fbpx
Wikipedia

SNX8

The SNX8 is a sorting nexin protein involved in intracellular molecular traffic from the early endosomes to the TGN. It is suggested that it acts as an adaptor protein in events related to immune response and cholesterol regulation, for example. As a protein of the SNXs family, the SNX8 is formed of 465 aminoacids and presents a BAR-domain and a PX-domain which are very relevant in relation to its functions. Furthermore, SNX8 study is motivated by its medical significance in relation to diseases such as Alzheimer's Disease, cancer, neurodevelopmental malformations and to its role in fighting against viral infections.

Tertiary structure of the SNX9 which gives an idea of the tertiary structure of the SNX8, since primary structure shares 20% sequence identity and 0.3 sequence similarity with SNX8 primary structure.[1]

Structure edit

Sorting nexins (SNXs) edit

SNX8 belongs to the sorting nexin family of proteins, which mainly contain two functional membrane-binding that allow SNXs to have different roles in endosomal sorting and protein trafficking thanks to its membrane curvature ability.[2] To begin with, SNX-PX is a distinct phosphoinositide (PI)-binding domain. The preferential interaction of this domain with membrane lipids makes the main function of SNX-PX the targeting of proteins to phosphatidylinositol-3-phosphate (PI(3)P) to endosomes. On the other hand, the BAR (Bin/amphiphysin/Rvs) domain is a key regulator of phosphoinositide-mediated, tubular-based endosomal sorting. Accordingly, this domain also dimerizes to sense, stabilize and induce membrane curvature. The SNX-BAR proteins that contain both domains are a part of phosphoinositide-enriched, high-curvature tubular micro-domains of the endo-lysosomal network.[3]

The mammalian genome contains 12 genes coding for SNX-BAR proteins (SNX1, SNX2, SNX4, SNX9, SNX18, SNX32 and SNX33). Other domains, such as PDZ (postsynaptic density protein-95, discs-large, zona occludens-1), SH3 (Src homology 3) and RA (Ras-associated), are involved in protein-protein interactions.

SNX8 edit

 
SNX8 primary structure highlighting relevant domains and amino acids.[2][4][5]

The SNX8 protein, even though is very similar to the other sorting nexins, presents a domain structure which resembles the most to SNX1's and SNX9's; for this reason, although its terciary structure remains unknown, it theoretically resembles that of SNX9 shown in the model above. Overall, the SNX8 protein is integrated by one unique peptide chain that has 465 amino acids with a molecular mass of 52.569 Da.[2]

PX Domain-containing N-terminus edit

SNX8 contains a PX domain in its N-terminus, which is located between amino acids 71 and 181. A homology domain with yeast's PX domain is localized between amino acids 75 and 178 within this same domain. As it is a phosphoinositide-binding domain, it is important to highlight amino acids 109, 135 and 148 as residues directly related to phosphatidylinositol 3-phosphate since being specific binding sites, constituting a phosphoinositid binding site with a span of 40 amino acids.[2][4] Furthermore, it includes a pair of phosphorylable tyrosines in positions 95 and 126 that are key in its function in the IFNγ-triggered IKKβ-mediated noncanonical signaling pathway.[5] Overall, the PX domain main function is to target SNX8 mainly to early endosomes and other membranes rich in phosphatidylinositol 3-phosphate phospholipids.[6]

BAR Domain-containing C-terminus edit

SNX contains a BAR domain in its C-terminus, which is located between amino acids 256 and 440. Its ability to form coatings in membranes in order to induce their curvature is key in SNX8 participation in tubular structures formation. Furthermore, SNX8 C-terminus contains a threonine in position 452 and a serine in position 456 which can go through post-traductional changes that induces its phosphorylation, resulting in a phosphothreonine and a phosphoserine. Therefore, there are classified as phosphorylation sites.[6]

MVP1 ortholog edit

SNX8 has a yeast ortholog protein, the MVP1 encoded by the also homolog gene Mvp1p, which also plays a role by mediating transport of cargo to the vacuolar and lysosomal compartments. For this reason, its investigation can lead to a better understanding of SNX8 functions in human cells.[7]

Biological functions and its molecular mechanisms edit

SNX8 is thought to be an adaptor protein involved in the endosome-to-Golgi transport pathway, participating in endocytosis and endosomal sorting and signaling. It downregulates retrograde transport of intracellular proteins from the early endosome compartment to the trans-Golgi network in a retromer-mediated manner. SNX8 is therefore localized in early endosomes, as its colocalization with components of the retromer such as SNX1, SNX2, Vps26 and Vps35 has been demonstrated by some studies (and also with EEA1).[7] Furthermore, the dynamics of endosomal structures with SNX8-enriched membrane domains are regulated by the opposite motor proteins dynein-1 containing LIC1 and kinesin-1, both of which allow SNX8-mediated cargo movement through the cytosol by exerting forces on these structures.[8] The biological functions of SNX8 that have been studied, all of which involve its role in intracellular endosomal transport, are explained in more detail in the following sections.

Innate immune response edit

Cytosolic RNA virus-triggered response edit

SNX8 is a key component in the induction of downstream effector antiviral gene transcription in response to RNA viruses, as it is necessary for RLR-mediated activation of the IFNβ promoter that leads to host defense in a dose-dependent manner. It positively regulates the aggregation and activation of VISA, a key adaptor protein involved in the innate immune response against this type of infections; this process is critical in the recruitment of other signaling components. Although SNX8's standard localization includes the cytoplasm and other compartments such as the ER, the ER-Golgi intermediate compartment, the Golgi apparatus or endosomes, it is partially localized at mitochondria. Furthermore, RNA viral infections cause the translocation of SNX8 from the cytosol to the mitochondria. During the early stage of the infection, SNX8 association with VISA increases above constitutive levels (degradation of VISA in later stages results in a reduction of this phenomenon). Structurally, the C-terminus transmembrane domain of VISA and both N-terminus PX domain and C-terminus of SNX8 are necessary for this linkage. The formation of the SNX8-VISA complex potentiates VISA prion-like polymerization and aggregation. After VISA activation, its accumulation allows the recruitment of signaling components which play a role in RLR-mediated antiviral response activation such as the intermediates of this pathway TBK1, IRF3, IκBα or ISRE. IRF3 cooperative activation with NF-κB transcription factor through phosphorylation leads to the induction of the IFNβ promoter transcription. Examples of genes whose RNA virus-triggered expression is stimulated by SNX8 are IFNB1, ISG56 and IL6 (being IL6 and IFNB1 related to cytokine secretion).[9]

In addition, SNX8 also plays a role in RIG-I containing CARD domain-mediated and MDA5-mediated activation of the IFNβ promoter, since VISA works as an intermediate for both signaling pathways. The mechanism for SNX8 recruitment to VISA remains unclear, although two options have been suggested: translocation of viral RNA-bound RIG-I or MDA5 to VISA may result in a conformational switch that would increase its affinity for SNX8 or the RNA virus may induce post-translational modifications of one of these proteins allowing the translocation of SNX8 to mitochondria for its interaction with VISA.[9]

Cytosolic DNA virus-triggered response edit

 
Simplified scheme of DNA-triggered SNX8-mediated association of MITA and VPS34, and its intracellular transport pathway from RE to perinuclear microsomes via Golgi apparatus.[10]

SNX8 is a key component in the induction of downstream effector antiviral gene transcription in response to DNA viruses , as it is necessary for MITA-mediated activation of the IFNβ promoter in a dose-dependent manner. It regulates the activation of MITA, a central adaptor protein in the innate immune response to DNA viral infections that activates and onsets the antiviral response, by allowing its association with the class III phosphatylinositol 3 kinase VPS34-containing translocon machinery to form the MITA-Vps34 translocation complex; this linkage is crucial for MITA transport from the ER to perinuclear microsomal punctuate structures, which are induced by infection of viruses such as HSV-1, via Golgi mediated intracellular traffic. This MITA transport pathway is vital for the immune response to start. SNX8 is suggested to play a role in MITA's activation through this transport pathway, modulating its phosphorylation at serine in position 366 and recruiting the transcription factor IRF3 whose activation is important in the activation of the IFNβ promoter transcription. This hypothesis is supported by the fact that MITA, SNX8 and VPS34 colocalize in the cytoplasm, and that SNX8 localization in ER, ER-Golgi intermediate compartment, Golgi and endosomes is similar to that of MITA. Examples of genes whose DNA virus-triggered expression is stimulated by SNX8 are IFNB1, ISG56, CXCL10 and IL6 (being IFNB1 and IL6 related to cytokine secretion).[10]

IFNγ-triggered IKKβ-mediated noncanonical signaling pathway edit

SNX8 is a component of an IKKβ-mediated noncanonical signaling pathway triggered by the interferon gamma, which takes places parallelly to the more known IFNγ-induced JAK-STAT1 mediated pathway. Overall, it participates in a series of chemical reactions and molecular interactions that lead to the selective promotion of a particular subset of downstream effector gene transcription that encode IFNγ-induced GTPases of the GBP family, which participate in host defense against intracellular pathogens, and secretion proteins of the chemokine family such as CXCL9, CXCL10, and CXCL11 which present direct antimicrobial activity. In addition, SNX8 enhances the IFNγ-induced activation of the IRF1 promoter in a dose-dependent manner.[5]

 
Simplified scheme of SNX8 participation in the IFNγ-triggered IKKβ-mediated noncanonical signaling pathway (autophosphorylation not shown).[5]

Specifically, the steps of this pathway that are regulated by SNX8 are the following. The reception of IFNγ activates Janus kinase 1, resulting in the stimulation of its association with Sxn8 above standard constitutive levels. The link between these two protein within the JAK1-SNX8 complex allows JAK1 to catalyse SNX8's tyrosines phosphorylation in positions 95 and 126. This phosphorylation activates the JAK1-SNX, and the SNX8 acts as an adaptor or scaffolding protein by permitting the recruitment of the inhibitor of nuclear factor-kappa-B kinase subunit beta (IKKβ) to JAK1 in the JAK1-SNX8-IKKβ complex. This linkage is essential for IKKβ activation through autophosphorylation at serine in position 177 (as SNX8 lacks enzymatic activity) and further dimerization and oligomerization.[5]

Cholesterol regulation edit

SNX8 protein regulates cholesterol levels as an activator of the SREBPs (Sterol Regulatory Element Binding Proteins), which is a family of transcription factors that control the expression of enzymes needed for the synthesis and uptake of fatty acids, endogenous cholesterol, triacylglycerides and phospholipids; this results in an overall regulation of intracellular lipid homeostasis. Although its precise mechanism of action remains unknown, data suggests that SNX8 produces changes in cholesterol distribution through regulation of the SREBP transcriptional activity by modulating intracellular traffic events rather than by interacting with proteins of the SREBP pathway like INSIG or SREBP cleavage-activating protein (SCAP). For example, it is unclear if SNX8 has a direct participation in the transport of SREBP pathway components or if it regulates endosomal and lysosomal compartmentalization through the production of cholesterol cargoes. This last possibility is supported by the fact that the ability of altering membrane curvature is shared by some proteins of the SNXs family.[3]

It has been observed that variation in cholesterol levels alter SNX8 transcription: it remains unaltered in conditions of moderate high concentrations of cholesterol, and it experiments a decrease in conditions of restricted levels of cholesterol due to the action of the fungus-derived cholesterol-lowering statin mevinolin and the drug U18666a which promotes cytosolic cholesterol clustering within the endosomal and lysosomal compartments by acting as an intracellular cholesterol transport inhibitor.[11] On the other hand, SNX8 overexpression induces intracellular clustering of cholesterol under conditions of high cellular cholesterol levels and aggravates abnormal distribution of cholesterol. In relation to the SREBP pathway, SNX8 is not able to successfully reduce the inhibitory impact of cholesterol on SREBP-mediated transcription, but it has an important effect that counteracts the block of this pathway triggered by the insulin-induced gene INSIG.[3]

Intracellular non-amyloidogenic APP traffic edit

 
Simplified scheme of APP traffic and its amyloidogenic and non-amyloidogenic proteolytic degradative pathways[12]

The amyloid precursor protein (APP) is consecutively transported from the ER after its synthesis to the plasma membrane via the trans-Golgi network; during this traffic, the neuroprotective soluble fragment sAPPα is produced as a product of APP cleavage by α-secretases. Lately, APP that reaches the membrane without being severed can be internalized into endosomes in order to be proteolytically processed through two parallel pathways: an amyloidogenic pathway via acidified late endosomes and a non-amyloidogenic or anti-myloidogenic retrograde pathway via Golgi apparatus. The amyloidogenic pathway leads to APP processing by γ-secretases and β-secretases such as BACE1, resulting in production of the neurotoxic amyloid beta (Aβ) peptide that accumulates in extracellular locations forming senile plaques.[12]

SNX8 promotes non-amyloidogenic transport from the Golgi apparatus to other cellular locations, leading to an increase of APP levels, a stimulated distribution throughout the outer face of cell membrane, an enhanced sAPPα secretion and a reduced production (specifically, production of Aβ40 and Aβ42) . In addition, SNX8 improves APP stability, which is responsible for a longer lifespan, without having any effect in its mRNA transcription. This hypothesis is supported by the fact that SNX8 mainly colocalizes with Rab5 at early endosomes and partially with Rab4 at recycling endosomes, with Rab7 at late endosomes and with Giantin at Golgi apparatus.[12]

Construction of tubular profiles edit

Although the endosomal compartment is composed of vesicular and tubular structures, it has been demonstrated that sorting involving recycling pathways is mainly tubule-mediated. Therefore, tubular structures building is essential for the activity of SNXs containing BAR domains (such as SNX1, SNX4 or SNX8) as phosphoinositide-mediated endosomal sorting proteins. This BAR domain allows them to assembly in a dose-dependent manner a helical coat with the capacity to detect, promote and stabilize the curvature of endosomal vesicular membranes into tubular profiles during the so-called incidence detection process, specially in phosphoinosited-enriched regions where they are localized thanks to the affinity of the PX domain for these membrane phospholipids. In particular, SNX8 colocalizes with Rab5 at early endosomes membranes and at the tubular endosomal network (TEN) around the endosomal vacuole, which is an important compartment for successful sorting of cargoes.[6]

It is also worth mentioning the existing coupling of tubular structures formation and the maduration processes from early endosomes to late endosomes, which is mainly characterized by a retrograde movement from the cell periphery to a juxtanuclear position, an acidification of early endosomes lumen and a switch from Rab5 GTPase to Rab7 GTPase. The construction of these tubules, which is important for SNX-mediated endosomal sorting, experiments an important acceleration during this transition process; therefore, it is suggested that SNX8 may be involved in the transport of endogenous acid environment-requiring cargo. In addition, as SNX8 interacts with elements of the retromer, it is important to highlight the role of both subcomplexes of the retromer in the coordinated relationship between endosomal maturation and the generation of tubular profiles from vesicular structures.[6]

Medical and clinical significance edit

Alzheimer's disease edit

Insoluble accumulations of β-amyloid peptide in brain regions related to memory and cognition are a defining characteristic of Alzheimer's Disease (AD).[13] SNX8 has a neuroprotective role related to AD: it enhances the non-amyloidogenic APP pathway and, hence, reduces the Aβ plaques and deposit accumulations and suppresses cognitive impairment caused by AD. Some studies found that SNX8 levels were drastically lower in patients with AD. Furthermore, it has been demonstrated that SNX8 overexpression mediated by adeno-associated virus (AVV) reduced levels and reversed cognitive impairment in AD mice.[12] It is also important mentioning that a pair of polymorphisms (rs2286206 and rs10249052) within the human SNX8 gene locus have also been associated with late-onset AD.[14]

Neurodevelopmental disorders edit

Several case studies studying deletions in the 7p22.3 genomic region, where the SNX8 gene is located, found that it contributes to neurodevelopmental problems with considerable impairments in the motor, cognitive and socio-emotional areas, with malformations in the heart and craniofacial structures, with developmental, intellectual and language delay, with mild intellectual disability and with cognitive impairment associated with autism in some cases.[15][16][17] This idea is supported by the fact that the overlapping deleted regions of the different patients of these studies contained SNX8, which suggests that it is one of the accountable genes.[18]

Antiviral activity edit

SNX8 plays an antiviral role against Listeria monocytogenes through the IFNγ-triggered IKKβ-mediated noncanonical signaling pathway; murine cells expressing SNX8 under this infection showed a higher expression and secretion of IFNβ and IL6 cytokines in blood and lower presence of bacteria in liver and spleens, which resulted in a reduction of Listeria monocytogenes lethality, in comparison to SNX8-negative induced murine cells.[5]

In addition, SNX8 plays an antiviral role against DNA viruses such as HSV-1 through the MITA-mediated activation of the IFNβ promoter; murine cells expressing SNX8 under this infection showed a higher expression and secretion of IFNβ and IL6 cytokines in blood and a decreased presence of cerebral viral titers, which resulted in a reduction of HSV-1 lethality, in comparison to SNX8-negative induced murine cells.[10]

Finally, SNX8 also plays an antiviral role against RNA viruses such as SeV (Sendai virus) through VISA-mediated activation of the IFNβ promoter; murine cells expressing SNX8 under this infection showed a higher expression and secretion of IFNβ and IL6 cytokines in blood and a reduced presence of viral accumulations, which resulted in a reduction of SeV lethality, in comparison to SNX8-negative induced murine cells.[9]

Cardiac malformations edit

It is suggested that SNX8 participates in the development of the embryonic cardiac tissue since the gene is expressed with cells within the area of heart. This hypothesis is supported by the fact that SNX8 activity has been associated to sortin nexin L, a protein of the same family encoded by the SNX21 gene, which plays a role in the development of the embryonic liver. Deletions of chromosome 7p22 that induce happloinsufficiency of SNX8 among other genes (FTSJ2, NUDTI and MAD1L1) seem to cause craniosynostosis, dysmorphic features and cardiac malformations encompassing tetralogy of Fallot, one of the most common cyanotic congenital heart defects. Nevertheless, evidence demonstrates the existence of patients with SNX8 deletion whose cardiac tissue development does not experience any alteration. Finally, although cardiac malformation requires SNX8 haploinsufficiency, its deletion is not enough to cause this malformations on its own.[17][19][15]

Relationship between cardiac malformations and cholesterol regulation edit

 
This bar plot shows the proportion of tumor samples from 15 cancer types that have any kind of altering mutation(s) in the given protein.[20]

SNX8 role in cholesterol levels regulation is markedly relevant since disruption of intracellular cholesterol metabolism and trafficking is the main cause of multiple human disorders. For example, some studies suggest that SNX8 deletions might produce cardiac malformations since its function in normal cholesterol levels regulations would be completely impaired.[21][22][3]

Neuropathic pain edit

SNPs (single nucleotide polymorphisms) in the SNX8 gene are related to neuropathy due to its role in endosomal content sorting. Neuropathic pain is a chronic debilitating pain condition caused by a nervous system lesion or dysfunction, which usually emerges after head and neck cancer treatment. This hypothesis is supported by the fact that other SNXs activity has also been identified with other pain conditions.[23][24]

Cancer edit

Some mutations of SNX8 have been related to certain types of cancer, specially to stomach and endometrial cancer. The bar plot on the right shows the proportion of tumor samples from 15 cancer types that have any kind of altering mutations in the given protein.[20] Moreover, some studies seem to draw an important relationship between different type of cancers and SNX8 expression; although most of the patients with colorectal, stomach or testis cancer showed high levels of SNX8, almost any patient with prostate, endometrial or carcinoid cancer presented low or any concentrations of SNX8. The rationale behind this differential phenotype of SNX8 synthesis remains unclear.[25]

References edit

  1. ^ "Q9Y5X2". SWISS-MODEL Repository.
  2. ^ a b c d "SNX8 - Sorting nexin-8 - Homo sapiens (Human) - SNX8 gene & protein". www.uniprot.org. Retrieved 25 October 2019.
  3. ^ a b c d Muirhead, Gillian; Dev, Kumlesh K. (2014). "The Expression of Neuronal Sorting Nexin 8 (SNX8) Exacerbates Abnormal Cholesterol Levels". Journal of Molecular Neuroscience. 53 (1): 125–134. doi:10.1007/s12031-013-0209-z. PMID 24362679. S2CID 14668542.
  4. ^ a b "SNX8 sorting nexin 8 [Homo sapiens (human)] - Gene - NCBI". www.ncbi.nlm.nih.gov. Retrieved 26 October 2019.
  5. ^ a b c d e f Wei, Jin; Guo, Wei; Lian, Huan; Yang, Qing; Lin, Heng; Li, Shu; Shu, Hong-Bing (5 December 2017). "SNX8 mediates IFNγ-triggered noncanonical signaling pathway and host defense against Listeria monocytogenes". Proceedings of the National Academy of Sciences of the United States of America. 114 (49): 13000–13005. Bibcode:2017PNAS..11413000W. doi:10.1073/pnas.1713462114. PMC 5724276. PMID 29180417.
  6. ^ a b c d van Weering, Jan R.T.; Verkade, Paul; Cullen, Peter J. (January 2012). "SNX-BAR-Mediated Endosome Tubulation is Co-ordinated with Endosome Maturation". Traffic. 13 (1): 94–107. doi:10.1111/j.1600-0854.2011.01297.x. PMID 21973056. S2CID 7295814.
  7. ^ a b Dyve, Anne Berit; Bergan, Jonas; Utskarpen, Audrun; Sandvig, Kirsten (December 2009). "Sorting nexin 8 regulates endosome-to-Golgi transport". Biochemical and Biophysical Research Communications. 390 (1): 109–114. doi:10.1016/j.bbrc.2009.09.076. PMID 19782049.
  8. ^ Hunt, Sylvie D.; Townley, Anna K.; Danson, Chris M.; Cullen, Peter J.; Stephens, David J. (1 June 2013). "Microtubule motors mediate endosomal sorting by maintaining functional domain organization". Journal of Cell Science. 126 (11): 2493–2501. doi:10.1242/jcs.122317. PMC 3679488. PMID 23549789.
  9. ^ a b c Guo, Wei; Wei, Jin; Zhong, Xuan; Zang, Ru; Lian, Huan; Hu, Ming-Ming; Li, Shu; Shu, Hong-Bing; Yang, Qing (11 September 2019). "SNX8 modulates the innate immune response to RNA viruses by regulating the aggregation of VISA". Cellular & Molecular Immunology. 17 (11): 1126–1135. doi:10.1038/s41423-019-0285-2. PMC 7784681. PMID 31511639. S2CID 202558274.
  10. ^ a b c Wei, Jin; Lian, Huan; Guo, Wei; Chen, Yun-Da; Zhang, Xia-Nan; Zang, Ru; Zhong, Li; Yang, Qing; Hu, Ming-Ming; Luo, Wei-Wei; Shu, Hong-Bing; Li, Shu; Feng, Pinghui (15 October 2018). "SNX8 modulates innate immune response to DNA virus by mediating trafficking and activation of MITA". PLOS Pathogens. 14 (10): e1007336. doi:10.1371/journal.ppat.1007336. PMC 6188873. PMID 30321235.
  11. ^ Liang, Xiao-Dong; Zhang, Yun-Na; Liu, Chun-Chun; Chen, Jing; Chen, Xiong-Nan; Sattar Baloch, Abdul; Zhou, Bin (1 November 2019). "U18666A inhibits classical swine fever virus replication through interference with intracellular cholesterol trafficking". Veterinary Microbiology. 238: 108436. doi:10.1016/j.vetmic.2019.108436. PMID 31648726. S2CID 204882235.
  12. ^ a b c d Xie, Yongzhuang; Niu, Mengxi; Ji, Chengxiang; Huang, Timothy Y.; Zhang, Cuilin; Tian, Ye; Shi, Zhun; Wang, Chen; Zhao, Yingjun; Luo, Hong; Can, Dan; Xu, Huaxi; Zhang, Yun-wu; Zhang, Xian (6 September 2019). "SNX8 Enhances Non-amyloidogenic APP Trafficking and Attenuates Aβ Accumulation and Memory Deficits in an AD Mouse". Frontiers in Cellular Neuroscience. 13: 410. doi:10.3389/fncel.2019.00410. PMC 6743354. PMID 31551717.
  13. ^ Glenner, George G.; Wong, Caine W. (May 1984). "Alzheimer's disease: Initial report of the purification and characterization of a novel cerebrovascular amyloid protein". Biochemical and Biophysical Research Communications. 120 (3): 885–890. doi:10.1016/S0006-291X(84)80190-4. PMID 6375662.
  14. ^ Rosenthal, Samantha L; Wang, Xingbin; Demirci, F Yesim; Barmada, Michael M; Ganguli, Mary; Lopez, Oscar L; Kamboh, M Ilyas (13 August 2012). "Beta-amyloid toxicity modifier genes and the risk of Alzheimer's disease". American Journal of Neurodegenerative Disease. 1 (2): 191–198. PMC 3560458. PMID 22984654.
  15. ^ a b Richards, Elliott G.; Zaveri, Hitisha P.; Wolf, Varina L.; Kang, Sung-Hae Lee; Scott, Daryl A. (July 2011). "Delineation of a less than 200 kb minimal deleted region for cardiac malformations on chromosome 7p22". American Journal of Medical Genetics Part A. 155 (7): 1729–1734. doi:10.1002/ajmg.a.34041. PMID 21671376. S2CID 7570369.
  16. ^ Rendu, John; Satre, Véronique; Testard, Hervé; Devillard, Francoise; Vieville, Gaëlle; Fauré, Julien; Amblard, Florence; Jouk, Pierre-Simon; Coutton, Charles (August 2014). "7p22.3 microdeletion disrupting SNX8 in a patient presenting with intellectual disability but no tetralogy of Fallot". American Journal of Medical Genetics Part A. 164 (8): 2133–2135. doi:10.1002/ajmg.a.36566. PMID 24715298. S2CID 8191354.
  17. ^ a b Vanzo, Rena J.; Martin, Megan M.; Sdano, Mallory R.; Teta, Kathie; Aggarwal, Vimla; South, Sarah T. (February 2014). "SNX8: A candidate gene for 7p22 cardiac malformations including tetralogy of fallot". American Journal of Medical Genetics Part A. 164 (2): 554–556. doi:10.1002/ajmg.a.36242. PMID 24311514. S2CID 31142228.
  18. ^ Mastromoro, Gioia; Capalbo, Anna; Guido, Cristiana Alessia; Torres, Barbara; Fabbretti, Maria; Traversa, Alice; Giancotti, Antonella; Ventriglia, Flavia; Bernardini, Laura; Spalice, Alberto; Pizzuti, Antonio (September 2019). "Small 7p22.3 microdeletion: Case report of Snx8 haploinsufficiency and neurological findings". European Journal of Medical Genetics. 63 (4): 103772. doi:10.1016/j.ejmg.2019.103772. PMID 31568860. S2CID 203624241.
  19. ^ Vanzo, Rena J.; Martin, Megan M.; Sdano, Mallory R.; Teta, Kathie; South, Sarah T. (August 2014). "Correspondence regarding SNX8 haploinsufficiency and its potential for cardiac anomalies including tetralogy of Fallot". American Journal of Medical Genetics Part A. 164 (8): 2136–2137. doi:10.1002/ajmg.a.36572. PMID 24733602. S2CID 39609715.
  20. ^ a b "SNX8 (human)". www.phosphosite.org. Retrieved 25 October 2019.
  21. ^ Chatterjee, Sandipan; Szustakowski, Joseph D.; Nanguneri, Nirmala R.; Mickanin, Craig; Labow, Mark A.; Nohturfft, Axel; Dev, Kumlesh K.; Sivasankaran, Rajeev; Xu, Aimin (21 April 2009). "Identification of Novel Genes and Pathways Regulating SREBP Transcriptional Activity". PLOS ONE. 4 (4): e5197. Bibcode:2009PLoSO...4.5197C. doi:10.1371/journal.pone.0005197. PMC 2668173. PMID 19381295.
  22. ^ Eberlé, Delphine; Hegarty, Bronwyn; Bossard, Pascale; Ferré, Pascal; Foufelle, Fabienne (November 2004). "SREBP transcription factors: master regulators of lipid homeostasis". Biochimie. 86 (11): 839–848. doi:10.1016/j.biochi.2004.09.018. PMID 15589694.
  23. ^ Reyes-Gibby, Cielito C.; Wang, Jian; Yeung, Sai-Ching J.; Chaftari, Patrick; Yu, Robert K.; Hanna, Ehab Y.; Shete, Sanjay (8 June 2018). "Genome-wide association study identifies genes associated with neuropathy in patients with head and neck cancer". Scientific Reports. 8 (1): 8789. Bibcode:2018NatSR...8.8789R. doi:10.1038/s41598-018-27070-4. PMC 5993794. PMID 29884837.
  24. ^ Lin, Tzer-Bin; Lai, Cheng-Yuan; Hsieh, Ming-Chun; Wang, Hsueh-Hsiao; Cheng, Jen-Kun; Chau, Yat-Pang; Chen, Gin-Den; Peng, Hsien-Yu (4 November 2015). "VPS26A–SNX27 Interaction-Dependent mGluR5 Recycling in Dorsal Horn Neurons Mediates Neuropathic Pain in Rats". Journal of Neuroscience. 35 (44): 14943–14955. doi:10.1523/JNEUROSCI.2587-15.2015. PMC 6605230. PMID 26538661.
  25. ^ "Expression of SNX8 in cancer". The Human Protein Atlas.

snx8, this, article, technical, most, readers, understand, please, help, improve, make, understandable, experts, without, removing, technical, details, october, 2019, learn, when, remove, this, message, sorting, nexin, protein, involved, intracellular, molecul. This article may be too technical for most readers to understand Please help improve it to make it understandable to non experts without removing the technical details October 2019 Learn how and when to remove this message The SNX8 is a sorting nexin protein involved in intracellular molecular traffic from the early endosomes to the TGN It is suggested that it acts as an adaptor protein in events related to immune response and cholesterol regulation for example As a protein of the SNXs family the SNX8 is formed of 465 aminoacids and presents a BAR domain and a PX domain which are very relevant in relation to its functions Furthermore SNX8 study is motivated by its medical significance in relation to diseases such as Alzheimer s Disease cancer neurodevelopmental malformations and to its role in fighting against viral infections Tertiary structure of the SNX9 which gives an idea of the tertiary structure of the SNX8 since primary structure shares 20 sequence identity and 0 3 sequence similarity with SNX8 primary structure 1 Contents 1 Structure 1 1 Sorting nexins SNXs 1 2 SNX8 1 2 1 PX Domain containing N terminus 1 2 2 BAR Domain containing C terminus 1 2 3 MVP1 ortholog 2 Biological functions and its molecular mechanisms 2 1 Innate immune response 2 1 1 Cytosolic RNA virus triggered response 2 1 2 Cytosolic DNA virus triggered response 2 1 3 IFNg triggered IKKb mediated noncanonical signaling pathway 2 2 Cholesterol regulation 2 3 Intracellular non amyloidogenic APP traffic 2 4 Construction of tubular profiles 3 Medical and clinical significance 3 1 Alzheimer s disease 3 2 Neurodevelopmental disorders 3 3 Antiviral activity 3 4 Cardiac malformations 3 4 1 Relationship between cardiac malformations and cholesterol regulation 3 5 Neuropathic pain 3 6 Cancer 4 ReferencesStructure editSorting nexins SNXs edit Main article Sorting nexin SNX8 belongs to the sorting nexin family of proteins which mainly contain two functional membrane binding that allow SNXs to have different roles in endosomal sorting and protein trafficking thanks to its membrane curvature ability 2 To begin with SNX PX is a distinct phosphoinositide PI binding domain The preferential interaction of this domain with membrane lipids makes the main function of SNX PX the targeting of proteins to phosphatidylinositol 3 phosphate PI 3 P to endosomes On the other hand the BAR Bin amphiphysin Rvs domain is a key regulator of phosphoinositide mediated tubular based endosomal sorting Accordingly this domain also dimerizes to sense stabilize and induce membrane curvature The SNX BAR proteins that contain both domains are a part of phosphoinositide enriched high curvature tubular micro domains of the endo lysosomal network 3 The mammalian genome contains 12 genes coding for SNX BAR proteins SNX1 SNX2 SNX4 SNX9 SNX18 SNX32 and SNX33 Other domains such as PDZ postsynaptic density protein 95 discs large zona occludens 1 SH3 Src homology 3 and RA Ras associated are involved in protein protein interactions SNX8 edit nbsp SNX8 primary structure highlighting relevant domains and amino acids 2 4 5 The SNX8 protein even though is very similar to the other sorting nexins presents a domain structure which resembles the most to SNX1 s and SNX9 s for this reason although its terciary structure remains unknown it theoretically resembles that of SNX9 shown in the model above Overall the SNX8 protein is integrated by one unique peptide chain that has 465 amino acids with a molecular mass of 52 569 Da 2 PX Domain containing N terminus edit Main article PX domain SNX8 contains a PX domain in its N terminus which is located between amino acids 71 and 181 A homology domain with yeast s PX domain is localized between amino acids 75 and 178 within this same domain As it is a phosphoinositide binding domain it is important to highlight amino acids 109 135 and 148 as residues directly related to phosphatidylinositol 3 phosphate since being specific binding sites constituting a phosphoinositid binding site with a span of 40 amino acids 2 4 Furthermore it includes a pair of phosphorylable tyrosines in positions 95 and 126 that are key in its function in the IFNg triggered IKKb mediated noncanonical signaling pathway 5 Overall the PX domain main function is to target SNX8 mainly to early endosomes and other membranes rich in phosphatidylinositol 3 phosphate phospholipids 6 BAR Domain containing C terminus edit Main article BAR domain SNX contains a BAR domain in its C terminus which is located between amino acids 256 and 440 Its ability to form coatings in membranes in order to induce their curvature is key in SNX8 participation in tubular structures formation Furthermore SNX8 C terminus contains a threonine in position 452 and a serine in position 456 which can go through post traductional changes that induces its phosphorylation resulting in a phosphothreonine and a phosphoserine Therefore there are classified as phosphorylation sites 6 MVP1 ortholog edit SNX8 has a yeast ortholog protein the MVP1 encoded by the also homolog gene Mvp1p which also plays a role by mediating transport of cargo to the vacuolar and lysosomal compartments For this reason its investigation can lead to a better understanding of SNX8 functions in human cells 7 Biological functions and its molecular mechanisms editSNX8 is thought to be an adaptor protein involved in the endosome to Golgi transport pathway participating in endocytosis and endosomal sorting and signaling It downregulates retrograde transport of intracellular proteins from the early endosome compartment to the trans Golgi network in a retromer mediated manner SNX8 is therefore localized in early endosomes as its colocalization with components of the retromer such as SNX1 SNX2 Vps26 and Vps35 has been demonstrated by some studies and also with EEA1 7 Furthermore the dynamics of endosomal structures with SNX8 enriched membrane domains are regulated by the opposite motor proteins dynein 1 containing LIC1 and kinesin 1 both of which allow SNX8 mediated cargo movement through the cytosol by exerting forces on these structures 8 The biological functions of SNX8 that have been studied all of which involve its role in intracellular endosomal transport are explained in more detail in the following sections Innate immune response edit Cytosolic RNA virus triggered response edit SNX8 is a key component in the induction of downstream effector antiviral gene transcription in response to RNA viruses as it is necessary for RLR mediated activation of the IFNb promoter that leads to host defense in a dose dependent manner It positively regulates the aggregation and activation of VISA a key adaptor protein involved in the innate immune response against this type of infections this process is critical in the recruitment of other signaling components Although SNX8 s standard localization includes the cytoplasm and other compartments such as the ER the ER Golgi intermediate compartment the Golgi apparatus or endosomes it is partially localized at mitochondria Furthermore RNA viral infections cause the translocation of SNX8 from the cytosol to the mitochondria During the early stage of the infection SNX8 association with VISA increases above constitutive levels degradation of VISA in later stages results in a reduction of this phenomenon Structurally the C terminus transmembrane domain of VISA and both N terminus PX domain and C terminus of SNX8 are necessary for this linkage The formation of the SNX8 VISA complex potentiates VISA prion like polymerization and aggregation After VISA activation its accumulation allows the recruitment of signaling components which play a role in RLR mediated antiviral response activation such as the intermediates of this pathway TBK1 IRF3 IkBa or ISRE IRF3 cooperative activation with NF kB transcription factor through phosphorylation leads to the induction of the IFNb promoter transcription Examples of genes whose RNA virus triggered expression is stimulated by SNX8 are IFNB1 ISG56 and IL6 being IL6 and IFNB1 related to cytokine secretion 9 In addition SNX8 also plays a role in RIG I containing CARD domain mediated and MDA5 mediated activation of the IFNb promoter since VISA works as an intermediate for both signaling pathways The mechanism for SNX8 recruitment to VISA remains unclear although two options have been suggested translocation of viral RNA bound RIG I or MDA5 to VISA may result in a conformational switch that would increase its affinity for SNX8 or the RNA virus may induce post translational modifications of one of these proteins allowing the translocation of SNX8 to mitochondria for its interaction with VISA 9 Cytosolic DNA virus triggered response edit nbsp Simplified scheme of DNA triggered SNX8 mediated association of MITA and VPS34 and its intracellular transport pathway from RE to perinuclear microsomes via Golgi apparatus 10 SNX8 is a key component in the induction of downstream effector antiviral gene transcription in response to DNA viruses as it is necessary for MITA mediated activation of the IFNb promoter in a dose dependent manner It regulates the activation of MITA a central adaptor protein in the innate immune response to DNA viral infections that activates and onsets the antiviral response by allowing its association with the class III phosphatylinositol 3 kinase VPS34 containing translocon machinery to form the MITA Vps34 translocation complex this linkage is crucial for MITA transport from the ER to perinuclear microsomal punctuate structures which are induced by infection of viruses such as HSV 1 via Golgi mediated intracellular traffic This MITA transport pathway is vital for the immune response to start SNX8 is suggested to play a role in MITA s activation through this transport pathway modulating its phosphorylation at serine in position 366 and recruiting the transcription factor IRF3 whose activation is important in the activation of the IFNb promoter transcription This hypothesis is supported by the fact that MITA SNX8 and VPS34 colocalize in the cytoplasm and that SNX8 localization in ER ER Golgi intermediate compartment Golgi and endosomes is similar to that of MITA Examples of genes whose DNA virus triggered expression is stimulated by SNX8 are IFNB1 ISG56 CXCL10 and IL6 being IFNB1 and IL6 related to cytokine secretion 10 IFNg triggered IKKb mediated noncanonical signaling pathway editSNX8 is a component of an IKKb mediated noncanonical signaling pathway triggered by the interferon gamma which takes places parallelly to the more known IFNg induced JAK STAT1 mediated pathway Overall it participates in a series of chemical reactions and molecular interactions that lead to the selective promotion of a particular subset of downstream effector gene transcription that encode IFNg induced GTPases of the GBP family which participate in host defense against intracellular pathogens and secretion proteins of the chemokine family such as CXCL9 CXCL10 and CXCL11 which present direct antimicrobial activity In addition SNX8 enhances the IFNg induced activation of the IRF1 promoter in a dose dependent manner 5 nbsp Simplified scheme of SNX8 participation in the IFNg triggered IKKb mediated noncanonical signaling pathway autophosphorylation not shown 5 Specifically the steps of this pathway that are regulated by SNX8 are the following The reception of IFNg activates Janus kinase 1 resulting in the stimulation of its association with Sxn8 above standard constitutive levels The link between these two protein within the JAK1 SNX8 complex allows JAK1 to catalyse SNX8 s tyrosines phosphorylation in positions 95 and 126 This phosphorylation activates the JAK1 SNX and the SNX8 acts as an adaptor or scaffolding protein by permitting the recruitment of the inhibitor of nuclear factor kappa B kinase subunit beta IKKb to JAK1 in the JAK1 SNX8 IKKb complex This linkage is essential for IKKb activation through autophosphorylation at serine in position 177 as SNX8 lacks enzymatic activity and further dimerization and oligomerization 5 Cholesterol regulation edit SNX8 protein regulates cholesterol levels as an activator of the SREBPs Sterol Regulatory Element Binding Proteins which is a family of transcription factors that control the expression of enzymes needed for the synthesis and uptake of fatty acids endogenous cholesterol triacylglycerides and phospholipids this results in an overall regulation of intracellular lipid homeostasis Although its precise mechanism of action remains unknown data suggests that SNX8 produces changes in cholesterol distribution through regulation of the SREBP transcriptional activity by modulating intracellular traffic events rather than by interacting with proteins of the SREBP pathway like INSIG or SREBP cleavage activating protein SCAP For example it is unclear if SNX8 has a direct participation in the transport of SREBP pathway components or if it regulates endosomal and lysosomal compartmentalization through the production of cholesterol cargoes This last possibility is supported by the fact that the ability of altering membrane curvature is shared by some proteins of the SNXs family 3 It has been observed that variation in cholesterol levels alter SNX8 transcription it remains unaltered in conditions of moderate high concentrations of cholesterol and it experiments a decrease in conditions of restricted levels of cholesterol due to the action of the fungus derived cholesterol lowering statin mevinolin and the drug U18666a which promotes cytosolic cholesterol clustering within the endosomal and lysosomal compartments by acting as an intracellular cholesterol transport inhibitor 11 On the other hand SNX8 overexpression induces intracellular clustering of cholesterol under conditions of high cellular cholesterol levels and aggravates abnormal distribution of cholesterol In relation to the SREBP pathway SNX8 is not able to successfully reduce the inhibitory impact of cholesterol on SREBP mediated transcription but it has an important effect that counteracts the block of this pathway triggered by the insulin induced gene INSIG 3 Intracellular non amyloidogenic APP traffic edit nbsp Simplified scheme of APP traffic and its amyloidogenic and non amyloidogenic proteolytic degradative pathways 12 The amyloid precursor protein APP is consecutively transported from the ER after its synthesis to the plasma membrane via the trans Golgi network during this traffic the neuroprotective soluble fragment sAPPa is produced as a product of APP cleavage by a secretases Lately APP that reaches the membrane without being severed can be internalized into endosomes in order to be proteolytically processed through two parallel pathways an amyloidogenic pathway via acidified late endosomes and a non amyloidogenic or anti myloidogenic retrograde pathway via Golgi apparatus The amyloidogenic pathway leads to APP processing by g secretases and b secretases such as BACE1 resulting in production of the neurotoxic amyloid beta Ab peptide that accumulates in extracellular locations forming senile plaques 12 SNX8 promotes non amyloidogenic transport from the Golgi apparatus to other cellular locations leading to an increase of APP levels a stimulated distribution throughout the outer face of cell membrane an enhanced sAPPa secretion and a reduced Ab production specifically production of Ab40 and Ab42 In addition SNX8 improves APP stability which is responsible for a longer lifespan without having any effect in its mRNA transcription This hypothesis is supported by the fact that SNX8 mainly colocalizes with Rab5 at early endosomes and partially with Rab4 at recycling endosomes with Rab7 at late endosomes and with Giantin at Golgi apparatus 12 Construction of tubular profiles edit Although the endosomal compartment is composed of vesicular and tubular structures it has been demonstrated that sorting involving recycling pathways is mainly tubule mediated Therefore tubular structures building is essential for the activity of SNXs containing BAR domains such as SNX1 SNX4 or SNX8 as phosphoinositide mediated endosomal sorting proteins This BAR domain allows them to assembly in a dose dependent manner a helical coat with the capacity to detect promote and stabilize the curvature of endosomal vesicular membranes into tubular profiles during the so called incidence detection process specially in phosphoinosited enriched regions where they are localized thanks to the affinity of the PX domain for these membrane phospholipids In particular SNX8 colocalizes with Rab5 at early endosomes membranes and at the tubular endosomal network TEN around the endosomal vacuole which is an important compartment for successful sorting of cargoes 6 It is also worth mentioning the existing coupling of tubular structures formation and the maduration processes from early endosomes to late endosomes which is mainly characterized by a retrograde movement from the cell periphery to a juxtanuclear position an acidification of early endosomes lumen and a switch from Rab5 GTPase to Rab7 GTPase The construction of these tubules which is important for SNX mediated endosomal sorting experiments an important acceleration during this transition process therefore it is suggested that SNX8 may be involved in the transport of endogenous acid environment requiring cargo In addition as SNX8 interacts with elements of the retromer it is important to highlight the role of both subcomplexes of the retromer in the coordinated relationship between endosomal maturation and the generation of tubular profiles from vesicular structures 6 Medical and clinical significance editAlzheimer s disease edit Insoluble accumulations of b amyloid peptide in brain regions related to memory and cognition are a defining characteristic of Alzheimer s Disease AD 13 SNX8 has a neuroprotective role related to AD it enhances the non amyloidogenic APP pathway and hence reduces the Ab plaques and deposit accumulations and suppresses cognitive impairment caused by AD Some studies found that SNX8 levels were drastically lower in patients with AD Furthermore it has been demonstrated that SNX8 overexpression mediated by adeno associated virus AVV reduced Ab levels and reversed cognitive impairment in AD mice 12 It is also important mentioning that a pair of polymorphisms rs2286206 and rs10249052 within the human SNX8 gene locus have also been associated with late onset AD 14 Neurodevelopmental disorders edit Several case studies studying deletions in the 7p22 3 genomic region where the SNX8 gene is located found that it contributes to neurodevelopmental problems with considerable impairments in the motor cognitive and socio emotional areas with malformations in the heart and craniofacial structures with developmental intellectual and language delay with mild intellectual disability and with cognitive impairment associated with autism in some cases 15 16 17 This idea is supported by the fact that the overlapping deleted regions of the different patients of these studies contained SNX8 which suggests that it is one of the accountable genes 18 Antiviral activity edit SNX8 plays an antiviral role against Listeria monocytogenes through the IFNg triggered IKKb mediated noncanonical signaling pathway murine cells expressing SNX8 under this infection showed a higher expression and secretion of IFNb and IL6 cytokines in blood and lower presence of bacteria in liver and spleens which resulted in a reduction of Listeria monocytogenes lethality in comparison to SNX8 negative induced murine cells 5 In addition SNX8 plays an antiviral role against DNA viruses such as HSV 1 through the MITA mediated activation of the IFNb promoter murine cells expressing SNX8 under this infection showed a higher expression and secretion of IFNb and IL6 cytokines in blood and a decreased presence of cerebral viral titers which resulted in a reduction of HSV 1 lethality in comparison to SNX8 negative induced murine cells 10 Finally SNX8 also plays an antiviral role against RNA viruses such as SeV Sendai virus through VISA mediated activation of the IFNb promoter murine cells expressing SNX8 under this infection showed a higher expression and secretion of IFNb and IL6 cytokines in blood and a reduced presence of viral accumulations which resulted in a reduction of SeV lethality in comparison to SNX8 negative induced murine cells 9 Cardiac malformations edit It is suggested that SNX8 participates in the development of the embryonic cardiac tissue since the gene is expressed with cells within the area of heart This hypothesis is supported by the fact that SNX8 activity has been associated to sortin nexin L a protein of the same family encoded by the SNX21 gene which plays a role in the development of the embryonic liver Deletions of chromosome 7p22 that induce happloinsufficiency of SNX8 among other genes FTSJ2 NUDTI and MAD1L1 seem to cause craniosynostosis dysmorphic features and cardiac malformations encompassing tetralogy of Fallot one of the most common cyanotic congenital heart defects Nevertheless evidence demonstrates the existence of patients with SNX8 deletion whose cardiac tissue development does not experience any alteration Finally although cardiac malformation requires SNX8 haploinsufficiency its deletion is not enough to cause this malformations on its own 17 19 15 Relationship between cardiac malformations and cholesterol regulation edit nbsp This bar plot shows the proportion of tumor samples from 15 cancer types that have any kind of altering mutation s in the given protein 20 SNX8 role in cholesterol levels regulation is markedly relevant since disruption of intracellular cholesterol metabolism and trafficking is the main cause of multiple human disorders For example some studies suggest that SNX8 deletions might produce cardiac malformations since its function in normal cholesterol levels regulations would be completely impaired 21 22 3 Neuropathic pain edit SNPs single nucleotide polymorphisms in the SNX8 gene are related to neuropathy due to its role in endosomal content sorting Neuropathic pain is a chronic debilitating pain condition caused by a nervous system lesion or dysfunction which usually emerges after head and neck cancer treatment This hypothesis is supported by the fact that other SNXs activity has also been identified with other pain conditions 23 24 Cancer edit Some mutations of SNX8 have been related to certain types of cancer specially to stomach and endometrial cancer The bar plot on the right shows the proportion of tumor samples from 15 cancer types that have any kind of altering mutations in the given protein 20 Moreover some studies seem to draw an important relationship between different type of cancers and SNX8 expression although most of the patients with colorectal stomach or testis cancer showed high levels of SNX8 almost any patient with prostate endometrial or carcinoid cancer presented low or any concentrations of SNX8 The rationale behind this differential phenotype of SNX8 synthesis remains unclear 25 References edit Q9Y5X2 SWISS MODEL Repository a b c d SNX8 Sorting nexin 8 Homo sapiens Human SNX8 gene amp protein www uniprot org Retrieved 25 October 2019 a b c d Muirhead Gillian Dev Kumlesh K 2014 The Expression of Neuronal Sorting Nexin 8 SNX8 Exacerbates Abnormal Cholesterol Levels Journal of Molecular Neuroscience 53 1 125 134 doi 10 1007 s12031 013 0209 z PMID 24362679 S2CID 14668542 a b SNX8 sorting nexin 8 Homo sapiens human Gene NCBI www ncbi nlm nih gov Retrieved 26 October 2019 a b c d e f Wei Jin Guo Wei Lian Huan Yang Qing Lin Heng Li Shu Shu Hong Bing 5 December 2017 SNX8 mediates IFNg triggered noncanonical signaling pathway and host defense against Listeria monocytogenes Proceedings of the National Academy of Sciences of the United States of America 114 49 13000 13005 Bibcode 2017PNAS 11413000W doi 10 1073 pnas 1713462114 PMC 5724276 PMID 29180417 a b c d van Weering Jan R T Verkade Paul Cullen Peter J January 2012 SNX BAR Mediated Endosome Tubulation is Co ordinated with Endosome Maturation Traffic 13 1 94 107 doi 10 1111 j 1600 0854 2011 01297 x PMID 21973056 S2CID 7295814 a b Dyve Anne Berit Bergan Jonas Utskarpen Audrun Sandvig Kirsten December 2009 Sorting nexin 8 regulates endosome to Golgi transport Biochemical and Biophysical Research Communications 390 1 109 114 doi 10 1016 j bbrc 2009 09 076 PMID 19782049 Hunt Sylvie D Townley Anna K Danson Chris M Cullen Peter J Stephens David J 1 June 2013 Microtubule motors mediate endosomal sorting by maintaining functional domain organization Journal of Cell Science 126 11 2493 2501 doi 10 1242 jcs 122317 PMC 3679488 PMID 23549789 a b c Guo Wei Wei Jin Zhong Xuan Zang Ru Lian Huan Hu Ming Ming Li Shu Shu Hong Bing Yang Qing 11 September 2019 SNX8 modulates the innate immune response to RNA viruses by regulating the aggregation of VISA Cellular amp Molecular Immunology 17 11 1126 1135 doi 10 1038 s41423 019 0285 2 PMC 7784681 PMID 31511639 S2CID 202558274 a b c Wei Jin Lian Huan Guo Wei Chen Yun Da Zhang Xia Nan Zang Ru Zhong Li Yang Qing Hu Ming Ming Luo Wei Wei Shu Hong Bing Li Shu Feng Pinghui 15 October 2018 SNX8 modulates innate immune response to DNA virus by mediating trafficking and activation of MITA PLOS Pathogens 14 10 e1007336 doi 10 1371 journal ppat 1007336 PMC 6188873 PMID 30321235 Liang Xiao Dong Zhang Yun Na Liu Chun Chun Chen Jing Chen Xiong Nan Sattar Baloch Abdul Zhou Bin 1 November 2019 U18666A inhibits classical swine fever virus replication through interference with intracellular cholesterol trafficking Veterinary Microbiology 238 108436 doi 10 1016 j vetmic 2019 108436 PMID 31648726 S2CID 204882235 a b c d Xie Yongzhuang Niu Mengxi Ji Chengxiang Huang Timothy Y Zhang Cuilin Tian Ye Shi Zhun Wang Chen Zhao Yingjun Luo Hong Can Dan Xu Huaxi Zhang Yun wu Zhang Xian 6 September 2019 SNX8 Enhances Non amyloidogenic APP Trafficking and Attenuates Ab Accumulation and Memory Deficits in an AD Mouse Frontiers in Cellular Neuroscience 13 410 doi 10 3389 fncel 2019 00410 PMC 6743354 PMID 31551717 Glenner George G Wong Caine W May 1984 Alzheimer s disease Initial report of the purification and characterization of a novel cerebrovascular amyloid protein Biochemical and Biophysical Research Communications 120 3 885 890 doi 10 1016 S0006 291X 84 80190 4 PMID 6375662 Rosenthal Samantha L Wang Xingbin Demirci F Yesim Barmada Michael M Ganguli Mary Lopez Oscar L Kamboh M Ilyas 13 August 2012 Beta amyloid toxicity modifier genes and the risk of Alzheimer s disease American Journal of Neurodegenerative Disease 1 2 191 198 PMC 3560458 PMID 22984654 a b Richards Elliott G Zaveri Hitisha P Wolf Varina L Kang Sung Hae Lee Scott Daryl A July 2011 Delineation of a less than 200 kb minimal deleted region for cardiac malformations on chromosome 7p22 American Journal of Medical Genetics Part A 155 7 1729 1734 doi 10 1002 ajmg a 34041 PMID 21671376 S2CID 7570369 Rendu John Satre Veronique Testard Herve Devillard Francoise Vieville Gaelle Faure Julien Amblard Florence Jouk Pierre Simon Coutton Charles August 2014 7p22 3 microdeletion disrupting SNX8 in a patient presenting with intellectual disability but no tetralogy of Fallot American Journal of Medical Genetics Part A 164 8 2133 2135 doi 10 1002 ajmg a 36566 PMID 24715298 S2CID 8191354 a b Vanzo Rena J Martin Megan M Sdano Mallory R Teta Kathie Aggarwal Vimla South Sarah T February 2014 SNX8 A candidate gene for 7p22 cardiac malformations including tetralogy of fallot American Journal of Medical Genetics Part A 164 2 554 556 doi 10 1002 ajmg a 36242 PMID 24311514 S2CID 31142228 Mastromoro Gioia Capalbo Anna Guido Cristiana Alessia Torres Barbara Fabbretti Maria Traversa Alice Giancotti Antonella Ventriglia Flavia Bernardini Laura Spalice Alberto Pizzuti Antonio September 2019 Small 7p22 3 microdeletion Case report of Snx8 haploinsufficiency and neurological findings European Journal of Medical Genetics 63 4 103772 doi 10 1016 j ejmg 2019 103772 PMID 31568860 S2CID 203624241 Vanzo Rena J Martin Megan M Sdano Mallory R Teta Kathie South Sarah T August 2014 Correspondence regarding SNX8 haploinsufficiency and its potential for cardiac anomalies including tetralogy of Fallot American Journal of Medical Genetics Part A 164 8 2136 2137 doi 10 1002 ajmg a 36572 PMID 24733602 S2CID 39609715 a b SNX8 human www phosphosite org Retrieved 25 October 2019 Chatterjee Sandipan Szustakowski Joseph D Nanguneri Nirmala R Mickanin Craig Labow Mark A Nohturfft Axel Dev Kumlesh K Sivasankaran Rajeev Xu Aimin 21 April 2009 Identification of Novel Genes and Pathways Regulating SREBP Transcriptional Activity PLOS ONE 4 4 e5197 Bibcode 2009PLoSO 4 5197C doi 10 1371 journal pone 0005197 PMC 2668173 PMID 19381295 Eberle Delphine Hegarty Bronwyn Bossard Pascale Ferre Pascal Foufelle Fabienne November 2004 SREBP transcription factors master regulators of lipid homeostasis Biochimie 86 11 839 848 doi 10 1016 j biochi 2004 09 018 PMID 15589694 Reyes Gibby Cielito C Wang Jian Yeung Sai Ching J Chaftari Patrick Yu Robert K Hanna Ehab Y Shete Sanjay 8 June 2018 Genome wide association study identifies genes associated with neuropathy in patients with head and neck cancer Scientific Reports 8 1 8789 Bibcode 2018NatSR 8 8789R doi 10 1038 s41598 018 27070 4 PMC 5993794 PMID 29884837 Lin Tzer Bin Lai Cheng Yuan Hsieh Ming Chun Wang Hsueh Hsiao Cheng Jen Kun Chau Yat Pang Chen Gin Den Peng Hsien Yu 4 November 2015 VPS26A SNX27 Interaction Dependent mGluR5 Recycling in Dorsal Horn Neurons Mediates Neuropathic Pain in Rats Journal of Neuroscience 35 44 14943 14955 doi 10 1523 JNEUROSCI 2587 15 2015 PMC 6605230 PMID 26538661 Expression of SNX8 in cancer The Human Protein Atlas Retrieved from https en wikipedia org w index php title SNX8 amp oldid 1188047801, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.