1
|
Sandhu A, Lyu X, Wan X, Meng X, Tang NH, Gonzalez G, Syed IN, Chen L, Jin Y, Chisholm AD. The microtubule regulator EFA-6 forms cortical foci dependent on its intrinsically disordered region and interactions with tubulins. Cell Rep 2024; 43:114776. [PMID: 39305484 DOI: 10.1016/j.celrep.2024.114776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/12/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
The EFA6 protein family, originally identified as Sec7 guanine nucleotide exchange factors, has also been found to regulate cortical microtubule (MT) dynamics. Here, we find that in the mature C. elegans epidermal epithelium, EFA-6 forms punctate foci in specific regions of the apical cortex, dependent on its intrinsically disordered region (IDR). The EFA-6 IDR can form biomolecular condensates in vitro. In genetic screens for mutants with altered GFP::EFA-6 localization, we identified a gain-of-function (gf) mutation in α-tubulin tba-1 that induces ectopic EFA-6 foci in multiple cell types. Lethality of tba-1(gf) is partially suppressed by loss of function in efa-6. The ability of TBA-1(gf) to trigger ectopic EFA-6 foci requires β-tubulin TBB-2 and the chaperon EVL-20/Arl2. tba-1(gf)-induced EFA-6 foci display slower turnover, contain the MT-associated protein TAC-1/TACC, and require the EFA-6 MT elimination domain (MTED). Our results reveal functionally important crosstalk between cellular tubulins and cortical MT regulators in vivo.
Collapse
Affiliation(s)
- Anjali Sandhu
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Xiaohui Lyu
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Xinghaoyun Wan
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Xuefeng Meng
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Ngang Heok Tang
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Gilberto Gonzalez
- Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ishana N Syed
- Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Lizhen Chen
- Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Andrew D Chisholm
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, San Diego, CA 92093, USA; Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
2
|
Lind MCH, Naimi WA, Chiarelli TJ, Sparrer T, Ghosh M, Shapiro L, Carlyon JA. Anaplasma phagocytophilum invasin AipA interacts with CD13 to elicit Src kinase signaling that promotes infection. mBio 2024; 15:e0156124. [PMID: 39324816 PMCID: PMC11481542 DOI: 10.1128/mbio.01561-24] [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: 05/28/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024] Open
Abstract
Host-microbe interactions that facilitate entry into mammalian cells are essential for obligate intracellular bacterial survival and pathogenesis. Anaplasma phagocytophilum is an obligate intracellular bacterium that invades neutrophils to cause granulocytic anaplasmosis. The invasin-receptor pairs and signaling events that induce Anaplasma uptake are inadequately defined. A. phagocytophilum invasion protein A orchestrates entry via residues 9-21 (AipA9-21) engaging an unknown receptor. Yeast two-hybrid screening suggested that AipA binds within C-terminal amino acids 851-967 of CD13 (aminopeptidase N), a multifunctional protein that, when crosslinked, initiates Src kinase and Syk signaling that culminates in endocytosis. Co-immunoprecipitation validated the interaction and confirmed that it requires the AipA N-terminus. CD13 ectopic expression on non-phagocytic cells increased susceptibility to A. phagocytophilum infection. Antibody blocking and enzymatic inhibition experiments found that the microbe exploits CD13 but not its ectopeptidase activity to infect myeloid cells. A. phagocytophilum induces Src and Syk phosphorylation during invasion. Inhibitor treatment established that Src is key for A. phagocytophilum infection, while Syk is dispensable and oriented the pathogen-invoked signaling pathway by showing that Src is activated before Syk. Disrupting the AipA-CD13 interaction with AipA9-21 or CD13781-967 antibody inhibited Src and Syk phosphorylation and also infection. CD13 crosslinking antibody that induces Src and Syk signaling restored infectivity of anti-AipA9-21-treated A. phagocytophilum. The bacterium poorly infected CD13 knockout mice, providing the first demonstration that CD13 is important for microbial infection in vivo. Overall, A. phagocytophilum AipA9-21 binds CD13 to induce Src signaling that mediates uptake into host cells, and CD13 is critical for infection in vivo. IMPORTANCE Diverse microbes engage CD13 to infect host cells. Yet invasin-CD13 interactions, the signaling they invoke for pathogen entry, and the relevance of CD13 to infection in vivo are underexplored. Dissecting these concepts would advance fundamental understanding of a convergently evolved infection strategy and could have translational benefits. Anaplasma phagocytophilum infects neutrophils to cause granulocytic anaplasmosis, an emerging disease for which there is no vaccine and few therapeutic options. We found that A. phagocytophilum uses its surface protein and recently identified protective immunogen, AipA, to bind CD13 to elicit Src kinase signaling, which is critical for infection. We elucidated the AipA CD13 binding domain, which CD13 region AipA engages, and established that CD13 is key for A. phagocytophilum infection in vivo. Disrupting the AipA-CD13 interaction could be utilized to prevent granulocytic anaplasmosis and offers a model that could be applied to protect against multiple infectious diseases.
Collapse
Affiliation(s)
- Mary Clark H. Lind
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Waheeda A. Naimi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Travis J. Chiarelli
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Tavis Sparrer
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Mallika Ghosh
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Linda Shapiro
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
3
|
Michaels TM, Essop MF, Joseph DE. Potential Effects of Hyperglycemia on SARS-CoV-2 Entry Mechanisms in Pancreatic Beta Cells. Viruses 2024; 16:1243. [PMID: 39205219 PMCID: PMC11358987 DOI: 10.3390/v16081243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The COVID-19 pandemic has revealed a bidirectional relationship between SARS-CoV-2 infection and diabetes mellitus. Existing evidence strongly suggests hyperglycemia as an independent risk factor for severe COVID-19, resulting in increased morbidity and mortality. Conversely, recent studies have reported new-onset diabetes following SARS-CoV-2 infection, hinting at a potential direct viral attack on pancreatic beta cells. In this review, we explore how hyperglycemia, a hallmark of diabetes, might influence SARS-CoV-2 entry and accessory proteins in pancreatic β-cells. We examine how the virus may enter and manipulate such cells, focusing on the role of the spike protein and its interaction with host receptors. Additionally, we analyze potential effects on endosomal processing and accessory proteins involved in viral infection. Our analysis suggests a complex interplay between hyperglycemia and SARS-CoV-2 in pancreatic β-cells. Understanding these mechanisms may help unlock urgent therapeutic strategies to mitigate the detrimental effects of COVID-19 in diabetic patients and unveil if the virus itself can trigger diabetes onset.
Collapse
Affiliation(s)
- Tara M. Michaels
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| | - M. Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa;
| | - Danzil E. Joseph
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| |
Collapse
|
4
|
Jiang C, Zhou P, Zhang X, Ma N, Hu Y, Zhang M, Ghonaim AH, Li H, Dong L, Zeng W, Li C, Lang Y, Sun Y, He Q, Li W. ARF6 promotes Streptococcus suis suilysin induced apoptosis in HBMECs. Int J Biol Macromol 2024; 268:131839. [PMID: 38663699 DOI: 10.1016/j.ijbiomac.2024.131839] [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: 02/21/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Abstract
Streptococcus suis (S. suis) is a significant zoonotic microorganism that causes a severe illness in both pigs and humans and is characterized by severe meningitis and septicemia. Suilysin (SLY), which is secreted by S. suis, plays a crucial role as a virulence factor in the disease. To date, the interaction between SLY and host cells is not fully understood. In this study, we identified the interacting proteins between SLY and human brain microvascular endothelial cells (HBMECs) using the TurboID-mediated proximity labeling method. 251 unique proteins were identified in TurboID-SLY treated group, of which six plasma membrane proteins including ARF6, GRK6, EPB41L5, DSC1, TJP2, and PNN were identified. We found that the proteins capable of interacting with SLY are ARF6 and PNN. Subsequent investigations revealed that ARF6 substantially increased the invasive ability of S. suis in HBMECs. Furthermore, ARF6 promoted SLY-induced the activation of p38 MAPK signaling pathway in HBMECs. Moreover, ARF6 promoted the apoptosis in HBMECs through the activation of p38 MAPK signaling pathway induced by SLY. Finally, we confirmed that ARF6 could increase the virulence of SLY in C57BL/6 mice. These findings offer valuable insights that contribute to a deeper understanding of the pathogenic mechanism of SLY.
Collapse
Affiliation(s)
- Changsheng Jiang
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Pei Zhou
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Xiaoqian Zhang
- China Institute of Veterinary Drug Control, Beijing 102629, China
| | - NingNing Ma
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Yaofang Hu
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Mengjia Zhang
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Ahmed H Ghonaim
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Desert Research Center, Cairo 11435, Egypt
| | - Huimin Li
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Ling Dong
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Wei Zeng
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Chang Li
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis, Ministry of Agriculture and Rural Affairs, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Yifei Lang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yumei Sun
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Qigai He
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China.
| | - Wentao Li
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China.
| |
Collapse
|
5
|
Fukaya M, Ibuchi K, Sugawara T, Itakura M, Ito A, Shiroshima T, Hara Y, Okamoto H, Luton F, Sakagami H. EFA6A, an Exchange Factor for Arf6, Regulates NGF-Dependent TrkA Recycling From Early Endosomes and Neurite Outgrowth in PC12 Cells. Traffic 2024; 25:e12936. [PMID: 38725127 DOI: 10.1111/tra.12936] [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: 03/22/2023] [Revised: 12/18/2023] [Accepted: 12/26/2023] [Indexed: 06/03/2024]
Abstract
Endosomal trafficking of TrkA is a critical process for nerve growth factor (NGF)-dependent neuronal cell survival and differentiation. The small GTPase ADP-ribosylation factor 6 (Arf6) is implicated in NGF-dependent processes in PC12 cells through endosomal trafficking and actin cytoskeleton reorganization. However, the regulatory mechanism for Arf6 in NGF signaling is largely unknown. In this study, we demonstrated that EFA6A, an Arf6-specific guanine nucleotide exchange factor, was abundantly expressed in PC12 cells and that knockdown of EFA6A significantly inhibited NGF-dependent Arf6 activation, TrkA recycling from early endosomes to the cell surface, prolonged ERK1/2 phosphorylation, and neurite outgrowth. We also demonstrated that EFA6A forms a protein complex with TrkA through its N-terminal region, thereby enhancing its catalytic activity for Arf6. Similarly, we demonstrated that EFA6A forms a protein complex with TrkA in cultured dorsal root ganglion (DRG) neurons. Furthermore, cultured DRG neurons from EFA6A knockout mice exhibited disturbed NGF-dependent TrkA trafficking compared with wild-type neurons. These findings provide the first evidence for EFA6A as a key regulator of NGF-dependent TrkA trafficking and signaling.
Collapse
Affiliation(s)
- Masahiro Fukaya
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kanta Ibuchi
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Takeyuki Sugawara
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Makoto Itakura
- Department of Biochemistry, Kitasato University School of Medicine, Sagamihara, Japan
| | - Akiko Ito
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Tomoko Shiroshima
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yoshinobu Hara
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hirotsugu Okamoto
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Frédéric Luton
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Université Côte d'Azur, Valbonne, France
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
6
|
Sandhu A, Lyu X, Wan X, Meng X, Tang NH, Gonzalez G, Syed IN, Chen L, Jin Y, Chisholm AD. The microtubule regulator EFA-6 forms spatially restricted cortical foci dependent on its intrinsically disordered region and interactions with tubulins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.14.588158. [PMID: 38645057 PMCID: PMC11030407 DOI: 10.1101/2024.04.14.588158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Microtubules (MTs) are dynamic components of the cytoskeleton and play essential roles in morphogenesis and maintenance of tissue and cell integrity. Despite recent advances in understanding MT ultrastructure, organization, and growth control, how cells regulate MT organization at the cell cortex remains poorly understood. The EFA-6/EFA6 proteins are recently identified membrane-associated proteins that inhibit cortical MT dynamics. Here, combining visualization of endogenously tagged C. elegans EFA-6 with genetic screening, we uncovered tubulin-dependent regulation of EFA-6 patterning. In the mature epidermal epithelium, EFA-6 forms punctate foci in specific regions of the apical cortex, dependent on its intrinsically disordered region (IDR). We further show the EFA-6 IDR is sufficient to form biomolecular condensates in vitro. In screens for mutants with altered GFP::EFA-6 localization, we identified a novel gain-of-function (gf) mutation in an α-tubulin tba-1 that induces ectopic EFA-6 foci in multiple cell types. tba-1(gf) animals exhibit temperature-sensitive embryonic lethality, which is partially suppressed by efa-6(lf), indicating the interaction between tubulins and EFA-6 is important for normal development. TBA-1(gf) shows reduced incorporation into filamentous MTs but has otherwise mild effects on cellular MT organization. The ability of TBA-1(gf) to trigger ectopic EFA-6 foci formation requires β-tubulin TBB-2 and the chaperon EVL-20/Arl2. The tba-1(gf)-induced EFA-6 foci display slower turnover, contain the MT-associated protein TAC-1/TACC, and require the EFA-6 MTED. Our results reveal a novel crosstalk between cellular tubulins and cortical MT regulators in vivo.
Collapse
Affiliation(s)
- Anjali Sandhu
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093 USA
| | - Xiaohui Lyu
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093 USA
| | - Xinghaoyun Wan
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093 USA
| | - Xuefeng Meng
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093 USA
| | - Ngang Heok Tang
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, CA 92093 USA
| | - Gilberto Gonzalez
- Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ishana N. Syed
- Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Lizhen Chen
- Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093 USA
| | - Andrew D. Chisholm
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, CA 92093 USA
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, CA 92093 USA
| |
Collapse
|
7
|
Ghosh M, McGurk F, Norris R, Dong A, Nair S, Jellison E, Murphy P, Verma R, Shapiro LH. The Implant-Induced Foreign Body Response Is Limited by CD13-Dependent Regulation of Ubiquitination of Fusogenic Proteins. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:663-676. [PMID: 38149920 PMCID: PMC10828181 DOI: 10.4049/jimmunol.2300688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/29/2023] [Indexed: 12/28/2023]
Abstract
Implanted medical devices, from artificial heart valves and arthroscopic joints to implantable sensors, often induce a foreign body response (FBR), a form of chronic inflammation resulting from the inflammatory reaction to a persistent foreign stimulus. The FBR is characterized by a subset of multinucleated giant cells (MGCs) formed by macrophage fusion, the foreign body giant cells (FBGCs), accompanied by inflammatory cytokines, matrix deposition, and eventually deleterious fibrotic implant encapsulation. Despite efforts to improve biocompatibility, implant-induced FBR persists, compromising the utility of devices and making efforts to control the FBR imperative for long-term function. Controlling macrophage fusion in FBGC formation presents a logical target to prevent implant failure, but the actual contribution of FBGCs to FBR-induced damage is controversial. CD13 is a molecular scaffold, and in vitro induction of CD13KO bone marrow progenitors generates many more MGCs than the wild type, suggesting that CD13 regulates macrophage fusion. In the mesh implant model of FBR, CD13KO mice produced significantly more peri-implant FBGCs with enhanced TGF-β expression and increased collagen deposition versus the wild type. Prior to fusion, increased protrusion and microprotrusion formation accompanies hyperfusion in the absence of CD13. Expression of fusogenic proteins driving cell-cell fusion was aberrantly sustained at high levels in CD13KO MGCs, which we show is due to a novel CD13 function, to our knowledge, regulating ubiquitin/proteasomal protein degradation. We propose CD13 as a physiologic brake limiting aberrant macrophage fusion and the FBR, and it may be a novel therapeutic target to improve the success of implanted medical devices. Furthermore, our data directly implicate FBGCs in the detrimental fibrosis that characterizes the FBR.
Collapse
Affiliation(s)
- Mallika Ghosh
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| | - Fraser McGurk
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| | - Rachael Norris
- Department of Cell Biology, University of Connecticut Medical School, Farmington, CT
| | - Andy Dong
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| | - Sreenidhi Nair
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| | - Evan Jellison
- Department of Immunology, University of Connecticut Medical School, Farmington, CT
| | - Patrick Murphy
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| | - Rajkumar Verma
- Department of Neuroscience, University of Connecticut Medical School, Farmington, CT
| | - Linda H. Shapiro
- Centers for Vascular Biology, University of Connecticut Medical School, Farmington, CT
| |
Collapse
|
8
|
Du T, Ma C, Wang Z, Hao Y, Zhang W. Distribution and Degradation of Pork Filamin during Postmortem Aging. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15287-15295. [PMID: 37788342 DOI: 10.1021/acs.jafc.3c04208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
The filamin C (FLNC) was hypothesized to be colocalized with its certain binding partners in pork tissues and calpain as well as caspase was assumed responsible for the postmortem degradation of FLNC. Therefore, the specific distribution of pork FLNC and its degradation pattern during postmortem aging were investigated in this study. The longissimus thoracis muscles from 12 pigs were removed from the carcasses and then aged at 4 °C for 1, 6, 12, 24, 72, and 168 h, respectively. The FLNC signals appeared to localize in subsarcolemmal areas by cross-sectional images, while the localization was found surrounding the myofibrils at the level of the Z-discs in longitudinal sections. FLNC displayed a highly overlapped spatial colocalization with actin or integrin. Western blot results showed that the intact 290 kDa FLNC was rapidly degraded to produce an approximately 280 kDa band. An almost overlapped distribution pattern was observed between FLNC and μ-calpain or caspase-3 in porcine skeletal muscle cells. Moreover, both the μ-calpain inhibitor and the caspase-3 inhibitor could inhibit the degradation of FLNC in porcine LT muscles during postmortem aging.
Collapse
Affiliation(s)
- Tongyao Du
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chao Ma
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zixu Wang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuejing Hao
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wangang Zhang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
9
|
Díaz-Alvarez L, Martínez-Sánchez ME, Gray E, Pérez-Figueroa E, Ortega E. Aminopeptidase N/CD13 Crosslinking Promotes the Activation and Membrane Expression of Integrin CD11b/CD18. Biomolecules 2023; 13:1488. [PMID: 37892170 PMCID: PMC10604325 DOI: 10.3390/biom13101488] [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: 08/23/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
The β2 integrin CD11b/CD18, also known as complement receptor 3 (CR3), and the moonlighting protein aminopeptidase N (CD13), are two myeloid immune receptors with overlapping activities: adhesion, migration, phagocytosis of opsonized particles, and respiratory burst induction. Given their common functions, shared physical location, and the fact that some receptors can activate a selection of integrins, we hypothesized that CD13 could induce CR3 activation through an inside-out signaling mechanism and possibly have an influence on its membrane expression. We revealed that crosslinking CD13 on the surface of human macrophages not only activates CR3 but also influences its membrane expression. Both phenomena are affected by inhibitors of Src, PLCγ, Syk, and actin polymerization. Additionally, after only 10 min at 37 °C, cells with crosslinked CD13 start secreting pro-inflammatory cytokines like interferons type 1 and 2, IL-12p70, and IL-17a. We integrated our data with a bioinformatic analysis to confirm the connection between these receptors and to suggest the signaling cascade linking them. Our findings expand the list of features of CD13 by adding the activation of a different receptor via inside-out signaling. This opens the possibility of studying the joint contribution of CD13 and CR3 in contexts where either receptor has a recognized role, such as the progression of some leukemias.
Collapse
Affiliation(s)
- Laura Díaz-Alvarez
- Instituto de Investigaciones Biomédicas, Departamento de Inmunología, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Mexico City 04510, Mexico
| | | | - Eleanor Gray
- London Centre for Nanotechnology, Department of Physics and Astronomy, University College London, London WC2R 2LS, UK
| | - Erandi Pérez-Figueroa
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| | - Enrique Ortega
- Instituto de Investigaciones Biomédicas, Departamento de Inmunología, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
10
|
Sun D, Guo Y, Tang P, Li H, Chen L. Arf6 as a therapeutic target: Structure, mechanism, and inhibitors. Acta Pharm Sin B 2023; 13:4089-4104. [PMID: 37799386 PMCID: PMC10547916 DOI: 10.1016/j.apsb.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/28/2023] [Accepted: 06/02/2023] [Indexed: 10/07/2023] Open
Abstract
ADP-ribosylation factor 6 (Arf6), a small G-protein of the Ras superfamily, plays pivotal roles in multiple cellular events, including exocytosis, endocytosis, actin remodeling, plasma membrane reorganization and vesicular transport. Arf6 regulates the progression of cancer through the activation of cell motility and invasion. Aberrant Arf6 activation is a potential therapeutic target. This review aims to understand the comprehensive function of Arf6 for future cancer therapy. The Arf6 GEFs, protein structure, and roles in cancer have been summarized. Comprehending the mechanism underlying Arf6-mediated cancer cell growth and survival is essential. The structural features of Arf6 and its efforts are discussed and may be contributed to the discovery of future novel protein-protein interaction inhibitors. In addition, Arf6 inhibitors and mechanism of action are listed in the table. This review further emphasizes the crucial roles in drug resistance and attempts to offer an outlook of Arf6 in cancer therapy.
Collapse
Affiliation(s)
- Dejuan Sun
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuanyuan Guo
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Piyu Tang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
11
|
Zhang J, Sun S, Liu J, Zhang L, Guo D, Zhang N, Zhao J, Kong D, Xu T, Wang X, Xu W, Li X, Jiang Y. Discovery of a Novel Ubenimex Derivative as a First-in-Class Dual CD13/Proteasome Inhibitor for the Treatment of Cancer. Molecules 2023; 28:6343. [PMID: 37687169 PMCID: PMC10489073 DOI: 10.3390/molecules28176343] [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: 07/19/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
The CD13 inhibitor ubenimex is used as an adjuvant drug with chemotherapy for the treatment of cancer due to its function as an immunoenhancer, but it has limitations in its cytotoxic efficacy. The proteasome inhibitor ixazomib is a landmark drug in the treatment of multiple myeloma with a high anti-cancer activity. Herein, we conjugated the pharmacophore of ubenimex and the boric acid of ixazomib to obtain a dual CD13 and proteasome inhibitor 7 (BC-05). BC-05 exhibited potent inhibitory activity on both human CD13 (IC50 = 0.13 μM) and the 20S proteasome (IC50 = 1.39 μM). Although BC-05 displayed lower anti-proliferative activity than that of ixazomib in vitro, an advantage was established in the in vivo anti-cancer efficacy and prolongation of survival time, which may be due to its anti-metastatic and immune-stimulating activity. A pharmacokinetic study revealed that BC-05 is a potentially orally active agent with an F% value of 24.9%. Moreover, BC-05 showed more favorable safety profiles than those of ixazomib in preliminary toxicity studies. Overall, the results indicate that BC-05 is a promising drug candidate for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Jian Zhang
- College of Pharmacy, Weifang Medical University, Weifang 261053, China; (J.Z.)
| | - Simin Sun
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China (X.L.)
| | - Jinyu Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China (X.L.)
| | - Liang Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China (X.L.)
| | - Di Guo
- College of Pharmacy, Weifang Medical University, Weifang 261053, China; (J.Z.)
| | - Naixin Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jun Zhao
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Tongqiang Xu
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Xuejian Wang
- College of Pharmacy, Weifang Medical University, Weifang 261053, China; (J.Z.)
| | - Wenfang Xu
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Xiaoyang Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China (X.L.)
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Yuqi Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China (X.L.)
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| |
Collapse
|
12
|
Niu J, Ma Y, Yang Y, Lv H, Wang J, Wang T, Liu F, Xu S, Jiang Z, Lin W. Lighting up the changes of plasma membranes during apoptosis with fluorescent probes. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
13
|
Integrin receptor trafficking in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:271-302. [PMID: 36813362 DOI: 10.1016/bs.pmbts.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Integrins are a family of 24 different heterodimers that are indispensable for multicellular life. Cell polarity, adhesion and migration are controlled by integrins delivered to the cell surface which in turn is regulated by the exo- and endocytic trafficking of integrins. The deep integration between trafficking and cell signaling determines the spatial and temporal output from any biochemical cue. Integrin trafficking plays a key role in development and many pathological conditions, especially cancer. Several novel regulators of integrin traffic have been discovered in recent times, including a novel class of integrin carrying vesicles, the intracellular nanovesicles (INVs). The tight regulation of trafficking pathways by cell signaling, where kinases phosphorylate key small GTPases in the trafficking pathway enable coordination of cell response to the extracellular milieu. Integrin heterodimer expression and trafficking differ in different tissues and contexts. In this Chapter, we discuss recent studies on integrin trafficking and its contribution to normal physiological and pathophysiological states.
Collapse
|
14
|
Pérez-Figueroa E, Álvarez-Carrasco P, Ortega E. Crosslinking of membrane CD13 in human neutrophils mediates phagocytosis and production of reactive oxygen species, neutrophil extracellular traps and proinflammatory cytokines. Front Immunol 2022; 13:994496. [PMID: 36439182 PMCID: PMC9686367 DOI: 10.3389/fimmu.2022.994496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/19/2022] [Indexed: 09/08/2023] Open
Abstract
Aminopeptidase N, or CD13, is a cell membrane ectopeptidase highly expressed in myeloid cells. Through its enzymatic activity, CD13 regulates the activity of several bioactive peptides, such as endorphins and enkephalins, chemotactic peptides like MCP-1 and IL-8, angiotensin III, bradikinin, etc. In recent years, it has been appreciated that independently of its peptidase activity, CD13 can activate signal transduction pathways and mediate effector functions such as phagocytosis and cytokine secretion in monocytes and macrophages. Although neutrophils are known to express CD13 on its membrane, it is currently unknown if CD13 can mediate effector functions in these cells. Here, we show that in human neutrophils CD13 can mediate phagocytosis, which is dependent on a signaling pathway that involves Syk, and PI3-K. Phagocytosis mediated by CD13 is associated with production of reactive oxygen species (ROS). The level of phagocytosis and ROS production mediated by CD13 are similar to those through FcγRIII (CD16b), a widely studied receptor of human neutrophils. Also, CD13 ligation induces the release of neutrophil extracellular traps (NETs) as well as cytokine secretion from neutrophils. These results support the hypothesis that CD13 is a membrane receptor able to activate effector functions in human neutrophils.
Collapse
Affiliation(s)
| | | | - Enrique Ortega
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de Mexico, Mexico
| |
Collapse
|
15
|
Kadefors M, Rolandsson Enes S, Åhrman E, Michaliková B, Löfdahl A, Dellgren G, Scheding S, Westergren-Thorsson G. CD105 +CD90 +CD13 + identifies a clonogenic subset of adventitial lung fibroblasts. Sci Rep 2021; 11:24417. [PMID: 34952905 PMCID: PMC8709856 DOI: 10.1038/s41598-021-03963-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/13/2021] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal cells are important components of specified niches in the lung, and can mediate a wide range of processes including tissue regeneration and repair. Dysregulation of these processes can lead to improper remodeling of tissue as observed in several lung diseases. The mesenchymal cells responsible remain poorly described, partially due to the heterogenic nature of the mesenchymal compartment and the absence of appropriate markers. Here, we describe that CD105+CD90+ mesenchymal cells can be divided into two populations based on their expression of CD13/aminopeptidase N (CD105+CD90+CD13− and CD105+CD90+CD13+). By prospective isolation using FACS, we show that both these populations give rise to clonogenic fibroblast-like cells, but with an increased clonogenic and proliferative capacity of CD105+CD90+CD13+ cells. Transcriptomic and spatial analysis pinpoints an adventitial fibroblast subset as the origin of CD105+CD90+CD13+ clonogenic mesenchymal cells in human lung.
Collapse
Affiliation(s)
- Måns Kadefors
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | | | - Emma Åhrman
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | - Anna Löfdahl
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Göran Dellgren
- Department of Cardiothoracic Surgery and Transplant Institute, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Stefan Scheding
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden.,Department of Hematology, Skåne University Hospital Lund, Lund, Sweden
| | | |
Collapse
|
16
|
Yoshihara M, Mizutani S, Kato Y, Matsumoto K, Mizutani E, Mizutani H, Fujimoto H, Osuka S, Kajiyama H. Recent Insights into Human Endometrial Peptidases in Blastocyst Implantation via Shedding of Microvesicles. Int J Mol Sci 2021; 22:13479. [PMID: 34948276 PMCID: PMC8708926 DOI: 10.3390/ijms222413479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/02/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022] Open
Abstract
Blastocyst implantation involves multiple interactions with numerous molecules expressed in endometrial epithelial cells (EECs) during the implantation window; however, there is limited information regarding the molecular mechanism underlying the crosstalk. In blastocysts, fibronectin plays a major role in the adhesion of various types of cells by binding to extracellular matrix proteins via the Arg-Gly-Asp (RGD) motif. In EECs, RGD-recognizing integrins are important bridging receptors for fibronectin, whereas the non-RGD binding of fibronectin includes interactions with dipeptidyl peptidase IV (DPPIV)/cluster of differentiation (CD) 26. Fibronectin may also bind to aminopeptidase N (APN)/CD13, and in the endometrium, these peptidases are present in plasma membranes and lysosomal membranes. Blastocyst implantation is accompanied by lysosome exocytosis, which transports various peptidases and nutrients into the endometrial cavity to facilitate blastocyst implantation. Both DPPIV and APN are released into the uterine cavity via shedding of microvesicles (MVs) from EECs. Recently, extracellular vesicles derived from endometrial cells have been proposed to act on trophectoderm cells to promote implantation. MVs are also secreted from embryonal stem cells and may play an active role in implantation. Thus, crosstalk between the blastocyst and endometrium via extracellular vesicles is a new insight into the fundamental molecular basis of blastocyst implantation.
Collapse
Affiliation(s)
- Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (H.M.); (H.F.); (S.O.); (H.K.)
| | - Shigehiko Mizutani
- Daiyabilding Lady’s Clinic, 1-1-17 Meieki, Nishi-ku, Nagoya 451-0045, Japan;
| | - Yukio Kato
- Department of Molecular Pharmacotherapeutics, Faculty of Pharmacy, Kanazawa University, Kanazawa 920-1192, Japan;
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Japan;
| | - Eita Mizutani
- Daiyabilding Lady’s Clinic, 1-1-17 Meieki, Nishi-ku, Nagoya 451-0045, Japan;
| | - Hidesuke Mizutani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (H.M.); (H.F.); (S.O.); (H.K.)
| | - Hiroki Fujimoto
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (H.M.); (H.F.); (S.O.); (H.K.)
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Satoko Osuka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (H.M.); (H.F.); (S.O.); (H.K.)
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (H.M.); (H.F.); (S.O.); (H.K.)
| |
Collapse
|
17
|
López-Cortés GI, Díaz-Alvarez L, Ortega E. Leukocyte Membrane Enzymes Play the Cell Adhesion Game. Front Immunol 2021; 12:742292. [PMID: 34887854 PMCID: PMC8650063 DOI: 10.3389/fimmu.2021.742292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/29/2021] [Indexed: 12/16/2022] Open
Abstract
For a long time, proteins with enzymatic activity have not been usually considered to carry out other functions different from catalyzing chemical reactions within or outside the cell. Nevertheless, in the last few years several reports have uncovered the participation of numerous enzymes in other processes, placing them in the category of moonlighting proteins. Some moonlighting enzymes have been shown to participate in complex processes such as cell adhesion. Cell adhesion plays a physiological role in multiple processes: it enables cells to establish close contact with one another, allowing communication; it is a key step during cell migration; it is also involved in tightly binding neighboring cells in tissues, etc. Importantly, cell adhesion is also of great importance in pathophysiological scenarios like migration and metastasis establishment of cancer cells. Cell adhesion is strictly regulated through numerous switches: proteins, glycoproteins and other components of the cell membrane. Recently, several cell membrane enzymes have been reported to participate in distinct steps of the cell adhesion process. Here, we review a variety of examples of membrane bound enzymes participating in adhesion of immune cells.
Collapse
Affiliation(s)
- Georgina I López-Cortés
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Laura Díaz-Alvarez
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Enrique Ortega
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
18
|
Yue K, Hou X, Jia G, Zhang L, Zhang J, Tan L, Wang X, Zhang Z, Li P, Xu W, Li X, Jiang Y. Design, synthesis and biological evaluation of hybrid of ubenimex-fluorouracil for hepatocellular carcinoma therapy. Bioorg Chem 2021; 116:105343. [PMID: 34544027 DOI: 10.1016/j.bioorg.2021.105343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/28/2021] [Accepted: 09/06/2021] [Indexed: 10/20/2022]
Abstract
In our previous study, we discovered a ubenimex-fluorouracil (5FU) conjugates BC-02, which displays significant in vivo anti-tumor activity, however, the instability of BC-02 in plasma limits its further development as a drug candidate. Herein, we designed and synthesized four novel ubenimex-5FU conjugates by optimizing the linkers between ubenimex and 5FU based on BC-02. Representative compound 20 is more stable than BC-02 in human plasma and displays about 100 times higher CD13 inhibitory activity than the positive control ubenimex. Meanwhile, the antiproliferative activity of 20 was comparable with 5FU in vitro. The preliminary mechanism study indicated that compound 20 exhibited significant anti-invasion and anti-angiogenesis activities in vitro. Furthermore, compound 20 obviously inhibits tumor growth and metastasis in vivo and prolong the survival time of tumor-bearing mice. Our study may have an important implication reference for the design of more druglike mutual prodrug, and compound 20 can be used as a lead compound for further design and development.
Collapse
Affiliation(s)
- Kairui Yue
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Xiaohan Hou
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Geng Jia
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Liang Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Jian Zhang
- College of Pharmacy, Weifang Medical University, 261053 Wei'fang, Shandong, PR China
| | - Leqiao Tan
- Weifang Bochuang International Biological Medicinal Institute, Weifang, Shandong 261061, PR China
| | - Xuejian Wang
- College of Pharmacy, Weifang Medical University, 261053 Wei'fang, Shandong, PR China
| | - Zhaolin Zhang
- Weifang Bochuang International Biological Medicinal Institute, Weifang, Shandong 261061, PR China
| | - Peixia Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China
| | - Wenfang Xu
- Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong 266071, PR China
| | - Xiaoyang Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, PR China.
| | - Yuqi Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, PR China.
| |
Collapse
|
19
|
CD13 orients the apical-basal polarity axis necessary for lumen formation. Nat Commun 2021; 12:4697. [PMID: 34349123 PMCID: PMC8338993 DOI: 10.1038/s41467-021-24993-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 07/09/2021] [Indexed: 02/07/2023] Open
Abstract
Polarized epithelial cells can organize into complex structures with a characteristic central lumen. Lumen formation requires that cells coordinately orient their polarity axis so that the basolateral domain is on the outside and apical domain inside epithelial structures. Here we show that the transmembrane aminopeptidase, CD13, is a key determinant of epithelial polarity orientation. CD13 localizes to the apical membrane and associates with an apical complex with Par6. CD13-deficient cells display inverted polarity in which apical proteins are retained on the outer cell periphery and fail to accumulate at an intercellular apical initiation site. Here we show that CD13 is required to couple apical protein cargo to Rab11-endosomes and for capture of endosomes at the apical initiation site. This role in polarity utilizes the short intracellular domain but is independent of CD13 peptidase activity.
Collapse
|
20
|
CD13 is a critical regulator of cell-cell fusion in osteoclastogenesis. Sci Rep 2021; 11:10736. [PMID: 34031489 PMCID: PMC8144195 DOI: 10.1038/s41598-021-90271-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/07/2021] [Indexed: 12/04/2022] Open
Abstract
The transmembrane aminopeptidase CD13 is highly expressed in cells of the myeloid lineage, regulates dynamin-dependent receptor endocytosis and recycling and is a necessary component of actin cytoskeletal organization. Here, we show that CD13-deficient mice present a low bone density phenotype with increased numbers of osteoclasts per bone surface, but display a normal distribution of osteoclast progenitor populations in the bone marrow and periphery. In addition, the bone formation and mineral apposition rates are similar between genotypes, indicating a defect in osteoclast-specific function in vivo. Lack of CD13 led to exaggerated in vitro osteoclastogenesis as indicated by significantly enhanced fusion of bone marrow-derived multinucleated osteoclasts in the presence of M-CSF and RANKL, resulting in abnormally large cells containing remarkably high numbers of nuclei. Mechanistically, while expression levels of the fusion-regulatory proteins dynamin and DC-STAMP1 must be downregulated for fusion to proceed, these are aberrantly sustained at high levels even in CD13-deficient mature multi-nucleated osteoclasts. Further, the stability of fusion-promoting proteins is maintained in the absence of CD13, implicating CD13 in protein turnover mechanisms. Together, we conclude that CD13 may regulate cell–cell fusion by controlling the expression and localization of key fusion regulatory proteins that are critical for osteoclast fusion.
Collapse
|
21
|
Sabo Y, de Los Santos K, Goff SP. IQGAP1 Negatively Regulates HIV-1 Gag Trafficking and Virion Production. Cell Rep 2021; 30:4065-4081.e4. [PMID: 32209469 PMCID: PMC7199802 DOI: 10.1016/j.celrep.2020.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/29/2020] [Accepted: 02/28/2020] [Indexed: 01/21/2023] Open
Abstract
IQGAP1 is a master regulator of many cellular processes, including intracellular vesicle trafficking and endocytosis. We show that depletion of IQGAP1 in a variety of cell types increases the release of HIV-1 infectious virions and that overexpression diminishes virion production, with neither affecting the early stages of infection. IQGAP1 negatively regulates the steady-state levels of HIV-1 Gag at the plasma membrane, the site of assembly. We establish that IQGAP1 interacts with both the nucleocapsid and p6 domains of Gag, and interaction with either domain is sufficient for its regulatory function. Finally, we demonstrate that IQGAP1 regulation is independent of HIV-1 Gag “late-domains” sequences required by the virus to recruit the cellular ESCRT machinery. Thus, we provide evidence that IQGAP1 is a negative regulatory factor inhibiting efficient budding of HIV-1 by reducing Gag accumulation at the plasma membrane. IQGAP1 is a ubiquitously expressed master regulator of many cellular processes, including intracellular trafficking. Sabo et al. demonstrate that in a variety of cell types, IQGAP1 acts as a negative regulator of HIV-1 viral particle release by reducing accumulation of the Gag viral structural protein at the plasma membrane.
Collapse
Affiliation(s)
- Yosef Sabo
- Howard Hughes Medical Institute, Columbia University, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University, New York, NY 10032, USA; Department of Medicine, Division of Infectious Diseases, Columbia University, New York, NY 10032, USA
| | - Kenia de Los Santos
- Howard Hughes Medical Institute, Columbia University, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University, New York, NY 10032, USA
| | - Stephen P Goff
- Howard Hughes Medical Institute, Columbia University, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
22
|
Devarakonda CKV, Meredith E, Ghosh M, Shapiro LH. Coronavirus Receptors as Immune Modulators. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:923-929. [PMID: 33380494 PMCID: PMC7889699 DOI: 10.4049/jimmunol.2001062] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/04/2020] [Indexed: 12/11/2022]
Abstract
The Coronaviridae family includes the seven known human coronaviruses (CoV) that cause mild to moderate respiratory infections (HCoV-229E, HCoV-NL63, HCoV-OC43, HCoV-HKU1) as well as severe illness and death (MERS-CoV, SARS-CoV, SARS-CoV-2). Severe infections induce hyperinflammatory responses that are often intensified by host adaptive immune pathways to profoundly advance disease severity. Proinflammatory responses are triggered by CoV entry mediated by host cell surface receptors. Interestingly, five of the seven strains use three cell surface metallopeptidases (CD13, CD26, and ACE2) as receptors, whereas the others employ O-acetylated-sialic acid (a key feature of metallopeptidases) for entry. Why CoV evolved to use peptidases as their receptors is unknown, but the peptidase activities of the receptors are dispensable, suggesting the virus uses/benefits from other functions of these molecules. Indeed, these receptors participate in the immune modulatory pathways that contribute to the pathological hyperinflammatory response. This review will focus on the role of CoV receptors in modulating immune responses.
Collapse
Affiliation(s)
| | - Emily Meredith
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Mallika Ghosh
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Linda H Shapiro
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT 06030
| |
Collapse
|
23
|
Peng X, Wang T, Gao H, Yue X, Bian W, Mei J, Zhang Y. The interplay between IQGAP1 and small GTPases in cancer metastasis. Biomed Pharmacother 2021; 135:111243. [PMID: 33434854 DOI: 10.1016/j.biopha.2021.111243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/27/2020] [Accepted: 12/31/2020] [Indexed: 01/07/2023] Open
Abstract
The metastatic spread of tumor cells to distant anatomical locations is a critical cause for disease progression and leads to more than 90 % of cancer-related deaths. IQ motif-containing GTPase-activating protein 1 (IQGAP1), a prominent regulator in the cancer metastasis process, is a scaffold protein that interacts with components of the cytoskeleton. As a critical node within the small GTPase network, IQGAP1 acts as a binding partner of several small GTPases, which in turn function as molecular switches to control most cellular processes, including cell migration and invasion. Given the significant interaction between IQGAP1 and small GTPases in cancer metastasis, we briefly elucidate the role of IQGAP1 in regulating cancer metastasis and the varied interactions existing between IQGAP1 and small GTPases. In addition, the potential regulators for IQGAP1 activity and its interaction with small GTPases are also incorporated in this review. Overall, we comprehensively summarize the role of IQGAP1 in cancer tumorigenicity and metastasis, which may be a potential anti-tumor target to restrain cancer progression.
Collapse
Affiliation(s)
- Xiafeng Peng
- Department of Gynecology and Obstetrics, Wuxi Maternal and Child Health Hospital, the Affiliated Hospital to Nanjing Medical University, Wuxi, 214023, China; First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China.
| | - Tiejun Wang
- Department of Gynecology and Obstetrics, Wuxi Maternal and Child Health Hospital, the Affiliated Hospital to Nanjing Medical University, Wuxi, 214023, China.
| | - Han Gao
- School of Medicine, Jiangnan University, Wuxi, 214122, China.
| | - Xin Yue
- First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China.
| | - Weiqi Bian
- First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China.
| | - Jie Mei
- Department of Gynecology and Obstetrics, Wuxi Maternal and Child Health Hospital, the Affiliated Hospital to Nanjing Medical University, Wuxi, 214023, China; Wuxi Clinical Medical College, Nanjing Medical University, Wuxi, 214023, China.
| | - Yan Zhang
- Department of Gynecology and Obstetrics, Wuxi Maternal and Child Health Hospital, the Affiliated Hospital to Nanjing Medical University, Wuxi, 214023, China.
| |
Collapse
|
24
|
Hu HF, Xu WW, Li YJ, He Y, Zhang WX, Liao L, Zhang QH, Han L, Yin XF, Zhao XX, Pan YL, Li B, He QY. Anti-allergic drug azelastine suppresses colon tumorigenesis by directly targeting ARF1 to inhibit IQGAP1-ERK-Drp1-mediated mitochondrial fission. Am J Cancer Res 2021; 11:1828-1844. [PMID: 33408784 PMCID: PMC7778598 DOI: 10.7150/thno.48698] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
This study aimed to screen novel anticancer strategies from FDA-approved non-cancer drugs and identify potential biomarkers and therapeutic targets for colorectal cancer (CRC). Methods: A library consisting of 1056 FDA-approved drugs was screened for anticancer agents. WST-1, colony-formation, flow cytometry, and tumor xenograft assays were used to determine the anticancer effect of azelastine. Quantitative proteomics, confocal imaging, Western blotting and JC-1 assays were performed to examine the effects on mitochondrial pathways. The target protein of azelastine was analyzed and confirmed by DARTS, WST-1, Biacore and tumor xenograft assays. Immunohistochemistry, gain- and loss-of-function experiments, WST-1, colony-formation, immunoprecipitation, and tumor xenograft assays were used to examine the functional and clinical significance of ARF1 in colon tumorigenesis. Results: Azelastine, a current anti-allergic drug, was found to exert a significant inhibitory effect on CRC cell proliferation in vitro and in vivo, but not on ARF1-deficient or ARF1-T48S mutant cells. ARF1 was identified as a direct target of azelastine. High ARF1 expression was associated with advanced stages and poor survival of CRC. ARF1 promoted colon tumorigenesis through its interaction with IQGAP1 and subsequent activation of ERK signaling and mitochondrial fission by enhancing the interaction of IQGAP1 with MEK and ERK. Mechanistically, azelastine bound to Thr-48 in ARF1 and repressed its activity, decreasing Drp1 phosphorylation. This, in turn, inhibited mitochondrial fission and suppressed colon tumorigenesis by blocking IQGAP1-ERK signaling. Conclusions: This study provides the first evidence that azelastine may be novel therapeutics for CRC treatment. ARF1 promotes colon tumorigenesis, representing a promising biomarker and therapeutic target in CRC.
Collapse
|
25
|
Nieuwenhuis B, Barber AC, Evans RS, Pearson CS, Fuchs J, MacQueen AR, van Erp S, Haenzi B, Hulshof LA, Osborne A, Conceicao R, Khatib TZ, Deshpande SS, Cave J, Ffrench‐Constant C, Smith PD, Okkenhaug K, Eickholt BJ, Martin KR, Fawcett JW, Eva R. PI 3-kinase delta enhances axonal PIP 3 to support axon regeneration in the adult CNS. EMBO Mol Med 2020; 12:e11674. [PMID: 32558386 PMCID: PMC7411663 DOI: 10.15252/emmm.201911674] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/27/2022] Open
Abstract
Peripheral nervous system (PNS) neurons support axon regeneration into adulthood, whereas central nervous system (CNS) neurons lose regenerative ability after development. To better understand this decline whilst aiming to improve regeneration, we focused on phosphoinositide 3-kinase (PI3K) and its product phosphatidylinositol (3,4,5)-trisphosphate (PIP3 ). We demonstrate that adult PNS neurons utilise two catalytic subunits of PI3K for axon regeneration: p110α and p110δ. However, in the CNS, axonal PIP3 decreases with development at the time when axon transport declines and regenerative competence is lost. Overexpressing p110α in CNS neurons had no effect; however, expression of p110δ restored axonal PIP3 and increased regenerative axon transport. p110δ expression enhanced CNS regeneration in both rat and human neurons and in transgenic mice, functioning in the same way as the hyperactivating H1047R mutation of p110α. Furthermore, viral delivery of p110δ promoted robust regeneration after optic nerve injury. These findings establish a deficit of axonal PIP3 as a key reason for intrinsic regeneration failure and demonstrate that native p110δ facilitates axon regeneration by functioning in a hyperactive fashion.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for NeuroscienceRoyal Netherlands Academy of Arts and Sciences (KNAW)AmsterdamThe Netherlands
| | - Amanda C Barber
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Rachel S Evans
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Craig S Pearson
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Joachim Fuchs
- Institute of BiochemistryCharité – Universitätsmedizin BerlinBerlinGermany
| | - Amy R MacQueen
- Laboratory of Lymphocyte Signalling and DevelopmentBabraham InstituteCambridgeUK
| | - Susan van Erp
- MRC Centre for Regenerative MedicineUniversity of EdinburghEdinburghUK
| | - Barbara Haenzi
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Lianne A Hulshof
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Andrew Osborne
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Raquel Conceicao
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Tasneem Z Khatib
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Sarita S Deshpande
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Joshua Cave
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | | | | | | | - Britta J Eickholt
- Institute of BiochemistryCharité – Universitätsmedizin BerlinBerlinGermany
| | - Keith R Martin
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Centre for Eye Research AustraliaRoyal Victorian Eye and Ear HospitalMelbourneVic.Australia
- OphthalmologyDepartment of SurgeryUniversity of MelbourneMelbourneVic.Australia
| | - James W Fawcett
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Centre of Reconstructive NeuroscienceInstitute of Experimental MedicineCzech Academy of SciencesPragueCzech Republic
| | - Richard Eva
- John Van Geest Centre for Brain RepairDepartment of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| |
Collapse
|
26
|
Lu C, Amin MA, Fox DA. CD13/Aminopeptidase N Is a Potential Therapeutic Target for Inflammatory Disorders. THE JOURNAL OF IMMUNOLOGY 2020; 204:3-11. [PMID: 31848300 DOI: 10.4049/jimmunol.1900868] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/01/2019] [Indexed: 02/05/2023]
Abstract
CD13/aminopeptidase N is a widely expressed ectoenzyme with multiple functions. As an enzyme, CD13 regulates activities of numerous cytokines by cleaving their N-terminals and is involved in Ag processing by trimming the peptides bound to MHC class II. Independent of its enzymatic activity, cell membrane CD13 functions by cross-linking-induced signal transduction, regulation of receptor recycling, enhancement of FcγR-mediated phagocytosis, and acting as a receptor for cytokines. Moreover, soluble CD13 has multiple proinflammatory roles mediated by binding to G-protein-coupled receptors. CD13 not only modulates development and activities of immune-related cells, but also regulates functions of inflammatory mediators. Therefore, CD13 is important in the pathogenesis of various inflammatory disorders. Inhibitors of CD13 have shown impressive anti-inflammatory effects, but none of them has yet been used for clinical therapy of human inflammatory diseases. We reevaluate CD13's regulatory role in inflammation and suggest that CD13 could be a potential therapeutic target for inflammatory disorders.
Collapse
Affiliation(s)
- Chenyang Lu
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109; and.,Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mohammad A Amin
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - David A Fox
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109; and
| |
Collapse
|
27
|
The Correct Localization of Borealin in Midbody during Cytokinesis Depends on IQGAP1. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6231697. [PMID: 32685508 PMCID: PMC7334785 DOI: 10.1155/2020/6231697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/31/2020] [Accepted: 06/08/2020] [Indexed: 11/17/2022]
Abstract
Borealin is a key component of chromosomal passenger complex, which is vital in cytokinesis. IQ domain-containing GTPase-activating protein 1 (IQGAP1) also participates in cytokinesis. The correlation between Borealin and IQGAP1 during cytokinesis is not yet clear. Here, we used mass spectrometry and endogenous coimmunoprecipitation experiments to investigate the interaction between IQGAP1 and Borealin. Results of the current study showed that Borealin interacted directly with IQGAP1 both in vitro and in vivo. Knockdown of IQGAP1 resulted in an abnormal location of Borealin in the midbody. Knocking down Borealin alone, IQGAP1 alone, or Borealin and IQGAP1 at the same time inhibited the completion of cytokinesis and formed multinucleated cells. Our results indicated that IQGAP1 interacts with Borealin during cytokinesis, and the correct localization of Borealin in the midbody during cytokinesis is determined by IQGAP1, and IQGAP1 may play an important role in regulating Borealin function in cytokinesis.
Collapse
|
28
|
Kadry YA, Calderwood DA. Chapter 22: Structural and signaling functions of integrins. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183206. [PMID: 31991120 PMCID: PMC7063833 DOI: 10.1016/j.bbamem.2020.183206] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 02/06/2023]
Abstract
The integrin family of transmembrane adhesion receptors is essential for sensing and adhering to the extracellular environment. Integrins are heterodimers composed of non-covalently associated α and β subunits that engage extracellular matrix proteins and couple to intracellular signaling and cytoskeletal complexes. Humans have 24 different integrin heterodimers with differing ligand binding specificities and non-redundant functions. Complex structural rearrangements control the ability of integrins to engage ligands and to activate diverse downstream signaling networks, modulating cell adhesion and dynamics, processes which are crucial for metazoan life and development. Here we review the structural and signaling functions of integrins focusing on recent advances which have enhanced our understanding of how integrins are activated and regulated, and the cytoplasmic signaling networks downstream of integrins.
Collapse
Affiliation(s)
- Yasmin A Kadry
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, United States of America
| | - David A Calderwood
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, United States of America; Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, United States of America..
| |
Collapse
|
29
|
Ghosh M, Shapiro LH. CD13 regulation of membrane recycling: implications for cancer dissemination. Mol Cell Oncol 2019; 6:e1648024. [PMID: 31692781 DOI: 10.1080/23723556.2019.1648024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 10/26/2022]
Abstract
Membrane recycling is critical to numerous cell functions and its dysregulation contributes to cancer and metastasis. We established that activation of the transmembrane molecule aminopeptidase N (ANPEP, also known as CD13) tethers the IQ motif containing, guanosine triphosphate hydrolase activating protein 1 (IQGAP1) scaffolding protein at the plasma membrane, thus stimulating the recycling regulator ADP-ribosylation factor 6 (ARF6) to ensure proper recycling of β1-integrin and other membrane components impacting cell attachment.
Collapse
Affiliation(s)
- Mallika Ghosh
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Linda H Shapiro
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|