2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Pannexin 1 Transgenic Mice: Human Diseases and Sleep-Wake Function Revision

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          In humans and other vertebrates pannexin protein family was discovered by homology to invertebrate gap junction proteins. Several biological functions were attributed to three vertebrate pannexins members. Six clinically significant independent variants of the PANX1 gene lead to human infertility and oocyte development defects, and the Arg217His variant was associated with pronounced symptoms of primary ovarian failure, severe intellectual disability, sensorineural hearing loss, and kyphosis. At the same time, only mild phenotypes were observed in Panx1 knockout mice. In addition, a passenger mutation was identified in a popular line of Panx1 knockout mice, questioning even those effects. Using CRISPR/Cas9, we created a new line of Panx1 knockout mice and a new line of mice with the clinically significant Panx1 substitution (Arg217His). In both cases, we observed no significant changes in mouse size, weight, or fertility. In addition, we attempted to reproduce a previous study on sleep/wake and locomotor activity functions in Panx1 knockout mice and found that previously reported effects were probably not caused by the Panx1 knockout itself. We consider that the pathological role of Arg217His substitution in Panx1, and some Panx1 functions in general calls for a re-evaluation.

          Related collections

          Most cited references55

          • Record: found
          • Abstract: found
          • Article: not found

          High-throughput profiling of off-target DNA cleavage reveals RNA-programmed Cas9 nuclease specificity

          The RNA-programmable Cas9 endonuclease cleaves double-stranded DNA at sites complementary to a 20-base-pair guide RNA. The Cas9 system has been used to modify genomes in multiple cells and organisms, demonstrating its potential as a facile genome-engineering tool. We used in vitro selection and high-throughput sequencing to determine the propensity of eight Cas9:guide RNA complexes to cleave each of 10^12 potential off-target DNA sequences. The selection results predicted five off-target sites in the human genome that were confirmed to undergo genome cleavage in HEK293T cells upon expression of one of two Cas9:guide RNA complexes. In contrast to previous models, our results show that Cas9:guide RNA specificity extends past a 7- to 12-base pair seed sequence. Our results also suggest a tradeoff between activity and specificity both in vitro and in cells as a shorter, less-active guide RNA is more specific then a longer, more-active guide RNA. High concentrations of Cas9:guide RNA complexes can cleave off-target sites containing mutations near or within the PAM that are not cleaved when enzyme concentrations are limiting.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Connexin 26 mutations in hereditary non-syndromic sensorineural deafness.

            Severe deafness or hearing impairment is the most prevalent inherited sensory disorder, affecting about 1 in 1,000 children. Most deafness results from peripheral auditory defects that occur as a consequence of either conductive (outer or middle ear) or sensorineuronal (cochlea) abnormalities. Although a number of mutant genes have been identified that are responsible for syndromic (multiple phenotypic disease) deafness such as Waardenburg syndrome and Usher 1B syndrome, little is known about the genetic basis of non-syndromic (single phenotypic disease) deafness. Here we study a pedigree containing cases of autosomal dominant deafness and have identified a mutation in the gene encoding the gap-junction protein connexin 26 (Cx26) that segregates with the profound deafness in the family. Cx26 mutations resulting in premature stop codons were also found in three autosomal recessive non-syndromic sensorineuronal deafness pedigrees, genetically linked to chromosome 13q11-12 (DFNB1), where the Cx26 gene is localized. Immunohistochemical staining of human cochlear cells for Cx26 demonstrated high levels of expression. To our knowledge, this is the first non-syndromic sensorineural autosomal deafness susceptibility gene to be identified, which implicates Cx26 as an important component of the human cochlea.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Pannexins, a family of gap junction proteins expressed in brain.

              Database search has led to the identification of a family of proteins, the pannexins, which share some structural features with the gap junction forming proteins of invertebrates and vertebrates. The function of these proteins has remained unclear so far. To test the possibility that pannexins underlie electrical communication in the brain, we have investigated their tissue distribution and functional properties. Here, we show that two of these genes, pannexin 1 (Px1) and Px2, are abundantly expressed in the CNS. In many neuronal cell populations, including hippocampus, olfactory bulb, cortex and cerebellum, there is coexpression of both pannexins, whereas in other brain regions, e.g., white matter, only Px1-positive cells were found. On expression in Xenopus oocytes, Px1, but not Px2 forms functional hemichannels. Coinjection of both pannexin RNAs results in hemichannels with functional properties that are different from those formed by Px1 only. In paired oocytes, Px1, alone and in combination with Px2, induces the formation of intercellular channels. The functional characteristics of homomeric Px1 versus heteromeric Px1/Px2 channels and the different expression patterns of Px1 and Px2 in the brain indicate that pannexins form cell type-specific gap junctions with distinct properties that may subserve different functions.
                Bookmark

                Author and article information

                Contributors
                Role: Academic Editor
                Journal
                Int J Mol Sci
                Int J Mol Sci
                ijms
                International Journal of Molecular Sciences
                MDPI
                1422-0067
                17 May 2021
                May 2021
                : 22
                : 10
                : 5269
                Affiliations
                [1 ]Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; korablevalexeyn@ 123456gmail.com (A.K.); irina_serova2004@ 123456mail.ru (I.S.)
                [2 ]Laboratory of Mammal Behavior and Behavioral Ecology, Severtsov Institute Ecology and Evolution, Russian Academy of Sciences, 119071 Moscow, Russia; kovalzon@ 123456sevin.ru
                [3 ]Laboratory for the Study of Information Processes at the Cellular and Molecular Levels, Institute for Information Transmission Problems, Russian Academy of Sciences, 119333 Moscow, Russia; kcyu@ 123456yandex.ru (O.O.K.); ypanchin@ 123456yahoo.com (Y.P.)
                [4 ]Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia; marta.peckovva@ 123456gmail.com (M.G.P.); k.bogotskoy@ 123456yandex.com (K.A.B.); ost.msu@ 123456gmail.com (O.S.T.)
                [5 ]Department of Mathematical Methods in Biology, Belozersky Institute, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
                Author notes
                [* ]Correspondence: battulin@ 123456gmail.com
                Author information
                https://orcid.org/0000-0003-0611-0203
                https://orcid.org/0000-0002-0941-2242
                Article
                ijms-22-05269
                10.3390/ijms22105269
                8155943
                50ac8b2f-8aef-4b2c-a2a9-ac2084976e20
                © 2021 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( https://creativecommons.org/licenses/by/4.0/).

                History
                : 08 April 2021
                : 14 May 2021
                Categories
                Article

                Molecular biology
                pannexin 1,crispr/cas9 method,sleep-wake cycle
                Molecular biology
                pannexin 1, crispr/cas9 method, sleep-wake cycle

                Comments

                Comment on this article