1
|
van Haaren MJH, Steller LB, Vastert SJ, Calis JJA, van Loosdregt J. Get Spliced: Uniting Alternative Splicing and Arthritis. Int J Mol Sci 2024; 25:8123. [PMID: 39125692 PMCID: PMC11311815 DOI: 10.3390/ijms25158123] [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: 06/25/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Immune responses demand the rapid and precise regulation of gene protein expression. Splicing is a crucial step in this process; ~95% of protein-coding gene transcripts are spliced during mRNA maturation. Alternative splicing allows for distinct functional regulation, as it can affect transcript degradation and can lead to alternative functional protein isoforms. There is increasing evidence that splicing can directly regulate immune responses. For several genes, immune cells display dramatic changes in isoform-level transcript expression patterns upon activation. Recent advances in long-read RNA sequencing assays have enabled an unbiased and complete description of transcript isoform expression patterns. With an increasing amount of cell types and conditions that have been analyzed with such assays, thousands of novel transcript isoforms have been identified. Alternative splicing has been associated with autoimmune diseases, including arthritis. Here, GWASs revealed that SNPs associated with arthritis are enriched in splice sites. In this review, we will discuss how alternative splicing is involved in immune responses and how the dysregulation of alternative splicing can contribute to arthritis pathogenesis. In addition, we will discuss the therapeutic potential of modulating alternative splicing, which includes examples of spliceform-based biomarkers for disease severity or disease subtype, splicing manipulation using antisense oligonucleotides, and the targeting of specific immune-related spliceforms using antibodies.
Collapse
Affiliation(s)
- Maurice J. H. van Haaren
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Levina Bertina Steller
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Sebastiaan J. Vastert
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Division of Pediatric Rheumatology and Immunology, Wilhelmina Children’s Hospital, 3584 CX Utrecht, The Netherlands
| | - Jorg J. A. Calis
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Jorg van Loosdregt
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
2
|
Kakinuma T, Toh BH, Sentry JW. Human autoantibodies as reagents in biomedical research. Mod Rheumatol 2014; 13:15-21. [PMID: 24387111 DOI: 10.3109/s101650300002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Abstract Autoantibodies are typically associated with autoimmune diseases. In some instances the association of specific autoantibodies to a specific autoimmune disease have made their detection invaluable in clinical diagnosis. However, certain autoantibodies show no specific disease association and therefore have limited clinical utility. Nevertheless, autoantibodies are powerful tools for identification, characterization, and functional studies of their cognate antoantigens. In addition, the study of autoantibodies and their cognate autoantigens in human disease and in experimental animal models can provide valuable insight into disease mechanisms and the factors that ameliorate or reverse disease. This review will focus on three specific sets of autoantibodies, which were initially selected for investigation purely on the basis of their novel patterns of reactivity. These were observed when they were applied to a diagnostic HEp-2 test slide for antinuclear antibody detection by indirect immunofluorescence. The target autoantigens were identified as the trans-Golgi network protein GOLGA4 (Golgin 245 or p230), the early endosome antigen-1 (EEA1) and a yet to be identified and fully characterized phosphoepitope(s) restricted to chromosomal arms of condensed mitotic/meiotic chromosomes (MCA1). This laboratory has exploited sera which are reactive to these autoantigens for their identification and characterization, and in functional studies. This review highlights the uses of autoantibodies that may have limited diagnostic or prognostic utility, but are nonetheless novel reagents in the prosecution of molecular cell biology.
Collapse
Affiliation(s)
- T Kakinuma
- Department of Orthopaedic Surgery, Faculty of Medicine, Kyoto University , Kyoto , Japan
| | | | | |
Collapse
|
3
|
Herbert N, Haferkamp A, Schmitz-Winnenthal HF, Zöller M. Concomitant tumor and autoantigen vaccination supports renal cell carcinoma rejection. THE JOURNAL OF IMMUNOLOGY 2010; 185:902-16. [PMID: 20548033 DOI: 10.4049/jimmunol.0902683] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Efficient tumor vaccination frequently requires adjuvant. Concomitant induction of an autoimmune response is discussed as a means to strengthen a weak tumor Ag-specific response. We asked whether the efficacy of dendritic cell (DC) vaccination with the renal cell carcinoma Ags MAGE-A9 (MAGE9) and G250 could be strengthened by covaccination with the renal cell carcinoma autoantigen GOLGA4. BALB/c mice were vaccinated with DC loaded with MHC class I-binding peptides of MAGE9 or G250 or tumor lysate, which sufficed for rejection of low-dose RENCA-MAGE9 and RENCA-G250 tumor grafts, but only retarded tumor growth at 200 times the tumor dose at which 100% of animals will develop a tumor. Instead, 75-100% of mice prevaccinated concomitantly with Salmonella typhimurium transformed with GOLGA4 cDNA in a eukaryotic expression vector rejected 200 times the tumor dose at which 100% of animals will develop tumor. In a therapeutic setting, the survival rate increased from 20-40% by covaccination with S. typhimurium-GOLGA4. Autoantigen covaccination significantly strengthened tumor Ag-specific CD4(+) and CD8(+) T cell expansion, particularly in peptide-loaded DC-vaccinated mice. Covaccination was accompanied by an increase in inflammatory cytokines, boosted IL-12 and IFN-gamma expression, and promoted a high tumor Ag-specific CTL response. Concomitant autoantigen vaccination also supported CCR6, CXCR3, and CXCR4 upregulation and T cell recruitment into the tumor. It did not affect regulatory T cells, but slightly increased myeloid-derived suppressor cells. Thus, tumor cell eradication was efficiently strengthened by concomitant induction of an immune response against a tumor Ag and an autoantigen expressed by the tumor cell. Activation of autoantigen-specific Th cells strongly supports tumor-specific Th cells and thereby CTL activation.
Collapse
Affiliation(s)
- Nicolás Herbert
- Department of Tumor Cell Biology, University Hospital of Surgery, University of Heidelberg, Germany
| | | | | | | |
Collapse
|
4
|
Xia Q, Lu F, Yan HP, Wang HX, Feng X, Zhao Y, Liu BY, Wang J, Li P, Xue Y, Hu MR, Qian L, Guo N, Yang SC, Li MY, Ma YF, Li BA, Zhang XM, Shen BF. Autoantibody profiling of Chinese patients with autoimmune hepatitis using immunoproteomic analysis. J Proteome Res 2008; 7:1963-70. [PMID: 18355017 DOI: 10.1021/pr700861s] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In the present study, immunoproteomic analysis was utilized to systemically characterize global autoantibody profiles in autoimmune hepatitis (AIH). Sera from 21 patients with AIH and 15 healthy controls were analyzed for the antibody reactivity against the protein antigens of HepG2, a human hepatoma cell line. The lysates of HepG2 cells were separated by two-dimensional electrophoresis and then immunoblotted with each serum sample. Matrix-assisted laser desorption/ionization mass spectrometry or/and nanoelectrospray ionization MS/MS were then used to identify antigens, among which a bifunctional enzyme in mitochondrial, fumarate hydratase (FH), was further analyzed by ELISA using recombinant FH as a coating antigen. A total of 18 immunoreactive spots were identified as 13 proteins, 8 of which have not been reported in AIH. Immune reactivity to FH was detected in 66.67% of patients with AIH, 19.35% of patients with primary biliary cirrhosis (PBC), 12.31% of patients with chronic hepatitis B (CHB), 6.35% of patients with chronic hepatitis C (CHC), 11.32% of patients with systemic lupus erythematosus (SLE), and 3.57% of normal individuals. The differences of prevalence between AIH patients and healthy controls as well as other diseases were of statistical significance (P<0.001). These data demonstrate the serological heterogeneity in AIH and suggest the diversity of the mechanisms underlying AIH. FH, recognized mainly in AIH rather than in viral hepatitis and other autoimmune diseases, may have utility in improved diagnosis of AIH.
Collapse
Affiliation(s)
- Qing Xia
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Taiping Road 27, Beijing 100850, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Williams PH, Cobb BL, Namjou B, Scofield RH, Sawalha AH, Harley JB. Horizons in Sjögren's syndrome genetics. Clin Rev Allergy Immunol 2008; 32:201-9. [PMID: 17963047 DOI: 10.1007/s12016-007-8002-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Sjögren's syndrome (SS) is a complex polygenic autoimmune disorder. A few major genetic effects have been identified. Historically, HLA and non-HLA genetic associations have been reported. Recently, the HLA region continued to reveal association findings. A new susceptibility region has been suggested by a study of a D6S349 microsatellite marker. Among non-HLA studies, recent association of immunoglobulin kappa chain allotype KM1 with anti-La autoantibodies in primary Sjögren's syndrome confirms findings in a study from two decades ago. Meanwhile, mouse models have been employed to study the genetic contribution to salivary lymphadenitis or dry eyes and mouth. Gene transfer exploration in mouse models shows promise. The authors review the HLA and non-HLA association studies and the mouse model work that has been reported. Newly developed genomic capacity will provide, in the future, a much closer approximation of the true picture of the genetic architecture of Sjögren's syndrome.
Collapse
Affiliation(s)
- Pamela H Williams
- Arthritis and Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City 73104, USA
| | | | | | | | | | | |
Collapse
|
6
|
Hayes GM, Carrigan PE, Dong M, Reubi JC, Miller LJ. A novel secretin receptor splice variant potentially useful for early diagnosis of pancreatic carcinoma. Gastroenterology 2007; 133:853-61. [PMID: 17678920 DOI: 10.1053/j.gastro.2007.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2006] [Accepted: 06/11/2007] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Pancreatic and bile duct carcinomas represent highly aggressive malignancies that evolve from secretin receptor-rich ductular cells. With premessenger RNA splicing abnormalities common in cancer, we evaluated whether an abnormal secretin receptor spliceoform were present, characterized it, and developed a serum assay for it. METHODS Cancer cell lines and healthy and neoplastic tissue were studied by nested reverse-transcription polymerase chain reaction and sequencing. A promising spliceoform was isolated and characterized, and monoclonal antibodies were raised to 2 distinct regions. A dual antibody enzyme-linked immunosorbent assay was developed and applied to blinded serum samples from 26 patients with pancreatic carcinoma, 10 patients with chronic pancreatitis, and 14 controls. RESULTS Each of 9 pancreatic cancer specimens and no normal tissue expressed a secretin receptor variant with exons 3 and 4 deleted. This encoded a 111-residue peptide with its first 43 residues identical to wild-type receptor, but, subsequent to a shift in coding frame and early truncation, the next 68 residues were unique in the transcriptome/proteome. This nonfunctional soluble protein did not bind or signal in response to secretin and was secreted from transfected MiaPaCa-2 cells. Elevated serum levels of this variant were present in 69% of pancreatic cancer patients, 60% of chronic pancreatitis patients, and 1 of 14 controls. CONCLUSIONS We identified a novel abnormal spliceoform of the secretin receptor in pancreatic and bile duct cancers and developed a dual antibody sandwich enzyme-linked immunosorbent assay to measure it in the circulation. Initial application of this assay in patients with pancreatic cancer and chronic pancreatitis was promising, but additional validation will be required to evaluate its clinical utility.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/immunology
- Base Sequence
- Biomarkers/metabolism
- Case-Control Studies
- Cell Line
- Cell Line, Tumor
- Cholangiocarcinoma/diagnosis
- Cholangiocarcinoma/metabolism
- Female
- Humans
- Male
- Mice
- Mice, Inbred BALB C
- Middle Aged
- Molecular Sequence Data
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/metabolism
- Pancreatitis, Chronic/diagnosis
- Pancreatitis, Chronic/metabolism
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/immunology
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Gastrointestinal Hormone/genetics
- Receptors, Gastrointestinal Hormone/immunology
- Receptors, Gastrointestinal Hormone/metabolism
Collapse
Affiliation(s)
- Gregory M Hayes
- Mayo Clinic, Cancer Center and Department of Molecular Pharmacology and Experimental Therapeutics, Scottsdale, Arizona 85259, USA
| | | | | | | | | |
Collapse
|
7
|
Dunster K, Lai FPL, Sentry JW. Limkain b1, a novel human autoantigen localized to a subset of ABCD3 and PXF marked peroxisomes. Clin Exp Immunol 2005; 140:556-63. [PMID: 15932519 PMCID: PMC1809386 DOI: 10.1111/j.1365-2249.2005.02774.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Detection of self-reactive antibodies has an established role in the diagnosis and monitoring of many human autoimmune diseases. Autoantibodies with restricted reactivity to cytoplasmic compartments and structures are an occasional incidental finding following routine examination of serum for antinuclear antibody reactivity. A prerequisite for rational exploitation of self-reactive antibodies, in either clinical or research settings, is the establishment of the molecular identity of the target autoantigen(s). Here we report on the identification of a novel autoantigen that co-localizes with a subset of cytoplasmic microbodies marked by ABCD3 (PMP-70) and/or PXF (PEX19). Immunoscreening a HeLa cell cDNA expression library with a human autoimmune serum identified two clones that encode fragments of limkain b1 (LKAP). We demonstrate that mouse polyclonal antibodies raised against a bacterially expressed fragment of limkain b1 mark the same cytoplasmic structures as human serum, as does an EGFP:LKAPCT429 fusion protein expressed in HeLa cells. An immunoblot screen against a bacterially expressed MBP:LKAPCT429 fusion protein substrate, using a cohort of 16 additional human sera that display Hep 2 cell cytoplasmic staining patterns similar to the prototype serum, identified three additional sera reactive to limkain b1. This is the first report establishing the molecular identity of a peroxisomal autoantigen. Preliminary results suggest that limkain b1 may be a relatively common target of human autoantibodies reactive to cytoplasmic vesicle-like structures.
Collapse
Affiliation(s)
- K Dunster
- Alfred Pathology Service, Alfred Hospital, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
8
|
Kakinuma T, Ichikawa H, Tsukada Y, Nakamura T, Toh BH. Interaction between p230 and MACF1 is associated with transport of a glycosyl phosphatidyl inositol-anchored protein from the Golgi to the cell periphery. Exp Cell Res 2004; 298:388-98. [PMID: 15265687 DOI: 10.1016/j.yexcr.2004.04.047] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2004] [Revised: 04/17/2004] [Indexed: 11/22/2022]
Abstract
The molecular basis by which proteins are transported along cytoskeletal tracts from the trans-Golgi network (TGN) to the cell periphery remains poorly understood. Previously, using human autoimmune sera, we identified and characterized a TGN protein, p230/Golgin-245, an extensively coiled-coil protein with flexible amino- and carboxyl-terminal ends, that is anchored to TGN membranes and TGN-derived vesicles by its carboxyl-terminal GRIP domain. To identify molecules that interact with the flexible amino-terminal end of p230, we used this domain as bait to screen a human brain cDNA library in a yeast two-hybrid assay. We found that this domain interacts with the carboxyl-terminal domain of MACF1, a protein that cross-links microtubules to the actin cytoskeleton. The interaction was confirmed by co-immunoprecipitation, an in vitro binding assay, double immunofluorescence images demonstrating overlapped localization in HeLa cells, and co-localization of FLAG-tagged constructs containing the interacting domains of these two proteins with their endogenous partners. Expression in HeLa cells of FLAG-tagged constructs containing the interacting domains of p230 and MACF1 disrupted transport of the glycosyl phosphatidyl inositol-anchored marker protein conjugated with yellow fluorescent protein (YFP-SP-GPI), while trafficking of the transmembrane marker protein, vesicular stomatitis virus glycoprotein conjugated with YFP (VSVG3-GL-YFP), was unaffected. Our results suggest that p230, through its interaction with MACF1, provides the molecular link for transport of GPI-anchored proteins along the microtubule and actin cytoskeleton from the TGN to the cell periphery.
Collapse
Affiliation(s)
- Takumi Kakinuma
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan.
| | | | | | | | | |
Collapse
|
9
|
Abstract
The trans-Golgi network is the major sorting compartment of the secretory pathway for protein, lipid and membrane traffic. There is a constant flow of membrane and cargo to and from this compartment. Evidence is emerging that the trans-Golgi network has multiple biochemically and functionally distinct subdomains, each of which contributes to the combined sorting and transport requirements of this dynamic compartment. The recruitment of distinct arrays of protein complexes to trans-Golgi network membranes is likely to produce the diversity of structure and biochemistry observed amongst subdomains that serve to generate different carriers or maintain resident trans-Golgi network components. This review discusses how these subdomains may be formed and examines the molecular players involved, including G proteins, clathrin adaptors and golgin tethers. Diversity within these protein families is highlighted and shown to be critical for the functionality of the trans-Golgi network, as a mediator of protein sorting and membrane transport, and for the maintenance of Golgi structure.
Collapse
Affiliation(s)
- Paul A Gleeson
- The Russell Grimwade School of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia.
| | | | | | | |
Collapse
|
10
|
Hong HS, Chung WH, Hung SI, Chen MJ, Lee SH, Yang LC. Clinical association of anti-golgi autoantibodies and their autoantigens. Scand J Immunol 2004; 59:79-87. [PMID: 14723625 DOI: 10.1111/j.0300-9475.2004.01353.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Anti-Golgi autoantibodies (AGAs) and their targets have been reported from several diseases. However, the association of AGAs, selective autoantigens and related clinical diseases is still obscure. In this study, the presence of AGAs in the sera of 5983 patients was screened to explore the association of AGAs and clinical diseases. By means of indirect immunofluorescence using HEp-2 cells, sera of 12 patients bearing AGAs were identified. The location of recognized Golgi autoantigen(s) was confirmed by the treatment of monensin and double immunostaining using beta-COP. Using the immunoelectron microscopy, AGA immunoreactivity was clearly demonstrated at a stack structure, characteristic of the Golgi complex. Furthermore, analysis of the 12 AGA-positive sera by Western blot revealed at least 15 components of Golgi antigens with relative molecular weights ranging from 54 to 350 kDa, and several Golgi autoantigens identified may be novel. Notably, over half of the AGA-positive cases found belong to non-autoimmune diseases, particularly hepatic disorder. This study presents the association of AGAs, components of the Golgi complex and clinical diseases.
Collapse
Affiliation(s)
- H-S Hong
- Department of Dermatology, Chang Gung Memorial Hospital, Taipei, Taiwan.
| | | | | | | | | | | |
Collapse
|
11
|
Sawalha AH, Potts R, Schmid WR, Scofield RH, Harley JB. The genetics of primary Sjögren's syndrome. Curr Rheumatol Rep 2004; 5:324-32. [PMID: 14531961 DOI: 10.1007/s11926-003-0012-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Primary Sjögren's syndrome is an autoimmune disease characterized clinically by dryness of the eyes and mouth. The use of different classification criteria for primary Sjögren's syndrome has led to dramatically different estimates of prevalence and incidence. Despite this, several genetic and environmental factors are thought to play a role in the susceptibility to primary Sjögren's syndrome, as is the current conceptual formulation of the pathogenesis of many other autoimmune maladies. Primary Sjögren's syndrome appears a complicated polygenic disorder with many genes interacting with environmental factors. Similar to many other polygenic autoimmune rheumatic diseases, human leukocyte antigen associations have been reported and confirmed. Additionally, other non-human leukocyte antigen candidate genes have been reported to reveal association with primary Sjögren's syndrome, but, in general, these effects are not confirmed. The authors review the human leukocyte antigen and non-human leukocyte antigen genetic associations herewith, knowing that new technologies are providing access to the entire genome for association studies. No doubt a much more comprehensive description of the genetics of this disorder will soon emerge.
Collapse
Affiliation(s)
- Amr H Sawalha
- Arthritis and Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | | | | | | | | |
Collapse
|
12
|
Dunster K, Toh BH, Sentry JW. Early endosomes, late endosomes, and lysosomes display distinct partitioning strategies of inheritance with similarities to Golgi-derived membranes. Eur J Cell Biol 2002; 81:117-24. [PMID: 11998863 DOI: 10.1078/0171-9335-00232] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The pattern of inheritance of compartments of the endocytic pathway has been rarely reported, and the precise mechanism(s) are yet to be elucidated. We used antibodies reactive to early endosomes (anti-EEA1), late endosomes (anti-LBPA and anti-LAMP-1), lysosomes (anti-LAMP-1) and trans-Golgi network (TGN) (anti-GOLGA4) to examine the inheritance of these compartments in fixed human HEp-2 cells. Prior to entering M phase, these compartments display a perinuclear bias in their cytoplasmic distribution with areas of local accumulation juxtaposed to the centrosome. The location of these compartments during mitosis was examined relative to each other, the chromosomes, centrosomes and the microtubule network. During M phase early endosomes and TGN-derived compartments share overlapping subcellular distributions. A portion of these compartments display discernible clustering around the separated and migrating centrosomes in prophase. At metaphase these compartments co-localise with the mitotic spindle, are absent at the metaphase plate and do not overlay the astral microtubules. At anaphase these compartments are concentrated between shortening kinetochore microtubules and centrosomes. In addition, they appear distributed over the elongating polar microtubules in the body of the cell. From telophase and into cytokinesis these compartments concentrate around the minus ends of the constricted remnants of polar spindle microtubules and re-establish a prominent presence juxtaposed to the centrosome. In contrast, there is little evidence of movement of late endosomes and lysosomes with migrating centrosomes in prophase, and these compartments are excluded from the mitotic spindle at metaphase. However, by the end of telophase, the subcellular distribution of a portion of late endosomes and lysosomes share overlapping distributions with that of early endosomes. We conclude a portion of endosomal compartments and Golgi-derived membranes undergo ordered partitioning based on the centrosome and mitotic spindle.
Collapse
Affiliation(s)
- Kate Dunster
- Department of Pathology and Immunology, Monash Medical School, Prahran, Victoria, Australia
| | | | | |
Collapse
|