1
|
Kunii M, Harada A. Molecular mechanisms of polarized transport to the apical plasma membrane. Front Cell Dev Biol 2024; 12:1477173. [PMID: 39445332 PMCID: PMC11497131 DOI: 10.3389/fcell.2024.1477173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Cell polarity is essential for cellular function. Directional transport within a cell is called polarized transport, and it plays an important role in cell polarity. In this review, we will introduce the molecular mechanisms of polarized transport, particularly apical transport, and its physiological importance.
Collapse
Affiliation(s)
| | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, The University of Osaka, Osaka, Japan
| |
Collapse
|
2
|
Dooley SA, Kolobova E, Burman A, Kaji I, Digrazia JR, Stubler R, Goldstein A, Packirisamy C, Coutts AW, Saqui-Salces M, Gao N, Engevik MA, Shub MD, Goldenring JR, Engevik AC. Myosin Vb Traffics P-Glycoprotein to the Apical Membrane of Intestinal Epithelial Cells. Gastroenterology 2024:S0016-5085(24)05459-3. [PMID: 39299401 DOI: 10.1053/j.gastro.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND & AIMS The xenobiotic efflux pump P-glycoprotein is highly expressed on the apical membrane of the gastrointestinal tract, where it regulates the levels of intracellular substrates. P-glycoprotein is altered in disease, but the mechanisms that regulate the levels of P-glycoprotein are still being explored. The molecular motor myosin Vb (Myo5b) traffics diverse cargo to the apical membrane of intestinal epithelial cells. We hypothesized that Myo5b was responsible for the delivery of P-glycoprotein to the apical membrane of enterocytes. METHODS We used multiple murine models that lack functional Myo5b or the myosin binding partner Rab11a to analyze P-glycoprotein localization. Pig and human tissue were analyzed to determine P-glycoprotein localization in the setting of MYO5B mutations. Intestinal organoids were used to examine P-glycoprotein trafficking and to assay P-glycoprotein function when MYO5 is inhibited. RESULTS In mice lacking Myo5b or the binding partner Rab11a, P-glycoprotein was improperly trafficked and had decreased presence in the brush border of enterocytes. Immunostaining of a pig model lacking functional Myo5b and human biopsies from a patient with an inactivating mutation in Myo5b also showed altered localization of intestinal P-glycoprotein. Human intestinal organoids expressing the motorless MYO5B tail domain had colocalization with P-glycoprotein, confirming that P-glycoprotein was trafficked by MYO5B in human enterocytes. Inhibition of MYO5 in human intestinal cell lines and organoids resulted in decreased P-glycoprotein capacity. Additionally, inhibition of MYO5 in human colon cancer cells diminished P-glycoprotein activity and increased cell death in response to a chemotherapeutic drug. CONCLUSIONS Collectively, these data demonstrate that Myo5b is necessary for the apical delivery of P-glycoprotein.
Collapse
Affiliation(s)
- Sarah A Dooley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Elena Kolobova
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Andreanna Burman
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jessica R Digrazia
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Rachel Stubler
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Anna Goldstein
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Charulekha Packirisamy
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | | | - Milena Saqui-Salces
- Department of Animal Science, University of Minnesota, Saint Paul, Minnesota
| | - Nan Gao
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Melinda A Engevik
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina; Department of Microbiology and Immunology, Medical University of South Carolina, South Carolina
| | - Mitchell D Shub
- Division of Gastroenterology, Phoenix Children's Hospital, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| | - Amy C Engevik
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
3
|
Szabó L, Pollio AR, Vogel GF. Intracellular Trafficking Defects in Congenital Intestinal and Hepatic Diseases. Traffic 2024; 25:e12954. [PMID: 39187475 DOI: 10.1111/tra.12954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/11/2024] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
Enterocytes and liver cells fulfill important metabolic and barrier functions and are responsible for crucial vectorial secretive and absorptive processes. To date, genetic diseases affecting metabolic enzymes or transmembrane transporters in the intestine and the liver are better comprehended than mutations affecting intracellular trafficking. In this review, we explore the emerging knowledge on intracellular trafficking defects and their clinical manifestations in both the intestine and the liver. We provide a detailed overview including more investigated diseases such as the canonical, variant and associated forms of microvillus inclusion disease, as well as recently described pathologies, highlighting the complexity and disease relevance of several trafficking pathways. We give examples of how intracellular trafficking hubs, such as the apical recycling endosome system, the trans-Golgi network, lysosomes, or the Golgi-to-endoplasmic reticulum transport are involved in the pathomechanism and lead to disease. Ultimately, understanding these processes could spark novel therapeutic approaches, which would greatly improve the quality of life of the affected patients.
Collapse
Affiliation(s)
- Luca Szabó
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Adam R Pollio
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Friedrich Vogel
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
Kaji I, Thiagarajah JR, Goldenring JR. Modeling the cell biology of monogenetic intestinal epithelial disorders. J Cell Biol 2024; 223:e202310118. [PMID: 38683247 PMCID: PMC11058565 DOI: 10.1083/jcb.202310118] [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: 10/24/2023] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024] Open
Abstract
Monogenetic variants are responsible for a range of congenital human diseases. Variants in genes that are important for intestinal epithelial function cause a group of disorders characterized by severe diarrhea and loss of nutrient absorption called congenital diarrheas and enteropathies (CODEs). CODE-causing genes include nutrient transporters, enzymes, structural proteins, and vesicular trafficking proteins in intestinal epithelial cells. Several severe CODE disorders result from the loss-of-function in key regulators of polarized endocytic trafficking such as the motor protein, Myosin VB (MYO5B), as well as STX3, STXBP2, and UNC45A. Investigations of the cell biology and pathophysiology following loss-of-function in these genes have led to an increased understanding of both homeostatic and pathological vesicular trafficking in intestinal epithelial cells. Modeling different CODEs through investigation of changes in patient tissues, coupled with the development of animal models and patient-derived enteroids, has provided critical insights into the enterocyte differentiation and function. Linking basic knowledge of cell biology with the phenotype of specific patient variants is a key step in developing effective treatments for rare monogenetic diseases. This knowledge can also be applied more broadly to our understanding of common epithelial disorders.
Collapse
Affiliation(s)
- Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jay R. Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Congenital Enteropathy Program, Boston Children’s Hospital, Boston, MA, USA
- Harvard Digestive Disease Center, Boston, MA, USA
| | - James R. Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
- Nashville VA Medical Center, Nashville, TN, USA
| |
Collapse
|
5
|
Xu S, Cao B, Xuan G, Xu S, An Z, Zhu C, Li L, Tang C. Function and regulation of Rab GTPases in cancers. Cell Biol Toxicol 2024; 40:28. [PMID: 38695990 PMCID: PMC11065922 DOI: 10.1007/s10565-024-09866-5] [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: 01/21/2024] [Accepted: 04/23/2024] [Indexed: 05/05/2024]
Abstract
The Rab small GTPases are characterized by the distinct intracellular localization and modulate various endocytic, transcytic and exocytic transport pathways. Rab proteins function as scaffolds that connect signaling pathways and intracellular membrane trafficking processes through the recruitment of effectors, such as tethering factors, phosphatases, motors and kinases. In different cancers, Rabs play as either an onco-protein or a tumor suppressor role, highly dependending on the context. The molecular mechanistic research has revealed that Rab proteins are involved in cancer progression through influences on migration, invasion, metabolism, exosome secretion, autophagy, and drug resistance of cancer cells. Therefore, targeting Rab GTPases to recover the dysregulated vesicle transport systems may provide potential strategy to restrain cancer progression. In this review, we discuss the regulation of Rab protein level and activity in modulating pathways involved in tumor progression, and propose that Rab proteins may serve as a prognostic factor in different cancers.
Collapse
Affiliation(s)
- Shouying Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Bin Cao
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Ge Xuan
- Department of Gynaecology, Ningbo Women and Children's Hospital, No.339 Liuting Road, Ningbo, 315012, China
| | - Shu Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Zihao An
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Chongying Zhu
- The Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Lin Li
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China.
| | - Chao Tang
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China.
| |
Collapse
|
6
|
Lapierre LA, Roland JT, Manning EH, Caldwell C, Glenn HL, Vidalain PO, Tangy F, Hogue BG, de Haan CAM, Goldenring JR. Coronavirus M Protein Trafficking in Epithelial Cells Utilizes a Myosin Vb Splice Variant and Rab10. Cells 2024; 13:126. [PMID: 38247817 PMCID: PMC10814003 DOI: 10.3390/cells13020126] [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: 12/02/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
The membrane (M) glycoprotein of coronaviruses (CoVs) serves as the nidus for virion assembly. Using a yeast two-hybrid screen, we identified the interaction of the cytosolic tail of Murine Hepatitis Virus (MHV-CoV) M protein with Myosin Vb (MYO5B), specifically with the alternative splice variant of cellular MYO5B including exon D (MYO5B+D), which mediates interaction with Rab10. When co-expressed in human lung epithelial A549 and canine kidney epithelial MDCK cells, MYO5B+D co-localized with the MHV-CoV M protein, as well as with the M proteins from Porcine Epidemic Diarrhea Virus (PEDV-CoV), Middle East Respiratory Syndrome (MERS-CoV) and Severe Acute Respiratory Syndrome 2 (SARS-CoV-2). Co-expressed M proteins and MYO5B+D co-localized with endogenous Rab10 and Rab11a. We identified point mutations in MHV-CoV M that blocked the interaction with MYO5B+D in yeast 2-hybrid assays. One of these point mutations (E121K) was previously shown to block MHV-CoV virion assembly and its interaction with MYO5B+D. The E to K mutation at homologous positions in PEDV-CoV, MERS-CoV and SARS-CoV-2 M proteins also blocked colocalization with MYO5B+D. The knockdown of Rab10 blocked the co-localization of M proteins with MYO5B+D and was rescued by re-expression of CFP-Rab10. Our results suggest that CoV M proteins traffic through Rab10-containing systems, in association with MYO5B+D.
Collapse
Affiliation(s)
- Lynne A. Lapierre
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (L.A.L.); (J.T.R.); (E.H.M.); (C.C.)
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Nashville VA Medical Center, Nashville, TN 37212, USA
| | - Joseph T. Roland
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (L.A.L.); (J.T.R.); (E.H.M.); (C.C.)
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Elizabeth H. Manning
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (L.A.L.); (J.T.R.); (E.H.M.); (C.C.)
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Nashville VA Medical Center, Nashville, TN 37212, USA
| | - Catherine Caldwell
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (L.A.L.); (J.T.R.); (E.H.M.); (C.C.)
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Nashville VA Medical Center, Nashville, TN 37212, USA
| | - Honor L. Glenn
- Biodesign Institute Center for Immunotherapy, Vaccines & Virotherapy, Tempe, AZ 85287, USA; (H.L.G.); (B.G.H.)
| | - Pierre-Olivier Vidalain
- Equipe Infections Virales, Métabolisme et Immunité, Centre International de Recherche en Infectiologie (CIRI), Univ. Lyon, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France;
- Unité Génomique Virale et Vaccination, Institut Pasteur, CNRS UMR3569, 75015 Paris, France
| | - Frederic Tangy
- Viral Genomics and Vaccination Unit, Department of Virology, Institut Pasteur, CNRS UMR3569, 75015 Paris, France;
| | - Brenda G. Hogue
- Biodesign Institute Center for Immunotherapy, Vaccines & Virotherapy, Tempe, AZ 85287, USA; (H.L.G.); (B.G.H.)
- Center for Applied Structural Discovery, Biodesign Institute, Tempe, AZ 85287, USA
- School of Life Sciences, Arizona State University, Phoenix, AZ 85004, USA
| | - C. A. M. de Haan
- Faculty of Veterinary Medicine, Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Section Virology, University of Utrecht, 3584 CS Utrecht, The Netherlands;
| | - James R. Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (L.A.L.); (J.T.R.); (E.H.M.); (C.C.)
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Nashville VA Medical Center, Nashville, TN 37212, USA
- Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
7
|
Frank D, Moussi CJ, Ulferts S, Lorenzen L, Schwan C, Grosse R. Vesicle-Associated Actin Assembly by Formins Promotes TGFβ-Induced ANGPTL4 Trafficking, Secretion and Cell Invasion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204896. [PMID: 36691769 PMCID: PMC10037683 DOI: 10.1002/advs.202204896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/01/2022] [Indexed: 06/17/2023]
Abstract
Vesicle trafficking has emerged as an important process driving tumor progression through various mechanisms. Transforming growth factor beta (TGFβ)-mediated secretion of Angiopoietin-like 4 (ANGPTL4) is important for cancer development. Here, Formin-like 2 (FMNL2) is identified to be necessary for ANGPTL4 trafficking and secretion in response to TGFβ. Protein kinase C (PKC)-dependent phosphorylation of FMNL2 downstream of TGFβ stimulation is required for cancer cell invasion as well as ANGPTL4 vesicle trafficking and secretion. Moreover, using super resolution microscopy, ANGPTL4 trafficking is actin-dependent with FMNL2 directly polymerizing actin at ANGPTL4-containing vesicles, which are associated with Rab8a and myosin Vb. This work uncovers a formin-controlled mechanism that transiently polymerizes actin directly at intracellular vesicles to facilitate their mobility. This mechanism may be important for the regulation of cancer cell metastasis and tumor progression.
Collapse
Affiliation(s)
- Dennis Frank
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
| | - Christel Jessica Moussi
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
- Deutsche Forschungsgemeinschaft Research Training GroupMembrane Plasticity in Tissue Development and RemodelingUniversity of Marburg35037MarburgGermany
| | - Svenja Ulferts
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
| | - Lina Lorenzen
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
| | - Carsten Schwan
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
| | - Robert Grosse
- Institute of Experimental and Clinical Pharmacology and ToxicologyMedical FacultyUniversity of Freiburg79104FreiburgGermany
- Centre for Integrative Biological Signalling Studies – CIBSS79104FreiburgGermany
| |
Collapse
|
8
|
Weng X, Wang H. Apical vesicles: Social networking at the pollen tube tip. REPRODUCTION AND BREEDING 2022. [DOI: 10.1016/j.repbre.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
9
|
Vij M, Shah V. Compound Heterozygous Myosin 5B (Myo5b) Mutation with Early Onset Progressive Cholestasis and No Intestinal Failure. Fetal Pediatr Pathol 2022; 41:811-817. [PMID: 34338607 DOI: 10.1080/15513815.2021.1959690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Background: Exome sequencing studies have recently identified novel genes implicated in normal or low GGT pediatric cholestasis including myosin 5B (MYO5B). Case report: We identified novel compound heterozygote mutations in exon 14 and exon 19 of the MYO5B gene in an 18-month-old Indian child with history of fluctuating jaundice and severe pruritus. His liver biopsy showed portal and perivenular fibrosis with focal bridging septa and mild activity. He is currently on UDCA, cholestyramine and vitamin supplements. There is no history of diarrhea. His asymptomatic mother showed heterozygous mutation in exon 19 of the MYO5B gene and his asymptomatic father showed heterozygous mutation in exon 14 of the MYO5B gene. Conclusion: Our report confirms that patients with compound heterozygote mutations in MYO5B develop progressive cholestasis with no intestinal disease.
Collapse
Affiliation(s)
- Mukul Vij
- Department of Pathology, Dr Rela Institute and Medical Centre, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Vaibhav Shah
- Gujarat Superspeciality Clinic Pharmacy, Ahmedabad, Gujarat, India
| |
Collapse
|
10
|
Rathan-Kumar S, Roland JT, Momoh M, Goldstein A, Lapierre LA, Manning E, Mitchell L, Norman J, Kaji I, Goldenring JR. Rab11FIP1-deficient mice develop spontaneous inflammation and show increased susceptibility to colon damage. Am J Physiol Gastrointest Liver Physiol 2022; 323:G239-G254. [PMID: 35819177 PMCID: PMC9423785 DOI: 10.1152/ajpgi.00042.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/23/2022] [Accepted: 06/29/2022] [Indexed: 01/31/2023]
Abstract
The small GTPase, Rab11a, regulates vesicle trafficking and cell polarity in epithelial cells through interaction with Rab11 family-interacting proteins (Rab11-FIPs). We hypothesized that deficiency of Rab11-FIP1 would affect mucosal integrity in the intestine. Global Rab11FIP1 knockout (KO) mice were generated by deletion of the second exon. Pathology of intestinal tissues was analyzed by immunostaining of colonic sections and RNA-sequencing of isolated colonic epithelial cells. A low concentration of dextran sodium sulfate (DSS, 2%) was added to drinking water for 5 days, and injury score was compared between Rab11FIP1 KO, Rab11FIP2 KO, and heterozygous littermates. Rab11FIP1 KO mice showed normal fertility and body weight gain. More frequent lymphoid patches and infiltration of macrophages and neutrophils were identified in Rab11FIP1 KO mice before the development of rectal prolapse compared with control mice. The population of trefoil factor 3 (TFF3)-positive goblet cells was significantly lower, and the ratio of proliferative to nonproliferative cells was higher in Rab11FIP1 KO colons. Transcription signatures indicated that Rab11FIP1 deletion downregulated genes that mediate stress tolerance response, whereas genes mediating the response to infection were significantly upregulated, consistent with the inflammatory responses in the steady state. Lack of Rab11FIP1 also resulted in abnormal accumulation of subapical vesicles in colonocytes and the internalization of transmembrane mucin, MUC13, with Rab14. After DSS treatment, Rab11FIP1 KO mice showed greater body weight loss and more severe mucosal damage than those in heterozygous littermates. These findings suggest that Rab11FIP1 is important for cytoprotection mechanisms and for the maintenance of colonic mucosal integrity.NEW & NOTEWORTHY Although Rab11FIP1 is important in membrane trafficking in epithelial cells, the gastrointestinal phenotype of Rab11FIP1 knockout (KO) mice had never been reported. This study demonstrated that Rab11FIP1 loss induces mistrafficking of Rab14 and MUC13 and decreases in colonic goblet cells, resulting in impaired mucosal integrity.
Collapse
Affiliation(s)
- Sudiksha Rathan-Kumar
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joseph T Roland
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael Momoh
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anna Goldstein
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lynne A Lapierre
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Elizabeth Manning
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Louise Mitchell
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Jim Norman
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| |
Collapse
|
11
|
Larocque G, Royle SJ. Integrating intracellular nanovesicles into integrin trafficking pathways and beyond. Cell Mol Life Sci 2022; 79:335. [PMID: 35657500 PMCID: PMC9166830 DOI: 10.1007/s00018-022-04371-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/28/2022] [Accepted: 05/11/2022] [Indexed: 12/24/2022]
Abstract
Membrane traffic controls the movement of proteins and lipids from one cellular compartment to another using a system of transport vesicles. Intracellular nanovesicles (INVs) are a newly described class of transport vesicles. These vesicles are small, carry diverse cargo, and are involved in multiple trafficking steps including anterograde traffic and endosomal recycling. An example of a biological process that they control is cell migration and invasion, due to their role in integrin recycling. In this review, we describe what is known so far about these vesicles. We discuss how INVs may integrate into established membrane trafficking pathways using integrin recycling as an example. We speculate where in the cell INVs have the potential to operate and we identify key questions for future investigation.
Collapse
Affiliation(s)
| | - Stephen J Royle
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Gibbet Hill Road, Coventry, CV4 7AL, UK.
| |
Collapse
|
12
|
Bowman DM, Kaji I, Goldenring JR. Altered MYO5B Function Underlies Microvillus Inclusion Disease: Opportunities for Intervention at a Cellular Level. Cell Mol Gastroenterol Hepatol 2022; 14:553-565. [PMID: 35660026 PMCID: PMC9304615 DOI: 10.1016/j.jcmgh.2022.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/31/2022] [Accepted: 04/29/2022] [Indexed: 12/10/2022]
Abstract
Microvillus inclusion disease (MVID) is a congenital diarrheal disorder resulting in life-threatening secretory diarrhea in newborns. Inactivating and nonsense mutations in myosin Vb (MYO5B) have been identified in MVID patients. Work using patient tissues, cell lines, mice, and pigs has led to critical insights into the pathology of MVID and a better understanding of both apical trafficking in intestinal enterocytes and intestinal stem cell differentiation. These studies have demonstrated that loss of MYO5B or inactivating mutations lead to loss of apical sodium and water transporters, without loss of apical CFTR, accounting for the major pathology of the disease. In addition, loss of MYO5B expression induces the formation of microvillus inclusions through apical bulk endocytosis that utilizes dynamin and PACSIN2 and recruits tight junction proteins to the sites of bulk endosome formation. Importantly, formation of microvillus inclusions is not required for the induction of diarrhea. Recent investigations have demonstrated that administration of lysophosphatidic acid (LPA) can partially reestablish apical ion transporters in enterocytes of MYO5B KO mice. In addition, further studies have shown that MYO5B loss induces an imbalance in Wnt/Notch signaling pathways that can lead to alterations in enterocyte maturation and tuft cell lineage differentiation. Inhibition of Notch signaling leads to improvements in those cell differentiation deficits. These studies demonstrate that directed strategies through LPA receptor activation and Notch inhibition can bypass the inhibitory effects of MYO5B loss. Thus, effective strategies may be successful in MVID patients and other congenital diarrhea syndromes to reestablish proper apical membrane absorption of sodium and water in enterocytes and ameliorate life-threatening congenital diarrhea.
Collapse
Affiliation(s)
- Deanna M Bowman
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Nashville VA Medical Center, Nashville, Tennessee.
| |
Collapse
|
13
|
Wang X, Li X, Wang J, Wang J, Hu C, Zeng J, Shi A, Lin L. SMGL-1/NBAS acts as a RAB-8 GEF to regulate unconventional protein secretion. J Cell Biol 2022; 221:213235. [PMID: 35604368 PMCID: PMC9129922 DOI: 10.1083/jcb.202111125] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 04/13/2022] [Accepted: 05/04/2022] [Indexed: 01/07/2023] Open
Abstract
Unconventional protein secretion (UPS) pathways are conserved across species. However, the underlying mechanisms that regulate Golgi-bypassing UPS of integral proteins remain elusive. In this study, we show that RAB-8 and SMGL-1/NBAS are required for the UPS of integral proteins in C. elegans intestine. SMGL-1 resides in the ER-Golgi intermediate compartment and adjacent RAB-8-positive structures, and NRZ complex component CZW-1/ZW10 is required for this residency. Notably, SMGL-1 acts as a guanine nucleotide exchange factor for RAB-8, ensuring UPS of integral proteins by driving the activation of RAB-8. Furthermore, we show that Pseudomonas aeruginosa infection elevated the expression of SMGL-1 and RAB-8. Loss of SMGL-1 or RAB-8 compromised resistance to environmental colchicine, arsenite, and pathogenic bacteria. These results suggest that the SMGL-1/RAB-8-mediated UPS could integrate environmental signals to serve as a host defense response. Together, by establishing the C. elegans intestine as a multicellular model, our findings provide insights into RAB-8-dependent Golgi-bypassing UPS, especially in the context of epithelia in vivo.
Collapse
Affiliation(s)
- Xianghong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xinxin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junkai Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiabin Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Can Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jia Zeng
- Department of Biochemistry and Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Anbing Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China,Correspondence to Anbing Shi:
| | - Long Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China,Long Lin:
| |
Collapse
|
14
|
Sakai T, Choo YY, Sato O, Ikebe R, Jeffers A, Idell S, Tucker T, Ikebe M. Myo5b Transports Fibronectin-Containing Vesicles and Facilitates FN1 Secretion from Human Pleural Mesothelial Cells. Int J Mol Sci 2022; 23:ijms23094823. [PMID: 35563212 PMCID: PMC9101030 DOI: 10.3390/ijms23094823] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 12/13/2022] Open
Abstract
Pleural mesothelial cells (PMCs) play a central role in the progression of pleural fibrosis. As pleural injury progresses to fibrosis, PMCs transition to mesenchymal myofibroblast via mesothelial mesenchymal transition (MesoMT), and produce extracellular matrix (ECM) proteins including collagen and fibronectin (FN1). FN1 plays an important role in ECM maturation and facilitates ECM-myofibroblast interaction, thus facilitating fibrosis. However, the mechanism of FN1 secretion is poorly understood. We report here that myosin 5b (Myo5b) plays a critical role in the transportation and secretion of FN1 from human pleural mesothelial cells (HPMCs). TGF-β significantly increased the expression and secretion of FN1 from HPMCs and facilitates the close association of Myo5B with FN1 and Rab11b. Moreover, Myo5b directly binds to GTP bound Rab11b (Rab11b-GTP) but not GDP bound Rab11b. Myo5b or Rab11b knockdown via siRNA significantly attenuated the secretion of FN1 without changing FN1 expression. TGF-β also induced Rab11b-GTP formation, and Rab11b-GTP but not Rab11b-GDP significantly activated the actin-activated ATPase activity of Myo5B. Live cell imaging revealed that Myo5b- and FN1-containing vesicles continuously moved together in a single direction. These results support that Myo5b and Rab11b play an important role in FN1 transportation and secretion from HPMCs, and consequently may contribute to the development of pleural fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mitsuo Ikebe
- Correspondence: ; Tel.: +1-(903)-877-7785; Fax: +1-(903)-877-5438
| |
Collapse
|
15
|
Borghesan E, Smith EP, Myeni S, Binder K, Knodler LA, Celli J. A Brucella effector modulates the Arf6-Rab8a GTPase cascade to promote intravacuolar replication. EMBO J 2021; 40:e107664. [PMID: 34423453 PMCID: PMC8488576 DOI: 10.15252/embj.2021107664] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 01/15/2023] Open
Abstract
Remodeling of host cellular membrane transport pathways is a common pathogenic trait of many intracellular microbes that is essential to their intravacuolar life cycle and proliferation. The bacterium Brucella abortus generates a host endoplasmic reticulum‐derived vacuole (rBCV) that supports its intracellular growth, via VirB Type IV secretion system‐mediated delivery of effector proteins, whose functions and mode of action are mostly unknown. Here, we show that the effector BspF specifically promotes Brucella replication within rBCVs by interfering with vesicular transport between the trans‐Golgi network (TGN) and recycling endocytic compartment. BspF targeted the recycling endosome, inhibited retrograde traffic to the TGN, and interacted with the Arf6 GTPase‐activating Protein (GAP) ACAP1 to dysregulate Arf6‐/Rab8a‐dependent transport within the recycling endosome, which resulted in accretion of TGN‐associated vesicles by rBCVs and enhanced bacterial growth. Altogether, these findings provide mechanistic insight into bacterial modulation of membrane transport used to promote their own proliferation within intracellular vacuoles.
Collapse
Affiliation(s)
- Elizabeth Borghesan
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Erin P Smith
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Sebenzile Myeni
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kelsey Binder
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Leigh A Knodler
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Jean Celli
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA.,Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
16
|
Advanced Microscopy for Liver and Gut Ultrastructural Pathology in Patients with MVID and PFIC Caused by MYO5B Mutations. J Clin Med 2021; 10:jcm10091901. [PMID: 33924896 PMCID: PMC8125609 DOI: 10.3390/jcm10091901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Mutations in the actin motor protein myosinVb (myo5b) cause aberrant apical cargo transport and the congenital enteropathy microvillus inclusion disease (MVID). Recently, missense mutations in myo5b were also associated with progressive familial intrahepatic cholestasis (MYO5B-PFIC). Here, we thoroughly characterized the ultrastructural and immuno-cytochemical phenotype of hepatocytes and duodenal enterocytes from a unique case of an adult MYO5B-PFIC patient who showed constant hepatopathy but only periodic enteric symptoms. Selected data from two other patients supported the findings. Advanced methods such as cryo-fixation, freeze-substitution, immuno-gold labeling, electron tomography and immuno-fluorescence microscopy complemented the standard procedures. Liver biopsies showed mislocalization of Rab11 and bile canalicular membrane proteins. Rab11-positive vesicles clustered around bile canaliculi and resembled subapical clusters of aberrant recycling endosomes in enterocytes from MVID patients. The adult patient studied in detail showed a severe, MVID-specific enterocyte phenotype, despite only a mild clinical intestinal presentation. This included mislocalization of numerous proteins essential for apical cargo transport and morphological alterations. We characterized the heterogeneous population of large catabolic organelles regarding their complex ultrastructure and differential distribution of autophagic and lysosomal marker proteins. Finally, we generated duodenal organoids/enteroids from biopsies that recapitulated all MVID hallmarks, demonstrating the potential of this disease model for personalized medicine.
Collapse
|
17
|
Platenkamp A, Detmar E, Sepulveda L, Ritz A, Rogers SL, Applewhite DA. The Drosophila melanogaster Rab GAP RN-tre cross-talks with the Rho1 signaling pathway to regulate nonmuscle myosin II localization and function. Mol Biol Cell 2020; 31:2379-2397. [PMID: 32816624 PMCID: PMC7851959 DOI: 10.1091/mbc.e20-03-0181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
To identify novel regulators of nonmuscle myosin II (NMII) we performed an image-based RNA interference screen using stable Drosophila melanogaster S2 cells expressing the enhanced green fluorescent protein (EGFP)-tagged regulatory light chain (RLC) of NMII and mCherry-Actin. We identified the Rab-specific GTPase-activating protein (GAP) RN-tre as necessary for the assembly of NMII RLC into contractile actin networks. Depletion of RN-tre led to a punctate NMII phenotype, similar to what is observed following depletion of proteins in the Rho1 pathway. Depletion of RN-tre also led to a decrease in active Rho1 and a decrease in phosphomyosin-positive cells by immunostaining, while expression of constitutively active Rho or Rho-kinase (Rok) rescues the punctate phenotype. Functionally, RN-tre depletion led to an increase in actin retrograde flow rate and cellular contractility in S2 and S2R+ cells, respectively. Regulation of NMII by RN-tre is only partially dependent on its GAP activity as overexpression of constitutively active Rabs inactivated by RN-tre failed to alter NMII RLC localization, while a GAP-dead version of RN-tre partially restored phosphomyosin staining. Collectively, our results suggest that RN-tre plays an important regulatory role in NMII RLC distribution, phosphorylation, and function, likely through Rho1 signaling and putatively serving as a link between the secretion machinery and actomyosin contractility.
Collapse
Affiliation(s)
| | - Elizabeth Detmar
- Department of Biology & Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280
| | - Liz Sepulveda
- Department of Biology, Reed College, Portland, OR 97202
| | - Anna Ritz
- Department of Biology, Reed College, Portland, OR 97202
| | - Stephen L Rogers
- Department of Biology & Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280
| | | |
Collapse
|
18
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
19
|
Kehl A, Göser V, Reuter T, Liss V, Franke M, John C, Richter CP, Deiwick J, Hensel M. A trafficome-wide RNAi screen reveals deployment of early and late secretory host proteins and the entire late endo-/lysosomal vesicle fusion machinery by intracellular Salmonella. PLoS Pathog 2020; 16:e1008220. [PMID: 32658937 PMCID: PMC7377517 DOI: 10.1371/journal.ppat.1008220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 07/23/2020] [Accepted: 05/19/2020] [Indexed: 12/17/2022] Open
Abstract
The intracellular lifestyle of Salmonella enterica is characterized by the formation of a replication-permissive membrane-bound niche, the Salmonella-containing vacuole (SCV). As a further consequence of the massive remodeling of the host cell endosomal system, intracellular Salmonella establish a unique network of various Salmonella-induced tubules (SIT). The bacterial repertoire of effector proteins required for the establishment for one type of these SIT, the Salmonella-induced filaments (SIF), is rather well-defined. However, the corresponding host cell proteins are still poorly understood. To identify host factors required for the formation of SIF, we performed a sub-genomic RNAi screen. The analyses comprised high-resolution live cell imaging to score effects on SIF induction, dynamics and morphology. The hits of our functional RNAi screen comprise: i) The late endo-/lysosomal SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complex, consisting of STX7, STX8, VTI1B, and VAMP7 or VAMP8, which is, in conjunction with RAB7 and the homotypic fusion and protein sorting (HOPS) tethering complex, a complete vesicle fusion machinery. ii) Novel interactions with the early secretory GTPases RAB1A and RAB1B, providing a potential link to coat protein complex I (COPI) vesicles and reinforcing recently identified ties to the endoplasmic reticulum. iii) New connections to the late secretory pathway and/or the recycling endosome via the GTPases RAB3A, RAB8A, and RAB8B and the SNAREs VAMP2, VAMP3, and VAMP4. iv) An unprecedented involvement of clathrin-coated structures. The resulting set of hits allowed us to characterize completely new host factor interactions, and to strengthen observations from several previous studies. The facultative intracellular pathogen Salmonella enterica serovar Typhimurium induces the reorganization of the endosomal system of mammalian host cells. This activity is dependent on translocated effector proteins of the pathogen. The host cell factors required for endosomal remodeling are only partially known. To identify such factors for the formation and dynamics of endosomal compartments in Salmonella-infected cells, we performed a live cell imaging-based RNAi screen to investigate the role of 496 mammalian proteins involved in cellular logistics. We identified that endosomal remodeling by intracellular Salmonella is dependent on host factors in the following functional classes: i) the late endo-/lysosomal SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complex, ii) the early secretory pathway, represented by regulator GTPases RAB1A and RAB1B, iii) the late secretory pathway and/or recycling endosomes represented by GTPases RAB3A, RAB8A, RAB8B, and the SNAREs VAMP2, VAMP3, and VAMP4, and iv) clathrin-coated structures. The identification of these new host factors provides further evidence for the complex manipulation of host cell transport functions by intracellular Salmonella and should enable detailed follow-up studies on the mechanisms involved.
Collapse
Affiliation(s)
- Alexander Kehl
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
- Division of Biophysics, University of Osnabrück, Osnabrück, Germany
- * E-mail: (AK); (MH)
| | - Vera Göser
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Tatjana Reuter
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Viktoria Liss
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Maximilian Franke
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Christopher John
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | | | - Jörg Deiwick
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Michael Hensel
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
- CellNanOs–Center for Cellular Nanoanalytics, Fachbereich Biologie/Chemie, Universität Osnabrück, Osnabrück, Germany
- * E-mail: (AK); (MH)
| |
Collapse
|
20
|
Wang Y, Guo S, Li D, Tang Y, Li L, Su L, Liu X. YIPF2 promotes chemotherapeutic agent-mediated apoptosis via enhancing TNFRSF10B recycling to plasma membrane in non-small cell lung cancer cells. Cell Death Dis 2020; 11:242. [PMID: 32303681 PMCID: PMC7165181 DOI: 10.1038/s41419-020-2436-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/19/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the most common histological type of lung cancer, and the identification of the apoptotic process of NSCLC is vital for its treatment. Usually, both the expression level and the cell surface level of TNFRSF10B (TNF Receptor superfamily member 10B) will increase after treatment with some chemotherapeutic agents, which plays a critical role in the apoptosis induction. However, the exact molecular mechanism underlying TNFRSF10B regulation remains largely elusive. Here, we found that TNFRSF10B, along with a vesicular trafficking regulator protein, YIPF2, were upregulated after treatment with pemetrexed (PEM) in NSCLC cells. Besides, YIPF2 increased the surface level of TNFRF10B, while YIPF2 knockdown inhibited the upregulation of TNFRSF10B and its recycling to plasma membrane. In addition, RAB8 decreased the cell surface TNFRSF10B by promoting its removing from plasma membrane to cytoplasm. Furthermore, we found that YIPF2, RAB8 and TNFRSF10B proteins interacted physically with each other. YIPF2 could further inhibit the physical interaction between TNFRSF10B and RAB8, thereby suppressing the removing of TNFRSF10B from plasma membrane to cytoplasm mediated by RAB8 and maintaining its high level on cell surface. Finally, using bioinformatics database, the YIPF2-TNFRSF10B axis was confirmed to be associated with the malignant progression of lung cancer. Taken together, we show that YIPF2 promotes chemotherapeutic agent-mediated apoptosis via enhancing TNFRSF10B recycling to plasma membrane in NSCLC cells. These findings may be beneficial for the development of potential prognostic markers of NSCLC and may provide effective treatment strategy.
Collapse
Affiliation(s)
- Yingying Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Sen Guo
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Dongmei Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yongkang Tang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Lei Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Ling Su
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.
| | - Xiangguo Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.
| |
Collapse
|
21
|
Bibeau JP, Furt F, Mousavi SI, Kingsley JL, Levine MF, Tüzel E, Vidali L. In vivo interactions between myosin XI, vesicles and filamentous actin are fast and transient in Physcomitrella patens. J Cell Sci 2020; 133:jcs.234682. [PMID: 31964706 DOI: 10.1242/jcs.234682] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 01/07/2020] [Indexed: 12/25/2022] Open
Abstract
The actin cytoskeleton and active membrane trafficking machinery are essential for polarized cell growth. To understand the interactions between myosin XI, vesicles and actin filaments in vivo, we performed fluorescence recovery after photobleaching and showed that the dynamics of myosin XIa at the tip of the spreading earthmoss Physcomitrella patens caulonemal cells are actin-dependent and that 50% of myosin XI is bound to vesicles. To obtain single-particle information, we used variable-angle epifluorescence microscopy in protoplasts to demonstrate that protein myosin XIa and VAMP72-labeled vesicles localize in time and space over periods lasting only a few seconds. By tracking data with Hidden Markov modeling, we showed that myosin XIa and VAMP72-labeled vesicles exhibit short runs of actin-dependent directed transport. We also found that the interaction of myosin XI with vesicles is short-lived. Together, this vesicle-bound fraction, fast off-rate and short average distance traveled seem be crucial for the dynamic oscillations observed at the tip, and might be vital for regulation and recycling of the exocytosis machinery, while simultaneously promoting vesicle focusing and vesicle secretion at the tip, necessary for cell wall expansion.
Collapse
Affiliation(s)
- Jeffrey P Bibeau
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Fabienne Furt
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - S Iman Mousavi
- Department of Physics, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - James L Kingsley
- Department of Physics, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Max F Levine
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Erkan Tüzel
- Department of Physics, Worcester Polytechnic Institute, Worcester, MA 01609, USA.,Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA 01609, USA.,Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Luis Vidali
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, USA .,Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| |
Collapse
|
22
|
Duan X, Li Y, Yi K, Guo F, Wang H, Wu PH, Yang J, Mair DB, Morales EA, Kalab P, Wirtz D, Sun SX, Li R. Dynamic organelle distribution initiates actin-based spindle migration in mouse oocytes. Nat Commun 2020; 11:277. [PMID: 31937754 PMCID: PMC6959240 DOI: 10.1038/s41467-019-14068-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 12/05/2019] [Indexed: 12/26/2022] Open
Abstract
Migration of meiosis-I (MI) spindle from the cell center to a sub-cortical location is a critical step for mouse oocytes to undergo asymmetric meiotic cell division. In this study, we investigate the mechanism by which formin-2 (FMN2) orchestrates the initial movement of MI spindle. By defining protein domains responsible for targeting FMN2, we show that spindle-periphery localized FMN2 is required for spindle migration. The spindle-peripheral FMN2 nucleates short actin bundles from vesicles derived likely from the endoplasmic reticulum (ER) and concentrated in a layer outside the spindle. This layer is in turn surrounded by mitochondria. A model based on polymerizing actin filaments pushing against mitochondria, thus generating a counter force on the spindle, demonstrated an inherent ability of this system to break symmetry and evolve directional spindle motion. The model is further supported through experiments involving spatially biasing actin nucleation via optogenetics and disruption of mitochondrial distribution and dynamics. Mammalian oocytes divide asymmetrically during meiotic maturation. Here, the authors show that spindle movement away from oocyte center depends on actin filaments nucleated from the spindle periphery pushing against surrounding mitochondria, which polarizes spontaneously to produce directional spindle motion.
Collapse
Affiliation(s)
- Xing Duan
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA.,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Yizeng Li
- Department of Mechanical Engineering, Kennesaw State University, Marietta, GA, 30060, USA.,Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Kexi Yi
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO, 64110, USA
| | - Fengli Guo
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO, 64110, USA
| | - HaiYang Wang
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA.,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jing Yang
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Devin B Mair
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA.,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Edwin Angelo Morales
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Sean X Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Rong Li
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD, 21205, USA. .,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA. .,Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore, 117411, Singapore.
| |
Collapse
|
23
|
Unconventional Myosins: How Regulation Meets Function. Int J Mol Sci 2019; 21:ijms21010067. [PMID: 31861842 PMCID: PMC6981383 DOI: 10.3390/ijms21010067] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 01/24/2023] Open
Abstract
Unconventional myosins are multi-potent molecular motors that are assigned important roles in fundamental cellular processes. Depending on their mechano-enzymatic properties and structural features, myosins fulfil their roles by acting as cargo transporters along the actin cytoskeleton, molecular anchors or tension sensors. In order to perform such a wide range of roles and modes of action, myosins need to be under tight regulation in time and space. This is achieved at multiple levels through diverse regulatory mechanisms: the alternative splicing of various isoforms, the interaction with their binding partners, their phosphorylation, their applied load and the composition of their local environment, such as ions and lipids. This review summarizes our current knowledge of how unconventional myosins are regulated, how these regulatory mechanisms can adapt to the specific features of a myosin and how they can converge with each other in order to ensure the required tight control of their function.
Collapse
|
24
|
Goldenring JR. The challenge of personalized cell biology: The example of microvillus inclusion disease. Traffic 2019; 21:169-171. [PMID: 31596022 DOI: 10.1111/tra.12703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 11/30/2022]
Abstract
Whole exome sequencing now provides a tool for rapid analysis of patients manifesting congenital diseases. Congenital diarrheal diseases provide a critical example of the challenges of combining identification of genetic mutations responsible for disease with characterization of the cell biological and cell physiological deficits observed in patients. Recent studies exploring the cellular events associated with loss of functional Myosin 5B (MYO5B) have demonstrated the importance of cell biological and physiological analyses to provide a greater understanding of the implications of pathological mutations. Development of enteroids derived from biopsies of patients with complex congenital diarrheal diseases provides a critical resource for evaluation of the cell biological impact of specific monogenic mutations on enterocyte function. The ability to identify putative causative mutations for congenital disease now provides an opportunity to coordinate the efforts of physicians and cell biologists in an effort to provide patients with personalized cell biology analysis to improve patient diagnosis and treatment.
Collapse
Affiliation(s)
- James R Goldenring
- Department of Surgery and Cell and Developmental Biology, and the Epithelial Biology Center, Vanderbilt University Medical Center, and the Nashville VA Medical Center, Nashville, Tennessee
| |
Collapse
|
25
|
Blondelle J, Marrocco V, Clark M, Desmond P, Myers S, Nguyen J, Wright M, Bremner S, Pierantozzi E, Ward S, Estève E, Sorrentino V, Ghassemian M, Lange S. Murine obscurin and Obsl1 have functionally redundant roles in sarcolemmal integrity, sarcoplasmic reticulum organization, and muscle metabolism. Commun Biol 2019; 2:178. [PMID: 31098411 PMCID: PMC6509138 DOI: 10.1038/s42003-019-0405-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/28/2019] [Indexed: 12/19/2022] Open
Abstract
Biological roles of obscurin and its close homolog Obsl1 (obscurin-like 1) have been enigmatic. While obscurin is highly expressed in striated muscles, Obsl1 is found ubiquitously. Accordingly, obscurin mutations have been linked to myopathies, whereas mutations in Obsl1 result in 3M-growth syndrome. To further study unique and redundant functions of these closely related proteins, we generated and characterized Obsl1 knockouts. Global Obsl1 knockouts are embryonically lethal. In contrast, skeletal muscle-specific Obsl1 knockouts show a benign phenotype similar to obscurin knockouts. Only deletion of both proteins and removal of their functional redundancy revealed their roles for sarcolemmal stability and sarcoplasmic reticulum organization. To gain unbiased insights into changes to the muscle proteome, we analyzed tibialis anterior and soleus muscles by mass spectrometry, uncovering additional changes to the muscle metabolism. Our analyses suggest that all obscurin protein family members play functions for muscle membrane systems.
Collapse
Affiliation(s)
- Jordan Blondelle
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Valeria Marrocco
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Madison Clark
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Patrick Desmond
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Stephanie Myers
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Jim Nguyen
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Matthew Wright
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Shannon Bremner
- Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Enrico Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, 53100 Italy
| | - Samuel Ward
- Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Eric Estève
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
- Université Grenoble Alpes, HP2, Grenoble, 38706 France
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, 53100 Italy
| | - Majid Ghassemian
- Department of Chemistry and Biochemistry, University of California, San Diego, 92093 CA USA
| | - Stephan Lange
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, 413 45 Sweden
| |
Collapse
|
26
|
Sakai R, Fukuda R, Unida S, Aki M, Ono Y, Endo A, Kusumi S, Koga D, Fukushima T, Komada M, Okiyoneda T. The integral function of the endocytic recycling compartment is regulated by RFFL-mediated ubiquitylation of Rab11 effectors. J Cell Sci 2019; 132:jcs.228007. [PMID: 30659120 DOI: 10.1242/jcs.228007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 01/03/2019] [Indexed: 12/11/2022] Open
Abstract
Endocytic trafficking is regulated by ubiquitylation (also known as ubiquitination) of cargoes and endocytic machineries. The role of ubiquitylation in lysosomal delivery has been well documented, but its role in the recycling pathway is largely unknown. Here, we report that the ubiquitin (Ub) ligase RFFL regulates ubiquitylation of endocytic recycling regulators. An RFFL dominant-negative (DN) mutant induced clustering of endocytic recycling compartments (ERCs) and delayed endocytic cargo recycling without affecting lysosomal traffic. A BioID RFFL interactome analysis revealed that RFFL interacts with the Rab11 effectors EHD1, MICALL1 and class I Rab11-FIPs. The RFFL DN mutant strongly captured these Rab11 effectors and inhibited their ubiquitylation. The prolonged interaction of RFFL with Rab11 effectors was sufficient to induce the clustered ERC phenotype and to delay cargo recycling. RFFL directly ubiquitylates these Rab11 effectors in vitro, but RFFL knockout (KO) only reduced the ubiquitylation of Rab11-FIP1. RFFL KO had a minimal effect on the ubiquitylation of EHD1, MICALL1, and Rab11-FIP2, and failed to delay transferrin recycling. These results suggest that multiple Ub ligases including RFFL regulate the ubiquitylation of Rab11 effectors, determining the integral function of the ERC.
Collapse
Affiliation(s)
- Ryohei Sakai
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Ryosuke Fukuda
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Shin Unida
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Misaki Aki
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Yuji Ono
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Akinori Endo
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Satoshi Kusumi
- Division of Morphological Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Daisuke Koga
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University, Asahikawa 078-8510, Hokkaido, Japan
| | - Toshiaki Fukushima
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Masayuki Komada
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Tsukasa Okiyoneda
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| |
Collapse
|
27
|
Regulation of Myosin-5b by Rab11a and the Rab11 family interacting protein 2. Biosci Rep 2019; 39:BSR20181252. [PMID: 30545898 PMCID: PMC6328864 DOI: 10.1042/bsr20181252] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/06/2018] [Accepted: 12/12/2018] [Indexed: 12/21/2022] Open
Abstract
Mammalian myosin-5b (Myo5b) plays a critical role in the recycling of endosomes to the plasma membrane via the interactions with Rab11a and the Rab11 family interacting protein 2 (FIP2). However, it remains unclear on how Rab11a and FIP2 are coordinated in tethering Myo5b with the vesicles and activating the motor function of Myo5b. In the present study, we show that Rab11a binds to the globular tail domain (GTD) of Myo5b and this binding abolishes the head–GTD interaction of Myo5b, thus activating the motor function of Myo5b. On the other hand, FIP2 directly interacts with both Rab11a and the tail of Myo5b, and the binding of FIP2 to Myo5b does not affect Myo5b motor function. Moreover, Rab11a displays higher affinity to FIP2 than to Myo5b, suggesting that Rab11a binds preferentially to FIP2 than to Myo5b. Based on the current findings, we propose that the association of Myo5b with vesicles is mediated by FIP2, which bridges Myo5b and the membrane-bound Rab11a, whereas the motor function of Myo5b is regulated by Rab11a.
Collapse
|
28
|
Pelaseyed T, Bretscher A. Regulation of actin-based apical structures on epithelial cells. J Cell Sci 2018; 131:131/20/jcs221853. [PMID: 30333133 DOI: 10.1242/jcs.221853] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cells of transporting epithelia are characterized by the presence of abundant F-actin-based microvilli on their apical surfaces. Likewise, auditory hair cells have highly reproducible rows of apical stereocilia (giant microvilli) that convert mechanical sound into an electrical signal. Analysis of mutations in deaf patients has highlighted the critical components of tip links between stereocilia, and related structures that contribute to the organization of microvilli on epithelial cells have been found. Ezrin/radixin/moesin (ERM) proteins, which are activated by phosphorylation, provide a critical link between the plasma membrane and underlying actin cytoskeleton in surface structures. Here, we outline recent insights into how microvilli and stereocilia are built, and the roles of tip links. Furthermore, we highlight how ezrin is locally regulated by phosphorylation, and that this is necessary to maintain polarity. Localized phosphorylation is achieved through an intricate coincidence detection mechanism that requires the membrane lipid phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] and the apically localized ezrin kinase, lymphocyte-oriented kinase (LOK, also known as STK10) or Ste20-like kinase (SLK). We also discuss how ezrin-binding scaffolding proteins regulate microvilli and how, despite these significant advances, it remains to be discovered how the cell polarity program ultimately interfaces with these processes.
Collapse
Affiliation(s)
- Thaher Pelaseyed
- Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, University of Gothenburg, SE-405 30 Gothenburg, Sweden
| | - Anthony Bretscher
- Weill Institute for Cell and Molecular Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
29
|
Zhu S, Bhat S, Syan S, Kuchitsu Y, Fukuda M, Zurzolo C. Rab11a-Rab8a cascade regulates the formation of tunneling nanotubes through vesicle recycling. J Cell Sci 2018; 131:jcs.215889. [PMID: 30209134 DOI: 10.1242/jcs.215889] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 08/29/2018] [Indexed: 01/02/2023] Open
Abstract
Tunneling nanotubes (TNTs) are actin-enriched membranous channels enabling cells to communicate over long distances. TNT-like structures form between various cell types and mediate the exchange of different cargos, such as ions, vesicles, organelles and pathogens; thus, they may play a role in physiological conditions and diseases (e.g. cancer and infection). TNTs also allow the intercellular passage of protein aggregates related to neurodegenerative diseases, thus propagating protein misfolding. Understanding the mechanism of TNT formation is mandatory in order to reveal the mechanism of disease propagation and to uncover their physiological function. Vesicular transport controlled by the small GTPases Rab11a and Rab8a can promote the formation of different plasma membrane protrusions (filopodia, cilia and neurites). Here, we report that inhibiting membrane recycling reduces the number of TNT-connected cells and that overexpression of Rab11a and Rab8a increases the number of TNT-connected cells and the propagation of vesicles between cells in co-culture. We demonstrate that these two Rab GTPases act in a cascade in which Rab11a activation of Rab8a is independent of Rabin8. We also show that VAMP3 acts downstream of Rab8a to regulate TNT formation.
Collapse
Affiliation(s)
- Seng Zhu
- Unit of Membrane Trafficking and Pathogenesis, Department of Cell Biology and Infection, Pasteur Institute, Paris 75015, France
| | - Shaarvari Bhat
- Unit of Membrane Trafficking and Pathogenesis, Department of Cell Biology and Infection, Pasteur Institute, Paris 75015, France
| | - Sylvie Syan
- Unit of Membrane Trafficking and Pathogenesis, Department of Cell Biology and Infection, Pasteur Institute, Paris 75015, France
| | - Yoshihiko Kuchitsu
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Chiara Zurzolo
- Unit of Membrane Trafficking and Pathogenesis, Department of Cell Biology and Infection, Pasteur Institute, Paris 75015, France
| |
Collapse
|
30
|
Kjos I, Vestre K, Guadagno NA, Borg Distefano M, Progida C. Rab and Arf proteins at the crossroad between membrane transport and cytoskeleton dynamics. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2018; 1865:1397-1409. [PMID: 30021127 DOI: 10.1016/j.bbamcr.2018.07.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/05/2018] [Accepted: 07/13/2018] [Indexed: 01/04/2023]
Abstract
The intracellular movement and positioning of organelles and vesicles is mediated by the cytoskeleton and molecular motors. Small GTPases like Rab and Arf proteins are main regulators of intracellular transport by connecting membranes to cytoskeleton motors or adaptors. However, it is becoming clear that interactions between these small GTPases and the cytoskeleton are important not only for the regulation of membrane transport. In this review, we will cover our current understanding of the mechanisms underlying the connection between Rab and Arf GTPases and the cytoskeleton, with special emphasis on the double role of these interactions, not only in membrane trafficking but also in membrane and cytoskeleton remodeling. Furthermore, we will highlight the most recent findings about the fine control mechanisms of crosstalk between different members of Rab, Arf, and Rho families of small GTPases in the regulation of cytoskeleton organization.
Collapse
Affiliation(s)
- Ingrid Kjos
- Department of Biosciences, University of Oslo, Norway
| | | | | | | | | |
Collapse
|
31
|
Naslavsky N, Caplan S. The enigmatic endosome - sorting the ins and outs of endocytic trafficking. J Cell Sci 2018; 131:131/13/jcs216499. [PMID: 29980602 DOI: 10.1242/jcs.216499] [Citation(s) in RCA: 202] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The early endosome (EE), also known as the sorting endosome (SE) is a crucial station for the sorting of cargoes, such as receptors and lipids, through the endocytic pathways. The term endosome relates to the receptacle-like nature of this organelle, to which endocytosed cargoes are funneled upon internalization from the plasma membrane. Having been delivered by the fusion of internalized vesicles with the EE or SE, cargo molecules are then sorted to a variety of endocytic pathways, including the endo-lysosomal pathway for degradation, direct or rapid recycling to the plasma membrane, and to a slower recycling pathway that involves a specialized form of endosome known as a recycling endosome (RE), often localized to the perinuclear endocytic recycling compartment (ERC). It is striking that 'the endosome', which plays such essential cellular roles, has managed to avoid a precise description, and its characteristics remain ambiguous and heterogeneous. Moreover, despite the rapid advances in scientific methodologies, including breakthroughs in light microscopy, overall, the endosome remains poorly defined. This Review will attempt to collate key characteristics of the different types of endosomes and provide a platform for discussion of this unique and fascinating collection of organelles. Moreover, under-developed, poorly understood and important open questions will be discussed.
Collapse
Affiliation(s)
- Naava Naslavsky
- The Department of Biochemistry and Molecular Biology, The University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Steve Caplan
- The Department of Biochemistry and Molecular Biology, The University of Nebraska Medical Center, Omaha, NE 68198, USA .,The Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
32
|
Bourke AM, Bowen AB, Kennedy MJ. New approaches for solving old problems in neuronal protein trafficking. Mol Cell Neurosci 2018; 91:48-66. [PMID: 29649542 DOI: 10.1016/j.mcn.2018.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 11/16/2022] Open
Abstract
Fundamental cellular properties are determined by the repertoire and abundance of proteins displayed on the cell surface. As such, the trafficking mechanisms for establishing and maintaining the surface proteome must be tightly regulated for cells to respond appropriately to extracellular cues, yet plastic enough to adapt to ever-changing environments. Not only are the identity and abundance of surface proteins critical, but in many cases, their regulated spatial positioning within surface nanodomains can greatly impact their function. In the context of neuronal cell biology, surface levels and positioning of ion channels and neurotransmitter receptors play essential roles in establishing important properties, including cellular excitability and synaptic strength. Here we review our current understanding of the trafficking pathways that control the abundance and localization of proteins important for synaptic function and plasticity, as well as recent technological advances that are allowing the field to investigate protein trafficking with increasing spatiotemporal precision.
Collapse
Affiliation(s)
- Ashley M Bourke
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Aaron B Bowen
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States.
| |
Collapse
|
33
|
Banworth MJ, Li G. Consequences of Rab GTPase dysfunction in genetic or acquired human diseases. Small GTPases 2018. [PMID: 29239692 DOI: 10.1080/215412481397833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2023] Open
Abstract
Rab GTPases are important regulators of intracellular membrane trafficking in eukaryotes. Both activating and inactivating mutations in Rab genes have been identified and implicated in human diseases ranging from neurological disorders to cancer. In addition, altered Rab expression is often associated with disease prognosis. As such, the study of diseases associated with Rabs or Rab-interacting proteins has shed light on the important role of intracellular membrane trafficking in disease etiology. In this review, we cover recent advances in the field with an emphasis on cellular mechanisms.
Collapse
Affiliation(s)
- Marcellus J Banworth
- a Department of Biochemistry and Molecular Biology , University of Oklahoma Health Sciences Center , Oklahoma City , OK , USA
| | - Guangpu Li
- a Department of Biochemistry and Molecular Biology , University of Oklahoma Health Sciences Center , Oklahoma City , OK , USA
| |
Collapse
|
34
|
Schneeberger K, Roth S, Nieuwenhuis EES, Middendorp S. Intestinal epithelial cell polarity defects in disease: lessons from microvillus inclusion disease. Dis Model Mech 2018; 11:11/2/dmm031088. [PMID: 29590640 PMCID: PMC5894939 DOI: 10.1242/dmm.031088] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelium is a highly organized tissue. The establishment of epithelial cell polarity, with distinct apical and basolateral plasma membrane domains, is pivotal for both barrier formation and for the uptake and vectorial transport of nutrients. The establishment of cell polarity requires a specialized subcellular machinery to transport and recycle proteins to their appropriate location. In order to understand and treat polarity-associated diseases, it is necessary to understand epithelial cell-specific trafficking mechanisms. In this Review, we focus on cell polarity in the adult mammalian intestine. We discuss how intestinal epithelial polarity is established and maintained, and how disturbances in the trafficking machinery can lead to a polarity-associated disorder, microvillus inclusion disease (MVID). Furthermore, we discuss the recent developments in studying MVID, including the creation of genetically manipulated cell lines, mouse models and intestinal organoids, and their uses in basic and applied research. Summary: Microvillus inclusion disease serves as a useful model to enhance our understanding of the intestinal trafficking and polarity machinery in health and disease.
Collapse
Affiliation(s)
- Kerstin Schneeberger
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Sabrina Roth
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Edward E S Nieuwenhuis
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Sabine Middendorp
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands .,Regenerative Medicine Center Utrecht, University Medical Centre (UMC) Utrecht, 3584 CT, Utrecht, The Netherlands
| |
Collapse
|
35
|
Schlegel C, Weis VG, Knowles BC, Lapierre LA, Martin MG, Dickman P, Goldenring JR, Shub MD. Apical Membrane Alterations in Non-intestinal Organs in Microvillus Inclusion Disease. Dig Dis Sci 2018; 63:356-365. [PMID: 29218485 PMCID: PMC5797493 DOI: 10.1007/s10620-017-4867-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/22/2017] [Indexed: 12/09/2022]
Abstract
OBJECTIVES Microvillus inclusion disease (MVID) is a severe form of neonatal diarrhea, caused mainly by mutations in MYO5B. Inactivating mutations in MYO5B causes depolarization of enterocytes in the small intestine, which gives rise to chronic, unremitting secretory diarrhea. While the pathology of the small intestine in MVID patients is well described, little is known about extraintestinal effects of MYO5B mutation. METHODS We examined stomach, liver, pancreas, colon, and kidney in Navajo MVID patients, who share a single homozygous MYO5B-P660L (1979C>T p.Pro660Leu, exon 16). Sections were stained for markers of the apical membrane to assess polarized trafficking. RESULTS Navajo MVID patients showed notable changes in H/K-ATPase-containing tubulovesicle structure in the stomach parietal cells. Colonic mucosa was morphologically normal, but did show losses in apical ezrin and Syntaxin 3. Hepatocytes in the MVID patients displayed aberrant canalicular expression of the essential transporters MRP2 and BSEP. The pancreas showed small fragmented islets and a decrease in apical ezrin in pancreatic ducts. Kidney showed normal primary cilia. CONCLUSIONS These findings indicate that the effects of the P660L mutation in MYO5B in Navajo MVID patients are not limited to the small intestine, but that certain tissues may be able to compensate functionally for alterations in apical trafficking.
Collapse
Affiliation(s)
- Cameron Schlegel
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Victoria G Weis
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Byron C Knowles
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Lynne A Lapierre
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Martin G Martin
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Paul Dickman
- Division of Pathology and Laboratory Medicine, Phoenix Children's Hospital, Phoenix, AZ, USA
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - James R Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
| | - Mitchell D Shub
- Division of Gastroenterology, Phoenix Children's Hospital, Phoenix, AZ, USA
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| |
Collapse
|
36
|
Tebar F, Enrich C, Rentero C, Grewal T. GTPases Rac1 and Ras Signaling from Endosomes. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2018; 57:65-105. [PMID: 30097772 DOI: 10.1007/978-3-319-96704-2_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The endocytic compartment is not only the functional continuity of the plasma membrane but consists of a diverse collection of intracellular heterogeneous complex structures that transport, amplify, sustain, and/or sort signaling molecules. Over the years, it has become evident that early, late, and recycling endosomes represent an interconnected vesicular-tubular network able to form signaling platforms that dynamically and efficiently translate extracellular signals into biological outcome. Cell activation, differentiation, migration, death, and survival are some of the endpoints of endosomal signaling. Hence, to understand the role of the endosomal system in signal transduction in space and time, it is therefore necessary to dissect and identify the plethora of decoders that are operational in the different steps along the endocytic pathway. In this chapter, we focus on the regulation of spatiotemporal signaling in cells, considering endosomes as central platforms, in which several small GTPases proteins of the Ras superfamily, in particular Ras and Rac1, actively participate to control cellular processes like proliferation and cell mobility.
Collapse
Affiliation(s)
- Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain.
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
37
|
Banworth MJ, Li G. Consequences of Rab GTPase dysfunction in genetic or acquired human diseases. Small GTPases 2017; 9:158-181. [PMID: 29239692 DOI: 10.1080/21541248.2017.1397833] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Rab GTPases are important regulators of intracellular membrane trafficking in eukaryotes. Both activating and inactivating mutations in Rab genes have been identified and implicated in human diseases ranging from neurological disorders to cancer. In addition, altered Rab expression is often associated with disease prognosis. As such, the study of diseases associated with Rabs or Rab-interacting proteins has shed light on the important role of intracellular membrane trafficking in disease etiology. In this review, we cover recent advances in the field with an emphasis on cellular mechanisms.
Collapse
Affiliation(s)
- Marcellus J Banworth
- a Department of Biochemistry and Molecular Biology , University of Oklahoma Health Sciences Center , Oklahoma City , OK , USA
| | - Guangpu Li
- a Department of Biochemistry and Molecular Biology , University of Oklahoma Health Sciences Center , Oklahoma City , OK , USA
| |
Collapse
|
38
|
Loss of Myosin Vb in colorectal cancer is a strong prognostic factor for disease recurrence. Br J Cancer 2017; 117:1689-1701. [PMID: 29024942 PMCID: PMC5729446 DOI: 10.1038/bjc.2017.352] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/24/2017] [Accepted: 09/08/2017] [Indexed: 02/06/2023] Open
Abstract
Background: Selecting the most beneficial treatment regimens for colorectal cancer (CRC) patients remains challenging due to a lack of prognostic markers. Members of the Myosin family, proteins recognised to have a major role in trafficking and polarisation of cells, have recently been reported to be closely associated with several types of cancer and might thus serve as potential prognostic markers in the context of CRC. Methods: We used a previously established meta-analysis of publicly available gene expression data to analyse the expression of different members of the Myosin V family, namely MYO5A, 5B, and 5C, in CRC. Using laser-microdissected material as well as tissue microarrays from paired human CRC samples, we validated both RNA and protein expression of Myosin Vb (MYO5B) and its known adapter proteins (RAB8A and RAB25) in an independent patient cohort. Finally, we assessed the prognostic value of both MYO5B and its adapter-coupled combinatorial gene expression signatures. Results: The meta-analysis as well as an independent patient cohort study revealed a methylation-independent loss of MYO5B expression in CRC that matched disease progression. Although MYO5B mutations were identified in a small number of patients, these cannot be solely responsible for the common downregulation observed in CRC patients. Significantly, CRC patients with low MYO5B expression displayed shorter overall, disease-, and metastasis-free survival, a trend that was further reinforced when RAB8A expression was also taken into account. Conclusions: Our data identify MYO5B as a powerful prognostic biomarker in CRC, especially in early stages (stages I and II), which might help stratifying patients with stage II for adjuvant chemotherapy.
Collapse
|
39
|
Feng Q, Bonder EM, Engevik AC, Zhang L, Tyska MJ, Goldenring JR, Gao N. Disruption of Rab8a and Rab11a causes formation of basolateral microvilli in neonatal enteropathy. J Cell Sci 2017; 130:2491-2505. [PMID: 28596241 DOI: 10.1242/jcs.201897] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 06/01/2017] [Indexed: 12/15/2022] Open
Abstract
Misplaced formation of microvilli to basolateral domains and intracellular inclusions in enterocytes are pathognomonic features in congenital enteropathy associated with mutation of the apical plasma membrane receptor syntaxin 3 (STX3). Although the demonstrated binding of Myo5b to the Rab8a and Rab11a small GTPases in vitro implicates cytoskeleton-dependent membrane sorting, the mechanisms underlying the microvillar location defect remain unclear. By selective or combinatory disruption of Rab8a and Rab11a membrane traffic in vivo, we demonstrate that transport of distinct cargo to the apical brush border rely on either individual or both Rab regulators, whereas certain basolateral cargos are redundantly transported by both factors. Enterocyte-specific Rab8a and Rab11a double-knockout mouse neonates showed immediate postnatal lethality and more severe enteropathy than single knockouts, with extensive formation of microvilli along basolateral surfaces. Notably, following an inducible Rab11a deletion from neonatal enterocytes, basolateral microvilli were induced within 3 days. These data identify a potentially important and distinct mechanism for a characteristic microvillus defect exhibited by enterocytes of patients with neonatal enteropathy.
Collapse
Affiliation(s)
- Qiang Feng
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Amy C Engevik
- Department of Surgery, and Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lanjing Zhang
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA.,Department of Pathology, University Medical Center of Princeton, Plainsboro, NJ 08536, USA.,Rutgers Cancer Institute of New Jersey, Rutgers University, Piscataway, NJ 08903, USA
| | - Matthew J Tyska
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - James R Goldenring
- Department of Surgery, and Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Nashville VA Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA .,Rutgers Cancer Institute of New Jersey, Rutgers University, Piscataway, NJ 08903, USA
| |
Collapse
|
40
|
Vogel GF, Janecke AR, Krainer IM, Gutleben K, Witting B, Mitton SG, Mansour S, Ballauff A, Roland JT, Engevik AC, Cutz E, Müller T, Goldenring JR, Huber LA, Hess MW. Abnormal Rab11-Rab8-vesicles cluster in enterocytes of patients with microvillus inclusion disease. Traffic 2017; 18:453-464. [PMID: 28407399 DOI: 10.1111/tra.12486] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/05/2017] [Accepted: 04/05/2017] [Indexed: 12/14/2022]
Abstract
Microvillus inclusion disease (MVID) is a congenital enteropathy characterized by accumulation of vesiculo-tubular endomembranes in the subapical cytoplasm of enterocytes, historically termed "secretory granules." However, neither their identity nor pathophysiological significance is well defined. Using immunoelectron microscopy and tomography, we studied biopsies from MVID patients (3× Myosin 5b mutations and 1× Syntaxin3 mutation) and compared them to controls and genome-edited CaCo2 cell models, harboring relevant mutations. Duodenal biopsies from 2 patients with novel Myosin 5b mutations and typical clinical symptoms showed unusual ultrastructural phenotypes: aberrant subapical vesicles and tubules were prominent in the enterocytes, though other histological hallmarks of MVID were almost absent (ectopic intra-/intercellular microvilli, brush border atrophy). We identified these enigmatic vesiculo-tubular organelles as Rab11-Rab8-positive recycling compartments of altered size, shape and location harboring the apical SNARE Syntaxin3, apical transporters sodium-hydrogen exchanger 3 (NHE3) and cystic fibrosis transmembrane conductance regulator. Our data strongly indicate that in MVID disrupted trafficking between cargo vesicles and the apical plasma membrane is the primary cause of a defect of epithelial polarity and subsequent facultative loss of brush border integrity, leading to malabsorption. Furthermore, they support the notion that mislocalization of transporters, such as NHE3 substantially contributes to the reported sodium loss diarrhea.
Collapse
Affiliation(s)
- Georg F Vogel
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria.,Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria.,Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas R Janecke
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Iris M Krainer
- Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria.,Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Karin Gutleben
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Witting
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Sahar Mansour
- Human Genetics Research Center, St. George's University of London, London, UK
| | | | - Joseph T Roland
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Amy C Engevik
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ernest Cutz
- Division of Pathology, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Lukas A Huber
- Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael W Hess
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
41
|
Qiu Y, Gong J, Feng J, Wang R, Han J, Liu T, Lu Y, Li L, Zhang M, Sheps JA, Wang N, Yan Y, Li J, Chen L, Borchers CH, Sipos B, Knisely A, Ling V, Xing Q, Wang J. Defects in myosin VB are associated with a spectrum of previously undiagnosed low γ-glutamyltransferase cholestasis. Hepatology 2017; 65:1655-1669. [PMID: 28027573 PMCID: PMC5413810 DOI: 10.1002/hep.29020] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 11/16/2016] [Accepted: 12/21/2016] [Indexed: 12/11/2022]
Abstract
Hereditary cholestasis in childhood and infancy with normal serum gamma-glutamyltransferase (GGT) activity is linked to several genes. Many patients, however, remain genetically undiagnosed. Defects in myosin VB (MYO5B; encoded by MYO5B) cause microvillus inclusion disease (MVID; MIM251850) with recurrent watery diarrhea. Cholestasis, reported as an atypical presentation in MVID, has been considered a side effect of parenteral alimentation. Here, however, we report on 10 patients who experienced cholestasis associated with biallelic, or suspected biallelic, mutations in MYO5B and who had neither recurrent diarrhea nor received parenteral alimentation. Seven of them are from two study cohorts, together comprising 31 undiagnosed low-GGT cholestasis patients; 3 are sporadic. Cholestasis in 2 patients was progressive, in 3 recurrent, in 2 transient, and in 3 uncategorized because of insufficient follow-up. Liver biopsy specimens revealed giant-cell change of hepatocytes and intralobular cholestasis with abnormal distribution of bile salt export pump (BSEP) at canaliculi, as well as coarse granular dislocation of MYO5B. Mass spectrometry of plasma demonstrated increased total bile acids, primary bile acids, and conjugated bile acids, with decreased free bile acids, similar to changes in BSEP-deficient patients. Literature review revealed that patients with biallelic mutations predicted to eliminate MYO5B expression were more frequent in typical MVID than in isolated-cholestasis patients (11 of 38 vs. 0 of 13). CONCLUSION MYO5B deficiency may underlie 20% of previously undiagnosed low-GGT cholestasis. MYO5B deficiency appears to impair targeting of BSEP to the canalicular membrane with hampered bile acid excretion, resulting in a spectrum of cholestasis without diarrhea. (Hepatology 2017;65:1655-1669).
Collapse
Affiliation(s)
- Yi‐Ling Qiu
- The Center for Pediatric Liver DiseasesChildren's Hospital of Fudan UniversityShanghaiChina
| | - Jing‐Yu Gong
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Jia‐Yan Feng
- Department of PathologyChildren's Hospital of Fudan UniversityShanghaiChina
| | | | - Jun Han
- University of Victoria−Genome BC Proteomics CentreUniversity of VictoriaVictoriaBritish ColumbiaCanada
| | - Teng Liu
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Yi Lu
- The Center for Pediatric Liver DiseasesChildren's Hospital of Fudan UniversityShanghaiChina
| | - Li‐Ting Li
- The Center for Pediatric Liver DiseasesChildren's Hospital of Fudan UniversityShanghaiChina
| | - Mei‐Hong Zhang
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | | | - Neng‐Li Wang
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Yan‐Yan Yan
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Jia‐Qi Li
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Lian Chen
- Department of PathologyChildren's Hospital of Fudan UniversityShanghaiChina
| | - Christoph H. Borchers
- University of Victoria−Genome BC Proteomics CentreUniversity of VictoriaVictoriaBritish ColumbiaCanada
| | - Bence Sipos
- Institute of General Pathology and NeuropathologyTübingen University HospitalTübingenGermany
| | - A.S. Knisely
- Institute of PathologyGraz Medical UniversityGrazAustria
| | - Victor Ling
- BC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Qing‐He Xing
- Institutes of Biomedical Sciences of Fudan UniversityShanghaiChina
| | - Jian‐She Wang
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
- Department of Infectious DiseasesChildren's Hospital of Fudan UniversityShanghaiChina
| |
Collapse
|
42
|
Welz T, Kerkhoff E. Exploring the iceberg: Prospects of coordinated myosin V and actin assembly functions in transport processes. Small GTPases 2017; 10:111-121. [PMID: 28394692 DOI: 10.1080/21541248.2017.1281863] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Spir actin nucleators and myosin V motor proteins were recently discovered to coexist in a protein complex. The direct interaction allows the coordinated activation of actin motor proteins and actin filament track generation at vesicle membranes. By now the cooperation of myosin V (MyoV) motors and Spir actin nucleation function has only been shown in the exocytic transport of Rab11 vesicles in metaphase mouse oocytes. Next to Rab11, myosin V motors however interact with a variety of Rab GTPases including Rab3, Rab8 and Rab10. As a common theme most of the MyoV interacting Rab GTPases function at different steps along the exocytic transport routes. We here summarize the different transport functions of class V myosins and provide as proof of principle data showing a colocalization of Spir actin nucleators and MyoVa at Rab8a vesicles. This suggests that besides Rab11/MyoV transport also the Rab8/MyoV and possibly other MyoV transport processes recruit Spir actin filament nucleation function.
Collapse
Affiliation(s)
- Tobias Welz
- a University Hospital Regensburg, Department of Neurology , Molecular Cell Biology Laboratory , Regensburg , Germany
| | - Eugen Kerkhoff
- a University Hospital Regensburg, Department of Neurology , Molecular Cell Biology Laboratory , Regensburg , Germany
| |
Collapse
|
43
|
Small GTPases Rab8a and Rab11a Are Dispensable for Rhodopsin Transport in Mouse Photoreceptors. PLoS One 2016; 11:e0161236. [PMID: 27529348 PMCID: PMC4987053 DOI: 10.1371/journal.pone.0161236] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 08/02/2016] [Indexed: 01/01/2023] Open
Abstract
Rab11a and Rab8a are ubiquitous small GTPases shown as required for rhodopsin transport in Xenopus laevis and zebrafish photoreceptors by dominant negative (dn) disruption of function. Here, we generated retina-specific Rab11a (retRab11a) and Rab8a (retRab8a) single and double knockout mice to explore the consequences in mouse photoreceptors. Rhodopsin and other outer segment (OS) membrane proteins targeted correctly to OS and electroretinogram (ERG) responses in all three mutant mouse lines were indistinguishable from wild-type (WT). Further, AAV (adeno-associated virus)-mediated expression of dnRab11b in retRab11a-/- retina, or expression of dnRab8b in retRab8a-/- retina did not cause OS protein mislocalization. Finally, a retRab8a-/- retina injected at one month of age with AAVs expressing dnRab11a, dnRab11b, dnRab8b, and dnRab10 (four dn viruses on Rab8a-/- background) and harvested three months later exhibited normal OS protein localization. In contrast to results obtained with dnRab GTPases in Xenopus and zebrafish, mouse Rab11a and Rab8a are dispensable for proper rhodopsin and outer segment membrane protein targeting. Absence of phenotype after expression of four dn Rab GTPases in a Rab8a-/- retina suggests that Rab8b and Rab11b paralogs maybe dispensable as well. Our data thus demonstrate significant interspecies variation in photoreceptor membrane protein and rhodopsin trafficking.
Collapse
|
44
|
Sidhaye J, Pinto CS, Dharap S, Jacob T, Bhargava S, Sonawane M. The zebrafish goosepimples/myosin Vb mutant exhibits cellular attributes of human microvillus inclusion disease. Mech Dev 2016; 142:62-74. [PMID: 27497746 PMCID: PMC5161235 DOI: 10.1016/j.mod.2016.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 08/01/2016] [Accepted: 08/02/2016] [Indexed: 12/20/2022]
Abstract
Microvillus inclusion disease (MVID) is a life-threatening enteropathy characterised by malabsorption and incapacitating fluid loss due to chronic diarrhoea. Histological analysis has revealed that enterocytes in MVID patients exhibit reduction of microvilli, presence of microvillus inclusion bodies and intestinal villus atrophy, whereas genetic linkage analysis has identified mutations in myosin Vb gene as the main cause of MVID. In order to understand the cellular basis of MVID and the associated formation of inclusion bodies, an animal model that develops ex utero and is tractable genetically as well as by microscopy would be highly useful. Here we report that the intestine of the zebrafish goosepimples (gsp)/myosin Vb (myoVb) mutant shows severe reduction in intestinal folds - structures similar to mammalian villi. The loss of folds is further correlated with changes in the shape of enterocytes. In striking similarity with MVID patients, zebrafish gsp/myoVb mutant larvae exhibit microvillus atrophy, microvillus inclusions and accumulation of secretory material in enterocytes. We propose that the zebrafish gsp/myoVb mutant is a valuable model to study the pathophysiology of MVID. Furthermore, owing to the advantages of zebrafish in screening libraries of small molecules, the gsp mutant will be an ideal tool to identify compounds having therapeutic value against MVID. myosin Vb is expressed in the zebrafish intestine. goosepimples/myosin Vb function is essential for epithelial morphogenesis in the zebrafish intestine. The goosepimples mutant recapitulates pathognomonic features of microvillus inclusion disease. The function of myosin Vb in the intestine is conserved between fish and mammals.
Collapse
Affiliation(s)
- Jaydeep Sidhaye
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Clyde Savio Pinto
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Shweta Dharap
- Department of Biotechnology, Abasaheb Garware College, Pune, India
| | - Tressa Jacob
- Indian Institute of Science Education and Research, Pune, India
| | - Shobha Bhargava
- Department of Zoology, University of Pune, Ganeshkhind, Pune, India
| | - Mahendra Sonawane
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India.
| |
Collapse
|
45
|
Bello OD, Cappa AI, de Paola M, Zanetti MN, Fukuda M, Fissore RA, Mayorga LS, Michaut MA. Rab3A, a possible marker of cortical granules, participates in cortical granule exocytosis in mouse eggs. Exp Cell Res 2016; 347:42-51. [PMID: 27423421 DOI: 10.1016/j.yexcr.2016.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 07/04/2016] [Accepted: 07/12/2016] [Indexed: 10/21/2022]
Abstract
Fusion of cortical granules with the oocyte plasma membrane is the most significant event to prevent polyspermy. This particular exocytosis, also known as cortical reaction, is regulated by calcium and its molecular mechanism is still not known. Rab3A, a member of the small GTP-binding protein superfamily, has been implicated in calcium-dependent exocytosis and is not yet clear whether Rab3A participates in cortical granules exocytosis. Here, we examine the involvement of Rab3A in the physiology of cortical granules, particularly, in their distribution during oocyte maturation and activation, and their participation in membrane fusion during cortical granule exocytosis. Immunofluorescence and Western blot analysis showed that Rab3A and cortical granules have a similar migration pattern during oocyte maturation, and that Rab3A is no longer detected after cortical granule exocytosis. These results suggested that Rab3A might be a marker of cortical granules. Overexpression of EGFP-Rab3A colocalized with cortical granules with a Pearson correlation coefficient of +0.967, indicating that Rab3A and cortical granules have almost a perfect colocalization in the egg cortical region. Using a functional assay, we demonstrated that microinjection of recombinant, prenylated and active GST-Rab3A triggered cortical granule exocytosis, indicating that Rab3A has an active role in this secretory pathway. To confirm this active role, we inhibited the function of endogenous Rab3A by microinjecting a polyclonal antibody raised against Rab3A prior to parthenogenetic activation. Our results showed that Rab3A antibody microinjection abolished cortical granule exocytosis in parthenogenetically activated oocytes. Altogether, our findings confirm that Rab3A might function as a marker of cortical granules and participates in cortical granule exocytosis in mouse eggs.
Collapse
Affiliation(s)
- Oscar Daniel Bello
- Instituto de Histología y Embriología, CONICET - Universidad Nacional de Cuyo, Av. Libertador 80, 5500 Mendoza, Argentina
| | - Andrea Isabel Cappa
- Instituto de Histología y Embriología, CONICET - Universidad Nacional de Cuyo, Av. Libertador 80, 5500 Mendoza, Argentina
| | - Matilde de Paola
- Instituto de Histología y Embriología, CONICET - Universidad Nacional de Cuyo, Av. Libertador 80, 5500 Mendoza, Argentina
| | - María Natalia Zanetti
- Instituto de Histología y Embriología, CONICET - Universidad Nacional de Cuyo, Av. Libertador 80, 5500 Mendoza, Argentina
| | - Mitsunori Fukuda
- Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Rafael A Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, MA 01003, USA
| | - Luis S Mayorga
- Instituto de Histología y Embriología, CONICET - Universidad Nacional de Cuyo, Av. Libertador 80, 5500 Mendoza, Argentina
| | - Marcela A Michaut
- Instituto de Histología y Embriología, CONICET - Universidad Nacional de Cuyo, Av. Libertador 80, 5500 Mendoza, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Argentina.
| |
Collapse
|
46
|
Shinde SR, Maddika S. A modification switch on a molecular switch: Phosphoregulation of Rab7 during endosome maturation. Small GTPases 2016; 7:164-7. [PMID: 27070490 DOI: 10.1080/21541248.2016.1175539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Rab GTPases, the highly conserved members of Ras GTPase superfamily are the pivotal regulators of vesicle-mediated trafficking. Rab GTPases, each with a specific subcellular localization, exert tremendous control over various aspects of vesicular transport, identity and dynamics. Several lines of research have established that GDI, GEFs and GAPs are the critical players to orchestrate Rab GTPase activity and function. The importance of post translational modifications in Rab GTPase functional regulation is poorly or not yet been addressed except for prenylation. Our recent study has revealed a novel dephosphorylation dependent regulatory mechanism for Rab7 activity and function. We have shown the importance of PTEN mediated dephosphorylation of Rab7 on highly conserved S72 and Y183 residues, which is essential for its GDI mediated membrane targeting and further activation by GEF. In conclusion, our study highlighted the importance of a phosphorylation/dephosphorylation switch in controlling timely Rab7 localization and activity on endosomes.
Collapse
Affiliation(s)
- Swapnil Rohidas Shinde
- a Laboratory of Cell Death & Cell Survival, Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally , Hyderabad , India.,b Graduate Studies, Manipal University , Manipal , India
| | - Subbareddy Maddika
- a Laboratory of Cell Death & Cell Survival, Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally , Hyderabad , India
| |
Collapse
|
47
|
Abstract
Fluorescence microscopy is employed to identify Kinesin-1 cargos. Recently, the heavy chain of Kinesin-1 (KIF5B) was shown to transport the nuclear transcription factor c-MYC for proteosomal degradation in the cytoplasm. The method described here involves the study of a motorless KIF5B mutant for fluorescence microscopy. The wild-type and motorless KIF5B proteins are tagged with the fluorescent protein tdTomato. The wild-type tdTomato-KIF5B appears homogenously in the cytoplasm, while the motorless tdTomato-KIF5B mutant forms aggregates in the cytoplasm. Aggregation of the motorless KIF5B mutant induces aggregation of its cargo c-MYC in the cytoplasm. Hence, this method provides a visual means to identify the cargos of Kinesin-1. A similar strategy can be utilized to identify cargos of other motor proteins.
Collapse
Affiliation(s)
- Clement M Lee
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai;
| |
Collapse
|
48
|
Weis VG, Knowles BC, Choi E, Goldstein AE, Williams JA, Manning EH, Roland JT, Lapierre LA, Goldenring JR. Loss of MYO5B in mice recapitulates Microvillus Inclusion Disease and reveals an apical trafficking pathway distinct to neonatal duodenum. Cell Mol Gastroenterol Hepatol 2016; 2:131-157. [PMID: 27019864 PMCID: PMC4806369 DOI: 10.1016/j.jcmgh.2015.11.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 11/25/2015] [Indexed: 12/10/2022]
Abstract
BACKGROUND AND AIMS Inactivating mutations in MYO5B cause severe neonatal diarrhea in Microvillus Inclusion Disease. Loss of active MYO5B causes the formation of pathognomonic inclusions and aberrations in brush border enzymes. METHODS We developed three mouse models of germline, constitutively intestinal targeted and inducible intestinal targeted deletion of MYO5B. The mice were evaluated for enterocyte cellular morphology. RESULTS Germline MYO5B KO mice showed early diarrhea and failure to thrive with evident microvillus inclusions and loss of apical transporters in the duodenum. IgG was present within inclusions. Apical transporters were lost and inclusions were present in the duodenum, but were nearly absent in the ileum. VillinCre;MYO5BF/F mice showed similar pathology and morphological changes in duodenal enterocytes. In contrast, when MYO5B KO was induced with tamoxifen treatment at 8 weeks of age, VillinCreERT2;MYO5BF/F mice developed severe diarrhea with loss of duodenal brush border enzymes, but few inclusions were observed in enterocytes. However, if tamoxifen is administered to 2-day-old VillinCreERT2;MYO5BF/F mice, prominent microvillus inclusions were observed. CONCLUSIONS The microvillus inclusions that develop after MYO5B loss reveal the presence of an unrecognized apical membrane trafficking pathway in neonatal duodenal enterocytes. However, the diarrheal pathology after MYO5B loss is due to deficits in transporter presentation at the apical membrane in duodenal enterocytes.
Collapse
Affiliation(s)
- Victoria G. Weis
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Byron C. Knowles
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Eunyoung Choi
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| | - Anna E. Goldstein
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Janice A. Williams
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Elizabeth H. Manning
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| | - Joseph T. Roland
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Lynne A. Lapierre
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| | - James R. Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| |
Collapse
|
49
|
Schafer JC, McRae RE, Manning EH, Lapierre LA, Goldenring JR. Rab11-FIP1A regulates early trafficking into the recycling endosomes. Exp Cell Res 2016; 340:259-73. [PMID: 26790954 PMCID: PMC4744548 DOI: 10.1016/j.yexcr.2016.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 12/19/2015] [Accepted: 01/10/2016] [Indexed: 12/31/2022]
Abstract
The Rab11 family of small GTPases, along with the Rab11-family interacting proteins (Rab11-FIPs), are critical regulators of intracellular vesicle trafficking and recycling. We have identified a point mutation of Threonine-197 site to an Alanine in Rab11-FIP1A, which causes a dramatic dominant negative phenotype when expressed in HeLa cells. The normally perinuclear distribution of GFP-Rab11-FIP1A was condensed into a membranous cisternum with almost no GFP-Rab11-FIP1A(T197A) remaining outside of this central locus. Also, this condensed GFP-FIP1A(T197A) altered the distribution of proteins in the Rab11a recycling pathway including endogenous Rab11a, Rab11-FIP1C, and transferrin receptor (CD71). Furthermore, this condensed GFP-FIP1A(T197A)-containing structure exhibited little movement in live HeLa cells. Expression of GFP-FIP1A(T197A) caused a strong blockade of transferrin recycling. Treatment of cells expressing GFP-FIP1A(T197A) with nocodazole did not disperse the Rab11a-containing recycling system. We also found that Rab5 and EEA1 were accumulated in membranes by GFP-Rab11-FIP1A but Rab4 was unaffected, suggesting that a direct pathway may exist from early endosomes into the Rab11a-containing recycling system. Our study of a potent inhibitory trafficking mutation in Rab11-FIP1A shows that Rab11-FIP1A associates with and regulates trafficking at an early step in the process of membrane recycling.
Collapse
Affiliation(s)
- Jenny C Schafer
- Departments of Surgery, Nashville, TN, USA; Epithelial Biology Center, Nashville, TN, USA
| | - Rebecca E McRae
- Departments of Surgery, Nashville, TN, USA; Cell & Developmental Biology, Nashville, TN, USA; Epithelial Biology Center, Nashville, TN, USA
| | - Elizabeth H Manning
- Departments of Surgery, Nashville, TN, USA; Epithelial Biology Center, Nashville, TN, USA
| | - Lynne A Lapierre
- Departments of Surgery, Nashville, TN, USA; Epithelial Biology Center, Nashville, TN, USA
| | - James R Goldenring
- Departments of Surgery, Nashville, TN, USA; Cell & Developmental Biology, Nashville, TN, USA; Epithelial Biology Center, Nashville, TN, USA; Vanderbilt University School of Medicine and the Nashville VA Medical Center, Nashville, TN, USA.
| |
Collapse
|
50
|
Bravo-Cordero JJ, Cordani M, Soriano SF, Díez B, Muñoz-Agudo C, Casanova-Acebes M, Boullosa C, Guadamillas MC, Ezkurdia I, González-Pisano D, del Pozo MA, Montoya MC. A novel high content analysis tool reveals Rab8-driven actin and FA reorganization through Rho GTPases and calpain/MT1. J Cell Sci 2016; 129:1734-49. [DOI: 10.1242/jcs.174920] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 02/29/2016] [Indexed: 01/31/2023] Open
Abstract
Rab8 is a small Ras-related GTPase that regulates polarized membrane transport to the plasma membrane. A high content analysis (HCA) tool developed to dissect Rab8-mediated actin and focal adhesion (FA) reorganization revealed that Rab8 activation significantly induced Rac1/Tiam1 to mediate cortical actin (CA) formation and RhoA-dependent stress fibre (SF) disassembly. Rab8 activation increased Rac1 activity, while its depletion activated RhoA, which led to reorganization of the actin cytoskeleton. Rab8 was also associated with FA, promoting their disassembly in a microtubule dependent manner. This Rab8 effect involved Calpain, MT1-MMP and Rho GTPases. Moreover, we demonstrate the role of Rab8 in the cell migration process. Indeed, Rab8 is required for EGF-induced cell polarization and chemotaxis as well as for the directional persistency of intrinsic cell motility. These data reveal that Rab8 drives cell motility by mechanisms both dependent and independent of Rho GTPases, thereby regulating the establishment of cell polarity, turnover of FA, and actin cytoskeleton rearrangements, thus determining the directionality of cell migration.
Collapse
Affiliation(s)
- José J. Bravo-Cordero
- Current Address: Division of Hematology and Oncology, Department of Medicine, Mount Sinai School of Medicine, Tisch Cancer Institute, New York, NY, Box 1079, USA
| | - Marco Cordani
- Integrin Signaling Laboratory, Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid, E28029, Spain
| | - Silvia F. Soriano
- Integrin Signaling Laboratory, Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid, E28029, Spain
| | - Begoña Díez
- Cellomics Unit. Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares CNIC. C/ Melchor Fernandez Almagro, 3. Madrid, E28029, Spain
| | - Carmen Muñoz-Agudo
- Cellomics Unit. Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares CNIC. C/ Melchor Fernandez Almagro, 3. Madrid, E28029, Spain
| | - María Casanova-Acebes
- Cellomics Unit. Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares CNIC. C/ Melchor Fernandez Almagro, 3. Madrid, E28029, Spain
| | - César Boullosa
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), C/Melchor Fernández Almagro 3, Madrid E28029, Spain
| | - Marta C. Guadamillas
- Integrin Signaling Laboratory, Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid, E28029, Spain
| | - Iakes Ezkurdia
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), C/Melchor Fernández Almagro 3, Madrid E28029, Spain
| | - David González-Pisano
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), C/Melchor Fernández Almagro 3, Madrid E28029, Spain
| | - Miguel A. del Pozo
- Integrin Signaling Laboratory, Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid, E28029, Spain
| | - María C. Montoya
- Cellomics Unit. Cell Biology & Physiology Program; Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares CNIC. C/ Melchor Fernandez Almagro, 3. Madrid, E28029, Spain
| |
Collapse
|