Haploinsufficient TNAP Mice Display Decreased Extracellular ATP Levels and Expression of Pannexin-1 Channels

Hypophosphatasia (HPP) is a rare heritable metabolic bone disease caused by hypomorphic mutations in the ALPL (in human) or Akp2 (in mouse) gene, encoding the tissue-nonspecific alkaline phosphatase (TNAP) enzyme. In addition to skeletal and dental malformations, severe forms of HPP are also characterized by the presence of spontaneous seizures. Initially, these seizures were attributed to an impairment of GABAergic neurotransmission caused by altered vitamin B6 metabolism. However, recent work by our group using knockout mice null for TNAP (TNAP-/-), a well-described model of infantile HPP, has revealed a deregulation of purinergic signaling contributing to the seizure phenotype. In the present study, we report that adult heterozygous (TNAP+/-) transgenic mice with decreased TNAP activity in the brain are more susceptible to adenosine 5′-triphosphate (ATP)-induced seizures. Interestingly, when we analyzed the extracellular levels of ATP in the cerebrospinal fluid, we found that TNAP+/- mice present lower levels than control mice. To elucidate the underlying mechanism, we evaluated the expression levels of other ectonucleotidases, as well as different proteins involved in ATP release, such as pannexin, connexins, and vesicular nucleotide transporter. Among these, Pannexin-1 (Panx1) was the only one showing diminished levels in the brains of TNAP+/- mice. Altogether, these findings suggest that a physiological regulation of extracellular ATP levels and Panx1 changes may compensate for the reduced TNAP activity in this model of HPP.

[1]  Álvaro Sebastián‐Serrano,et al.  Neuronal P2X7 Receptor: Involvement in Neuronal Physiology and Pathology , 2017, The Journal of Neuroscience.

[2]  Hui Yang,et al.  Expression of pannexin 1 and 2 in cortical lesions from intractable epilepsy patients with focal cortical dysplasia , 2016, Oncotarget.

[3]  Álvaro Sebastián‐Serrano,et al.  Neurodevelopmental alterations and seizures developed by mouse model of infantile hypophosphatasia are associated with purinergic signalling deregulation , 2016, Human molecular genetics.

[4]  G. Burnstock An introduction to the roles of purinergic signalling in neurodegeneration, neuroprotection and neuroregeneration , 2016, Neuropharmacology.

[5]  J. Millán,et al.  Alkaline Phosphatase and Hypophosphatasia , 2015, Calcified Tissue International.

[6]  D. Wenkert,et al.  Hypophosphatasia: validation and expansion of the clinical nosology for children from 25 years experience with 173 pediatric patients. , 2015, Bone.

[7]  N. Dale,et al.  Pannexin-1-mediated ATP release from area CA3 drives mGlu5-dependent neuronal oscillations , 2015, Neuropharmacology.

[8]  Â. R. Tomé,et al.  ATP as a multi-target danger signal in the brain , 2015, Front. Neurosci..

[9]  C. Cepeda,et al.  In Rasmussen Encephalitis, Hemichannels Associated with Microglial Activation are linked to Cortical Pyramidal Neuron Coupling: A Possible Mechanism for Cellular Hyperexcitability , 2015, CNS neuroscience & therapeutics.

[10]  G. Enikolopov,et al.  NTPDase2 and Purinergic Signaling Control Progenitor Cell Proliferation in Neurogenic Niches of the Adult Mouse Brain , 2015, Stem cells.

[11]  Álvaro Sebastián‐Serrano,et al.  Tissue-nonspecific Alkaline Phosphatase Regulates Purinergic Transmission in the Central Nervous System During Development and Disease , 2014, Computational and structural biotechnology journal.

[12]  B. Xiao,et al.  Altered expression of pannexin proteins in patients with temporal lobe epilepsy. , 2013, Molecular medicine reports.

[13]  G. Burnstock Introduction to purinergic signalling in the brain. , 2013, Advances in experimental medicine and biology.

[14]  D. Manahan‐Vaughan,et al.  Pannexin1 Stabilizes Synaptic Plasticity and Is Needed for Learning , 2012, PloS one.

[15]  B. Sperlágh,et al.  K+ depolarization evokes ATP, adenosine and glutamate release from glia in rat hippocampus: a microelectrode biosensor study , 2012, British journal of pharmacology.

[16]  D. Henshall,et al.  Seizure suppression and neuroprotection by targeting the purinergic P2X7 receptor during status epilepticus in mice , 2012, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[17]  L. A. Swayne,et al.  Pannexin 1 regulates postnatal neural stem and progenitor cell proliferation , 2012, Neural Development.

[18]  J. Velíšková,et al.  Targeting Pannexin1 Improves Seizure Outcome , 2011, PloS one.

[19]  Ji-Eun Kim,et al.  The P2X7 receptor-pannexin-1 complex decreases muscarinic acetylcholine receptor-mediated seizure susceptibility in mice. , 2011, The Journal of clinical investigation.

[20]  Roger J. Thompson,et al.  Pannexin channels are not gap junction hemichannels , 2011, Channels.

[21]  E. Lazarowski,et al.  Molecular mechanisms of purine and pyrimidine nucleotide release. , 2011, Advances in pharmacology.

[22]  Michael R. Elliott,et al.  Pannexin 1 channels mediate ‘find-me’ signal release and membrane permeability during apoptosis , 2010, Nature.

[23]  Roger J. Thompson,et al.  Supporting Online Material Materials and Methods Som Text Figs. S1 to S6 References Activation of Pannexin-1 Hemichannels Augments Aberrant Bursting in the Hippocampus , 2022 .

[24]  J. Schrader,et al.  Distribution of ectonucleotidases in the rodent brain revisited , 2008, Cell and Tissue Research.

[25]  N. Takahashi,et al.  Maxi-anion channel as a candidate pathway for osmosensitive ATP release from mouse astrocytes in primary culture , 2008, Cell Research.

[26]  Keisuke Sawada,et al.  Identification of a vesicular nucleotide transporter , 2008, Proceedings of the National Academy of Sciences.

[27]  D. Laird,et al.  Pannexin 1 and pannexin 3 are glycoproteins that exhibit many distinct characteristics from the connexin family of gap junction proteins , 2007, Journal of Cell Science.

[28]  J. Sévigny,et al.  Specificity of the ecto‐ATPase inhibitor ARL 67156 on human and mouse ectonucleotidases , 2007, British journal of pharmacology.

[29]  D. Spray,et al.  Connexin and pannexin mediated cell-cell communication. , 2007, Neuron glia biology.

[30]  N. Chaudhari,et al.  The role of pannexin 1 hemichannels in ATP release and cell–cell communication in mouse taste buds , 2007, Proceedings of the National Academy of Sciences.

[31]  H. Zimmermann Ectonucleotidases in the nervous system. , 2008, Novartis Foundation symposium.

[32]  G. Dubyak ATP Release Mechanisms , 2006 .

[33]  G. Burnstock,et al.  Nucleotides and regulation of bone cell function , 2006 .

[34]  J. Millán Structure, substrate specificity and functional relatedness to other members of a large superfamily of enzymes , 2006 .

[35]  L. Leung,et al.  Epilepsy-based changes in hippocampal excitability: causes and effects. , 2006, Advances in neurology.

[36]  S. Narisawa,et al.  Abnormal vitamin B6 metabolism in alkaline phosphatase knock‐out mice causes multiple abnormalities, but not the impaired bone mineralization , 2001, The Journal of pathology.

[37]  J. Rash,et al.  Connexins and gap junctions of astrocytes and oligodendrocytes in the CNS , 2000, Brain Research Reviews.

[38]  J. Millán,et al.  Inactivation of two mouse alkaline phosphatase genes and establishment of a model of infantile hypophosphatasia , 1997, Developmental dynamics : an official publication of the American Association of Anatomists.

[39]  T. Guilarte,et al.  Mice lacking tissue non–specific alkaline phosphatase die from seizures due to defective metabolism of vitamin B–6 , 1995, Nature Genetics.

[40]  L. Kerr,et al.  Mefloquine prophylaxis: an overview of spontaneous reports of severe psychiatric reactions and convulsions. , 1992, The Journal of tropical medicine and hygiene.

[41]  H. Wiśniewski,et al.  Localization of alkaline phosphatase activity in endothelia of developing and mature mouse blood-brain barrier. , 1986, Developmental neuroscience.