaptobrevin2 (Sb2). However, Sb2 vesicular arrangement is unclear. Here we use super-resolution fluorescence microscopy to study the prevalence and distribution of endogenous and exogenous Sb2 in single vesicles of astrocytes, the most abundant glial cells in the brain. We tag Sb2 protein at C- and N termini with a pair of fluorophores, which allows us to determine the Sb2 length and geometry. To estimate total number of Sb2 proteins per vesicle and the quantity necessary for the formation of fusion pores, we treat cells with ATP to stimulate Ca2+-dependent exocytosis, increase intracellular alkalinity to enhance the fluorescence presentation of yellow-shifted pHluorin (YpH), appended to the vesicle lumen domain of Sb2, and perform photobleaching of YpH fluorophores. Fluorescence intensity analysis reveals that the total number of endogenous Sb2 units or molecules per vesicle is ≤25.


Subject(s)
Astrocytes/physiology , Brain/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Adenosine Triphosphate/pharmacology , Animals , Brain/cytology , Cells, Cultured , Exocytosis/physiology , Female , Green Fluorescent Proteins , Membrane Fusion , Microscopy, Fluorescence , Photobleaching , Rats , SNARE Proteins/metabolism , Transport Vesicles/physiology
19.
Biomaterials ; 35(7): 2188-98, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24360578

ABSTRACT

We used a synthetic biology approach to produce myotubes from mammalian C2C12 myoblasts in non-differentiation growth conditions using the expression of basic helix-loop-helix transcription factors, MyoD and E12, in various combinations and configurations. Our approach not only recapitulated the basics of muscle development and physiology, as the obtained myotubes showed qualities similar to those seen in striated muscle fibers in vivo, but also allowed for the synthesis of populations of myotubes which assumed distinct morphology, myofibrillar development and Ca(2+) dynamics. This fashioned class of biomaterials is suitable for the building blocks of soft actuators in micro-scale biomimetic robotics. This production line strategy can be embraced in reparative medicine as synthetic human myotubes with predetermined morphological/functional properties could be obtained using this very approach. This methodology can be adopted beyond striated muscle for the engineering of other tissue components/cells whose differentiation is governed by the principles of basic helix-loop-helix transcription factors, as in the case, for example, of neural or immune cell types.


Subject(s)
Cell Differentiation , Muscle Fibers, Skeletal/metabolism , MyoD Protein/physiology , Transcription Factor 3/physiology , Transcription Factors/metabolism , Animals , Cell Line , Mice
20.
Gastroenterology ; 146(2): 497-507.e1, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24211490

ABSTRACT

BACKGROUND & AIMS: In the enteric nervous system, neurotransmitters initiate changes in calcium (Ca(2+) responses) in glia, but it is not clear how this process affects intestinal function. We investigated whether Ca(2+)-mediated responses in enteric glia are required to maintain gastrointestinal function. METHODS: We used in situ Ca(2+) imaging to monitor glial Ca(2+) responses, which were manipulated with pharmacologic agents or via glia-specific disruption of the gene encoding connexin-43 (Cx43) (hGFAP::CreER(T2+/-)/Cx43(f/f) mice). Gastrointestinal function was assessed based on pellet output, total gut transit, colonic bead expulsion, and muscle tension recordings. Proteins were localized and quantified by immunohistochemistry, immunoblot, and reverse transcription polymerase chain reaction analyses. RESULTS: Ca(2+) responses in enteric glia of mice were mediated by Cx43 hemichannels. Cx43 immunoreactivity was confined to enteric glia within the myenteric plexus of the mouse colon; the Cx43 inhibitors carbenoxolone and 43Gap26 inhibited the ability of enteric glia to propagate Ca(2+) responses. In vivo attenuation of Ca(2+) responses in the enteric glial network slowed gut transit overall and delayed colonic transit--these changes are also observed during normal aging. Altered motility with increasing age was associated with reduced glial Ca(2+)-mediated responses and changes in glial expression of Cx43 messenger RNA and protein. CONCLUSIONS: Ca(2+)-mediated responses in enteric glia regulate gastrointestinal function in mice. Altered intercellular signaling between enteric glia and neurons might contribute to motility disorders.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Colon/physiology , Connexin 43/metabolism , Enteric Nervous System/physiology , Gastrointestinal Transit/physiology , Neuroglia/metabolism , Animals , Biomarkers/metabolism , Blotting, Western , Connexin 43/antagonists & inhibitors , Connexin 43/deficiency , Connexin 43/genetics , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...