1
|
Cui Z, Wang H, Feng X, Wu C, Yi M, He R, Pan T, Gao R, Feng L, Zeng B, Huang G, Wang Y, Du Y, Zhang CJ, Xiao X, Wang C. MYO1F regulates T-cell activation and glycolytic metabolism by promoting the acetylation of GAPDH. Cell Mol Immunol 2025; 22:176-190. [PMID: 39668163 PMCID: PMC11782582 DOI: 10.1038/s41423-024-01247-6] [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: 09/05/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024] Open
Abstract
Proper cellular metabolism in T cells is critical for a productive immune response. However, when dysregulated by intrinsic or extrinsic metabolic factors, T cells may contribute to a wide spectrum of diseases, such as cancers and autoimmune diseases. However, the metabolic regulation of T cells remains incompletely understood. Here, we show that MYO1F is required for human and mouse T-cell activation after TCR stimulation and that T-cell-specific Myo1f knockout mice exhibit an increased tumor burden and attenuated EAE severity due to impaired T-cell activation in vivo. Mechanistically, after TCR stimulation, MYO1F is phosphorylated by LCK at tyrosines 607 and 634, which is critical for glyceraldehyde-3-phosphate dehydrogenase (GAPDH) acetylation at Lys84, 86 and 227 mediated by α-TAT1, which is an acetyltransferase, and these processes are important for its activation, cellular glycolysis and thus the effector function of T cells. Importantly, we show that a fusion protein of VAV1-MYO1F, a recurrent peripheral T-cell lymphoma (PTCL)-associated oncogenic protein, promotes hyperacetylation of GAPDH and its activation, which leads to aberrant glycolysis and T-cell proliferation, and that inhibition of the activity of GAPDH significantly limits T-cell activation and proliferation and extends the survival of hVAV1-MYO1F knock-in mice. Moreover, hyperacetylation of GAPDH was confirmed in human PTCL patient samples containing the VAV1-MYO1F gene fusion. Overall, this study revealed not only the mechanisms by which MYO1F regulates T-cell metabolism and VAV1-MYO1F fusion-induced PTCL but also promising therapeutic targets for the treatment of PTCL.
Collapse
Affiliation(s)
- Zhihui Cui
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074; Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Heping Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074; Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiong Feng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074; Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chuyu Wu
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ming Yi
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ruirui He
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ting Pan
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ru Gao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074; Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lingyun Feng
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Bo Zeng
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Guoling Huang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yuan Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yanyun Du
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China.
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| | - Cun-Jin Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Xue Xiao
- Department of Pathology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Chenhui Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China.
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Grundtner S, Sondermann JR, Xian F, Malzl D, Segelcke D, Pogatzki-Zahn EM, Menche J, Gómez-Varela D, Schmidt M. Deep proteomics and network pharmacology reveal sex- and age-shared neuropathic pain signatures in mouse dorsal root ganglia. Pharmacol Res 2025; 211:107552. [PMID: 39694124 DOI: 10.1016/j.phrs.2024.107552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Our understanding of how sex and age influence chronic pain at the molecular level is still limited with wide-reaching consequences for adolescent patients. Here, we leveraged deep proteome profiling of mouse dorsal root ganglia (DRG) from adolescent (4-week-old) and adult (12-week-old) male and female mice to investigate the establishment of neuropathic pain in the spared nerve injury (SNI)-model in parallel. We quantified over 12,000 proteins, including notable ion channels involved in pain, highlighting the sensitivity of our approach. Differential expression revealed sex- and age-dependent proteome changes upon nerve injury. In contrast to most previous studies, our comprehensive dataset enabled us to determine differentially expressed proteins (DEPs), which were shared between male and female mice of both age groups. Among these, the vast majority (94 %) were also expressed and, in part, altered in human DRG of neuropathic pain patients, indicating evolutionary conservation. Proteome signatures represented numerous targets of FDA-approved drugs comprising both (i) known pain therapeutics (e.g. Pregabalin and opioids) and, importantly, (ii) compounds with high potential for future re-purposing, e.g. Ptprc-modulators and Epoetins. Protein network and multidimensional analysis uncovered distinct hubs of sex- and age-shared biological pathways impacted by neuropathic pain, such as neuronal activity and synaptic function, DNA-damage, and neuroimmune interactions. Taken together, our results capture the complexity of nerve injury-associated DRG alterations in mice at the network level, moving beyond single-candidate studies. Consequently, we provide an innovative resource of the molecular landscape of neuropathic pain, enabling novel opportunities for translational pain research and network-based drug discovery.
Collapse
Affiliation(s)
- Sabrina Grundtner
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Julia R Sondermann
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Feng Xian
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Daniel Malzl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Department of Structural and Computational Biology, Center for Molecular Biology, University of Vienna, Vienna, Austria
| | - Daniel Segelcke
- Clinic for Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Muenster, Germany
| | - Esther M Pogatzki-Zahn
- Clinic for Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Muenster, Germany
| | - Jörg Menche
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Department of Structural and Computational Biology, Center for Molecular Biology, University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Network Medicine at the University of Vienna, Vienna, Austria; Faculty of Mathematics, University of Vienna, Vienna, Austria
| | - David Gómez-Varela
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Manuela Schmidt
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Yan K, Zhang Y, Li Y, Tang M, Xu Y, Yan X, Hu J, Wang Y. Immune Response of Silver Pomfret (Pampus argenteus) CC Chemokine Ligand Gene Family to Photobacterium damselae Subsp. Damselae and Nocardia seriolae Infections. JOURNAL OF FISH DISEASES 2025; 48:e14032. [PMID: 39440715 DOI: 10.1111/jfd.14032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/14/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Chemokines play a crucial role in immune responses by facilitating the migration of cells expressing corresponding chemokine receptors along concentration gradients. Photobacterium damselae subsp. Damselae (PDD) and Nocardia seriolae (NS) are known to induce substantial mortality in silver pomfret populations, yet there exists a dearth of research regarding the immune response of CCLs in PDD- or NS-infected silver pomfret. In our investigation, we identified 10 PaCCLs, which include one fish-specific CCL (PaCCL44). Phylogenetic analysis revealed considerable diversity in CCL types and copy numbers among various teleost fishes. Notably, silver pomfret lacks specific CCL genes, with most PaCCLs exhibiting heightened expression levels in immune-related organs such as the spleen and kidney, and some being expressed in mucosal immune-related organs like the skin and gills. Transcriptome analysis conducted on silver pomfret infected with NS and PDD elucidated that the expression changes of PaCCLs primarily manifested in the spleen during the initial stages of NS infection, shifting to the kidney in later stages. Conversely, the expression changes of PaCCLs following PDD infection predominantly occurred in the kidney. In vitro studies using silver pomfret spleen cell lines demonstrated an early peak in PaCCLs expression during infection, followed by gradual decline with NS treatment and rapid diminishment with PDD treatment. These findings suggest that PaCCLs primarily support the innate immunity of silver pomfret, potentially exhibiting chemotactic effects in the early infection stages, such as the synergistic action of PaCCL4 and PaCCL25, and later serving as direct antibacterial agents. NS invasion is characterised by a chronic infection affecting multiple organs, whereas PDD primarily inflicts severe damage to the kidney. PaCCL19a and PaCCL19b are specific to PDD, and their expression levels may decrease in the later stages of infection due to PDD immune escape. These data offer initial insights into understanding the mechanism underlying the innate immune response of the CCL gene family in silver pomfret and provide theoretical underpinnings for fish culture practices.
Collapse
Affiliation(s)
- Kaiheng Yan
- College of Marine Sciences, Ningbo University, Ningbo, China
- Institute of Marine Science and Technology, Shandong University, Qingdao, China
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
| | - Youyi Zhang
- College of Marine Sciences, Ningbo University, Ningbo, China
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
| | - Yuanbo Li
- College of Marine Sciences, Ningbo University, Ningbo, China
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
| | - Mengke Tang
- College of Marine Sciences, Ningbo University, Ningbo, China
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
| | - Yifan Xu
- Institute of Marine Science and Technology, Shandong University, Qingdao, China
| | - Xiaojun Yan
- College of Marine Sciences, Ningbo University, Ningbo, China
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
| | - Jiabao Hu
- College of Marine Sciences, Ningbo University, Ningbo, China
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
| | - Yajun Wang
- College of Marine Sciences, Ningbo University, Ningbo, China
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
| |
Collapse
|
4
|
Zhang Y, Ren Y, Zhou T, Qian Z, Bao Z. Vav family exchange factors: Potential regulator in atherosclerosis. Biochem Biophys Rep 2024; 40:101878. [PMID: 39649800 PMCID: PMC11625217 DOI: 10.1016/j.bbrep.2024.101878] [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: 08/02/2024] [Revised: 10/26/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024] Open
Abstract
The Vav family of guanosine nucleotide exchange factors (GEFs) regulates the phosphorylation of tyrosinase, influencing various physiological and pathological processes by modulating the binding of Rho GTPases to GDP/GTP. Recent research has highlighted the critical role of Vav family activation in tumorigenesis, neurological disorders, immune-related dysfunctions, and other diseases. This review offers a comprehensive overview of the structure and function of Vav proteins and their significant impact on the pathophysiology of atherosclerosis. In addition, we pay attention to the development of diagnostic and therapeutic targets centered around Vav proteins.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, China
| | - Yongwei Ren
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, China
| | - Tao Zhou
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, China
| | - Zhengtao Qian
- Department of Clinical Laboratory, Changshu Medicine Examination Institute, Changshu, 215500, China
| | - Zhengyang Bao
- Department of Internal Medicine, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, China
| |
Collapse
|
5
|
Ma Y, Dumesny C, Dong L, Ang CS, Asadi K, Zhan Y, Nikfarjam M, He H. Inhibition of P21-activated kinases 1 and 4 synergistically suppresses the growth of pancreatic cancer by stimulating anti-tumour immunity. Cell Commun Signal 2024; 22:287. [PMID: 38797819 PMCID: PMC11129409 DOI: 10.1186/s12964-024-01670-2] [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: 02/21/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal types of cancer, and KRAS oncogene occurs in over 90% of cases. P21-activated kinases (PAK), containing six members (PAK1 to 6), function downstream of KRAS. PAK1 and PAK4 play important roles in carcinogenesis, but their combinational effect remains unknown. In this study, we have determined the effect of dual inhibition of PAK1 and PAK4 in PDA progression using knockout (KO) cancer cell lines. METHODS Murine wild-type (WT) and PAK1KO pancreatic cancer cell lines were isolated from PAK1+/+ and PAK1-/- KPC (LSL-KrasG12D/+; LSL-Trp53 R172H/+; Pdx-1-Cre) mice. KPC PAK4KO and KPC PAK1&4 KO cell lines were generated from KPC WT and KPC PAK1KO cell lines respectively using the CRISPR-CAS9 gene knockout technique. PAK WT and KO cell lines were used in mouse models of pancreatic tumours. Cells and tumour tissue were also used in flow cytometry and proteomic studies. A human PDA tissue microarray was stained by immunohistochemistry. RESULTS Double knock out of PAK1 and PAK4 caused complete regression of tumour in a syngeneic mouse model. PAK4KO inhibited tumour growth by stimulating a rapid increase of cytotoxic CD8+ T cell infiltration. PAK1KO synergistically with PAK4KO increased cytotoxic CD8+ T cell infiltration and stimulated a sustained infiltration of CD8+ T cells at a later phase to overcome the immune evasion in the PAK4KO tumour. The human PDA tissue microarray study showed the important role of PAK1 and PAK4 in intra-tumoral T-cell function. CONCLUSION Our results demonstrated that dual inhibition of PAK1 and PAK4 synergistically suppressed PDA progression by stimulating cytotoxic CD8 + T cell response.
Collapse
Affiliation(s)
- Yi Ma
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
- Department of General Surgery, Monash Health, Clayton, VIC, Australia
| | - Chelsea Dumesny
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
| | - Li Dong
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
| | - Ching-Seng Ang
- Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Khashayar Asadi
- Department of Anatomical Pathology, Austin Health, Heidelberg, VIC, Australia
| | - Yifan Zhan
- Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Mehrdad Nikfarjam
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia
- Department of Hepato-Pancreato-Biliary Surgery, Austin Health, Heidelberg, VIC, Australia
| | - Hong He
- Department of Surgery, Austin Precinct, University of Melbourne, Level 8, Lance Townsend Building, Austin Hospital, 145 Studley Road, Heidelberg, VIC, Australia.
| |
Collapse
|
6
|
Marrocco R, Bernard I, Joulia E, Barascud R, Dejean AS, Lesourne R, Saoudi A. Positive regulation of Vav1 by Themis controls CD4 T cell pathogenicity in a mouse model of central nervous system inflammation. Cell Mol Life Sci 2024; 81:161. [PMID: 38565808 PMCID: PMC10987373 DOI: 10.1007/s00018-024-05203-5] [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: 10/13/2023] [Revised: 02/15/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024]
Abstract
The susceptibility to autoimmune diseases is conditioned by the association of modest genetic alterations which altogether weaken self-tolerance. The mechanism whereby these genetic interactions modulate T-cell pathogenicity remains largely uncovered. Here, we investigated the epistatic interaction of two interacting proteins involved in T Cell Receptor signaling and which were previously associated with the development of Multiple Sclerosis. To this aim, we used mice expressing an hypomorphic variant of Vav1 (Vav1R63W), combined with a T cell-conditional deletion of Themis. We show that the combined mutations in Vav1 and Themis induce a strong attenuation of the severity of Experimental Autoimmune Encephalomyelitis (EAE), contrasting with the moderate effect of the single mutation in each of those two proteins. This genotype-dependent gradual decrease of EAE severity correlates with decreased quantity of phosphorylated Vav1 in CD4 T cells, establishing that Themis promotes the development of encephalitogenic Tconv response by enhancing Vav1 activity. We also show that the cooperative effect of Themis and Vav1 on EAE severity is independent of regulatory T cells and unrelated to the impact of Themis on thymic selection. Rather, it results from decreased production of pro-inflammatory cytokines (IFN-γ, IL-17, TNF and GM-CSF) and reduced T cell infiltration in the CNS. Together, our results provide a rationale to study combination of related genes, in addition to single gene association, to better understand the genetic bases of human diseases.
Collapse
Affiliation(s)
- Remi Marrocco
- Institut Toulousain des Maladies Infectieuses Et Inflammatoires (Infinity), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), INSERM U1291, Université Paul Sabatier (UPS), CHU Purpan, BP 3028, 31024, Toulouse Cedex 3, France
- Division of Immune Regulation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Isabelle Bernard
- Institut Toulousain des Maladies Infectieuses Et Inflammatoires (Infinity), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), INSERM U1291, Université Paul Sabatier (UPS), CHU Purpan, BP 3028, 31024, Toulouse Cedex 3, France
| | - Emeline Joulia
- Institut Toulousain des Maladies Infectieuses Et Inflammatoires (Infinity), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), INSERM U1291, Université Paul Sabatier (UPS), CHU Purpan, BP 3028, 31024, Toulouse Cedex 3, France
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Rebecca Barascud
- Institut Toulousain des Maladies Infectieuses Et Inflammatoires (Infinity), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), INSERM U1291, Université Paul Sabatier (UPS), CHU Purpan, BP 3028, 31024, Toulouse Cedex 3, France
| | - Anne S Dejean
- Institut Toulousain des Maladies Infectieuses Et Inflammatoires (Infinity), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), INSERM U1291, Université Paul Sabatier (UPS), CHU Purpan, BP 3028, 31024, Toulouse Cedex 3, France
| | - Renaud Lesourne
- Institut Toulousain des Maladies Infectieuses Et Inflammatoires (Infinity), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), INSERM U1291, Université Paul Sabatier (UPS), CHU Purpan, BP 3028, 31024, Toulouse Cedex 3, France
| | - Abdelhadi Saoudi
- Institut Toulousain des Maladies Infectieuses Et Inflammatoires (Infinity), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), INSERM U1291, Université Paul Sabatier (UPS), CHU Purpan, BP 3028, 31024, Toulouse Cedex 3, France.
| |
Collapse
|
7
|
Sun M, Wei Y, Zhang C, Nian H, Du B, Wei R. Integrated DNA Methylation and Transcriptomics Analyses of Lacrimal Glands Identify the Potential Genes Implicated in the Development of Sjögren's Syndrome-Related Dry Eye. J Inflamm Res 2023; 16:5697-5714. [PMID: 38050559 PMCID: PMC10693829 DOI: 10.2147/jir.s440263] [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: 09/15/2023] [Accepted: 11/21/2023] [Indexed: 12/06/2023] Open
Abstract
Purpose Sjögren's syndrome-related dry eye (SS-related dry eye) is an intractable autoimmune disease characterized by chronic inflammation of lacrimal glands (LGs), where epigenetic factors are proven to play a crucial role in the pathogenesis of this disease. However, the alteration of DNA methylation in LGs and its role in the pathogenesis of SS-related dry eye is still unknown. Here, we performed an integrated analysis of DNA methylation and RNA-Seq data in LGs to identify novel DNA methylation-regulated differentially expressed genes (MeDEGs) in the pathogenesis of SS-related dry eye. Methods The DNA methylation and transcription profiles of LGs in NOD mice at different stages of SS-related dry eye (4-, 8-, 12- and 16 weeks old) were generated by reduced representation bisulfite sequencing (RRBS) and RNA-Seq. The differentially methylated genes (DMGs) and differentially expressed genes (DEGs) were analyzed by MethylKit R package and edgeR. Correlation analysis between methylation level and mRNA expression was conducted with R software. The functional correlation of DMGs and DEGs was analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Finally, LG tissues from another litter of NOD mice were collected for methylation-specific polymerase chain reaction (MSP) and quantitative real-time PCR (qRT-PCR) to validate the methylation and expression levels of key genes. CD4+ cell infiltration of LGs was detected by immunofluorescence staining. Results Hypermethylation of LGs was identified in NOD mice with the progression of SS-related dry eye and the DMGs were mainly enriched in the GTPases activation and Ras signaling pathway. RNA-seq analysis revealed 1321, 2549, and 3712 DEGs in the 8-, 12- and 16-week-old NOD mice compared with 4-week-old normal control mice. For GO analysis, the DEGs were mainly enriched in T cell immune responses. Further, a total of 140 MeDEGs were obtained by integrated analysis of methylome and transcriptome, which were primarily enriched in T cell activation, proliferation and differentiation. Based on the main GO terms and KEGG pathways of MeDEGs, 8 genes were screened out. The expression levels of these key genes, especially Itgal, Vav1, Irf4 and Icosl, were verified to elevate after the onset of SS-related dry eye in NOD mice and positively correlated with the extent of inflammatory cell infiltration in LGs. Immunofluorescence assay revealed that CD4+ cell infiltration dramatically increased in LGs of SS-related dry eye mice compared with the control mice. And the expression levels of four genes showed significantly positive correlation with the extent of CD4+ cell infiltration in LGs. MSP showed the hypomethylation of the Irf4 and Itgal promoters in NOD mice with SS-related dry eye compared to control group. Conclusion Our study revealed the critical role of epigenetic regulation of T cell immunity-related genes in the progression of SS-related dry eye and reminded us that DNA methylation-regulated genes such as Itgal, Vav1, Irf4 and Icosl may be used as new targets for SS-related dry eye therapy.
Collapse
Affiliation(s)
- Mei Sun
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People’s Republic of China
| | - Yankai Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People’s Republic of China
| | - Chengyuan Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People’s Republic of China
| | - Hong Nian
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People’s Republic of China
| | - Bei Du
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People’s Republic of China
| | - Ruihua Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People’s Republic of China
| |
Collapse
|
8
|
Abdelmoaty MM, Yeapuri P, Machhi J, Lu Y, Namminga KL, Kadry R, Lu E, Bhattarai S, Mosley RL, Gendelman HE. Immune senescence in aged APP/PS1 mice. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:317-330. [PMID: 38023614 PMCID: PMC10659760 DOI: 10.1515/nipt-2023-0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 12/01/2023]
Abstract
Objectives To evaluate the linkage between age and deficits in innate and adaptive immunity which heralds both Alzheimer's disease (AD) onset and progression. The pathobiological events which underlie and tie these outcomes remain not fully understood. Methods To investigate age-dependent immunity in AD, we evaluated innate and adaptive immunity in coordinate studies of regulatory T cell (Treg) function, T cell frequencies, and microglial integrity. These were assessed in blood, peripheral lymphoid tissues, and the hippocampus of transgenic (Tg) amyloid precursor protein/presenilin 1 (APP/PS1) against non-Tg mice. Additionally, immune arrays of hippocampal tissue were performed at 4, 6, 12, and 20 months of age. Results APP/PS1 mice showed progressive impairment of Treg immunosuppressive function with age. There was partial restoration of Treg function in 20-month-old mice. Ingenuity pathway analyses of hippocampal tissues were enriched in inflammatory, oxidative, and cellular activation pathways that paralleled advancing age and AD-pathobiology. Operative genes in those pathways included, but were not limited to triggering receptor on myeloid cells 1 (TREM1), T helper type 1 (Th1), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathways. Interleukin-17 (IL-17), nitric oxide, acute phase, and T cell receptor signaling pathways were also perturbed. Significant inflammation was observed at 6- and 12-months. However, at 20-months, age associated partial restoration of Treg function reduced inflammatory phenotype. Conclusions Impaired Treg function, inflammation and oxidative stress were associated with AD pathology. Age associated partial restoration of Treg function in old mice reduced the hippocampal inflammatory phenotype. Restoring Treg suppressive function can be a therapeutic modality for AD.
Collapse
Affiliation(s)
- Mai M. Abdelmoaty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Krista L. Namminga
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rana Kadry
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eugene Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shaurav Bhattarai
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rodney Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
9
|
Rump A, Ratas K, Lepasepp TK, Suurväli J, Smolander OP, Gross-Paju K, Toomsoo T, Kanellopoulos J, Rüütel Boudinot S. Sex-dependent expression levels of VAV1 and P2X7 in PBMC of multiple sclerosis patients. Scand J Immunol 2023; 98:e13283. [PMID: 38441379 DOI: 10.1111/sji.13283] [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: 10/15/2022] [Revised: 01/21/2023] [Accepted: 05/01/2023] [Indexed: 03/07/2024]
Abstract
Multiple sclerosis (MS) is an inflammatory autoimmune disorder of the central nervous system and the leading cause of progressive neurological disability in young adults. It decreases the patient's lifespan by about 10 years and affects women more than men. No medication entirely restricts or reverses neurological degradation. However, early diagnosis and treatment increase the possibility of a better outcome. To identify new MS biomarkers, we tested the expression of six potential markers (P2X4, P2X7, CXCR4, RGS1, RGS16 and VAV1) using qPCR in peripheral blood mononuclear cells (PBMC) of MS patients treated with interferon β (IFNβ), with glatiramer acetate (GA) or untreated. We showed that P2X7 and VAV1 are significantly induced in MS patients. In contrast, the expression of P2X4, CXCR4, RGS1 and RGS16 was not significantly modified by MS in PBMC. P2X7 and VAV1 are essentially induced in female patients, suggesting these markers are connected to sex-specific mechanisms. Strikingly, VAV1 expression is higher in healthy women than healthy men and IFNβ treatment of MS reduced VAV1 expression in female MS patients while it up-regulated VAV1 in male MS patients. Our data point to the differential, sex-dependent value of MS markers and treatment effects. Although rgs16 expression in PBMC was not a valid MS marker in patients, the strong upregulation of P2X4 and P2X7 induced in the spinal cord of WT mice by EAE was abrogated in rgs16KO mice suggesting that rgs16 is required for P2X4 and P2X7 induction by neurological diseases.
Collapse
Affiliation(s)
- Airi Rump
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Kristel Ratas
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Synlab, Tallinn, Estonia
| | - Tuuli Katarina Lepasepp
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Jaanus Suurväli
- Department of Biological Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Olli-Pekka Smolander
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Katrin Gross-Paju
- Department of Health Technologies, School of Information Technologies, Tallinn University of Technology, Tallinn, Estonia
- West-Tallinn Central Hospital, Tallinn, Estonia
| | - Toomas Toomsoo
- School of Natural Sciences and Health, Tallinn University, Tallinn, Estonia
- Confido Medical Center, Tallinn, Estonia
| | - Jean Kanellopoulos
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, France
| | - Sirje Rüütel Boudinot
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| |
Collapse
|
10
|
Shalom B, Salaymeh Y, Risling M, Katzav S. Unraveling the Oncogenic Potential of VAV1 in Human Cancer: Lessons from Mouse Models. Cells 2023; 12:cells12091276. [PMID: 37174676 PMCID: PMC10177506 DOI: 10.3390/cells12091276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
VAV1 is a hematopoietic signal transducer that possesses a GDP/GTP nucleotide exchange factor (GEF) that is tightly regulated by tyrosine phosphorylation, along with adapter protein domains, such as SH2 and SH3. Research on VAV1 has advanced over the years since its discovery as an in vitro activated oncogene in an NIH3T3 screen for oncogenes. Although the oncogenic form of VAV1 first identified in the screen has not been detected in human clinical tumors, its wild-type and mutant forms have been implicated in mammalian malignancies of various tissue origins, as well as those of the hematopoietic system. This review article addresses the activity of human VAV1 as an overexpressed or mutated gene and also describes the differences in the distribution of VAV1 mutations in the hematopoietic system and in other tissues. The knowledge accumulated thus far from GEMMs expressing VAV1 is described, with the conclusion that GEMMs of both wild-type VAV1 and mutant VAV1 do not form tumors, yet these will be generated when additional molecular insults, such as loss of p53 or KRAS mutation, occur.
Collapse
Affiliation(s)
- Batel Shalom
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
| | - Yaser Salaymeh
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
| | - Matan Risling
- Department of Military Medicine and "Tzameret", Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
- Medical Corps, Israel Defense Forces, Tel-Hashomer 02149, Israel
| | - Shulamit Katzav
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
| |
Collapse
|
11
|
Castellanos-Martínez R, León-Vega II, Guerrero-Fonseca IM, Vargas-Robles H, Jiménez-Camacho KE, Hernández-Galicia G, Ortiz-Navarrete VF, Rottner K, Medina-Contreras O, Schnoor M. T cell functions and organ infiltration by leukemic T cells require cortactin. J Leukoc Biol 2023; 113:315-325. [PMID: 36808495 DOI: 10.1093/jleuko/qiad001] [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: 11/07/2022] [Indexed: 01/21/2023] Open
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that is still fatal in many cases. T cell blasts are characterized by hyperactivation and strong proliferative and migratory capacities. The chemokine receptor CXCR4 is involved in mediating malignant T cell properties, and cortactin has been shown to control CXCR4 surface localization in T-ALL cells. We have previously shown that cortactin overexpression is correlated with organ infiltration and relapse in B-ALL. However, the role of cortactin in T cell biology and T-ALL remains elusive. Here, we analyzed the functional relevance of cortactin for T cell activation and migration and the implications for T-ALL development. We found that cortactin is upregulated in response to T cell receptor engagement and recruited to the immune synapse in normal T cells. Loss of cortactin caused reduced IL-2 production and proliferation. Cortactin-depleted T cells showed defects in immune synapse formation and migrated less due to impaired actin polymerization in response to T cell receptor and CXCR4 stimulation. Leukemic T cells expressed much higher levels of cortactin compared to normal T cells that correlated with greater migratory capacity. Xenotransplantation assays in NSG mice revealed that cortactin-depleted human leukemic T cells colonized the bone marrow significantly less and failed to infiltrate the central nervous system, suggesting that cortactin overexpression drives organ infiltration, which is a major complication of T-ALL relapse. Thus, cortactin could serve as a potential therapeutic target for T-ALL and other pathologies involving aberrant T cell responses.
Collapse
Affiliation(s)
- Ramón Castellanos-Martínez
- Department of Molecular Biomedicine, CINVESTAV-IPN, Av. IPN 2508, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - Iliana I León-Vega
- Department of Molecular Biomedicine, CINVESTAV-IPN, Av. IPN 2508, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - Idaira M Guerrero-Fonseca
- Department of Molecular Biomedicine, CINVESTAV-IPN, Av. IPN 2508, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - Hilda Vargas-Robles
- Department of Molecular Biomedicine, CINVESTAV-IPN, Av. IPN 2508, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - Karina E Jiménez-Camacho
- Department of Molecular Biomedicine, CINVESTAV-IPN, Av. IPN 2508, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - Gabriela Hernández-Galicia
- Department of Molecular Biomedicine, CINVESTAV-IPN, Av. IPN 2508, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - Vianney F Ortiz-Navarrete
- Department of Molecular Biomedicine, CINVESTAV-IPN, Av. IPN 2508, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, TU Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Oscar Medina-Contreras
- Department of Hemato-oncology, Hospital Infantil "Federico Gómez,", Av. Dr. Márquez 162, 06720 Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine, CINVESTAV-IPN, Av. IPN 2508, San Pedro Zacatenco, 07360 Mexico City, Mexico
| |
Collapse
|
12
|
Morrish E, Wartewig T, Kratzert A, Rosenbaum M, Steiger K, Ruland J. The fusion oncogene VAV1-MYO1F triggers aberrant T-cell receptor signaling in vivo and drives peripheral T-cell lymphoma in mice. Eur J Immunol 2023; 53:e2250147. [PMID: 36541400 DOI: 10.1002/eji.202250147] [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: 08/22/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
VAV1-MYO1F is a recently identified gain-of-function fusion protein of the proto-oncogene Vav guanine nucleotide exchange factor 1 (VAV1) that is recurrently detected in T-cell non-Hodgkin's lymphoma (T-NHL) patients. However, the pathophysiological functions of VAV1-MYO1F in lymphomagenesis are insufficiently defined. Therefore, we generated transgenic mouse models to conditionally express VAV1-MYO1F in T-cells in vivo. We demonstrate that VAV1-MYO1F triggers cell autonomous activation of T-cell signaling with an activation of the ERK, JNK, and AKT pathways. VAV1-MYO1F expression induces a T-cell activation phenotype with high surface expression of CD25, ICOS, CD44, PD-1, and decreased CD62L as well as aberrant T-cell differentiation, proliferation, and neoplastic transformation. Consequently, the VAV1-MYO1F expressing T-cells induce a malignant T lymphoproliferative disease with 100% penetrance in vivo that mimics key aspects of human peripheral T-cell lymphoma. These results demonstrate that the human T-cell oncogene VAV1-MYO1F is sufficient to trigger oncogenic T-cell signaling and neoplastic transformation, and moreover, it provides a new clinically relevant mouse model to explore the pathogenesis of and treatment concepts for human T-cell lymphoma.
Collapse
Affiliation(s)
- Emma Morrish
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Tim Wartewig
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
- Center of Molecular and Cellular Oncology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Andreas Kratzert
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Marc Rosenbaum
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Katja Steiger
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Comparative Experimental Pathology, Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| |
Collapse
|
13
|
Moringa oleifera leaf attenuate osteoporosis in ovariectomized rats by modulating gut microbiota composition and MAPK signaling pathway. Biomed Pharmacother 2023; 161:114434. [PMID: 36841025 DOI: 10.1016/j.biopha.2023.114434] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Moringa oleifera leaf (MLP) contains abundant complex nutrients with anti-osteoporosis potential. However, its efficacy and mechanisms against osteoporosis remain unknown. The purpose of this research is to investigate MLP's anti-osteoporotic effects and mechanisms. Animal experiments were used in this work to validate MLP's anti-osteoporotic efficacy. We investigated the mode of action of MLP, analyzed its impact on the gut microbiota, and predicted and validated its anti-osteoporosis-related molecular targets and pathways through network pharmacology, molecular docking, and western blotting. In an ovariectomized osteoporosis rat model, MLP significantly increased bone mineral density and improved bone metabolism-related indicators, bone microstructure, and lipid profile. Moreover, it improved gut microbiota composition and increased the expression of Occludin and Claudin-1 protein in the duodenum. Network pharmacology identified a total of 97 active ingredients and 478 core anti-osteoporosis targets. Of these, MAPK1 (also known as ERK2), MAPK3 (also known as ERK1), and MAPK8 (also known as JNK) were successfully docked with the active constituents of MLP. Interestingly, MLP increased ERK and VAV3 protein expression and decreased p-ERK and JNK protein expression in the femur. These findings confirm MLP's anti-osteoporotic efficacy, which could be mediated via regulation of gut microbiota and MAPK signaling.
Collapse
|
14
|
Hu L, Zhang X, Li H, Lin S, Zang S. Targeting TET2 as a Therapeutic Approach for Angioimmunoblastic T Cell Lymphoma. Cancers (Basel) 2022; 14:cancers14225699. [PMID: 36428791 PMCID: PMC9688210 DOI: 10.3390/cancers14225699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/22/2022] Open
Abstract
Angioimmunoblastic T-cell lymphoma (AITL), a type of malignant lymphoma with unique genomic aberrations, significant clinicopathological features, and poor prognosis, is characterized by immune system dysregulation. Recent sequencing studies have identified recurrent mutations and interactions in tet methylcytosine dioxygenase 2 (TET2), ras homology family member A (RHOA), DNA methyltransferase 3 alpha (DNMT3A), and mitochondrial isocitrate dehydrogenase II (IDH2). Notably, since B-cell lymphomas are frequently observed along with AITL, this review first summarizes its controversial mechanisms based on traditional and recent views. Epigenetic regulation represented by TET2 plays an increasingly important role in understanding the multi-step and multi-lineage tumorigenesis of AITL, providing new research directions and treatment strategies for patients with AITL. Here, we review the latest advances in our understanding of AITL and highlight relevant issues that have yet to be addressed in clinical practice.
Collapse
Affiliation(s)
- Lina Hu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xuanye Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Huifeng Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Suxia Lin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shengbing Zang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Correspondence: ; Tel.: +86-13559131526
| |
Collapse
|
15
|
Jie X, Wu H, Yang M, He M, Zhao G, Ling S, Huang Y, Yue B, Yang N, Zhang X. Whole genome bisulfite sequencing reveals DNA methylation roles in the adaptive response of wildness training giant pandas to wild environment. Front Genet 2022; 13:995700. [PMID: 36303550 PMCID: PMC9592921 DOI: 10.3389/fgene.2022.995700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
DNA methylation modification can regulate gene expression without changing the genome sequence, which helps organisms to rapidly adapt to new environments. However, few studies have been reported in non-model mammals. Giant panda (Ailuropoda melanoleuca) is a flagship species for global biodiversity conservation. Wildness and reintroduction of giant pandas are the important content of giant pandas’ protection. However, it is unclear how wildness training affects the epigenetics of giant pandas, and we lack the means to assess the adaptive capacity of wildness training giant pandas. We comparatively analyzed genome-level methylation differences in captive giant pandas with and without wildness training to determine whether methylation modification played a role in the adaptive response of wildness training pandas. The whole genome DNA methylation sequencing results showed that genomic cytosine methylation ratio of all samples was 5.35%–5.49%, and the methylation ratio of the CpG site was the highest. Differential methylation analysis identified 544 differentially methylated genes (DMGs). The results of KEGG pathway enrichment of DMGs showed that VAV3, PLCG2, TEC and PTPRC participated in multiple immune-related pathways, and may participate in the immune response of wildness training giant pandas by regulating adaptive immune cells. A large number of DMGs enriched in GO terms may also be related to the regulation of immune activation during wildness training of giant pandas. Promoter differentially methylation analysis identified 1,199 genes with differential methylation at promoter regions. Genes with low methylation level at promoter regions and high expression such as, CCL5, P2Y13, GZMA, ANP32A, VWF, MYOZ1, NME7, MRPS31 and TPM1 were important in environmental adaptation for wildness training giant pandas. The methylation and expression patterns of these genes indicated that wildness training giant pandas have strong immunity, blood coagulation, athletic abilities and disease resistance. The adaptive response of giant pandas undergoing wildness training may be regulated by their negatively related promoter methylation. We are the first to describe the DNA methylation profile of giant panda blood tissue and our results indicated methylation modification is involved in the adaptation of captive giant pandas when undergoing wildness training. Our study also provided potential monitoring indicators for the successful reintroduction of valuable and threatened animals to the wild.
Collapse
Affiliation(s)
- Xiaodie Jie
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Honglin Wu
- Key Laboratory of State Forestry and Grassland Administration on Conservation Biology of Rare Animals in the Giant Panda National Park, China Conservation and Research Center for the Giant Panda, Dujiangyan, China
| | - Miao Yang
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Ming He
- Key Laboratory of State Forestry and Grassland Administration on Conservation Biology of Rare Animals in the Giant Panda National Park, China Conservation and Research Center for the Giant Panda, Dujiangyan, China
| | - Guangqing Zhao
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Shanshan Ling
- Key Laboratory of State Forestry and Grassland Administration on Conservation Biology of Rare Animals in the Giant Panda National Park, China Conservation and Research Center for the Giant Panda, Dujiangyan, China
| | - Yan Huang
- Key Laboratory of State Forestry and Grassland Administration on Conservation Biology of Rare Animals in the Giant Panda National Park, China Conservation and Research Center for the Giant Panda, Dujiangyan, China
| | - Bisong Yue
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Nan Yang
- Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, China
- *Correspondence: Nan Yang, ; Xiuyue Zhang,
| | - Xiuyue Zhang
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, China
- *Correspondence: Nan Yang, ; Xiuyue Zhang,
| |
Collapse
|
16
|
Vav1 accelerates Ras-driven lung cancer and modulates its tumor microenvironment. Cell Signal 2022; 97:110395. [PMID: 35752351 DOI: 10.1016/j.cellsig.2022.110395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 11/23/2022]
Abstract
The potential impact of Vav1 on human cancer was only recently acknowledged, as it is detected as a mutant or an overexpressed gene in various cancers, including lung cancer. Vav1, which is normally and exclusively expressed in the hematopoietic system functions as a specific GDP/GTP nucleotide exchange factor (GEF), strictly regulated by tyrosine phosphorylation. To investigate whether Vav1 plays a causative or facilitating role in-vivo in lung cancer development and to examine whether it co-operates with other oncogenes, such as mutant K-Ras, we generated novel mouse strains that express: Vav1 or K-RasG12D in type II pneumocytes, as well as a transgenic mouse line that expresses both Vav1 and K-RasG12D in these cells. Coexpression of Vav1 and K-RasG12D in the lungs dramatically increased malignant lung cancer lesions, and did so significantly faster than K-RasG12D alone, strongly suggesting that these two oncogenes synergize to enhance lung tumor development. Vav1 expression alone had no apparent effects on lung tumorigenesis. The increase in lung cancer in K-RasG12D/Vav1 mice was accompanied by an increase in B-cell, T-cells, and monocyte infiltration in the tumor microenvironment. Concomitantly, ERK phosphorylation was highly elevated in the lungs of K-RasG12 D/Vav1 mice. Also, several cytokines such as IL-4 and IL-13 which play a significant role in the immune system, were elevated in lungs of Vav1 and K-RasG12 D/Vav1 mice. Our findings emphasize the contribution of Vav1 to lung tumor development through its signaling properties.
Collapse
|
17
|
Grassilli S, Brugnoli F, Cairo S, Bianchi N, Judde JG, Bertagnolo V. Vav1 Selectively Down-Regulates Akt2 through miR-29b in Certain Breast Tumors with Triple Negative Phenotype. J Pers Med 2022; 12:jpm12060993. [PMID: 35743776 PMCID: PMC9224635 DOI: 10.3390/jpm12060993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 12/24/2022] Open
Abstract
Triple negative breast cancer (TNBC) represents the most aggressive breast tumor, showing a high intrinsic variability in terms of both histopathological features and response to therapies. Blocking the Akt signaling pathway is a well-studied approach in the treatment of aggressive breast tumors. The high homology among the Akt isoforms and their distinct, and possibly opposite, oncogenic functions made it difficult to develop effective drugs. Here we investigated the role of Vav1 as a potential down-regulator of individual Akt isozymes. We revealed that the over-expression of Vav1 in triple negative MDA-MB-231 cells reduced only the Akt2 isoform, acting at the post-transcriptional level through the up-modulation of miR-29b. The Vav1/miR-29b dependent decrease in Akt2 was correlated with a reduced lung colonization of circulating MDA-MB-231 cells. In cell lines established from PDX, the Vav1 induced down-modulation of Akt2 is strongly dependent on miR-29b and occurs only in some TNBC tumors. These findings may contribute to better classify breast tumors having the triple negative phenotype, and suggest that the activation of the Vav1/miR-29b axis, precisely regulating the amount of an Akt isozyme crucial for tumor dissemination, could have great potential for driving more accurate therapies to TNBCs, often not eligible or resistant to treatments.
Collapse
Affiliation(s)
- Silvia Grassilli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.G.); (F.B.); (N.B.)
- LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Federica Brugnoli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.G.); (F.B.); (N.B.)
| | - Stefano Cairo
- Xentech, 91000 Evry, France; (S.C.); (J.-G.J.)
- Istituto di Ricerca Pediatrica, 35127 Padova, Italy
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.G.); (F.B.); (N.B.)
| | | | - Valeria Bertagnolo
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.G.); (F.B.); (N.B.)
- Correspondence:
| |
Collapse
|
18
|
Jassim BA, Lin J, Zhang ZY. PTPN22: Structure, Function, and Developments in Inhibitor Discovery with Applications for Immunotherapy. Expert Opin Drug Discov 2022; 17:825-837. [PMID: 35637605 PMCID: PMC9378720 DOI: 10.1080/17460441.2022.2084607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION While immunotherapy strategies such as immune checkpoint inhibition and adoptive T cell therapy have become commonplace in cancer therapy, they suffer from limitations, including lack of patient response and toxicity. To wield the maximum potential of the immune system, cancer immunotherapy must integrate novel targets and therapeutic strategies with potential to augment clinical efficacy of currently utilized immunotherapies. PTPN22, a member of the protein tyrosine phosphatase (PTP) superfamily that downregulates T cell signaling and proliferation, has recently emerged as a systemically druggable and novel immunotherapy target. AREAS COVERED This review describes the basics of PTPN22 structure and function and provides comprehensive insight into recent advances in small molecule PTPN22 inhibitor development and the immense potential of PTPN22 inhibition to synergize with current immunotherapies. EXPERT OPINION It is apparent that small molecule PTPN22 inhibitors have enormous potential to augment efficacy of current immunotherapy strategies such as checkpoint inhibition and adoptive cell transfer. Nevertheless, several constraints must be overcome before these inhibitors can be applied as useful therapeutics, namely selectivity, potency, and in vivo efficacy.
Collapse
Affiliation(s)
- Brenson A Jassim
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, West Lafayette
| | - Jianping Lin
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, West Lafayette
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, West Lafayette
| |
Collapse
|
19
|
Vav1 Promotes B-Cell Lymphoma Development. Cells 2022; 11:cells11060949. [PMID: 35326399 PMCID: PMC8946024 DOI: 10.3390/cells11060949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 03/06/2022] [Accepted: 03/08/2022] [Indexed: 02/07/2023] Open
Abstract
Vav1 is normally and exclusively expressed in the hematopoietic system where it functions as a specific GDP/GTP nucleotide exchange factor (GEF), firmly regulated by tyrosine phosphorylation. Mutations and overexpression of Vav1 in hematopoietic malignancies, and in human cancers of various histologic origins, are well documented. To reveal whether overexpression of Vav1 in different tissues suffices for promoting the development of malignant lesions, we expressed Vav1 in transgenic mice by using the ubiquitous ROSA26 promoter (Rosa Vav1). We detected Vav1 expression in epithelial tissues of various organs including pancreas, liver, and lung. While carcinomas did not develop in these organs, surprisingly, we noticed the development of B-cell lymphomas. Rac1-GTP levels did not change in tissues from Rosa Vav1 mice expressing the transgenic Vav1, while ERK phosphorylation increased in the lymphomas, suggesting that signaling pathways are evoked. One of the growth factors analyzed by us as a suspect candidate to mediate paracrine stimulation in the lymphocytes was CSF-1, which was highly expressed in the epithelial compartment of Rosa Vav1 mice. The expression of its specific receptor, CSF-1R, was found to be highly expressed in the B-cell lymphomas. Taken together, our results suggest a potential cross-talk between epithelial cells expressing Vav1, that secrete CSF-1, and the lymphocytes that express CSF-1R, thus leading to the generation of B-cell lymphomas. Our findings provide a novel mechanism by which Vav1 contributes to tumor propagation.
Collapse
|
20
|
Transient exposure of a buried phosphorylation site in an autoinhibited protein. Biophys J 2022; 121:91-101. [PMID: 34864046 PMCID: PMC8758417 DOI: 10.1016/j.bpj.2021.11.2890] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/25/2021] [Accepted: 11/29/2021] [Indexed: 01/07/2023] Open
Abstract
Autoinhibition is a mechanism used to regulate protein function, often by making functional sites inaccessible through the interaction with a cis-acting inhibitory domain. Such autoinhibitory domains often display a substantial degree of structural disorder when unbound, and only become structured in the inhibited state. These conformational dynamics make it difficult to study the structural origin of regulation, including effects of regulatory post-translational modifications. Here, we study the autoinhibition of the Dbl Homology domain in the protein Vav1 by the so-called acidic inhibitory domain. We use molecular simulations to study the process by which a mostly unstructured inhibitory domain folds upon binding and how transient exposure of a key buried tyrosine residue makes it accessible for phosphorylation. We show that the inhibitory domain, which forms a helix in the bound and inhibited stated, samples helical structures already before binding and that binding occurs via a molten-globule-like intermediate state. Together, our results shed light on key interactions that enable the inhibitory domain to sample a finely tuned equilibrium between an inhibited and a kinase-accessible state.
Collapse
|
21
|
Thelen F, Wissmann S, Ruef N, Stein JV. The Tec Kinase Itk Integrates Naïve T Cell Migration and In Vivo Homeostasis. Front Immunol 2021; 12:716405. [PMID: 34566971 PMCID: PMC8458560 DOI: 10.3389/fimmu.2021.716405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/19/2021] [Indexed: 11/13/2022] Open
Abstract
Naïve T cells (TN) constitutively recirculate through secondary lymphatic organs (SLOs), where they scan dendritic cells (DCs) for cognate peptide-loaded major histocompatibility complexes (pMHC). Continuous trafficking between SLOs not only enables rapid clonal selection but also ensures TN homeostasis by providing access to prosurvival signals from TCR, IL-7R, and the chemokine receptor CCR7. Inside the lymphoid tissue, CCR7-mediated TN motility is mainly driven by the Rac activator DOCK2, with a separate contribution by a phosphoinositide-3-kinase γ (PI3Kγ)-dependent pathway. Tec tyrosine kinases and the Rac activator Tiam1 constitute prominent downstream effectors of PI3K signaling. Yet, the precise role of Tec kinase versus Tiam1 signaling during CCR7-mediated TN migration and homeostasis remains incompletely understood. Here, we examined the function of the Tec family member interleukin-2-inducible T-cell kinase (Itk) and Tiam1 during TN migration in vitro and in vivo using intravital microscopy. Itk deficiency caused a mild decrease in CCR7-triggered TN migration, mirroring observations made with PI3Kγ;-/- T cells, while lack of Tiam1 did not affect TN motility. In silico modeling suggested that reduced migration in the absence of Itk does not result in a substantial decrease in the frequency of TN encounters with DCs within the lymphoid tissue. In contrast, Itk was important to maintain in vivo homeostasis of CD4+ TN, also in MHCII-deficient hosts. Taken together, our data suggest that Itk contributes to TN migration and survival by integrating chemokine receptor and TCR signaling pathways.
Collapse
Affiliation(s)
- Flavian Thelen
- Department of Medical Oncology and Hematology, University of Zürich and University Hospital Zürich, Zürich, Switzerland
| | - Stefanie Wissmann
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Nora Ruef
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
22
|
Boudria R, Laurienté V, Oudar A, Harouna-Rachidi S, Dondi E, Le Roy C, Gardano L, Varin-Blank N, Guittat L. Regulatory interplay between Vav1, Syk and β-catenin occurs in lung cancer cells. Cell Signal 2021; 86:110079. [PMID: 34252536 DOI: 10.1016/j.cellsig.2021.110079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 01/15/2023]
Abstract
Vav1 exhibits two signal transducing properties as an adaptor protein and a regulator of cytoskeleton organization through its Guanine nucleotide Exchange Factor module. Although the expression of Vav1 is restricted to the hematopoietic lineage, its ectopic expression has been unraveled in a number of solid tumors. In this study, we show that in lung cancer cells, as such in hematopoietic cells, Vav1 interacts with the Spleen Tyrosine Kinase, Syk. Likewise, Syk interacts with β-catenin and, together with Vav1, regulates the phosphorylation status of β-catenin. Depletion of Vav1, Syk or β-catenin inhibits Rac1 activity and decreases cell migration suggesting the interplay of the three effectors to a common signaling pathway. This model is further supported by the finding that in turn, β-catenin regulates the transcription of Syk gene expression. This study highlights the elaborated connection between Vav1, Syk and β-catenin and the contribution of the trio to cell migration.
Collapse
Affiliation(s)
- Rofia Boudria
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Vanessa Laurienté
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Antonin Oudar
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Souleymane Harouna-Rachidi
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Elisabetta Dondi
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Christine Le Roy
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Laura Gardano
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Nadine Varin-Blank
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France.
| | - Lionel Guittat
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France.
| |
Collapse
|
23
|
Minutolo A, Potestà M, Roglia V, Cirilli M, Iacovelli F, Cerva C, Fokam J, Desideri A, Andreoni M, Grelli S, Colizzi V, Muleo R, Montesano C. Plant microRNAs from Moringa oleifera Regulate Immune Response and HIV Infection. Front Pharmacol 2021; 11:620038. [PMID: 33643043 PMCID: PMC7905167 DOI: 10.3389/fphar.2020.620038] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/03/2020] [Indexed: 11/24/2022] Open
Abstract
Traditional medicine is often chosen due to its affordability, its familiarity with patient’s cultural practices, and its wider access to the local community. Plants play an important role in providing indispensable nutrients, while specific small RNAs can regulate human gene expression in a cross-kingdom manner. The aim of the study was to evaluate the effects of plant-enriched purified extract microRNAs from Moringa oleifera seeds (MO) on the immune response and on HIV infection. Bioinformatic analysis shows that plant microRNAs (p-miRs) from MO belonging to 18 conserved families, including p-miR160h, p-miR166, p-miR482b, p-miR159c, p-miR395d, p-miR2118a, p-miR393a, p-miR167f-3p, and p-miR858b are predicted to target with high affinity BCL2, IL2RA, TNF, and VAV1, all these being involved in the cell cycle, apoptosis, immune response and also in the regulation of HIV pathogenesis. The effects of MO p-miRs transfected into HIV+ PBMCs were analyzed and revealed a decrease in viability associated with an increase of apoptosis; an increase of T helper cells expressing Fas and a decrease of intracellular Bcl2 protein expression. Meanwhile no effects were detected in PBMCs from healthy donors. In CD4+ T cells, transfection significantly reduced cell activation and modified the T cell differentiation, thereby decreasing both central and effector memory cells while increasing terminal effector memory cells. Interestingly, the p-miRs transfection induces a reduction of intracellular HIV p24 protein and a reduction of viral DNA integration. Finally, we evaluated the effect of synthetic (mimic) p-miR858b whose sequence is present in the MO p-miR pool and predicted to target VAV1, a protein involved in HIV-Nef binding. This protein plays a pivotal role in T cell antigen receptor (TCR) signaling, so triggering the activation of various pathways. The transfection of HIV+ PBMCs with the synthetic p-miR858b showed a reduced expression of VAV1 and HIV p24 proteins. Overall, our evidence defines putative mechanisms underlying a supplementary benefit of traditional medicine, alongside current antiretroviral therapy, in managing HIV infection in resource-limited settings where MO remains widely available.
Collapse
Affiliation(s)
| | - Marina Potestà
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Valentina Roglia
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Marco Cirilli
- Department of Agricultural and Forestry Science, University of Tuscia, Viterbo, Italy.,Department of Agricultural and Environmental Sciences, University of Milan, Milan, Italy
| | | | - Carlotta Cerva
- Department of System Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Joseph Fokam
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | | | - Massimo Andreoni
- Department of System Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Sandro Grelli
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Vittorio Colizzi
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy.,Faculty of Sciences and Technology, Evangelic University of Cameroon, Bandjoun, Cameroon
| | - Rosario Muleo
- Department of Agricultural and Forestry Science, University of Tuscia, Viterbo, Italy
| | - Carla Montesano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
24
|
Kaminski S, Adjali O, Jacquet C, Garaude J, Keriel A, Lassaux A, Hipskind R, Sitbon M, Taylor N, Villalba M. The protooncogene Vav1 regulates murine leukemia virus-induced T-cell leukemogenesis. Oncoimmunology 2021; 1:600-608. [PMID: 22934252 PMCID: PMC3429564 DOI: 10.4161/onci.20225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Vav1 is expressed exclusively in hematopoietic cells and is required for T cell development and activation. Vav1-deficient mice show thymic hypocellularity due to a partial block during thymocyte development at the DN3 stage and between the double positive (DP) and single positive (SP) transition. Vav1 has been shown to play a significant role in several non-hematopoietic tumors but its role in leukemogenesis is unknown. To address this question, we investigated the role of Vav1 in retrovirus-induced T cell leukemogenesis. Infection of Vav1-deficient mice with the Moloney strain of murine leukemia virus (M-MuLV) significantly affected tumor phenotype without modulating tumor incidence or latency. M-MuLV-infected Vav1-deficient mice showed reduced splenomegaly, higher hematocrit levels and hypertrophic thymi. Notably, Vav1-deficient mice with M-MuLV leukemias presented with markedly lower TCRβ/CD3 levels, indicating that transformation occurred at an earlier stage of T cell development than in WT mice. Thus, impaired T cell development modulates the outcome of retrovirus-induced T cell leukemias, demonstrating a link between T cell development and T cell leukemogenesis.
Collapse
Affiliation(s)
- Sandra Kaminski
- Institut de Génétique Moléculaire de Montpellier; UMR 5535; CNRS; Montpellier, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Vav1 Sustains the In Vitro Differentiation of Normal and Tumor Precursors to Insulin Producing Cells Induced by all-Trans Retinoic Acid (ATRA). Stem Cell Rev Rep 2020; 17:673-684. [PMID: 33165749 PMCID: PMC8036226 DOI: 10.1007/s12015-020-10074-x] [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] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
All-trans retinoic acid (ATRA) promotes the development and the function of insulin producing cells and induces partial differentiation of pancreatic tumor cells. A number of evidences clearly indicate that the ATRA mediated signaling may have a substantial role in therapeutic approaches based on restoration of functional β-cells. Among the proteins up-regulated by ATRA, Vav1 is involved in maturation and function of haematopoietic cells and is essential for retinoids induced differentiation of tumor promyelocytes. The presence of Vav1 in solid tissues, including pancreas, is considered ectopic and no role in the differentiation of human epithelial cells has so far been described. We demonstrated here that Vav1 sustains the maturation to β-cells of the normal precursors human Biliary Tree Stem/progenitor Cells (hBTSCs) induced by a differentiation medium containing ATRA and that, in the mature normal pancreas, insulin-producing cells express variable levels of Vav1. Using pancreatic ductal adenocarcinoma (PDAC)-derived cells, we also revealed that the ATRA induced up-modulation of Vav1 is essential for the retinoid-induced trans-differentiation of neoplastic cells into insulin producing cells. The results of this study identify Vav1 as crucial molecule in ATRA induced maturation of insulin producing cells and suggest this protein as a marker for new strategies ended to restore functional β-cells. Graphical abstract ![]()
Collapse
|
26
|
Vav1 Down-Modulates Akt2 Expression in Cells from Pancreatic Ductal Adenocarcinoma: Nuclear Vav1 as a Potential Regulator of Akt Related Malignancy in Pancreatic Cancer. Biomedicines 2020; 8:biomedicines8100379. [PMID: 32993067 PMCID: PMC7600902 DOI: 10.3390/biomedicines8100379] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 09/24/2020] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most aggressive tumor malignancy worldwide, mainly due to uncontrolled metastasis. Among the numerous molecules deregulated in PDAC, different members of the Akt pathways are of great importance because they are involved in tumor cell proliferation, migration, and invasion. We have recently demonstrated that Vav1, ectopically expressed in solid tumors, is capable of down-modulating expression and/or activation of specific Akt isoforms in breast cancer cells. By using pancreatic cell lines expressing different basal levels of Vav1, we demonstrated here that Vav1 down-regulates the expression of Akt2, known to correlate with tumor metastases and resistance to therapy. In particular, while the silencing of Vav1 is sufficient to induce Akt2, its up-modulation reduces Akt2 levels only when Vav1 accumulates inside the nucleus of PDAC cells. Moreover, in PDAC tissues, we revealed that high nuclear levels of Vav1 correlate with low Akt2 expression. Although we cannot demonstrate the mechanisms involved, our results provide new insights into the role of Vav1 in PDAC and, as targeting specific members of the Akt family is a promising therapeutic chance in solid tumors, they suggest that Vav1, by down-modulating Akt2, has potential as a molecular target in PDAC.
Collapse
|
27
|
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
|
28
|
Chiba S, Sakata-Yanagimoto M. Advances in understanding of angioimmunoblastic T-cell lymphoma. Leukemia 2020; 34:2592-2606. [PMID: 32704161 PMCID: PMC7376827 DOI: 10.1038/s41375-020-0990-y] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/27/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022]
Abstract
It has been nearly half a century since angioimmunoblastic T-cell lymphoma (AITL) was characterized in the early 1970’s. Our understanding of the disease has dramatically changed due to multiple discoveries and insights. One of the key features of AITL is aberrant immune activity. Although AITL is now understood to be a neoplastic disease, pathologists appreciated that it was an inflammatory condition. The more we understand AITL at cellular and genetic levels, the more we view it as both a neoplastic and an inflammatory disease. Here, we review recent progress in our understanding of AITL, focusing on as yet unsolved questions.
Collapse
Affiliation(s)
- Shigeru Chiba
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Mamiko Sakata-Yanagimoto
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| |
Collapse
|
29
|
Löber J, Hitzing C, Münchhalfen M, Engels N. Vav family proteins constitute disparate branching points for distinct BCR signaling pathways. Eur J Immunol 2020; 50:1912-1928. [PMID: 32671844 DOI: 10.1002/eji.202048621] [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: 03/03/2020] [Revised: 05/18/2020] [Accepted: 07/10/2020] [Indexed: 01/16/2023]
Abstract
Antigen recognition by B-cell antigen receptors (BCRs) activates distinct intracellular signaling pathways that control the differentiation fate of activated B lymphocytes. BCR-proximal signaling enzymes comprise protein tyrosine kinases, phosphatases, and plasma membrane lipid-modifying enzymes, whose function is furthermore coordinated by catalytically inert adaptor proteins. Here, we show that an additional class of enzymatic activity provided by guanine-nucleotide exchange factors (GEFs) of the Vav family controls BCR-proximal Ca2+ mobilization, cytoskeletal actin reorganization, and activation of the PI3 kinase/Akt pathway. Whereas Vav1 and Vav3 supported all of those signaling processes to different extents in a human B-cell model system, Vav2 facilitated Actin remodeling, and activation of Akt but did not promote Ca2+ signaling. On BCR activation, Vav1 was directly recruited to the phosphorylated BCR and to the central adaptor protein SLP65 via its Src homology 2 domain. Pharmacological inhibition or genetic inactivation of the substrates of Vav GEFs, small G proteins of the Rho/Rac family, impaired BCR-induced Ca2+ mobilization, probably because phospholipase Cγ2 requires activated Rac proteins for optimal activity. Our findings show that Vav family members are key relays of the BCR signalosome that differentially control distinct signaling pathways both in a catalysis-dependent and -independent manner.
Collapse
Affiliation(s)
- Jens Löber
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Christoffer Hitzing
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Matthias Münchhalfen
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Niklas Engels
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
30
|
Younes N, Zhou L, Amatullah H, Mei SHJ, Herrero R, Lorente JA, Stewart DJ, Marsden P, Liles WC, Hu P, Dos Santos CC. Mesenchymal stromal/stem cells modulate response to experimental sepsis-induced lung injury via regulation of miR-27a-5p in recipient mice. Thorax 2020; 75:556-567. [PMID: 32546573 PMCID: PMC7361025 DOI: 10.1136/thoraxjnl-2019-213561] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 01/08/2020] [Accepted: 03/13/2020] [Indexed: 01/11/2023]
Abstract
Introduction Mesenchymal stromal cell (MSC) therapy mitigates lung injury and improves survival in murine models of sepsis. Precise mechanisms of therapeutic benefit remain poorly understood. Objectives To identify host-derived regulatory elements that may contribute to the therapeutic effects of MSCs, we profiled the microRNAome (miRNAome) and transcriptome of lungs from mice randomised to experimental polymicrobial sepsis-induced lung injury treated with either placebo or MSCs. Methods and results A total of 11 997 genes and 357 microRNAs (miRNAs) expressed in lungs were used to generate a statistical estimate of association between miRNAs and their putative mRNA targets; 1395 miRNA:mRNA significant association pairs were found to be differentially expressed (false discovery rate ≤0.05). MSC administration resulted in the downregulation of miR-27a-5p and upregulation of its putative target gene VAV3 (adjusted p=1.272E-161) in septic lungs. In human pulmonary microvascular endothelial cells, miR-27a-5p expression levels were increased while VAV3 was decreased following lipopolysaccharide (LPS) or tumour necrosis factor (TNF) stimulation. Transfection of miR-27a-5p mimic or inhibitor resulted in increased or decreased VAV3 message, respectively. Luciferase reporter assay demonstrated specific binding of miR-27a-5p to the 3′UTR of VAV3. miR27a-5p inhibition mitigated TNF-induced (1) delayed wound closure, increased (2) adhesion and (3) transendothelial migration but did not alter permeability. In vivo, cell infiltration was attenuated by intratracheal coinstillation of the miR-27a-5p inhibitor, but this did not protect against endotoxin-induced oedema formation. Conclusions Our data support involvement of miR-27a-5p and VAV3 in cellular adhesion and infiltration during acute lung injury and a potential role for miR-27a-based therapeutics for acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Nadim Younes
- Critical Care Medicine, The Keenan Research Centre for Biomedical Science of Saint Michael's Hospital, Toronto, Ontario, Canada
| | - Louis Zhou
- Critical Care Medicine, The Keenan Research Centre for Biomedical Science of Saint Michael's Hospital, Toronto, Ontario, Canada.,Institute of Medical Sciences and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hajera Amatullah
- Critical Care Medicine, The Keenan Research Centre for Biomedical Science of Saint Michael's Hospital, Toronto, Ontario, Canada.,Institute of Medical Sciences and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shirley H J Mei
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Raquel Herrero
- Critical Care Service, Hospital Universitario de Getafe-CIBER de Enfermedades Respiratorias (CIBERES), Getafe, Spain
| | - Jose Angel Lorente
- Critical Care Service, Hospital Universitario de Getafe-CIBER de Enfermedades Respiratorias (CIBERES), Getafe, Spain
| | - Duncan J Stewart
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Philip Marsden
- Critical Care Medicine, The Keenan Research Centre for Biomedical Science of Saint Michael's Hospital, Toronto, Ontario, Canada
| | - W Conrad Liles
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Claudia C Dos Santos
- Critical Care Medicine, The Keenan Research Centre for Biomedical Science of Saint Michael's Hospital, Toronto, Ontario, Canada .,Institute of Medical Sciences and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Salaymeh Y, Farago M, Sebban S, Shalom B, Pikarsky E, Katzav S. Vav1 and mutant K-Ras synergize in the early development of pancreatic ductal adenocarcinoma in mice. Life Sci Alliance 2020; 3:e202000661. [PMID: 32277014 PMCID: PMC7156281 DOI: 10.26508/lsa.202000661] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023] Open
Abstract
To explore the contribution of Vav1, a hematopoietic signal transducer, to pancreatic ductal adenocarcinoma (PDAC) development, we generated transgenic mouse lines expressing, Vav1, K-RasG12D, or both K-RasG12D and Vav1 in pancreatic acinar cells. Co-expression of Vav1 and K-RasG12D synergistically enhanced acinar-to-ductal metaplasia (ADM) formation, far exceeding the number of lesions developed in K-RasG12D mice. Mice expressing only Vav1 did not develop ADM. Moreover, the incidence of PDAC in K-RasG12D/Vav1 was significantly higher than in K-RasG12D mice. Discontinuing Vav1 expression in K-RasG12D/Vav1 mice elicited a marked regression of malignant lesions in the pancreas, demonstrating Vav1 is required for generation and maintenance of ADM. Rac1-GTP levels in the K-RasG12D/Vav1 mice pancreas clearly demonstrated an increase in Rac1 activity. Treatment of K-RasG12D and K-RasG12D/Vav1 mice with azathioprine, an immune-suppressor drug which inhibits Vav1's activity as a GDP/GTP exchange factor, dramatically reduced the number of malignant lesions. These results suggest that Vav1 plays a role in the development of PDAC when co-expressed with K-RasG12D via its activity as a GEF for Rac1GTPase.
Collapse
Affiliation(s)
- Yaser Salaymeh
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Marganit Farago
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Shulamit Sebban
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Batel Shalom
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Eli Pikarsky
- The Lautenberg Center for Immunology and Cancer Research and Department of Pathology, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Shulamit Katzav
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
32
|
Rodríguez-Fdez S, Fernández-Nevado L, Lorenzo-Martín LF, Bustelo XR. Lysine Acetylation Reshapes the Downstream Signaling Landscape of Vav1 in Lymphocytes. Cells 2020; 9:cells9030609. [PMID: 32143292 PMCID: PMC7140538 DOI: 10.3390/cells9030609] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022] Open
Abstract
Vav1 works both as a catalytic Rho GTPase activator and an adaptor molecule. These functions, which are critical for T cell development and antigenic responses, are tyrosine phosphorylation-dependent. However, it is not known whether other posttranslational modifications can contribute to the regulation of the biological activity of this protein. Here, we show that Vav1 becomes acetylated on lysine residues in a stimulation- and SH2 domain-dependent manner. Using a collection of both acetylation- and deacetylation-mimicking mutants, we show that the acetylation of four lysine residues (Lys222, Lys252, Lys587, and Lys716) leads to the downmodulation of the adaptor function of Vav1 that triggers the stimulation of the nuclear factor of activated T cells (NFAT). These sites belong to two functional subclasses according to mechanistic criteria. We have also unveiled additional acetylation sites potentially involved in either the stimulation (Lys782) or the downmodulation (Lys335, Lys374) of specific Vav1-dependent downstream responses. Collectively, these results indicate that Nε-lysine acetylation can play variegated roles in the regulation of Vav1 signaling. Unlike the case of the tyrosine phosphorylation step, this new regulatory layer is not conserved in other Vav family paralogs.
Collapse
Affiliation(s)
- Sonia Rodríguez-Fdez
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.-N.); (L.F.L.-M.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Lucía Fernández-Nevado
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.-N.); (L.F.L.-M.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - L. Francisco Lorenzo-Martín
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.-N.); (L.F.L.-M.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Xosé R. Bustelo
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.-N.); (L.F.L.-M.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Correspondence: ; Tel.: +34-663194634
| |
Collapse
|
33
|
The role of actin and myosin in antigen extraction by B lymphocytes. Semin Cell Dev Biol 2019; 102:90-104. [PMID: 31862219 DOI: 10.1016/j.semcdb.2019.10.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/14/2019] [Accepted: 10/31/2019] [Indexed: 12/14/2022]
Abstract
B cells must extract antigens attached to the surface of antigen presenting cells to generate high-affinity antibodies. Antigen extraction requires force, and recent studies have implicated actomyosin-dependent pulling forces generated within the B cell as the major driver of antigen extraction. These actomyosin-dependent pulling forces also serve to test the affinity of the B cell antigen receptor for antigen prior to antigen extraction. Such affinity discrimination is central to the process of antibody affinity maturation. Here we review the evidence that actomyosin-dependent pulling forces generated within the B cell promote affinity discrimination and power antigen extraction. Our take on these critical B cell functions is influenced significantly by the recent identification of formin-generated, myosin-rich, concentric actin arcs in the medial portion of the T cell immune synapse, as B cells appear to contain a similar contractile actomyosin structure.
Collapse
|
34
|
Rodríguez-Fdez S, Bustelo XR. The Vav GEF Family: An Evolutionary and Functional Perspective. Cells 2019; 8:E465. [PMID: 31100928 PMCID: PMC6562523 DOI: 10.3390/cells8050465] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 02/07/2023] Open
Abstract
Vav proteins play roles as guanosine nucleotide exchange factors for Rho GTPases and signaling adaptors downstream of protein tyrosine kinases. The recent sequencing of the genomes of many species has revealed that this protein family originated in choanozoans, a group of unicellular organisms from which animal metazoans are believed to have originated from. Since then, the Vav family underwent expansions and reductions in its members during the evolutionary transitions that originated the agnates, chondrichthyes, some teleost fish, and some neoaves. Exotic members of the family harboring atypical structural domains can be also found in some invertebrate species. In this review, we will provide a phylogenetic perspective of the evolution of the Vav family. We will also pay attention to the structure, signaling properties, regulatory layers, and functions of Vav proteins in both invertebrate and vertebrate species.
Collapse
Affiliation(s)
- Sonia Rodríguez-Fdez
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
- Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
| | - Xosé R Bustelo
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
- Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
| |
Collapse
|
35
|
Hammer JA, Wang JC, Saeed M, Pedrosa AT. Origin, Organization, Dynamics, and Function of Actin and Actomyosin Networks at the T Cell Immunological Synapse. Annu Rev Immunol 2019; 37:201-224. [PMID: 30576253 PMCID: PMC8343269 DOI: 10.1146/annurev-immunol-042718-041341] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The engagement of a T cell with an antigen-presenting cell (APC) or activating surface results in the formation within the T cell of several distinct actin and actomyosin networks. These networks reside largely within a narrow zone immediately under the T cell's plasma membrane at its site of contact with the APC or activating surface, i.e., at the immunological synapse. Here we review the origin, organization, dynamics, and function of these synapse-associated actin and actomyosin networks. Importantly, recent insights into the nature of these actin-based cytoskeletal structures were made possible in several cases by advances in light microscopy.
Collapse
Affiliation(s)
- John A Hammer
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Jia C Wang
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Mezida Saeed
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Antonio T Pedrosa
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| |
Collapse
|
36
|
Pesenacker AM, Chen V, Gillies J, Speake C, Marwaha AK, Sun A, Chow S, Tan R, Elliott T, Dutz JP, Tebbutt SJ, Levings MK. Treg gene signatures predict and measure type 1 diabetes trajectory. JCI Insight 2019; 4:123879. [PMID: 30730852 DOI: 10.1172/jci.insight.123879] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/05/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Multiple therapeutic strategies to restore immune regulation and slow type 1 diabetes (T1D) progression are in development and testing. A major challenge has been defining biomarkers to prospectively identify subjects likely to benefit from immunotherapy and/or measure intervention effects. We previously found that, compared with healthy controls, Tregs from children with new-onset T1D have an altered Treg gene signature (TGS), suggesting that this could be an immunoregulatory biomarker. METHODS nanoString was used to assess the TGS in sorted Tregs (CD4+CD25hiCD127lo) or peripheral blood mononuclear cells (PBMCs) from individuals with T1D or type 2 diabetes, healthy controls, or T1D recipients of immunotherapy. Biomarker discovery pipelines were developed and applied to various sample group comparisons. RESULTS Compared with controls, the TGS in isolated Tregs or PBMCs was altered in adult new-onset and cross-sectional T1D cohorts, with sensitivity or specificity of biomarkers increased by including T1D-associated SNPs in algorithms. The TGS was distinct in T1D versus type 2 diabetes, indicating disease-specific alterations. TGS measurement at the time of T1D onset revealed an algorithm that accurately predicted future rapid versus slow C-peptide decline, as determined by longitudinal analysis of placebo arms of START and T1DAL trials. The same algorithm stratified participants in a phase I/II clinical trial of ustekinumab (αIL-12/23p40) for future rapid versus slow C-peptide decline. CONCLUSION These data suggest that biomarkers based on measuring TGSs could be a new approach to stratify patients and monitor autoimmune activity in T1D. FUNDING JDRF (1-PNF-2015-113-Q-R, 2-PAR-2015-123-Q-R, 3-SRA-2016-209-Q-R, 3-PDF-2014-217-A-N), the JDRF Canadian Clinical Trials Network, the National Institute of Allergy and Infectious Diseases of the National Institutes of Health (UM1AI109565 and FY15ITN168), and BCCHRI.
Collapse
Affiliation(s)
- Anne M Pesenacker
- Department of Surgery, University of British Columbia (UBC), and BC Children's Hospital Research Institute (BCCHRI), Vancouver, British Columbia, Canada
| | - Virginia Chen
- Department of Medicine and Centre for Heart Lung Innovation, UBC, and Prevention of Organ Failure (PROOF) Centre of Excellence, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Jana Gillies
- Department of Surgery, University of British Columbia (UBC), and BC Children's Hospital Research Institute (BCCHRI), Vancouver, British Columbia, Canada
| | - Cate Speake
- Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, Washington, USA
| | - Ashish K Marwaha
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Annika Sun
- Department of Surgery, University of British Columbia (UBC), and BC Children's Hospital Research Institute (BCCHRI), Vancouver, British Columbia, Canada
| | - Samuel Chow
- Department of Dermatology, UBC, and BCCHRI, Vancouver, British Columbia, Canada
| | - Rusung Tan
- Department of Pathology, Sidra Medicine, Weill Cornell Medicine, Doha, Qatar
| | - Thomas Elliott
- Department of Medicine, UBC, and BCDiabetes, Vancouver, British Columbia, Canada
| | - Jan P Dutz
- Department of Dermatology, UBC, and BCCHRI, Vancouver, British Columbia, Canada
| | - Scott J Tebbutt
- Department of Medicine and Centre for Heart Lung Innovation, UBC, and Prevention of Organ Failure (PROOF) Centre of Excellence, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia (UBC), and BC Children's Hospital Research Institute (BCCHRI), Vancouver, British Columbia, Canada
| |
Collapse
|
37
|
Stierwalt HD, Ehrlicher SE, Bergman BC, Robinson MM, Newsom SA. Insulin-stimulated Rac1-GTP binding is not impaired by palmitate treatment in L6 myotubes. Physiol Rep 2018; 6:e13956. [PMID: 30592185 PMCID: PMC6308110 DOI: 10.14814/phy2.13956] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 11/28/2018] [Indexed: 01/01/2023] Open
Abstract
Ras-related C3 botulinum toxin substrate 1 (Rac1) is required for normal insulin-stimulated glucose transport in skeletal muscle and evidence indicates Rac1 may be negatively regulated by lipids. We investigated if insulin-stimulated activation of Rac1 (i.e., Rac1-GTP binding) is impaired by accumulation of diacylglycerols (DAG) and ceramides in cultured muscle cells. Treating L6 myotubes with 100 nmol/L insulin resulted in increased Rac1-GTP binding that was rapid (occurring within 2 min), relatively modest (+38 ± 19% vs. basal, P < 0.001), and short-lived, returning to near-basal levels within 15 min of continuous treatment. Incubating L6 myotubes overnight in 500 μmol/L palmitate increased the accumulation of DAG and ceramides (P < 0.05 vs. no fatty acid control). Despite significant accumulation of lipids, insulin-stimulated Rac1-GTP binding was not impaired during palmitate treatment (P = 0.39 vs. no fatty acid control). Nevertheless, phosphorylation of Rac1 effector protein p21-activated kinase (PAK) was attenuated in response to palmitate treatment (P = 0.02 vs. no fatty acid control). Palmitate treatment also increased inhibitory phosphorylation of insulin receptor substrate-1 and attenuated insulin-stimulated phosphorylation of Akt at both Thr308 and Ser473 (all P < 0.05 vs. no fatty acid control). Such signaling impairments resulted in near complete inhibition of insulin-stimulated translocation of glucose transporter protein 4 (GLUT4; P = 0.10 vs. basal during palmitate treatment). In summary, our finding suggests that Rac1 may not undergo negative regulation by DAG or ceramides. We instead provide evidence that attenuated PAK phosphorylation and impaired GLUT4 translocation during palmitate-induced insulin resistance can occur independent of defects in insulin-stimulated Rac1-GTP binding.
Collapse
Affiliation(s)
- Harrison D. Stierwalt
- School of Biological and Population Health SciencesCollege of Public Health and Human SciencesOregon State UniversityCorvallisOregon
| | - Sarah E. Ehrlicher
- School of Biological and Population Health SciencesCollege of Public Health and Human SciencesOregon State UniversityCorvallisOregon
| | - Bryan C. Bergman
- Division of Endocrinology, Metabolism and DiabetesSchool of MedicineUniversity of Colorado DenverDenverColorado
| | - Matthew M. Robinson
- School of Biological and Population Health SciencesCollege of Public Health and Human SciencesOregon State UniversityCorvallisOregon
| | - Sean A. Newsom
- School of Biological and Population Health SciencesCollege of Public Health and Human SciencesOregon State UniversityCorvallisOregon
| |
Collapse
|
38
|
Shifrut E, Carnevale J, Tobin V, Roth TL, Woo JM, Bui CT, Li PJ, Diolaiti ME, Ashworth A, Marson A. Genome-wide CRISPR Screens in Primary Human T Cells Reveal Key Regulators of Immune Function. Cell 2018; 175:1958-1971.e15. [PMID: 30449619 DOI: 10.1016/j.cell.2018.10.024] [Citation(s) in RCA: 365] [Impact Index Per Article: 52.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/08/2018] [Accepted: 10/08/2018] [Indexed: 12/11/2022]
Abstract
Human T cells are central effectors of immunity and cancer immunotherapy. CRISPR-based functional studies in T cells could prioritize novel targets for drug development and improve the design of genetically reprogrammed cell-based therapies. However, large-scale CRISPR screens have been challenging in primary human cells. We developed a new method, single guide RNA (sgRNA) lentiviral infection with Cas9 protein electroporation (SLICE), to identify regulators of stimulation responses in primary human T cells. Genome-wide loss-of-function screens identified essential T cell receptor signaling components and genes that negatively tune proliferation following stimulation. Targeted ablation of individual candidate genes characterized hits and identified perturbations that enhanced cancer cell killing. SLICE coupled with single-cell RNA sequencing (RNA-seq) revealed signature stimulation-response gene programs altered by key genetic perturbations. SLICE genome-wide screening was also adaptable to identify mediators of immunosuppression, revealing genes controlling responses to adenosine signaling. The SLICE platform enables unbiased discovery and characterization of functional gene targets in primary cells.
Collapse
Affiliation(s)
- Eric Shifrut
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Julia Carnevale
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Victoria Tobin
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Theodore L Roth
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jonathan M Woo
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Christina T Bui
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - P Jonathan Li
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Morgan E Diolaiti
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alan Ashworth
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexander Marson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA; UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
39
|
Bernard I, Sacquin A, Kassem S, Benamar M, Colacios C, Gador M, Pérals C, Fazilleau N, Saoudi A. A Natural Variant of the Signaling Molecule Vav1 Enhances Susceptibility to Myasthenia Gravis and Influences the T Cell Receptor Repertoire. Front Immunol 2018; 9:2399. [PMID: 30410484 PMCID: PMC6210741 DOI: 10.3389/fimmu.2018.02399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/27/2018] [Indexed: 01/01/2023] Open
Abstract
The guanine nucleotide exchange factor Vav1 is essential for transducing T cell receptor (TCR) signals and plays an important role in T cell development and activation. Previous genetic studies identified a natural variant of Vav1 characterized by the substitution of an arginine (R) residue by a tryptophane (W) at position 63 (Vav1R63W). This variant impacts Vav1 adaptor functions and controls susceptibility to T cell-mediated neuroinflammation. To assess the implication of this Vav1 variant on the susceptibility to antibody-mediated diseases, we used the animal model of myasthenia gravis, experimental autoimmune myasthenia gravis (EAMG). To this end, we generated a knock-in (KI) mouse model bearing a R to W substitution in the Vav1 gene (Vav1R63W) and immunized it with either torpedo acetylcholine receptor (tAChR) or the α146-162 immunodominant peptide. We observed that the Vav1R63W conferred increased susceptibility to EAMG, revealed by a higher AChR loss together with an increased production of effector cytokines (IFN-γ, IL-17A, GM-CSF) by antigen-specific CD4+ T cells, as well as an increased frequency of antigen-specific CD4+ T cells. This correlated with the emergence of a dominant antigen-specific T cell clone in KI mice that was not present in wild-type mice, suggesting an impact on thymic selection and/or a different clonal selection threshold following antigen encounter. Our results highlight the key role of Vav1 in the pathophysiology of EAMG and this was associated with an impact on the TCR repertoire of AChR reactive T lymphocytes.
Collapse
Affiliation(s)
- Isabelle Bernard
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Antoine Sacquin
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Sahar Kassem
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Mehdi Benamar
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Céline Colacios
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Mylène Gador
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Corine Pérals
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Nicolas Fazilleau
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Abdelhadi Saoudi
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| |
Collapse
|
40
|
Vav1 mutations identified in human cancers give rise to different oncogenic phenotypes. Oncogenesis 2018; 7:80. [PMID: 30297765 PMCID: PMC6175932 DOI: 10.1038/s41389-018-0091-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/27/2018] [Accepted: 09/09/2018] [Indexed: 01/26/2023] Open
Abstract
Vav1 is physiologically active as a GDP/GTP nucleotide exchange factor (GEF) in the hematopoietic system. Overexpression of Vav1 in multiple tumor types is known to enhance oncogenicity, yet whether or not Vav1 is a bona fide oncogene is still a matter of debate. Although mutations in Vav1 were recently identified in human cancers of various origins, the functional activities of these mutants are not known. We tested the transforming potential of three mutations identified in human lung adenocarcinoma: E59K, D517E, and L801P. Results from several assays indicative of transforming activities such as rate of proliferation, growth in agar, and generation of tumors in NOD/SCID mice clearly indicated that E59K and D517E are highly transforming but L801P at the SH3 domain is not. The acquired oncogenic activity of these mutants can be attributed to their enhanced activity as GEFs for Rho/Rac GTPases. Deciphering of the mechanisms leading to overactivity of the tested mutants revealed that the E59K mutation facilitates cleavage of a truncated protein that is uncontrollably active as a GEF, while D517E generates a highly stable overexpressed protein that is also more active as a GEF than wild-type Vav1. These findings support the classification of Vav1 as a bona fide oncogene in human cancer.
Collapse
|
41
|
Woo JS, Srikanth S, Kim KD, Elsaesser H, Lu J, Pellegrini M, Brooks DG, Sun Z, Gwack Y. CRACR2A-Mediated TCR Signaling Promotes Local Effector Th1 and Th17 Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:1174-1185. [PMID: 29987160 PMCID: PMC6081249 DOI: 10.4049/jimmunol.1800659] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/12/2018] [Indexed: 12/24/2022]
Abstract
Ca2+ release-activated Ca2+ channel regulator 2A (CRACR2A) is expressed abundantly in T cells and acts as a signal transmitter between TCR stimulation and activation of the Ca2+/NFAT and JNK/AP1 pathways. CRACR2A has been linked to human diseases in numerous genome-wide association studies and was shown to be one of the most sensitive targets of the widely used statin drugs. However, the physiological role of CRACR2A in T cell functions remains unknown. In this study, using transgenic mice for tissue-specific deletion, we show that CRACR2A promotes Th1 responses and effector function of Th17 cells. CRACR2A was abundantly expressed in Th1 and Th17 cells. In vitro, deficiency of CRACR2A decreased Th1 differentiation under nonpolarizing conditions, whereas the presence of polarizing cytokines compensated this defect. Transcript analysis showed that weakened TCR signaling by deficiency of CRACR2A failed to promote Th1 transcriptional program. In vivo, conditional deletion of CRACR2A in T cells alleviated Th1 responses to acute lymphocytic choriomeningitis virus infection and imparted resistance to experimental autoimmune encephalomyelitis. Analysis of CNS from experimental autoimmune encephalomyelitis-induced mice showed impaired effector functions of both Th1 and Th17 cell types, which correlated with decreased pathogenicity. Collectively, our findings demonstrate the requirement of CRACR2A-mediated TCR signaling in Th1 responses as well as pathogenic conversion of Th17 cells, which occurs at the site of inflammation.
Collapse
Affiliation(s)
- Jin Seok Woo
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Kyun-Do Kim
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Heidi Elsaesser
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 2M9, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario M5G 2M9, Canada
| | - Jing Lu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095; and
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095; and
| | - David G Brooks
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 2M9, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario M5G 2M9, Canada
| | - Zuoming Sun
- Division of Molecular Immunology, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095;
| |
Collapse
|
42
|
Gaud G, Roncagalli R, Chaoui K, Bernard I, Familiades J, Colacios C, Kassem S, Monsarrat B, Burlet-Schiltz O, de Peredo AG, Malissen B, Saoudi A. The costimulatory molecule CD226 signals through VAV1 to amplify TCR signals and promote IL-17 production by CD4 + T cells. Sci Signal 2018; 11:11/538/eaar3083. [PMID: 29991650 DOI: 10.1126/scisignal.aar3083] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The activation of T cells requires the guanine nucleotide exchange factor VAV1. Using mice in which a tag for affinity purification was attached to endogenous VAV1 molecules, we analyzed by quantitative mass spectrometry the signaling complex that assembles around activated VAV1. Fifty VAV1-binding partners were identified, most of which had not been previously reported to participate in VAV1 signaling. Among these was CD226, a costimulatory molecule of immune cells. Engagement of CD226 induced the tyrosine phosphorylation of VAV1 and synergized with T cell receptor (TCR) signals to specifically enhance the production of interleukin-17 (IL-17) by primary human CD4+ T cells. Moreover, co-engagement of the TCR and a risk variant of CD226 that is associated with autoimmunity (rs763361) further enhanced VAV1 activation and IL-17 production. Thus, our study reveals that a VAV1-based, synergistic cross-talk exists between the TCR and CD226 during both physiological and pathological T cell responses and provides a rational basis for targeting CD226 for the management of autoimmune diseases.
Collapse
Affiliation(s)
- Guillaume Gaud
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France
| | - Romain Roncagalli
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, Inserm, CNRS, 13288 Marseille, France
| | - Karima Chaoui
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS UMR 5089, 31077 Toulouse Cedex, France
| | - Isabelle Bernard
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France
| | - Julien Familiades
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France
| | - Céline Colacios
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France
| | - Sahar Kassem
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France
| | - Bernard Monsarrat
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS UMR 5089, 31077 Toulouse Cedex, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS UMR 5089, 31077 Toulouse Cedex, France
| | - Anne Gonzalez de Peredo
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS UMR 5089, 31077 Toulouse Cedex, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, Inserm, CNRS, 13288 Marseille, France.,Centre d'Immunophénomique, Aix-Marseille Université, Inserm, CNRS, 13288 Marseille, France
| | - Abdelhadi Saoudi
- Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, CNRS, Inserm, Toulouse 31300, France.
| |
Collapse
|
43
|
Grassilli S, Brugnoli F, Lattanzio R, Marchisio M, Perracchio L, Piantelli M, Bavelloni A, Capitani S, Bertagnolo V. Vav1 downmodulates Akt in different breast cancer subtypes: a new promising chance to improve breast cancer outcome. Mol Oncol 2018; 12:1012-1025. [PMID: 29658179 PMCID: PMC6026867 DOI: 10.1002/1878-0261.12203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 03/22/2018] [Accepted: 04/02/2018] [Indexed: 11/07/2022] Open
Abstract
Targeting different members of the Akt pathways is a promising therapeutic chance in solid tumors including breast cancer. The variable expression levels of Akt isoforms with opposite effects on tumor growth and metastasis, however, make it difficult to select the inhibitors to be used for specific breast tumor subtypes. Using in vitro and in vivo models, we demonstrated here that Vav1, ectopically expressed in invasive breast tumors derived cells, downmodulates Akt acting at expression and/or activation levels depending on tumor subtype. The decreased p‐Akt1 (Ser473) levels are a common effect of Vav1 upmodulation, suggesting that, in breast tumor‐derived cells and independently of their phenotype, Vav1 interferes with signaling pathways ended to specifically recruit Akt1. Only in ER‐negative cell lines, the silencing of Vav1 induced the expression but not the activation of Akt2. A retrospective analysis of early invasive breast tumors allowed to establish the prognostic significance of the p‐Akt/Vav1 relationship. In particular, low Vav1 levels negatively influence the follow‐up of patients with low p‐Akt in their primary tumors and subjected to adjuvant chemotherapy. As the use of specific or pan Akt inhibitors may not be sufficient or may even be detrimental, increasing the levels of Vav1 could be a new approach to improve breast cancer outcomes. This might be particularly relevant for tumors with a triple‐negative phenotype, for which target‐based therapies are not currently available.
Collapse
Affiliation(s)
- Silvia Grassilli
- Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy
| | - Federica Brugnoli
- Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy
| | - Rossano Lattanzio
- Department of Medical, Oral and Biotechnological Sciences, University 'G. d'Annunzio', Chieti, Italy.,Center on Aging Sciences and Translational Medicine (CeSI-MeT), University 'G. d'Annunzio', Chieti, Italy
| | - Marco Marchisio
- Center on Aging Sciences and Translational Medicine (CeSI-MeT), University 'G. d'Annunzio', Chieti, Italy.,Department of Medicine and Aging Sciences, University 'G. d'Annunzio', Chieti, Italy
| | | | | | - Alberto Bavelloni
- Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Silvano Capitani
- Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy.,LTTA Centre, University of Ferrara, Italy
| | - Valeria Bertagnolo
- Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy
| |
Collapse
|
44
|
GWAS analysis using interspecific backcross progenies reveals superior blue catfish alleles responsible for strong resistance against enteric septicemia of catfish. Mol Genet Genomics 2018; 293:1107-1120. [PMID: 29737402 DOI: 10.1007/s00438-018-1443-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 05/02/2018] [Indexed: 12/11/2022]
Abstract
Infectious diseases pose significant threats to the catfish industry. Enteric septicemia of catfish (ESC) caused by Edwardsiella ictaluri is the most devastating disease for catfish aquaculture, causing huge economic losses annually. Channel catfish and blue catfish exhibit great contrast in resistance against ESC, with channel catfish being highly susceptible and blue catfish being highly resistant. As such, the interspecific backcross progenies provide an ideal system for the identification of quantitative trait locus (QTL). We previously reported one significant QTL on linkage group (LG) 1 using the third-generation backcrosses, but the number of founders used to make the second- and third-generation backcross progenies was very small. Although the third-generation backcross progenies provided a greater power for fine mapping than the first-generation backcrosses, some major QTL for disease resistance may have been missing due to the small numbers of founders used to produce the higher generation backcrosses. In this study, we performed a genome-wide association study using first-generation backcrosses with the catfish 690 K SNP arrays to identify additional ESC disease resistance QTL, especially those at the species level. Two genomic regions on LG1 and LG23 were determined to be significantly associated with ESC resistance as revealed by a mixed linear model and family-based association test. Examination of the resistance alleles indicated their origin from blue catfish, indicating that at least two major disease resistance loci exist among blue catfish populations. Upon further validation, markers linked with major ESC disease resistance QTL should be useful for marker-assisted introgression, allowing development of highly ESC resistant breeds of catfish.
Collapse
|
45
|
Affiliation(s)
- Byron B. Au-Yeung
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Neel H. Shah
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA
| | - Lin Shen
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, California 94143, USA;,
| | - Arthur Weiss
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, California 94143, USA;,
- Howard Hughes Medical Institute, University of California, San Francisco, California 94143, USA
| |
Collapse
|
46
|
Abstract
Natural killer (NK) cells express an array of germ-line encoded receptors that are capable of triggering cytotoxicity. NK cells tend to express many members of a given family of signalling molecules. The presence of many activating receptors and many members of a given family of signalling molecules can enable NK cells to detect different kinds of target cells, and to mount different kinds of responses. This contributes also to the robustness of NK cells responses; cytotoxic functions of NK cells often remain unaffected in the absence of selected signalling molecules. NK cells express many MHC-I-specific inhibitory receptors. Signals from MHC-I-specific inhibitory receptors tightly control NK cell cytotoxicity and, paradoxically, maintain NK cells in a state of proper responsiveness. This review provides a brief overview of the events that underlie NK cell activation, and how signals from inhibitory receptors intercept NK cell activation to prevent inappropriate triggering of cytotoxicity.
Collapse
Affiliation(s)
- Santosh Kumar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, Telangana, India
| |
Collapse
|
47
|
Genome-wide CRISPR screen identifies FAM49B as a key regulator of actin dynamics and T cell activation. Proc Natl Acad Sci U S A 2018; 115:E4051-E4060. [PMID: 29632189 DOI: 10.1073/pnas.1801340115] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Despite decades of research, mechanisms controlling T cell activation remain only partially understood, which hampers T cell-based immune cancer therapies. Here, we performed a genome-wide CRISPR screen to search for genes that regulate T cell activation. Our screen confirmed many of the known regulators in proximal T cell receptor signaling and, importantly, also uncovered a previously uncharacterized regulator, FAM49B (family with sequence similarity 49 member B). FAM49B deficiency led to hyperactivation of Jurkat T cells following T cell receptor stimulation, as indicated by enhancement of CD69 induction, PAK phosphorylation, and actin assembly. FAM49B directly interacted with the active form of the small GTPase Rac, and genetic disruption of the FAM49B-Rac interaction compromised FAM49B function. Thus, FAM49B inhibits T cell activation by repressing Rac activity and modulating cytoskeleton reorganization.
Collapse
|
48
|
Abstract
CD28 superagonistic antibodies (CD28SAb) can preferentially activate and expand immunosuppressive regulatory T cells (Treg) in mice. However, pre-clinical trials assessing CD28SAbs for the therapy of autoimmune diseases reveal severe systemic inflammatory response syndrome in humans, thereby implying the existence of distinct signalling abilities between human and mouse CD28. Here, we show that a single amino acid variant within the C-terminal proline-rich motif of human and mouse CD28 (P212 in human vs. A210 in mouse) regulates CD28-induced NF-κB activation and pro-inflammatory cytokine gene expression. Moreover, this Y209APP212 sequence in humans is crucial for the association of CD28 with the Nck adaptor protein for actin cytoskeleton reorganisation events necessary for CD28 autonomous signalling. This study thus unveils different outcomes between human and mouse CD28 signalling to underscore the importance of species difference when transferring results from preclinical models to the bedside. CD28 transmits co-stimulatory signals for the activation of both mouse and human T cells, but in vivo hyperactivation of CD28 has opposite effects on system immunity. Here, the authors show that a single amino acid difference between mouse and human CD28 dictates this function distinction via differential recruitment of Nck.
Collapse
|
49
|
Acevedo A, González-Billault C. Crosstalk between Rac1-mediated actin regulation and ROS production. Free Radic Biol Med 2018; 116:101-113. [PMID: 29330095 DOI: 10.1016/j.freeradbiomed.2018.01.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/03/2018] [Accepted: 01/05/2018] [Indexed: 02/08/2023]
Abstract
The small RhoGTPase Rac1 is implicated in a variety of events related to actin cytoskeleton rearrangement. Remarkably, another event that is completely different from those related to actin regulation has the same relevance; the Rac1-mediated production of reactive oxygen species (ROS) through NADPH oxidases (NOX). Each outcome involves different Rac1 downstream effectors; on one hand, events related to the actin cytoskeleton require Rac1 to bind to WAVEs proteins and PAKs that ultimately promote actin branching and turnover, on the other, NOX-derived ROS production demands active Rac1 to be bound to a cytosolic activator of NOX. How Rac1-mediated signaling ends up promoting actin-related events, NOX-derived ROS, or both is poorly understood. Rac1 regulators, including scaffold proteins, are known to exert tight control over its functions. Hence, evidence of Rac1 regulatory events leading to both actin remodeling and NOX-mediated ROS generation are discussed. Moreover, cellular functions linked to physiological and pathological conditions that exhibit crosstalk between Rac1 outcomes are analyzed, while plausible roles in neuronal functions (and dysfunctions) are highlighted. Together, discussed evidence shed light on cellular mechanisms which requires Rac1 to direct either actin- and/or ROS-related events, helping to understand crucial roles of Rac1 dual functionality.
Collapse
Affiliation(s)
- Alejandro Acevedo
- FONDAP Geroscience Center for Brain Health and Metabolism, Santiago, Chile.
| | - Christian González-Billault
- FONDAP Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Department of Biology, Faculty of Sciences, Universidad de Chile, 7800024, Chile; The Buck Institute for Research on Aging, Novato, USA.
| |
Collapse
|
50
|
Kabanova A, Zurli V, Baldari CT. Signals Controlling Lytic Granule Polarization at the Cytotoxic Immune Synapse. Front Immunol 2018. [PMID: 29515593 PMCID: PMC5826174 DOI: 10.3389/fimmu.2018.00307] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cytotoxic immunity relies on specialized effector T cells, the cytotoxic T cells, which are endowed with specialized cytolytic machinery that permits them to induce death of their targets. Upon recognition of a target cell, cytotoxic T cells form a lytic immune synapse and by docking the microtubule-organizing center at the synaptic membrane get prepared to deliver a lethal hit of enzymes contained in lytic granules. New insights suggest that the directionality of lytic granule trafficking along the microtubules represents a fine means to tune the functional outcome of the encounter between a T cell and its target. Thus, mechanisms regulating the directionality of granule transport may have a major impact in settings characterized by evasion from the cytotoxic response, such as chronic infection and cancer. Here, we review our current knowledge on the signaling pathways implicated in the polarized trafficking at the immune synapse of cytotoxic T cells, complementing it with information on the regulation of this process in natural killer cells. Furthermore, we highlight some of the parameters which we consider critical in studying the polarized trafficking of lytic granules, including the use of freshly isolated cytotoxic T cells, and discuss some of the major open questions.
Collapse
Affiliation(s)
- Anna Kabanova
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Vanessa Zurli
- Department of Life Sciences, University of Siena, Siena, Italy
| | | |
Collapse
|