1
|
Byrd DT, Han ZC, Piggott CA, Jin Y. PACS-1 variant protein is aberrantly localized in Caenorhabditis elegans model of PACS1/PACS2 syndromes. Genetics 2024; 228:iyae118. [PMID: 39031646 PMCID: PMC11457933 DOI: 10.1093/genetics/iyae118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/22/2024] Open
Abstract
PACS (phosphofurin acidic cluster sorting) proteins are known for their roles in sorting cargo proteins to organelles and can physically interact with WD40 repeat-containing protein WDR37. PACS1, PACS2, and WDR37 variants are associated with multisystemic syndromes and neurodevelopmental disorders characterized by intellectual disability, seizures, developmental delays, craniofacial abnormalities, and autism spectrum disorder. However, the functional effects of syndromic variants at the cellular level remain unknown. Here, we report the expression pattern of Caenorhabditis elegans orthologs of PACS and WDR37 and their interaction. We show that cePACS-1 and ceWDR-37 colocalize to somatic cytoplasm of many types of cells and are mutually required for expression, supporting a conclusion that the intermolecular dependence of PACS1/PACS2/PACS-1 and WDR37/WDR-37 is evolutionarily conserved. We further show that editing in PACS1 and PACS2 variants in cePACS-1 changes protein localization in multiple cell types, including neurons. Moreover, expression of human PACS1 can functionally complement C. elegans PACS-1 in neurons, demonstrating conserved functions of the PACS-WDR37 axis in an invertebrate model system. Our findings reveal effects of human variants and suggest potential strategies to identify regulatory network components that may contribute to understanding molecular underpinnings of PACS/WDR37 syndromes.
Collapse
Affiliation(s)
- Dana T Byrd
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ziyuan Christina Han
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christopher A Piggott
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
2
|
Nishida S, Matovelo SA, Kajimoto T, Nakamura SI, Okada T. Involvement of sphingosine 1-phosphate signaling in insulin-like growth factor-II/mannose 6-phosphate receptor trafficking from endosome to the trans-Golgi network. Commun Biol 2024; 7:1182. [PMID: 39300315 DOI: 10.1038/s42003-024-06828-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 09/03/2024] [Indexed: 09/22/2024] Open
Abstract
The insulin-like growth factor II/mannose 6-phosphate (IGF-II/M6P) receptor is a multifunctional glycoprotein not only play roles in IGF-II degradation and pro-TGFβ activation but binding to and transport M6P-bearing lysosomal enzymes from the trans-Golgi network (TGN) or the cell surface to lysosomes. At present, information regarding a retrograde transport of IGF-II/M6P receptor from endosomes to the TGN is still limited. We show here that a continuous ligand-dependent activation of sphingosine 1-phosphate receptor type 3 (S1P3R) on the endosomal membranes is required for subsequent recycling back of cargo-unloaded IGF-II/M6P receptors to the TGN. We have further clarified that Gq coupled with S1P3R plays a critical role in the activation of casein kinase 2, which phosphorylates and keeps PACS1 connector protein active for the association with IGF-II/M6P receptors, which enables transport carrier formation with the aid of other adaptor proteins toward the TGN. These findings shed light on the molecular mechanism underlying how continuous activation of the S1P receptor and subsequent downstream Gq signaling regulates the retrograde transport of the empty IGF-II/M6P receptors back to the TGN.
Collapse
Affiliation(s)
- Susumu Nishida
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shubi Ambwene Matovelo
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Medical Biochemistry, School of Medicine and Dentistry, The University of Dodoma, Dodoma, Tanzania
| | - Taketoshi Kajimoto
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shun-Ichi Nakamura
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Taro Okada
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan.
| |
Collapse
|
3
|
Byrd DT, Han ZC, Piggott CA, Jin Y. PACS-1 variant protein is aberrantly localized in C. elegans model of PACS1/PACS2 syndromes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590644. [PMID: 38712144 PMCID: PMC11071410 DOI: 10.1101/2024.04.22.590644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
PACS (Phosphofurin Acidic Cluster Sorting Protein) proteins are known for their roles in sorting cargo proteins to organelles and can physically interact with WD40 repeat-containing protein WDR37. PACS1, PACS2, and WDR37 variants are associated with multisystemic syndromes and neurodevelopmental disorders characterized by intellectual disability, seizures, developmental delays, craniofacial abnormalities, and autism spectrum disorder. However, the effects of syndromic variants on function in vivo remains unknown. Here, we report the expression pattern of C. elegans orthologs of PACS and WDR37 and their interaction. We show that cePACS-1 and ceWDR-37 co-localize to somatic cytoplasm of many types of cells, and are mutually required for expression, supporting a conclusion that the intermolecular dependence of PACS1/PACS2/PACS-1 and WDR37/WDR-37 is evolutionarily conserved. We further show that editing in PACS1 and PACS2 variants in cePACS-1 changes protein localization in multiple cell types, including neurons. Moreover, expression of human PACS1 can functionally complement C. elegans PACS-1 in neurons, demonstrating conserved functions of the PACS-WDR37 axis in an invertebrate model system. Our findings reveal effects of human variants and suggest potential strategies to identify regulatory network components that may contribute to understanding molecular underpinnings of PACS/WDR37 syndromes.
Collapse
Affiliation(s)
- Dana T. Byrd
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093
| | - Ziyuan Christina Han
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093
| | - Christopher A. Piggott
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093
| |
Collapse
|
4
|
Rylaarsdam L, Rakotomamonjy J, Pope E, Guemez-Gamboa A. iPSC-derived models of PACS1 syndrome reveal transcriptional and functional deficits in neuron activity. Nat Commun 2024; 15:827. [PMID: 38280846 PMCID: PMC10821916 DOI: 10.1038/s41467-024-44989-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 01/11/2024] [Indexed: 01/29/2024] Open
Abstract
PACS1 syndrome is a neurodevelopmental disorder characterized by intellectual disability and distinct craniofacial abnormalities resulting from a de novo p.R203W variant in phosphofurin acidic cluster sorting protein 1 (PACS1). PACS1 is known to have functions in the endosomal pathway and nucleus, but how the p.R203W variant affects developing neurons is not fully understood. Here we differentiated stem cells towards neuronal models including cortical organoids to investigate the impact of the PACS1 syndrome-causing variant on neurodevelopment. While few deleterious effects were detected in PACS1(+/R203W) neural precursors, mature PACS1(+/R203W) glutamatergic neurons exhibited impaired expression of genes involved in synaptic signaling processes. Subsequent characterization of neural activity using calcium imaging and multielectrode arrays revealed the p.R203W PACS1 variant leads to a prolonged neuronal network burst duration mediated by an increased interspike interval. These findings demonstrate the impact of the PACS1 p.R203W variant on developing human neural tissue and uncover putative electrophysiological underpinnings of disease.
Collapse
Affiliation(s)
- Lauren Rylaarsdam
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jennifer Rakotomamonjy
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eleanor Pope
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alicia Guemez-Gamboa
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
5
|
Bruno LP, Doddato G, Baldassarri M, Rizzo CL, Resciniti S, Bruttini M, Mirjam L, Zguro K, Furini S, Mencarelli MA, Renieri A, Ariani F. Expanding the clinical spectrum associated with the PACS1 p.Arg203Trp mutational hot-spot: Two additional Italian patients. Am J Med Genet A 2023; 191:284-288. [PMID: 36210549 PMCID: PMC10092205 DOI: 10.1002/ajmg.a.62984] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/19/2022] [Accepted: 08/28/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Lucia Pia Bruno
- Medical GeneticsUniversity of SienaSienaItaly
- Med Biotech Hub and Competence Center, Department of Medical BiotechnologiesUniversity of SienaSienaItaly
| | - Gabriella Doddato
- Medical GeneticsUniversity of SienaSienaItaly
- Med Biotech Hub and Competence Center, Department of Medical BiotechnologiesUniversity of SienaSienaItaly
| | - Margherita Baldassarri
- Medical GeneticsUniversity of SienaSienaItaly
- Med Biotech Hub and Competence Center, Department of Medical BiotechnologiesUniversity of SienaSienaItaly
| | | | - Sara Resciniti
- Medical GeneticsUniversity of SienaSienaItaly
- Med Biotech Hub and Competence Center, Department of Medical BiotechnologiesUniversity of SienaSienaItaly
| | - Mirella Bruttini
- Medical GeneticsUniversity of SienaSienaItaly
- Med Biotech Hub and Competence Center, Department of Medical BiotechnologiesUniversity of SienaSienaItaly
- Genetica MedicaAzienda Ospedaliera Universitaria SeneseSienaItaly
| | - Lista Mirjam
- Medical GeneticsUniversity of SienaSienaItaly
- Med Biotech Hub and Competence Center, Department of Medical BiotechnologiesUniversity of SienaSienaItaly
| | - Kristina Zguro
- Med Biotech Hub and Competence Center, Department of Medical BiotechnologiesUniversity of SienaSienaItaly
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical BiotechnologiesUniversity of SienaSienaItaly
| | | | - Alessandra Renieri
- Medical GeneticsUniversity of SienaSienaItaly
- Med Biotech Hub and Competence Center, Department of Medical BiotechnologiesUniversity of SienaSienaItaly
- Genetica MedicaAzienda Ospedaliera Universitaria SeneseSienaItaly
| | - Francesca Ariani
- Medical GeneticsUniversity of SienaSienaItaly
- Med Biotech Hub and Competence Center, Department of Medical BiotechnologiesUniversity of SienaSienaItaly
- Genetica MedicaAzienda Ospedaliera Universitaria SeneseSienaItaly
| |
Collapse
|
6
|
Mani C, Tripathi K, Luan S, Clark DW, Andrews JF, Vindigni A, Thomas G, Palle K. The multifunctional protein PACS-1 is required for HDAC2- and HDAC3-dependent chromatin maturation and genomic stability. Oncogene 2020; 39:2583-2596. [PMID: 31988453 PMCID: PMC7085454 DOI: 10.1038/s41388-020-1167-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/18/2019] [Accepted: 01/15/2020] [Indexed: 12/21/2022]
Abstract
Phosphofurin acidic cluster sorting protein-1 (PACS-1) is a multifunctional membrane traffic regulator that plays important roles in organ homeostasis and disease. In this study, we elucidate a novel nuclear function for PACS-1 in maintaining chromosomal integrity. PACS-1 progressively accumulates in the nucleus during cell cycle progression, where it interacts with class I histone deacetylases 2 and 3 (HDAC2 and HDAC3) to regulate chromatin dynamics by maintaining the acetylation status of histones. PACS-1 knockdown results in the proteasome-mediated degradation of HDAC2 and HDAC3, compromised chromatin maturation, as indicated by elevated levels of histones H3K9 and H4K16 acetylation, and, consequently, increased replication stress-induced DNA damage and genomic instability.
Collapse
Affiliation(s)
- Chinnadurai Mani
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Centre, Lubbock, TX, 79430, USA.,Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Kaushlendra Tripathi
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Shan Luan
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.,University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15239, USA.,University of Pittsburgh School of Medicine, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - David W Clark
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Joel F Andrews
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Alessandro Vindigni
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Gary Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.,University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15239, USA.,University of Pittsburgh School of Medicine, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Komaraiah Palle
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Centre, Lubbock, TX, 79430, USA. .,Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA. .,Department of Surgery, Texas Tech University Health Sciences Centre, Lubbock, TX, 79430, USA.
| |
Collapse
|
7
|
Krzysiak TC, Thomas L, Choi YJ, Auclair S, Qian Y, Luan S, Krasnow SM, Thomas LL, Koharudin LMI, Benos PV, Marks DL, Gronenborn AM, Thomas G. An Insulin-Responsive Sensor in the SIRT1 Disordered Region Binds DBC1 and PACS-2 to Control Enzyme Activity. Mol Cell 2018; 72:985-998.e7. [PMID: 30415949 PMCID: PMC6309500 DOI: 10.1016/j.molcel.2018.10.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 08/13/2018] [Accepted: 10/04/2018] [Indexed: 12/16/2022]
Abstract
Current models of SIRT1 enzymatic regulation primarily consider the effects of fluctuating levels of its co-substrate NAD+, which binds to the stably folded catalytic domain. By contrast, the roles of the sizeable disordered N- and C-terminal regions of SIRT1 are largely unexplored. Here we identify an insulin-responsive sensor in the SIRT1 N-terminal region (NTR), comprising an acidic cluster (AC) and a 3-helix bundle (3HB), controlling deacetylase activity. The allosteric assistor DBC1 removes a distal N-terminal shield from the 3-helix bundle, permitting PACS-2 to engage the acidic cluster and the transiently exposed helix 3 of the 3-helix bundle, disrupting its structure and inhibiting catalysis. The SIRT1 activator (STAC) SRT1720 binds and stabilizes the 3-helix bundle, protecting SIRT1 from inhibition by PACS-2. Identification of the SIRT1 insulin-responsive sensor and its engagement by the DBC1 and PACS-2 regulatory hub provides important insight into the roles of disordered regions in enzyme regulation and the mode by which STACs promote metabolic fitness.
Collapse
Affiliation(s)
- Troy C Krzysiak
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Laurel Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - You-Jin Choi
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Sylvain Auclair
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Yiqi Qian
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Shan Luan
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Stephanie M Krasnow
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Laura L Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Leonardus M I Koharudin
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Panayiotis V Benos
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Daniel L Marks
- Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Angela M Gronenborn
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA; Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Gary Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
8
|
A Recurrent De Novo PACS2 Heterozygous Missense Variant Causes Neonatal-Onset Developmental Epileptic Encephalopathy, Facial Dysmorphism, and Cerebellar Dysgenesis. Am J Hum Genet 2018; 102:995-1007. [PMID: 29656858 DOI: 10.1016/j.ajhg.2018.03.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/27/2018] [Indexed: 11/24/2022] Open
Abstract
Developmental and epileptic encephalopathies (DEEs) represent a large clinical and genetic heterogeneous group of neurodevelopmental diseases. The identification of pathogenic genetic variants in DEEs remains crucial for deciphering this complex group and for accurately caring for affected individuals (clinical diagnosis, genetic counseling, impacting medical, precision therapy, clinical trials, etc.). Whole-exome sequencing and intensive data sharing identified a recurrent de novo PACS2 heterozygous missense variant in 14 unrelated individuals. Their phenotype was characterized by epilepsy, global developmental delay with or without autism, common cerebellar dysgenesis, and facial dysmorphism. Mixed focal and generalized epilepsy occurred in the neonatal period, controlled with difficulty in the first year, but many improved in early childhood. PACS2 is an important PACS1 paralog and encodes a multifunctional sorting protein involved in nuclear gene expression and pathway traffic regulation. Both proteins harbor cargo(furin)-binding regions (FBRs) that bind cargo proteins, sorting adaptors, and cellular kinase. Compared to the defined PACS1 recurrent variant series, individuals with PACS2 variant have more consistently neonatal/early-infantile-onset epilepsy that can be challenging to control. Cerebellar abnormalities may be similar but PACS2 individuals exhibit a pattern of clear dysgenesis ranging from mild to severe. Functional studies demonstrated that the PACS2 recurrent variant reduces the ability of the predicted autoregulatory domain to modulate the interaction between the PACS2 FBR and client proteins, which may disturb cellular function. These findings support the causality of this recurrent de novo PACS2 heterozygous missense in DEEs with facial dysmorphim and cerebellar dysgenesis.
Collapse
|
9
|
Brasacchio D, Busuttil RA, Noori T, Johnstone RW, Boussioutas A, Trapani JA. Down-regulation of a pro-apoptotic pathway regulated by PCAF/ADA3 in early stage gastric cancer. Cell Death Dis 2018; 9:442. [PMID: 29670108 PMCID: PMC5906598 DOI: 10.1038/s41419-018-0470-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 01/10/2018] [Accepted: 03/06/2018] [Indexed: 12/28/2022]
Abstract
The loss of p300/CBP-associated protein (PCAF) expression is associated with poor clinical outcome in gastric cancer, and a potential bio-marker for invasive and aggressive tumors. However, the mechanism linking loss of PCAF to the onset of gastric cancer has not been identified. Given that PCAF and its binding partner transcriptional adaptor protein 3 (ADA3) were recently shown to regulate the intrinsic (mitochondrial) pathway to apoptosis via epigenetic regulation of phosphofurin acidic cluster sorting proteins 1 and 2 (PACS1, PACS2), we analyzed PCAF, ADA3, and PACS1/2 expression in 99 patient-matched surgical samples ranging from normal gastric mucosa, through pre-malignant chronic gastritis and intestinal metaplasia to stage I–III invasive cancers. PCAF mRNA levels were not reduced in either pre-malignant state but were significantly down-regulated in all stages of gastric cancer, commencing at AJCC stage I (p < 0.05), thus linking reduced PCAF expression with early malignant change. Furthermore, patients with combined reduction of PCAF and PACS1 had significantly poorer overall survival (p = 0.0257), confirmed in an independent dataset of 359 patients (p = 5.8 × 10e-6). At the protein level, PCAF, ADA3, and PACS1 expression were all significantly down-regulated in intestinal-type gastric cancer, and correlated with reduced progression free survival. We conclude that a pro-apoptotic mechanism centered on the intrinsic (mitochondrial) pathway and regulated by PCAF/ADA3 can influence the progression from premalignant to malignant change, and thus act as a tumor suppression mechanism in gastric cancer.
Collapse
Affiliation(s)
- Daniella Brasacchio
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Rita A Busuttil
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Upper Gastrointestinal Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Tahereh Noori
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Ricky W Johnstone
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Cancer Therapeutics Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Alex Boussioutas
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Upper Gastrointestinal Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Joseph A Trapani
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia. .,Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
| |
Collapse
|
10
|
Makaraci P, Kim K. trans-Golgi network-bound cargo traffic. Eur J Cell Biol 2018; 97:137-149. [PMID: 29398202 DOI: 10.1016/j.ejcb.2018.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/15/2017] [Accepted: 01/16/2018] [Indexed: 12/19/2022] Open
Abstract
Cargo following the retrograde trafficking are sorted at endosomes to be targeted the trans-Golgi network (TGN), a central receiving organelle. Though molecular requirements and their interaction networks have been somewhat established, the complete understanding of the intricate nature of their action mechanisms in every step of the retrograde traffic pathway remains unachieved. This review focuses on elucidating known functions of key regulators, including scission factors at the endosome and tethering/fusion mediators at the receiving dock, TGN, as well as a diverse range of cargo.
Collapse
Affiliation(s)
- Pelin Makaraci
- Department of Biology, Missouri State University, 901 S National Ave., Springfield, MO 65807, USA
| | - Kyoungtae Kim
- Department of Biology, Missouri State University, 901 S National Ave., Springfield, MO 65807, USA.
| |
Collapse
|
11
|
Thomas G, Aslan JE, Thomas L, Shinde P, Shinde U, Simmen T. Caught in the act - protein adaptation and the expanding roles of the PACS proteins in tissue homeostasis and disease. J Cell Sci 2017; 130:1865-1876. [PMID: 28476937 PMCID: PMC5482974 DOI: 10.1242/jcs.199463] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Vertebrate proteins that fulfill multiple and seemingly disparate functions are increasingly recognized as vital solutions to maintaining homeostasis in the face of the complex cell and tissue physiology of higher metazoans. However, the molecular adaptations that underpin this increased functionality remain elusive. In this Commentary, we review the PACS proteins - which first appeared in lower metazoans as protein traffic modulators and evolved in vertebrates to integrate cytoplasmic protein traffic and interorganellar communication with nuclear gene expression - as examples of protein adaptation 'caught in the act'. Vertebrate PACS-1 and PACS-2 increased their functional density and roles as metabolic switches by acquiring phosphorylation sites and nuclear trafficking signals within disordered regions of the proteins. These findings illustrate one mechanism by which vertebrates accommodate their complex cell physiology with a limited set of proteins. We will also highlight how pathogenic viruses exploit the PACS sorting pathways as well as recent studies on PACS genes with mutations or altered expression that result in diverse diseases. These discoveries suggest that investigation of the evolving PACS protein family provides a rich opportunity for insight into vertebrate cell and organ homeostasis.
Collapse
Affiliation(s)
- Gary Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15239, USA
- University of Pittsburgh Cancer Institute, Pittsburgh, PA 15239, USA
| | - Joseph E Aslan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Laurel Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15239, USA
| | - Pushkar Shinde
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ujwal Shinde
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Thomas Simmen
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada T6G2H7
| |
Collapse
|
12
|
Stern D, Cho MT, Chikarmane R, Willaert R, Retterer K, Kendall F, Deardorff M, Hopkins S, Bedoukian E, Slavotinek A, Schrier Vergano S, Spangler B, McDonald M, McConkie-Rosell A, Burton BK, Kim KH, Oundjian N, Kronn D, Chandy N, Baskin B, Guillen Sacoto MJ, Wentzensen IM, McLaughlin HM, McKnight D, Chung WK. Association of the missense variant p.Arg203Trp in PACS1 as a cause of intellectual disability and seizures. Clin Genet 2017; 92:221-223. [PMID: 28111752 DOI: 10.1111/cge.12956] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/05/2016] [Accepted: 12/06/2016] [Indexed: 02/04/2023]
Abstract
Graphical abstract key: ADHD, attention deficit hyperactivity disorder; ASD, atrial septal defect; DD, developmental delay; EEG, electroencephalogram; Ht, height; ID, intellectual disability; OCD, obsessive-compulsive disorder; OFC, open fontanelle; PDA, patent ductus arteriosis; PFO, patent foramen ovale; VSD, ventricular septal defect; Wt, weight.
Collapse
Affiliation(s)
- D Stern
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - M T Cho
- GeneDx, Gaithersburg, MD, USA
| | | | | | | | - F Kendall
- VMP Genetics, Roswell, GA, USA.,Department of Kinesiology, University of Georgia, Athens, GA, USA
| | - M Deardorff
- Department of Pediatrics, Division of Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - S Hopkins
- Division of Neurology, The Children's Hospital of Philadelphia, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - E Bedoukian
- Individualized Medical Genetics Center, Division of Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - A Slavotinek
- Department of Pediatrics, UCSF Benioff Children's Hospital, University of California, San Francisco, CA, USA
| | - S Schrier Vergano
- Division of Medical Genetics and Metabolism, Children's Hospital of The King's Daughters, Norfolk, VA, USA
| | - B Spangler
- Division of Medical Genetics and Metabolism, Children's Hospital of The King's Daughters, Norfolk, VA, USA
| | - M McDonald
- Division of Medical Genetics, Duke University Medical Center, Durham, NC, USA
| | - A McConkie-Rosell
- Division of Medical Genetics, Duke University Medical Center, Durham, NC, USA
| | - B K Burton
- Division of Genetics, Birth Defects & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - K H Kim
- Division of Genetics, Birth Defects & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | | | - D Kronn
- New York Medical College, Valhalla, NY, USA
| | - N Chandy
- New York Medical College, Valhalla, NY, USA
| | | | | | | | | | | | - W K Chung
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA.,Department of Medicine, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
13
|
Pereira EA, daSilva LLP. HIV-1 Nef: Taking Control of Protein Trafficking. Traffic 2016; 17:976-96. [PMID: 27161574 DOI: 10.1111/tra.12412] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/05/2016] [Accepted: 05/05/2016] [Indexed: 12/25/2022]
Abstract
The Nef protein of the human immunodeficiency virus is a crucial determinant of viral pathogenesis and disease progression. Nef is abundantly expressed early in infection and is thought to optimize the cellular environment for viral replication. Nef controls expression levels of various cell surface molecules that play important roles in immunity and virus life cycle, by directly interfering with the itinerary of these proteins within the endocytic and late secretory pathways. To exert these functions, Nef physically interacts with host proteins that regulate protein trafficking. In recent years, considerable progress was made in identifying host-cell-interacting partners for Nef, and the molecular machinery used by Nef to interfere with protein trafficking has started to be unraveled. Here, we briefly review the knowledge gained and discuss new findings regarding the mechanisms by which Nef modifies the intracellular trafficking pathways to prevent antigen presentation, facilitate viral particle release and enhance the infectivity of HIV-1 virions.
Collapse
Affiliation(s)
- Estela A Pereira
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luis L P daSilva
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
14
|
Abstract
Protein phosphatase 2A (PP2A) plays a critical multi-faceted role in the regulation of the cell cycle. It is known to dephosphorylate over 300 substrates involved in the cell cycle, regulating almost all major pathways and cell cycle checkpoints. PP2A is involved in such diverse processes by the formation of structurally distinct families of holoenzymes, which are regulated spatially and temporally by specific regulators. Here, we review the involvement of PP2A in the regulation of three cell signaling pathways: wnt, mTOR and MAP kinase, as well as the G1→S transition, DNA synthesis and mitotic initiation. These processes are all crucial for proper cell survival and proliferation and are often deregulated in cancer and other diseases.
Collapse
Affiliation(s)
- Nathan Wlodarchak
- a McArdle Laboratory for Cancer Research, University of Wisconsin-Madison , Madison , WI , USA
| | - Yongna Xing
- a McArdle Laboratory for Cancer Research, University of Wisconsin-Madison , Madison , WI , USA
| |
Collapse
|
15
|
Pawlak EN, Dikeakos JD. HIV-1 Nef: a master manipulator of the membrane trafficking machinery mediating immune evasion. Biochim Biophys Acta Gen Subj 2015; 1850:733-41. [PMID: 25585010 DOI: 10.1016/j.bbagen.2015.01.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/09/2014] [Accepted: 01/06/2015] [Indexed: 11/25/2022]
Abstract
BACKGROUND Many viral genomes encode a limited number of proteins, illustrating their innate efficiency in bypassing host immune surveillance. This concept of genomic efficiency is exemplified by the 9 kb RNA genome of human immunodeficiency virus 1 (HIV-1), encoding 15 proteins sub-divided according to function. The enzymatic group includes proteins such as the drug targets reverse transcriptase and protease. In contrast, the accessory proteins lack any known enzymatic or structural function, yet are essential for viral fitness and HIV-1 pathogenesis. Of these, the HIV-1 accessory protein Nef is a master manipulator of host cellular processes, ensuring efficient counterattack against the host immune response, as well as long-term evasion of immune surveillance. In particular, the ability of Nef to downmodulate major histocompatibility complex class I (MHC-I) is a key cellular event that enables HIV-1 to bypass the host's defenses by evading the adaptive immune response. SCOPE OF REVIEW In this article, we briefly review how various pathogenic viruses control cell-surface MHC-I, and then focus on the mechanisms and implications of HIV-1 Nef-mediated MHC-I downregulation via modulation of the host membrane trafficking machinery. CONCLUSION The extensive interaction network formed between Nef and numerous membrane trafficking regulators suggests that Nef's role in evading the immune surveillance system intersects multiple host membrane trafficking pathways. SIGNIFICANCE Nef's ability to evade the immune surveillance system is linked to AIDS pathogenesis. Thus, a complete understanding of the molecular pathways that are subverted by Nef in order to downregulate MHC-I will enhance our understanding of HIV-1's progression to AIDS.
Collapse
Affiliation(s)
- Emily N Pawlak
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Jimmy D Dikeakos
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1.
| |
Collapse
|
16
|
Zainabadi K, Jain AV, Donovan FX, Elashoff D, Rao NP, Murty VV, Chandrasekharappa SC, Srivatsan ES. One in four individuals of African-American ancestry harbors a 5.5kb deletion at chromosome 11q13.1. Genomics 2014; 103:276-87. [PMID: 24412158 DOI: 10.1016/j.ygeno.2014.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 11/11/2013] [Accepted: 01/02/2014] [Indexed: 01/11/2023]
Abstract
Cloning and sequencing of 5.5 kb deletion at chromosome 11q13.1 from the HeLa cells, tumorigenic hybrids and two fibroblast cell lines have revealed homologous recombination between AluSx and AluY resulting in the deletion of intervening sequences. Long-range PCR of the 5.5 kb sequence in 494 normal lymphocyte samples showed heterozygous deletion in 28.3% of African-American ancestry samples but only in 4.8% of Caucasian samples (p<0.0001). This observation is strengthened by the copy number variation (CNV) data of the HapMap samples which showed that this deletion occurs in 27% of YRI (Yoruba--West African) population but none in non-African populations. The HapMap analysis further identified strong linkage disequilibrium between 5 single nucleotide polymorphisms and the 5.5 kb deletion in people of African ancestry. Computational analysis of 175 kb sequence surrounding the deletion site revealed enhanced flexibility, low thermodynamic stability, high repetitiveness, and stable stem-loop/hairpin secondary structures that are hallmarks of common fragile sites.
Collapse
Affiliation(s)
- Kayvan Zainabadi
- Division of General Surgery, Department of Surgery, VAGLAHS West Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, CA 90073, USA
| | - Anuja V Jain
- Division of General Surgery, Department of Surgery, VAGLAHS West Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, CA 90073, USA
| | - Frank X Donovan
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Elashoff
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90025, USA
| | - Nagesh P Rao
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90025, USA
| | - Vundavalli V Murty
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Settara C Chandrasekharappa
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eri S Srivatsan
- Division of General Surgery, Department of Surgery, VAGLAHS West Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, CA 90073, USA.
| |
Collapse
|
17
|
Bonnemaison ML, Eipper BA, Mains RE. Role of adaptor proteins in secretory granule biogenesis and maturation. Front Endocrinol (Lausanne) 2013; 4:101. [PMID: 23966980 PMCID: PMC3743005 DOI: 10.3389/fendo.2013.00101] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 07/31/2013] [Indexed: 12/29/2022] Open
Abstract
In the regulated secretory pathway, secretory granules (SGs) store peptide hormones that are released on demand. SGs are formed at the trans-Golgi network and must undergo a maturation process to become responsive to secretagogues. The production of mature SGs requires concentrating newly synthesized soluble content proteins in granules whose membranes contain the appropriate integral membrane proteins. The mechanisms underlying the sorting of soluble and integral membrane proteins destined for SGs from other proteins are not yet well understood. For soluble proteins, luminal pH and divalent metals can affect aggregation and interaction with surrounding membranes. The trafficking of granule membrane proteins can be controlled by both luminal and cytosolic factors. Cytosolic adaptor proteins (APs), which recognize the cytosolic domains of proteins that span the SG membrane, have been shown to play essential roles in the assembly of functional SGs. Adaptor protein 1A (AP-1A) is known to interact with specific motifs in its cargo proteins and with the clathrin heavy chain, contributing to the formation of a clathrin coat. AP-1A is present in patches on immature SG membranes, where it removes cargo and facilitates SG maturation. AP-1A recruitment to membranes can be modulated by Phosphofurin Acidic Cluster Sorting protein 1 (PACS-1), a cytosolic protein which interacts with both AP-1A and cargo that has been phosphorylated by casein kinase II. A cargo/PACS-1/AP-1A complex is necessary to drive the appropriate transport of several cargo proteins within the regulated secretory pathway. The Golgi-localized, γ-ear containing, ADP-ribosylation factor binding (GGA) family of APs serve a similar role. We review the functions of AP-1A, PACS-1, and GGAs in facilitating the retrieval of proteins from immature SGs and review examples of cargo proteins whose trafficking within the regulated secretory pathway is governed by APs.
Collapse
Affiliation(s)
- Mathilde L. Bonnemaison
- Department of Molecular, Microbial and Structural Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Betty A. Eipper
- Department of Molecular, Microbial and Structural Biology, University of Connecticut Health Center, Farmington, CT, USA
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Richard E. Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
- *Correspondence: Richard E. Mains, Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-3401, USA e-mail:
| |
Collapse
|
18
|
Schuurs-Hoeijmakers J, Oh E, Vissers L, Swinkels M, Gilissen C, Willemsen M, Holvoet M, Steehouwer M, Veltman J, de Vries B, van Bokhoven H, de Brouwer A, Katsanis N, Devriendt K, Brunner H. Recurrent de novo mutations in PACS1 cause defective cranial-neural-crest migration and define a recognizable intellectual-disability syndrome. Am J Hum Genet 2012; 91:1122-7. [PMID: 23159249 DOI: 10.1016/j.ajhg.2012.10.013] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 09/05/2012] [Accepted: 10/04/2012] [Indexed: 10/27/2022] Open
Abstract
We studied two unrelated boys with intellectual disability (ID) and a striking facial resemblance suggestive of a hitherto unappreciated syndrome. Exome sequencing in both families identified identical de novo mutations in PACS1, suggestive of causality. To support these genetic findings and to understand the pathomechanism of the mutation, we studied the protein in vitro and in vivo. Altered PACS1 forms cytoplasmic aggregates in vitro with concomitant increased protein stability and shows impaired binding to an isoform-specific variant of TRPV4, but not the full-length protein. Furthermore, consistent with the human pathology, expression of mutant PACS1 mRNA in zebrafish embryos induces craniofacial defects most likely in a dominant-negative fashion. This phenotype is driven by aberrant specification and migration of SOX10-positive cranial, but not enteric, neural-crest cells. Our findings suggest that PACS1 is necessary for the formation of craniofacial structures and that perturbation of its functions results in a specific syndromic ID phenotype.
Collapse
|
19
|
Cancino J, Luini A. Signaling Circuits on the Golgi Complex. Traffic 2012; 14:121-34. [DOI: 10.1111/tra.12022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/12/2012] [Accepted: 10/12/2012] [Indexed: 01/21/2023]
|
20
|
Huang Y, Cavanaugh A, Breitwieser GE. Regulation of stability and trafficking of calcium-sensing receptors by pharmacologic chaperones. ADVANCES IN PHARMACOLOGY 2012; 62:143-73. [PMID: 21907909 DOI: 10.1016/b978-0-12-385952-5.00007-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gain- or loss-of-function mutations and polymorphisms of the calcium-sensing receptor (CaSR) cause Ca(2+) handling diseases. Altered expression and/or signaling of wild-type CaSR can also contribute to pathology. Recent studies have demonstrated that a significant proportion of mutations cause altered targeting and/or trafficking of CaSR to the plasma membrane. Pharmacological approaches to rescue of CaSR function include treatment with allosteric modulators, which potentiate the effects of the orthosteric agonist Ca(2+). Dissection of the mechanism(s) contributing to allosteric agonist-mediated rescue of loss-of-function CaSR mutants has demonstrated pharmacologic chaperone actions coincident with CaSR biosynthesis. The distinctive responses to the allosteric agonist (NPS R-568), which promotes CaSR stability, and the allosteric antagonist (NPS 2143), which promotes CaSR degradation, have led to a model for a conformational checkpoint during CaSR biosynthesis. The conformational checkpoint would "tune" CaSR biosynthesis to cellular signaling state. Navigation of a distinct checkpoint for endoplasmic release can also be augmented by pharmacologic chaperones. The diverse, post-endoplasmic reticulum quality control site(s) for pharmacologic chaperone modulation of CaSR stability and trafficking redefines the role(s) of allosteric modulators in regulation of overall GPCR function.
Collapse
Affiliation(s)
- Ying Huang
- Cancer Drug Research Laboratory, McGill University, Royal Victoria Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
21
|
Wolfenstetter S, Wirsching P, Dotzauer D, Schneider S, Sauer N. Routes to the tonoplast: the sorting of tonoplast transporters in Arabidopsis mesophyll protoplasts. THE PLANT CELL 2012; 24:215-32. [PMID: 22253225 PMCID: PMC3289566 DOI: 10.1105/tpc.111.090415] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 12/22/2011] [Accepted: 12/28/2011] [Indexed: 05/05/2023]
Abstract
Vacuoles perform a multitude of functions in plant cells, including the storage of amino acids and sugars. Tonoplast-localized transporters catalyze the import and release of these molecules. The mechanisms determining the targeting of these transporters to the tonoplast are largely unknown. Using the paralogous Arabidopsis thaliana inositol transporters INT1 (tonoplast) and INT4 (plasma membrane), we performed domain swapping and mutational analyses and identified a C-terminal di-leucine motif responsible for the sorting of higher plant INT1-type transporters to the tonoplast in Arabidopsis mesophyll protoplasts. We demonstrate that this motif can reroute other proteins, such as INT4, SUCROSE TRANSPORTER2 (SUC2), or SWEET1, to the tonoplast and that the position of the motif relative to the transmembrane helix is critical. Rerouted INT4 is functionally active in the tonoplast and complements the growth phenotype of an int1 mutant. In Arabidopsis plants defective in the β-subunit of the AP-3 adaptor complex, INT1 is correctly localized to the tonoplast, while sorting of the vacuolar sucrose transporter SUC4 is blocked in cis-Golgi stacks. Moreover, we demonstrate that both INT1 and SUC4 trafficking to the tonoplast is sensitive to brefeldin A. Our data show that plants possess at least two different Golgi-dependent targeting mechanisms for newly synthesized transporters to the tonoplast.
Collapse
Affiliation(s)
| | | | | | | | - Norbert Sauer
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Molecular Plant Physiology and ECROPS (Erlangen Center of Plant Science), D-91058 Erlangen, Germany
| |
Collapse
|
22
|
Liebau MC, Höpker K, Müller RU, Schmedding I, Zank S, Schairer B, Fabretti F, Höhne M, Bartram MP, Dafinger C, Hackl M, Burst V, Habbig S, Zentgraf H, Blaukat A, Walz G, Benzing T, Schermer B. Nephrocystin-4 regulates Pyk2-induced tyrosine phosphorylation of nephrocystin-1 to control targeting to monocilia. J Biol Chem 2011; 286:14237-45. [PMID: 21357692 PMCID: PMC3077625 DOI: 10.1074/jbc.m110.165464] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nephronophthisis is the most common genetic cause of end-stage renal failure during childhood and adolescence. Genetic studies have identified disease-causing mutations in at least 11 different genes (NPHP1–11), but the function of the corresponding nephrocystin proteins remains poorly understood. The two evolutionarily conserved proteins nephrocystin-1 (NPHP1) and nephrocystin-4 (NPHP4) interact and localize to cilia in kidney, retina, and brain characterizing nephronophthisis and associated pathologies as result of a ciliopathy. Here we show that NPHP4, but not truncating patient mutations, negatively regulates tyrosine phosphorylation of NPHP1. NPHP4 counteracts Pyk2-mediated phosphorylation of three defined tyrosine residues of NPHP1 thereby controlling binding of NPHP1 to the trans-Golgi sorting protein PACS-1. Knockdown of NPHP4 resulted in an accumulation of NPHP1 in trans-Golgi vesicles of ciliated retinal epithelial cells. These data strongly suggest that NPHP4 acts upstream of NPHP1 in a common pathway and support the concept of a role for nephrocystin proteins in intracellular vesicular transport.
Collapse
Affiliation(s)
- Max C Liebau
- Renal Division, Department of Medicine and Center for Molecular Medicine, University of Cologne, 50937 Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Russo J, Snider K, Pereira JS, Russo IH. Estrogen induced breast cancer is the result in the disruption of the asymmetric cell division of the stem cell. Horm Mol Biol Clin Investig 2010; 1:53-65. [PMID: 21258630 PMCID: PMC3024544 DOI: 10.1515/hmbci.2010.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There is evidence that in the human breast there is a stem cell population that can give rise to many different cell types and have the unique potential to divide asymmetrically. In this way stem cells maintain the stem cell pool and simultaneously generate committed cells that reconstitute the organ for example for preparing the breast for a new pregnancy after the involution from a previous pregnancy and lactation process. In addition to the in vivo models of mammary morphogenesis there are in vitro systems that are more amenable to study in critically determined conditions the ductulogenic pattern of growth of the breast epithelia. Primary mammary epithelial cells grown in collagen matrix are able to form tree-like structures resembling in vivo ductulogenesis. The human breast epithelial cells MCF-10F formed tubules when grown in type I collagen and we demonstrated that treatment of these cells with 17β-estradiol (E(2)) induces phonotypical changes indicative of neoplastic transformation. The transformation of MCF-10F by E(2) is associated with impaired ductal morphogenesis by altering the stem cells unique potential to divide asymmetrically inducing formation of solid masses mimicking intraductal carcinoma that progress to invasive and tumorigenic phenotype. In the present work we present evidence for the mechanism of cell asymmetry leading to normal ductulogenesis and how the normal stem cell is transformed to cancer stem cell by altering this process. Furthermore, we demonstrate that the carcinogenic agent, in this case E(2), induces a defect in the asymmetric cell division program of the normal mammary stem cell.
Collapse
Affiliation(s)
- Jose Russo
- Breast Cancer Research laboratory, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
24
|
Alvarado-Díaz CP, Tapia JC, Antonelli M, Moreno RD. Differential localization of α’ and β subunits of protein kinase CK2 during rat spermatogenesis. Cell Tissue Res 2009; 338:139-49. [DOI: 10.1007/s00441-009-0847-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Accepted: 07/10/2009] [Indexed: 11/28/2022]
|
25
|
Jenkins PM, Zhang L, Thomas G, Martens JR. PACS-1 mediates phosphorylation-dependent ciliary trafficking of the cyclic-nucleotide-gated channel in olfactory sensory neurons. J Neurosci 2009; 29:10541-51. [PMID: 19710307 PMCID: PMC2749268 DOI: 10.1523/jneurosci.1590-09.2009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Revised: 06/25/2009] [Accepted: 07/20/2009] [Indexed: 02/02/2023] Open
Abstract
Impaired ciliary protein transport in olfactory sensory neurons (OSNs) leads to anosmia, and is a newly recognized clinical manifestation of a class of human disorders called ciliopathies. Surprisingly little is known regarding the mechanisms controlling trafficking to this unique neuronal compartment. Here, we show a novel role for phosphofurin acidic cluster-sorting protein 1 (PACS-1) in the ciliary trafficking of the olfactory cyclic-nucleotide-gated (CNG) channel. PACS-1 is an intracellular sorting protein that mediates its effects through the binding of acidic clusters on cargo protein. This interaction is dependent on CK2 phosphorylation of both PACS-1 and its cargo. We show that CNGB1b contains two putative PACS-1 binding sites, which are phosphorylated by the serine/threonine protein kinase, CK2. Additionally, we show that PACS-1 is expressed in OSNs and interacts in complex with the CNG channel. CK2 inhibition in native OSNs causes a loss of CNG channel from cilia and subsequent olfactory dysfunction, while adenoviral expression of mutant PACS-1 causes similar mislocalization. These results provide a mechanism for the subunit-dependent ciliary trafficking of the CNG channel and offer insight into the mechanisms of ciliary transport.
Collapse
Affiliation(s)
- Paul M. Jenkins
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109-5632, and
| | - Lian Zhang
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109-5632, and
| | - Gary Thomas
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239
| | - Jeffrey R. Martens
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109-5632, and
| |
Collapse
|
26
|
Chen J, Wang J, Meyers KR, Enns CA. Transferrin-directed internalization and cycling of transferrin receptor 2. Traffic 2009; 10:1488-501. [PMID: 19682329 DOI: 10.1111/j.1600-0854.2009.00961.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Transferrin receptor 2 (TfR2) is a homologue of transferrin receptor 1 (TfR1) but has distinct functions from TfR1 in iron homeostasis. In keeping with its proposed role in iron sensing, previous studies showed that TfR2 has a short half-life and that holo-Tf stabilizes TfR2 by redirecting it from a degradative pathway to a recycling pathway. In this study, we characterized how the endocytosis, recycling and degradation of TfR2 relates to its function and differs from TfR1. TfR2 endocytosis was adaptor protein-2 (AP-2) dependent. Flow cytometry analysis showed that TfR1 and TfR2 utilized the same endocytic pathway only in the presence of holo-Tf, indicating that holo-Tf alters the interaction of TfR2 with the endocytic machinery. Unlike TfR1, phosphofurin acidic cluster sorting protein 1 (PACS-1) binds to the cytoplasmic domain of TfR2 and data suggest that PACS-1 is involved in the TfR2 recycling. Depletion of TSG101 by siRNA or expression of a dominant negative Vps4 inhibited TfR2 degradation, indicating that TfR2 degradation occurs through a multivesicular body (MVB) pathway. TfR2 degradation is not mediated through ubiquitination on the single lysine (K31) in the cytoplasmic domain or on the amino terminal residue. No ubiquitination of TfR2 by HA-ubiquitin was detected, indicating a lack of direct TfR2 ubiquitination involvement in its degradation.
Collapse
Affiliation(s)
- Juxing Chen
- Department of Cell and Developmental Biology L215, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
27
|
At the crossroads of homoeostasis and disease: roles of the PACS proteins in membrane traffic and apoptosis. Biochem J 2009; 421:1-15. [PMID: 19505291 DOI: 10.1042/bj20081016] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The endomembrane system in mammalian cells has evolved over the past two billion years from a simple endocytic pathway in a single-celled primordial ancestor to complex networks supporting multicellular structures that form metazoan tissue and organ systems. The increased organellar complexity of metazoan cells requires additional trafficking machinery absent in yeast or other unicellular organisms to maintain organ homoeostasis and to process the signals that control proliferation, differentiation or the execution of cell death programmes. The PACS (phosphofurin acidic cluster sorting) proteins are one such family of multifunctional membrane traffic regulators that mediate organ homoeostasis and have important roles in diverse pathologies and disease states. This review summarizes our current knowledge of the PACS proteins, including their structure and regulation in cargo binding, their genetics, their roles in secretory and endocytic pathway traffic, interorganellar communication and how cell-death signals reprogramme the PACS proteins to regulate apoptosis. We also summarize our current understanding of how PACS genes are dysregulated in cancer and how viral pathogens ranging from HIV-1 to herpesviruses have evolved to usurp the PACS sorting machinery to promote virus assembly, viral spread and immunoevasion.
Collapse
|
28
|
Caron D, Winstall É, Inaguma Y, Michaud S, Lettre F, Bourassa S, Kelly I, Poirier GG, Faure RL, Tanguay RM. Proteomic Characterization of Mouse Cytosolic and Membrane Prostate Fractions: High Levels of Free SUMO Peptides Are Androgen-Regulated. J Proteome Res 2008; 7:4492-9. [DOI: 10.1021/pr8002497] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Danielle Caron
- Department of Pediatrics, Proteomic platform, CHUL Research Center, Québec G1V 4G2, Canada, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan, and Laboratory of Cellular and Developmental Genetics, Department of Medicine and CREFSIP, Université Laval, Québec G1K 7P4, Canada
| | - Éric Winstall
- Department of Pediatrics, Proteomic platform, CHUL Research Center, Québec G1V 4G2, Canada, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan, and Laboratory of Cellular and Developmental Genetics, Department of Medicine and CREFSIP, Université Laval, Québec G1K 7P4, Canada
| | - Yutaka Inaguma
- Department of Pediatrics, Proteomic platform, CHUL Research Center, Québec G1V 4G2, Canada, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan, and Laboratory of Cellular and Developmental Genetics, Department of Medicine and CREFSIP, Université Laval, Québec G1K 7P4, Canada
| | - Sébastien Michaud
- Department of Pediatrics, Proteomic platform, CHUL Research Center, Québec G1V 4G2, Canada, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan, and Laboratory of Cellular and Developmental Genetics, Department of Medicine and CREFSIP, Université Laval, Québec G1K 7P4, Canada
| | - Francine Lettre
- Department of Pediatrics, Proteomic platform, CHUL Research Center, Québec G1V 4G2, Canada, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan, and Laboratory of Cellular and Developmental Genetics, Department of Medicine and CREFSIP, Université Laval, Québec G1K 7P4, Canada
| | - Sylvie Bourassa
- Department of Pediatrics, Proteomic platform, CHUL Research Center, Québec G1V 4G2, Canada, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan, and Laboratory of Cellular and Developmental Genetics, Department of Medicine and CREFSIP, Université Laval, Québec G1K 7P4, Canada
| | - Isabelle Kelly
- Department of Pediatrics, Proteomic platform, CHUL Research Center, Québec G1V 4G2, Canada, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan, and Laboratory of Cellular and Developmental Genetics, Department of Medicine and CREFSIP, Université Laval, Québec G1K 7P4, Canada
| | - Guy G. Poirier
- Department of Pediatrics, Proteomic platform, CHUL Research Center, Québec G1V 4G2, Canada, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan, and Laboratory of Cellular and Developmental Genetics, Department of Medicine and CREFSIP, Université Laval, Québec G1K 7P4, Canada
| | - Robert L. Faure
- Department of Pediatrics, Proteomic platform, CHUL Research Center, Québec G1V 4G2, Canada, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan, and Laboratory of Cellular and Developmental Genetics, Department of Medicine and CREFSIP, Université Laval, Québec G1K 7P4, Canada
| | - Robert M. Tanguay
- Department of Pediatrics, Proteomic platform, CHUL Research Center, Québec G1V 4G2, Canada, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi 480-0392, Japan, and Laboratory of Cellular and Developmental Genetics, Department of Medicine and CREFSIP, Université Laval, Québec G1K 7P4, Canada
| |
Collapse
|
29
|
Han J, Wang Y, Wang S, Chi C. Interaction of Mint3 with Furin regulates the localization of Furin in the trans-Golgi network. J Cell Sci 2008; 121:2217-23. [PMID: 18544638 DOI: 10.1242/jcs.019745] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Furin is a proprotein convertase that cycles between the plasma membrane, endosomes and the trans-Golgi network (TGN), maintaining a predominant distribution in the latter. Mint3, a member of the Mint protein family, is involved in the signaling and trafficking of membrane proteins. Until now, little has been known about the roles of Mint3 in the localization or trafficking of Furin. Here, using co-immunoprecipitation and immunofluorescence assays, we show that Mint3 interacts with Furin in the Golgi compartment of HeLa cells. Knockdown of endogenous Mint3 expression by RNA interference disrupts the TGN-specific localization of Furin and increases its distribution in endosomes. We further demonstrate that the phosphotyrosine-binding (PTB) domain of Mint3 is essential for the binding of Furin and that this binding affects the TGN-specific localization of Furin. Moreover, mutation studies of Furin indicate that Mint3 regulates Furin distribution mainly through interaction with the acidic peptide signal of Furin. Collectively, these data suggest that the interaction between the PTB domain of Mint3 and the acidic peptide signal of Furin regulates the specific localization of Furin in the TGN.
Collapse
Affiliation(s)
- Jinbo Han
- Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, People's Republic of China
| | | | | | | |
Collapse
|
30
|
Eichhorn PJA, Creyghton MP, Bernards R. Protein phosphatase 2A regulatory subunits and cancer. Biochim Biophys Acta Rev Cancer 2008; 1795:1-15. [PMID: 18588945 DOI: 10.1016/j.bbcan.2008.05.005] [Citation(s) in RCA: 274] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Revised: 05/20/2008] [Accepted: 05/21/2008] [Indexed: 01/06/2023]
Abstract
The serine/threonine protein phosphatase (PP2A) is a trimeric holoenzyme that plays an integral role in the regulation of a number of major signaling pathways whose deregulation can contribute to cancer. The specificity and activity of PP2A are highly regulated through the interaction of a family of regulatory B subunits with the substrates. Accumulating evidence indicates that PP2A acts as a tumor suppressor. In this review we summarize the known effects of specific PP2A holoenzymes and their roles in cancer relevant pathways. In particular we highlight PP2A function in the regulation of MAPK and Wnt signaling.
Collapse
Affiliation(s)
- Pieter J A Eichhorn
- Division of Molecular Carcinogenesis, Center for Cancer Genomics and Center for Biomedical Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | |
Collapse
|
31
|
Boutilier J, Ceni C, Pagdala PC, Forgie A, Neet KE, Barker PA. Proneurotrophins require endocytosis and intracellular proteolysis to induce TrkA activation. J Biol Chem 2008; 283:12709-16. [PMID: 18299325 PMCID: PMC2442317 DOI: 10.1074/jbc.m710018200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 02/14/2008] [Indexed: 01/19/2023] Open
Abstract
The uncleaved, pro-form of nerve growth factor (proNGF) functions as a pro-apoptotic ligand for the p75 neurotrophin receptor (p75NTR). However, some reports have indicated that proneurotrophins bind and activate Trk receptors. In this study, we have examined proneurotrophin receptor binding and activation properties in an attempt to reconcile these findings. We show that proNGF readily binds p75NTR expressed in HEK293T cells but does not interact with TrkA expressed under similar circumstances. Importantly, proNGF activates TrkA tyrosine phosphorylation, induces Erk and Akt activation, and causes PC12 cell differentiation. We show that inhibiting endocytosis or furin activity reduced TrkA activation induced by proNGF but not that induced by mature NGF and that proNGF123, a mutant form of NGF lacking dibasic cleavage sites in the prodomain, does not induce TrkA phosphorylation in PC12 cells. Therefore, endocytosis and cleavage appear to be prerequisites for proNGF-induced TrkA activity. We also found that proBDNF induces activation of TrkB in cerebellar granule neurons and that proBDNF cleavage by furin and metalloproteases facilitates this effect. Taken together, these data indicate that under physiological conditions, proneurotrophins do not directly bind or activate Trk receptors. However, endocytosis and cleavage of proneurotrophins produce processed forms of neurotrophins that are capable of inducing Trk activation.
Collapse
Affiliation(s)
- Jacqueline Boutilier
- Department of Neurology and Neurosurgery, McGill University, Montreal Neurological Institute, Montreal H3A 2B4, Canada
| | | | | | | | | | | |
Collapse
|
32
|
Atkins KM, Thomas L, Youker RT, Harriff MJ, Pissani F, You H, Thomas G. HIV-1 Nef binds PACS-2 to assemble a multikinase cascade that triggers major histocompatibility complex class I (MHC-I) down-regulation: analysis using short interfering RNA and knock-out mice. J Biol Chem 2008; 283:11772-84. [PMID: 18296443 DOI: 10.1074/jbc.m707572200] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human immunodeficiency virus, type 1, negative factor (Nef) initiates down-regulation of cell-surface major histocompatibility complex-I (MHC-I) by assembling an Src family kinase (SFK)-ZAP70/Syk-phosphoinositide 3-kinase (PI3K) cascade through the sequential actions of two sites, Nef EEEE(65) and PXXP(75). The internalized MHC-I molecules are then sequestered in endosomal compartments by a process requiring Nef Met(20). How Nef assembles the multikinase cascade to trigger the MHC-I down-regulation pathway is unknown. Here we report that EEEE(65)-dependent binding to the sorting protein PACS-2 targets Nef to the paranuclear region, enabling PXXP(75) to bind and activate a trans-Golgi network (TGN)-localized SFK. This SFK then phosphorylates ZAP-70 to recruit class I PI3K by interaction with the p85 C-terminal Src homology 2 domain. Using splenocytes and embryonic fibroblasts from PACS-2(-/-) mice, we confirm genetically that Nef requires PACS-2 to localize to the paranuclear region and assemble the multikinase cascade. Moreover, genetic loss of PACS-2 or inhibition of class I PI3K prevents Nef-mediated MHC-I down-regulation, demonstrating that short interfering RNA knockdown of PACS-2 phenocopies the gene knock-out. This PACS-2-dependent targeting pathway is not restricted to Nef, because PACS-2 is also required for trafficking of an endocytosed cation-independent mannose 6-phosphate receptor reporter from early endosomes to the TGN. Together, these results demonstrate PACS-2 is required for Nef action and sorting of itinerant membrane cargo in the TGN/endosomal system.
Collapse
|
33
|
Arrigoni G, Pagano MA, Sarno S, Cesaro L, James P, Pinna LA. Mass spectrometry analysis of a protein kinase CK2beta subunit interactome isolated from mouse brain by affinity chromatography. J Proteome Res 2008; 7:990-1000. [PMID: 18220339 DOI: 10.1021/pr070500s] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
CK2, an acronym derived from the misnomer "casein kinase 2", denotes a ubiquitous and extremely pleiotropic Ser/Thr protein kinase, the holoenzyme of which is composed of two catalytic (alpha and/or alpha') and two noncatalytic beta subunits acting as a docking platform and the multifarious functions of which are still incompletely understood. By combining affinity chromatography and mass spectrometry, we have identified 144 mouse brain proteins that associate with immobilized CK2beta. A large proportion (60%) of the identified proteins had been previously reported to be functionally related to CK2, and a similar proportion have been classified as phosphoproteins with approximately half of these having the features of CK2 targets. A large number of the identified proteins ( approximately 40%) either are nuclear or shuttle between the nucleus and cytoplasm, and the biggest functional classes of CK2beta interactors are committed to protein synthesis and degradation (32 proteins) and RNA/DNA interaction (20 proteins). Also well represented are the categories of cytoskeletal/structural proteins (19), trafficking proteins (17), and signaling proteins (14). The identified proteins are examined in relation to their functions and potential as targets and/or regulators of CK2, disclosing in some cases unanticipated links between this kinase and a variety of biochemical events.
Collapse
Affiliation(s)
- Giorgio Arrigoni
- Department of Biological Chemistry and CNR Institute of Neurosciences, University of Padova, Padova, Italy.
| | | | | | | | | | | |
Collapse
|
34
|
Bujny MV, Popoff V, Johannes L, Cullen PJ. The retromer component sorting nexin-1 is required for efficient retrograde transport of Shiga toxin from early endosome to the trans Golgi network. J Cell Sci 2007; 120:2010-21. [PMID: 17550970 DOI: 10.1242/jcs.003111] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The mammalian retromer complex is a multi-protein complex that regulates retrograde transport of the cation-independent mannose 6-phosphate receptor (CI-MPR) from early endosomes to the trans Golgi network (TGN). It consists of two subcomplexes: a membrane-bound coat comprising sorting nexin-1 (SNX1) and possibly sorting nexin-2 (SNX2), and a cargo-selective subcomplex, composed of VPS26, VPS29 and VPS35. In addition to the retromer, a variety of other protein complexes has been suggested to regulate endosome-to-TGN transport of not only the CI-MPR but a wide range of other cargo proteins. Here, we have examined the role of SNX1 and SNX2 in endosomal sorting of Shiga and cholera toxins, two toxins that undergo endosome-to-TGN transport en route to their cellular targets located within the cytosol. By using small interfering RNA (siRNA)-mediated silencing combined with single-cell fluorescent-toxin-uptake assays and well-established biochemical assays to analyze toxin delivery to the TGN, we have established that suppression of SNX1 leads to a significant reduction in the efficiency of endosome-to-TGN transport of the Shiga toxin B-subunit. Furthermore, we show that for the B subunit of cholera toxin, retrograde endosome-to-TGN transport is less reliant upon SNX1. Overall, our data establish a role for SNX1 in the endosome-to-TGN transport of Shiga toxin and are indicative for a fundamental difference between endosomal sorting of Shiga and cholera toxins into endosome-to-TGN retrograde transport pathways.
Collapse
Affiliation(s)
- Miriam V Bujny
- The Henry Wellcome Integrated Signalling Laboratories, Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | | | | | | |
Collapse
|
35
|
Lubben NB, Sahlender DA, Motley AM, Lehner PJ, Benaroch P, Robinson MS. HIV-1 Nef-induced down-regulation of MHC class I requires AP-1 and clathrin but not PACS-1 and is impeded by AP-2. Mol Biol Cell 2007; 18:3351-65. [PMID: 17581864 PMCID: PMC1951775 DOI: 10.1091/mbc.e07-03-0218] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Revised: 05/24/2007] [Accepted: 06/11/2007] [Indexed: 12/13/2022] Open
Abstract
Major histocompatibility complex class I is down-regulated from the surface of human immunodeficiency virus (HIV)-1-infected cells by Nef, a virally encoded protein that is thought to reroute MHC-I to the trans-Golgi network (TGN) in a phosphofurin acidic cluster sorting protein (PACS) 1, adaptor protein (AP)-1, and clathrin-dependent manner. More recently, an alternative model has been proposed, in which Nef uses AP-1 to direct MHC-I to endosomes and lysosomes. Here, we show that knocking down either AP-1 or clathrin with small interfering RNA inhibits the down-regulation of HLA-A2 (an MHC-I isotype) by Nef in HeLa cells. However, knocking down PACS-1 has no effect, not only on Nef-induced down-regulation of HLA-A2 but also on the localization of other proteins containing acidic cluster motifs. Surprisingly, knocking down AP-2 actually enhances Nef activity. Immuno-electron microscopy labeling of Nef-expressing cells indicates that HLA-A2 is rerouted not to the TGN, but to endosomes. In AP-2-depleted cells, more of the HLA-A2 localizes to the inner vesicles of multivesicular bodies. We propose that depleting AP-2 potentiates Nef activity by altering the membrane composition and dynamics of endosomes and causing increased delivery of HLA-A2 to a prelysosomal compartment.
Collapse
Affiliation(s)
- Nienke B. Lubben
- *University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom; and
| | - Daniela A. Sahlender
- *University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom; and
| | - Alison M. Motley
- *University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom; and
| | - Paul J. Lehner
- *University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom; and
| | | | - Margaret S. Robinson
- *University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom; and
| |
Collapse
|
36
|
Longin S, Zwaenepoel K, Martens E, Louis JV, Rondelez E, Goris J, Janssens V. Spatial control of protein phosphatase 2A (de)methylation. Exp Cell Res 2007; 314:68-81. [PMID: 17803990 DOI: 10.1016/j.yexcr.2007.07.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Revised: 07/20/2007] [Accepted: 07/21/2007] [Indexed: 10/23/2022]
Abstract
Reversible methylation of the protein phosphatase 2A catalytic subunit (PP2A(C)(1)) is an important regulatory mechanism playing a crucial role in the selective recruitment of regulatory B subunits. Here, we investigated the subcellular localization of leucine carboxyl methyltransferase (LCMT1) and protein phosphatase methylesterase (PME-1), the two enzymes catalyzing this process. The results show that PME-1 is predominantly localized in the nucleus and harbors a functional nuclear localization signal, whereas LCMT1 is underrepresented in the nucleus and mainly localizes to the cytoplasm, Golgi region and late endosomes. Indirect immunofluorescence with methylation-sensitive anti-PP2A(C) antibodies revealed a good correlation with the methylation status of PP2A(C), demethylated PP2A(C) being substantially nuclear. Throughout mitosis, demethylated PP2A(C) is associated with the mitotic spindle and during cytokinesis with the cleavage furrow. Overexpression of PME-1, but not of an inactive mutant, results in increased demethylation of PP2A(C) in the nucleus, whereas overexpression of a cytoplasmic PME-1 mutant lacking the NLS results in increased demethylation in the cytoplasm-in all cases, however, without any obvious functional consequences. PME-1 associates with an inactive PP2A population, regardless of its esterase activity or localization. We propose that stabilization of this inactive, nuclear PP2A pool is a major in vivo function of PME-1.
Collapse
Affiliation(s)
- Sari Longin
- Department of Molecular Cell Biology, Faculty of Medicine, KU Leuven, Herestraat 49 bus 901, B-3000 Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
37
|
Hung CH, Thomas L, Ruby CE, Atkins KM, Morris NP, Knight ZA, Scholz I, Barklis E, Weinberg AD, Shokat KM, Thomas G. HIV-1 Nef assembles a Src family kinase-ZAP-70/Syk-PI3K cascade to downregulate cell-surface MHC-I. Cell Host Microbe 2007; 1:121-33. [PMID: 18005690 DOI: 10.1016/j.chom.2007.03.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2006] [Revised: 01/25/2007] [Accepted: 03/20/2007] [Indexed: 11/24/2022]
Abstract
HIV-1 Nef, which is required for the efficient onset of AIDS, enhances viral replication and infectivity by exerting multiple effects on infected cells. Nef downregulates cell-surface MHC-I molecules by an uncharacterized PI3K pathway requiring the actions of two Nef motifs-EEEE(65) and PXXP(75). We report that the Nef EEEE(65) targeting motif enables Nef PXXP(75) to bind and activate a trans-Golgi network-localized Src family tyrosine kinase (SFK). The Nef/SFK complex then recruits and phosphorylates the tyrosine kinase ZAP-70, which binds class I PI3K to trigger MHC-I downregulation in primary CD4+ T cells. In promonocytic cells, Nef/SFK recruits the ZAP-70 homolog Syk to downregulate MHC-I, implicating this PI3K pathway in multiple HIV-1 reservoirs. Isoform-specific PI3K inhibitors repress MHC-I downregulation, identifying them as potential therapeutic agents to combat HIV-1. The discovery of this Nef-SFK-ZAP-70/Syk-PI3K signaling pathway explains the hierarchal role of the Nef motifs in effecting immunoevasion.
Collapse
Affiliation(s)
- Chien-Hui Hung
- Vollum Institute, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Scott GK, Fei H, Thomas L, Medigeshi GR, Thomas G. A PACS-1, GGA3 and CK2 complex regulates CI-MPR trafficking. EMBO J 2006; 25:4423-35. [PMID: 16977309 PMCID: PMC1589982 DOI: 10.1038/sj.emboj.7601336] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Accepted: 08/16/2006] [Indexed: 11/09/2022] Open
Abstract
The cation-independent mannose-6-phosphate receptor (CI-MPR) follows a highly regulated sorting itinerary to deliver hydrolases from the trans-Golgi network (TGN) to lysosomes. Cycling of CI-MPR between the TGN and early endosomes is mediated by GGA3, which directs TGN export, and PACS-1, which directs endosome-to-TGN retrieval. Despite executing opposing sorting steps, GGA3 and PACS-1 bind to an overlapping CI-MPR trafficking motif and their sorting activity is controlled by the CK2 phosphorylation of their respective autoregulatory domains. However, how CK2 coordinates these opposing roles is unknown. We report a CK2-activated phosphorylation cascade controlling PACS-1- and GGA3-mediated CI-MPR sorting. PACS-1 links GGA3 to CK2, forming a multimeric complex required for CI-MPR sorting. PACS-1-bound CK2 stimulates GGA3 phosphorylation, releasing GGA3 from CI-MPR and early endosomes. Bound CK2 also phosphorylates PACS-1Ser(278), promoting binding of PACS-1 to CI-MPR to retrieve the receptor to the TGN. Our results identify a CK2-controlled cascade regulating hydrolase trafficking and sorting of itinerant proteins in the TGN/endosomal system.
Collapse
Affiliation(s)
| | - Hao Fei
- Vollum Institute, Portland, OR, USA
| | | | | | - Gary Thomas
- Vollum Institute, Portland, OR, USA
- Vollum Institute, Oregon Health & Science University, Oregon Health Sciences University, L-474, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, 97239, USA. Tel.: +1 503 494 6955; Fax: +1 503 494 1218; E-mail:
| |
Collapse
|
39
|
Schermer B, Höpker K, Omran H, Ghenoiu C, Fliegauf M, Fekete A, Horvath J, Köttgen M, Hackl M, Zschiedrich S, Huber TB, Kramer-Zucker A, Zentgraf H, Blaukat A, Walz G, Benzing T. Phosphorylation by casein kinase 2 induces PACS-1 binding of nephrocystin and targeting to cilia. EMBO J 2005; 24:4415-24. [PMID: 16308564 PMCID: PMC1356326 DOI: 10.1038/sj.emboj.7600885] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2005] [Accepted: 11/03/2005] [Indexed: 11/09/2022] Open
Abstract
Mutations in proteins localized to cilia and basal bodies have been implicated in a growing number of human diseases. Access of these proteins to the ciliary compartment requires targeting to the base of the cilia. However, the mechanisms involved in transport of cilia proteins to this transitional zone are elusive. Here we show that nephrocystin, a ciliary protein mutated in the most prevalent form of cystic kidney disease in childhood, is expressed in respiratory epithelial cells and accumulates at the base of cilia, overlapping with markers of the basal body area and the transition zone. Nephrocystin interacts with the phosphofurin acidic cluster sorting protein (PACS)-1. Casein kinase 2 (CK2)-mediated phosphorylation of three critical serine residues within a cluster of acidic amino acids in nephrocystin mediates PACS-1 binding, and is essential for colocalization of nephrocystin with PACS-1 at the base of cilia. Inhibition of CK2 activity abrogates this interaction and results in the loss of correct nephrocystin targeting. These data suggest that CK2-dependent transport processes represent a novel pathway of targeting proteins to the cilia.
Collapse
Affiliation(s)
| | - Katja Höpker
- Renal Division, University Hospital Freiburg, Freiburg, Germany
| | - Heymut Omran
- Childrens Hospital, University Hospital Freiburg, Freiburg, Germany
| | | | - Manfred Fliegauf
- Childrens Hospital, University Hospital Freiburg, Freiburg, Germany
| | - Andrea Fekete
- Childrens Hospital, University Hospital Freiburg, Freiburg, Germany
| | - Judit Horvath
- Childrens Hospital, University Hospital Freiburg, Freiburg, Germany
| | - Michael Köttgen
- Renal Division, University Hospital Freiburg, Freiburg, Germany
| | - Matthias Hackl
- Renal Division, University Hospital Freiburg, Freiburg, Germany
| | | | - Tobias B Huber
- Renal Division, University Hospital Freiburg, Freiburg, Germany
| | | | | | - Andree Blaukat
- Department of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Gerd Walz
- Renal Division, University Hospital Freiburg, Freiburg, Germany
| | - Thomas Benzing
- Renal Division, University Hospital Freiburg, Freiburg, Germany
- Renal Division, University Hospital, Hugstetterstrasse 55, 79106 Freiburg, Germany. Tel.: +49 761 270 3559; Fax: +49 761 270 3270; E-mail:
| |
Collapse
|
40
|
Simmen T, Aslan JE, Blagoveshchenskaya AD, Thomas L, Wan L, Xiang Y, Feliciangeli SF, Hung CH, Crump CM, Thomas G. PACS-2 controls endoplasmic reticulum-mitochondria communication and Bid-mediated apoptosis. EMBO J 2005; 24:717-29. [PMID: 15692567 PMCID: PMC549619 DOI: 10.1038/sj.emboj.7600559] [Citation(s) in RCA: 462] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2004] [Accepted: 12/15/2004] [Indexed: 01/07/2023] Open
Abstract
The endoplasmic reticulum (ER) and mitochondria form contacts that support communication between these two organelles, including synthesis and transfer of lipids, and the exchange of calcium, which regulates ER chaperones, mitochondrial ATP production, and apoptosis. Despite the fundamental roles for ER-mitochondria contacts, little is known about the molecules that regulate them. Here we report the identification of a multifunctional sorting protein, PACS-2, that integrates ER-mitochondria communication, ER homeostasis, and apoptosis. PACS-2 controls the apposition of mitochondria with the ER, as depletion of PACS-2 causes BAP31-dependent mitochondria fragmentation and uncoupling from the ER. PACS-2 also controls formation of ER lipid-synthesizing centers found on mitochondria-associated membranes and ER homeostasis. However, in response to apoptotic inducers, PACS-2 translocates Bid to mitochondria, which initiates a sequence of events including the formation of mitochondrial truncated Bid, the release of cytochrome c, and the activation of caspase-3, thereby causing cell death. Together, our results identify PACS-2 as a novel sorting protein that links the ER-mitochondria axis to ER homeostasis and the control of cell fate, and provide new insights into Bid action.
Collapse
Affiliation(s)
| | | | | | | | - Lei Wan
- Vollum Institute, Portland, OR, USA
| | | | | | | | | | | |
Collapse
|
41
|
Maresova L, Pasieka TJ, Homan E, Gerday E, Grose C. Incorporation of three endocytosed varicella-zoster virus glycoproteins, gE, gH, and gB, into the virion envelope. J Virol 2005; 79:997-1007. [PMID: 15613328 PMCID: PMC538533 DOI: 10.1128/jvi.79.2.997-1007.2005] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The cytoplasmic tails of all three major varicella-zoster virus (VZV) glycoproteins, gE, gH, and gB, harbor functional tyrosine-based endocytosis motifs that mediate internalization. The aim of the present study was to examine whether endocytosis from the plasma membrane is a cellular route by which VZV glycoproteins are delivered to the final envelopment compartment. In this study, we demonstrated that internalization of the glycoproteins occurred in the first 24 h postinfection but was reduced later in infection. Using surface biotinylation of VZV-infected cells followed by a glutathione cleavage assay, we showed that endocytosis was independent of antibody binding to gE, gH, and gB. Subsequently, with this assay, we demonstrated that biotinylated gE, gH, and gB retrieved from the cell surface were incorporated into nascent virus particles isolated after density gradient sedimentation. To confirm and extend this finding, we repeated the above sedimentation step and specifically detected envelopes decorated with Streptavidin-conjugated gold beads on a majority of complete virions through examination by transmission electron microscopy. In addition, a gE-gI complex and a gE-gH complex were found on the virions. Therefore, the above studies established that VZV subsumed a postendocytosis trafficking pathway as one mechanism by which to deliver viral glycoproteins to the site of virion assembly in the cytoplasm. Furthermore, since a recombinant VZV genome lacking only endocytosis-competent gE cannot replicate, these results supported the conclusion that the endocytosis-envelopment pathway is an essential component of the VZV life cycle.
Collapse
Affiliation(s)
- Lucie Maresova
- University of Iowa Hospital/2501 JCP, 200 Hawkins Dr., Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
42
|
Farr CD, Gafken PR, Norbeck AD, Doneanu CE, Stapels MD, Barofsky DF, Minami M, Saugstad JA. Proteomic analysis of native metabotropic glutamate receptor 5 protein complexes reveals novel molecular constituents. J Neurochem 2004; 91:438-50. [PMID: 15447677 PMCID: PMC2747775 DOI: 10.1111/j.1471-4159.2004.02735.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We used a proteomic approach to identify novel proteins that may regulate metabotropic glutamate receptor 5 (mGluR5) responses by direct or indirect protein interactions. This approach does not rely on the heterologous expression of proteins and offers the advantage of identifying protein interactions in a native environment. The mGluR5 protein was immunoprecipitated from rat brain lysates; co-immunoprecipitating proteins were analyzed by mass spectrometry and identified peptides were matched to protein databases to determine the correlating parent proteins. This proteomic approach revealed the interaction of mGluR5 with known regulatory proteins, as well as novel proteins that reflect previously unidentified molecular constituents of the mGluR5-signaling complex. Immunoblot analysis confirmed the interaction of high confidence proteins, such as phosphofurin acidic cluster sorting protein 1, microtubule-associated protein 2a and dynamin 1, as mGluR5-interacting proteins. These studies show that a proteomic approach can be used to identify candidate interacting proteins. This approach may be particularly useful for neurobiology applications where distinct protein interactions within a signaling complex can dramatically alter the outcome of the response to neurotransmitter release, or the disruption of normal protein interactions can lead to severe neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Carol D. Farr
- Robert S. Dow Neurobiology Laboratories, Legacy Research, Portland, Oregon, USA
| | - Philip R. Gafken
- Proteomics Facility, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Angela D. Norbeck
- Proteomics Facility, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Catalin E. Doneanu
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, USA
| | - Martha D. Stapels
- Department of Chemistry, Oregon State University, Corvallis, Oregon, USA
| | - Douglas F. Barofsky
- Department of Chemistry, Oregon State University, Corvallis, Oregon, USA
- Mass Spectrometry Core Facility of the Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon, USA
| | - Manabu Minami
- Robert S. Dow Neurobiology Laboratories, Legacy Research, Portland, Oregon, USA
| | - Julie A. Saugstad
- Robert S. Dow Neurobiology Laboratories, Legacy Research, Portland, Oregon, USA
| |
Collapse
|