1
|
Kumar S, Mahendiran S, Nair RS, Vyas H, Singh SK, Srivastava P, Jha S, Rana B, Rana A. A mechanistic, functional, and clinical perspective on targeting CD70 in cancer. Cancer Lett 2024; 611:217428. [PMID: 39725151 DOI: 10.1016/j.canlet.2024.217428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
The oncoimmunology research has witnessed notable advancements in recent years. Reshaping the tumor microenvironment (TME) approach is an effective method to improve antitumor immune response. The T cell-mediated antitumor response is crucial for favorable therapeutic outcomes in several cancers. The United States Food and Drug Administration (FDA) has approved immune checkpoint inhibitors (ICIs) for targeting the immune checkpoint proteins (ICPs) expressed in various hematological and solid malignancies. The ICPs are T cell co-inhibitory molecules that block T cell activation and, thus, antitumor response. Currently, most of the FDA-approved ICIs are antagonistic antibodies of programmed death-ligand 1 (PD-L1), programmed cell death protein 1 (PD-1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). In contrast to ICPs, the T cell costimulatory molecules are required for T cell activation, expansion, and effector function. However, the abrupt expression of these costimulatory molecules in tumors presents a concern for T cell-mediated antitumor response. One of the T cell costimulatory molecules, the cluster of differentiation 70 (CD70), has emerged as a druggable target in various hematological and solid malignancies due to its role in T cell effector function and immune evasion. The present review describes the expression of CD70, factors affecting the CD70 expression, the physiological and clinical relevance of CD70, and the current approaches to target CD70 in hematological and solid malignancies.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois Chicago, Chicago, IL, 60612, USA.
| | - Sowdhamini Mahendiran
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Rakesh Sathish Nair
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Harsh Vyas
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Piush Srivastava
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Saket Jha
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois Chicago, Chicago, IL, 60612, USA; Research Unit, Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois Chicago, Chicago, IL, 60612, USA; Research Unit, Jesse Brown VA Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
2
|
Koo J, Sim WJ, Lim W, Lim TG. Activation of mixed lineage kinase 3 by fine particulate matter induces skin inflammation in human keratinocytes. Toxicol Lett 2024; 402:38-43. [PMID: 39547318 DOI: 10.1016/j.toxlet.2024.11.002] [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: 06/30/2024] [Revised: 10/18/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Fine particulate matter (PM2.5) induces a range of diseases, including skin disorders, through inflammatory responses. In this study, we investigated the novel mechanisms by which PM2.5 causes skin inflammation in human keratinocytes HaCaT. We observed increased protein expression of cyclooxygenase-2 (COX-2) and the production of prostaglandin E2 (PGE2) in PM2.5-treated HaCaT cells. To identify the pathways promoting the expression of these inflammatory proteins, we conducted a phospho-kinase antibody array and confirmed that the phosphorylation levels of JNK and p38 were increased by PM2.5-treated HaCaT cells. Further investigation of the phosphorylation levels of mitogen-activated protein kinases (MAPKs) and upstream signals revealed that PM2.5 activated the MKK4/7-JNK-c-Jun and MKK3/6-p38-p70S6K signaling pathways, while the phosphorylation level of ERK1/2 remained unchanged. HaCaT cells treated with PM2.5 phosphorylated Mixed-lineage kinase 3 (MLK3), an upstream regulator of p38 and JNK. Furthermore, inhibition of ROS production by N-Acetylcysteine (NAC) treatment inhibited MLK3 phosphorylation. Taken together, ROS production induced by PM2.5 activated the MLK3 signaling pathway and induced skin inflammation.
Collapse
Affiliation(s)
- Jamyeong Koo
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Woo-Jin Sim
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Wonchul Lim
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea; Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea.
| | - Tae-Gyu Lim
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea; Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea.
| |
Collapse
|
3
|
Ke R, Kumar S, Singh SK, Rana A, Rana B. Molecular insights into the role of mixed lineage kinase 3 in cancer hallmarks. Biochim Biophys Acta Rev Cancer 2024; 1879:189157. [PMID: 39032538 DOI: 10.1016/j.bbcan.2024.189157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Mixed-lineage kinase 3 (MLK3) is a serine/threonine kinase of the MAPK Kinase kinase (MAP3K) family that plays critical roles in various biological processes, including cancer. Upon activation, MLK3 differentially activates downstream MAPKs, such as JNK, p38, and ERK. In addition, it regulates various non-canonical signaling pathways, such as β-catenin, AMPK, Pin1, and PAK1, to regulate cell proliferation, apoptosis, invasion, and metastasis. Recent studies have also uncovered other potentially diverse roles of MLK3 in malignancy, which include metabolic reprogramming, cancer-associated inflammation, and evasion of cancer-related immune surveillance. The role of MLK3 in cancer is complex and cancer-specific, and an understanding of its function at the molecular level aligned specifically with the cancer hallmarks will have profound therapeutic implications for diagnosing and treating MLK3-dependent cancers. This review summarizes the current knowledge about the effect of MLK3 on the hallmarks of cancer, providing insights into its potential as a promising anticancer drug target.
Collapse
Affiliation(s)
- Rong Ke
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
4
|
Li H, Terrando N, Gelbard HA. Infectious Diseases. ADVANCES IN NEUROBIOLOGY 2024; 37:423-444. [PMID: 39207706 PMCID: PMC11556852 DOI: 10.1007/978-3-031-55529-9_24] [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] [Indexed: 09/04/2024]
Abstract
Microglia, brain-resident innate immune cells, have been extensively studied in neurodegenerative contexts like Alzheimer's disease. The Coronavirus disease 2019 (COVID-19) pandemic highlighted how peripheral infection and inflammation can be detrimental to the neuroimmune milieu and initiate microgliosis driven by peripheral inflammation. Microglia can remain deleterious to brain health by sustaining inflammation in the central nervous system even after the clearance of the original immunogenic agents. In this chapter, we discuss how pulmonary infection with Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) can lead to neurovascular and neuroimmune inflammation causing the neurological syndrome of post-acute sequelae of COVID-19 (PASC). Further, we incorporate lessons from the Human Immunodeficiency Virus' (HIV's) effects on microglial functioning in the era of combined antiretroviral therapies (cART) that contribute to HIV-1 associated neurocognitive disorders (HAND). Finally, we describe roles for mixed lineage kinase 3 (MLK3) and leucine-rich repeat kinase (LRRK2) as key regulators of multiple inflammatory and apoptotic pathways important to the pathogenesis of PASC and HAND. Inhibition of these pathways provides a therapeutically synergistic method of treating both PASC and HAND.
Collapse
Affiliation(s)
- Herman Li
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Niccolò Terrando
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Harris A Gelbard
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
5
|
cIAP1/TRAF2 interplay promotes tumor growth through the activation of STAT3. Oncogene 2023; 42:198-208. [PMID: 36400972 DOI: 10.1038/s41388-022-02544-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022]
Abstract
Cellular inhibitor of apoptosis-1 (cIAP1) is a signaling regulator with oncogenic properties. It is involved in the regulation of signaling pathways controlling inflammation, cell survival, proliferation, differentiation and motility. It is recruited into membrane-receptor-associated signaling complexes thanks to the molecular adaptor TRAF2. However, the cIAP1/TRAF2 complex exists, independently of receptor engagement, in several subcellular compartments. The present work strengthens the importance of TRAF2 in the oncogenic properties of cIAP1. cIAPs-deficient mouse embryonic fibroblasts (MEFs) were transformed using the HRas-V12 oncogene. Re-expression of cIAP1 enhanced tumor growth in a nude mice xenograft model, and promoted lung tumor nodes formation. Deletion or mutation of the TRAF2-binding site completely abolished the oncogenic properties of cIAP1. Further, cIAP1 mediated the clustering of TRAF2, which was sufficient to stimulate tumor growth. Our TRAF2 interactome analysis showed that cIAP1 was critical for TRAF2 to bind to its protein partners. Thus, cIAP1 and TRAF2 would be two essential subunits of a signaling complex promoting a pro-tumoral signal. cIAP1/TRAF2 promoted the activation of the canonical NF-κB and ERK1/2 signaling pathways. NF-κB-dependent production of IL-6 triggered the activation of the JAK/STAT3 axis in an autocrine manner. Inhibition or downregulation of STAT3 specifically compromised the growth of cIAP1-restored MEFs but not that of MEFs expressing a cIAP1-mutant and treating mice with the STAT3 inhibitor niclosamide completely abrogated cIAP1/TRAF2-mediated tumor growth. Altogether, we demonstrate that cIAP1/TRAF2 binding is essential to promote tumor growth via the activation of the JAK/STAT3 signaling pathway.
Collapse
|
6
|
Siegmund D, Wagner J, Wajant H. TNF Receptor Associated Factor 2 (TRAF2) Signaling in Cancer. Cancers (Basel) 2022; 14:cancers14164055. [PMID: 36011046 PMCID: PMC9406534 DOI: 10.3390/cancers14164055] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/05/2022] [Accepted: 08/19/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Tumor necrosis factor (TNF) receptor associated factor-2 (TRAF2) is an intracellular adapter protein with E3 ligase activity, which interacts with a plethora of other signaling proteins, including plasma membrane receptors, kinases, phosphatases, other E3 ligases, and deubiquitinases. TRAF2 is involved in various cancer-relevant cellular processes, such as the activation of transcription factors of the NFκB family, stimulation of mitogen-activated protein (MAP) kinase cascades, endoplasmic reticulum (ER) stress signaling, autophagy, and the control of cell death programs. In a context-dependent manner, TRAF2 promotes tumor development but it can also act as a tumor suppressor. Based on a general description, how TRAF2 in concert with TRAF2-interacting proteins and other TRAF proteins act at the molecular level is discussed for its importance for tumor development and its potential usefulness as a therapeutic target in cancer therapy. Abstract Tumor necrosis factor (TNF) receptor associated factor-2 (TRAF2) has been originally identified as a protein interacting with TNF receptor 2 (TNFR2) but also binds to several other receptors of the TNF receptor superfamily (TNFRSF). TRAF2, often in concert with other members of the TRAF protein family, is involved in the activation of the classical NFκB pathway and the stimulation of various mitogen-activated protein (MAP) kinase cascades by TNFRSF receptors (TNFRs), but is also required to inhibit the alternative NFκB pathway. TRAF2 has also been implicated in endoplasmic reticulum (ER) stress signaling, the regulation of autophagy, and the control of cell death programs. TRAF2 fulfills its functions by acting as a scaffold, bringing together the E3 ligase cellular inhibitor of apoptosis-1 (cIAP1) and cIAP2 with their substrates and various regulatory proteins, e.g., deubiquitinases. Furthermore, TRAF2 can act as an E3 ligase by help of its N-terminal really interesting new gene (RING) domain. The finding that TRAF2 (but also several other members of the TRAF family) interacts with the latent membrane protein 1 (LMP1) oncogene of the Epstein–Barr virus (EBV) indicated early on that TRAF2 could play a role in the oncogenesis of B-cell malignancies and EBV-associated non-keratinizing nasopharyngeal carcinoma (NPC). TRAF2 can also act as an oncogene in solid tumors, e.g., in colon cancer by promoting Wnt/β-catenin signaling. Moreover, tumor cell-expressed TRAF2 has been identified as a major factor-limiting cancer cell killing by cytotoxic T-cells after immune checkpoint blockade. However, TRAF2 can also be context-dependent as a tumor suppressor, presumably by virtue of its inhibitory effect on the alternative NFκB pathway. For example, inactivating mutations of TRAF2 have been associated with tumor development, e.g., in multiple myeloma and mantle cell lymphoma. In this review, we summarize the various TRAF2-related signaling pathways and their relevance for the oncogenic and tumor suppressive activities of TRAF2. Particularly, we discuss currently emerging concepts to target TRAF2 for therapeutic purposes.
Collapse
|
7
|
Cedeno-Rosario L, Honda D, Sunderland AM, Lewandowski MD, Taylor WR, Chadee DN. Phosphorylation of mixed lineage kinase MLK3 by cyclin-dependent kinases CDK1 and CDK2 controls ovarian cancer cell division. J Biol Chem 2022; 298:102263. [PMID: 35843311 PMCID: PMC9399292 DOI: 10.1016/j.jbc.2022.102263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 11/03/2022] Open
Abstract
Mixed lineage kinase 3 (MLK3) is a serine/threonine mitogen-activated protein kinase kinase kinase that promotes the activation of multiple mitogen-activated protein kinase pathways and is required for invasion and proliferation of ovarian cancer cells. Inhibition of MLK activity causes G2/M arrest in HeLa cells; however, the regulation of MLK3 during ovarian cancer cell cycle progression is not known. Here, we found that MLK3 is phosphorylated in mitosis and that inhibition of cyclin-dependent kinase 1 (CDK1) prevented MLK3 phosphorylation. In addition, we observed that c-Jun N-terminal kinase, a downstream target of MLK3 and a direct target of MKK4 (SEK1), was activated in G2 phase when CDK2 activity is increased and then inactivated at the beginning of mitosis concurrent with the increase in CDK1 and MLK3 phosphorylation. Using in vitro kinase assays and phosphomutants, we determined that CDK1 phosphorylates MLK3 on Ser548 and decreases MLK3 activity during mitosis, whereas CDK2 phosphorylates MLK3 on Ser770 and increases MLK3 activity during G1/S and G2 phases. We also found that MLK3 inhibition causes a reduction in cell proliferation and a cell cycle arrest in ovarian cancer cells, suggesting that MLK3 is required for ovarian cancer cell cycle progression. Taken together, our results suggest that phosphorylation of MLK3 by CDK1 and CDK2 is important for the regulation of MLK3 and c-Jun N-terminal kinase activities during G1/S, G2, and M phases in ovarian cancer cell division.
Collapse
Affiliation(s)
- Luis Cedeno-Rosario
- Department of Biological Sciences, College of Natural Sciences and Mathematics, The University of Toledo, Toledo, Ohio, USA
| | - David Honda
- Department of Biological Sciences, College of Natural Sciences and Mathematics, The University of Toledo, Toledo, Ohio, USA
| | - Autumn M Sunderland
- Department of Biological Sciences, College of Natural Sciences and Mathematics, The University of Toledo, Toledo, Ohio, USA
| | - Mark D Lewandowski
- Department of Biological Sciences, College of Natural Sciences and Mathematics, The University of Toledo, Toledo, Ohio, USA
| | - William R Taylor
- Department of Biological Sciences, College of Natural Sciences and Mathematics, The University of Toledo, Toledo, Ohio, USA
| | - Deborah N Chadee
- Department of Biological Sciences, College of Natural Sciences and Mathematics, The University of Toledo, Toledo, Ohio, USA.
| |
Collapse
|
8
|
Calamaras TD, Pande S, Baumgartner RA, Kim SK, McCarthy JC, Martin GL, Tam K, McLaughlin AL, Wang GR, Aronovitz MJ, Lin W, Aguirre JI, Baca P, Liu P, Richards DA, Davis RJ, Karas RH, Jaffe IZ, Blanton RM. MLK3 mediates impact of PKG1α on cardiac function and controls blood pressure through separate mechanisms. JCI Insight 2021; 6:e149075. [PMID: 34324442 PMCID: PMC8492323 DOI: 10.1172/jci.insight.149075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
cGMP-dependent protein kinase 1α (PKG1α) promotes left ventricle (LV) compensation after pressure overload. PKG1-activating drugs improve heart failure (HF) outcomes but are limited by vasodilation-induced hypotension. Signaling molecules that mediate PKG1α cardiac therapeutic effects but do not promote PKG1α-induced hypotension could therefore represent improved therapeutic targets. We investigated roles of mixed lineage kinase 3 (MLK3) in mediating PKG1α effects on LV function after pressure overload and in regulating BP. In a transaortic constriction HF model, PKG activation with sildenafil preserved LV function in MLK3+/+ but not MLK3-/- littermates. MLK3 coimmunoprecipitated with PKG1α. MLK3-PKG1α cointeraction decreased in failing LVs. PKG1α phosphorylated MLK3 on Thr277/Ser281 sites required for kinase activation. MLK3-/- mice displayed hypertension and increased arterial stiffness, though PKG stimulation with sildenafil or the soluble guanylate cyclase (sGC) stimulator BAY41-2272 still reduced BP in MLK3-/- mice. MLK3 kinase inhibition with URMC-099 did not affect BP but induced LV dysfunction in mice. These data reveal MLK3 as a PKG1α substrate mediating PKG1α preservation of LV function but not acute PKG1α BP effects. Mechanistically, MLK3 kinase-dependent effects preserved LV function, whereas MLK3 kinase-independent signaling regulated BP. These findings suggest augmenting MLK3 kinase activity could preserve LV function in HF but avoid hypotension from PKG1α activation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kelly Tam
- Molecular Cardiology Research Institute and
| | | | | | | | - Weiyu Lin
- Molecular Cardiology Research Institute and
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | | | - Paulina Baca
- Molecular Cardiology Research Institute and
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Peiwen Liu
- Molecular Cardiology Research Institute and
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | | | - Roger J. Davis
- University of Massachusetts School of Medicine, Worchester, Massachusetts, USA
| | | | - Iris Z. Jaffe
- Molecular Cardiology Research Institute and
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Robert M. Blanton
- Molecular Cardiology Research Institute and
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Transmembrane TNF and Its Receptors TNFR1 and TNFR2 in Mycobacterial Infections. Int J Mol Sci 2021; 22:ijms22115461. [PMID: 34067256 PMCID: PMC8196896 DOI: 10.3390/ijms22115461] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
Tumor necrosis factor (TNF) is one of the main cytokines regulating a pro-inflammatory environment. It has been related to several cell functions, for instance, phagocytosis, apoptosis, proliferation, mitochondrial dynamic. Moreover, during mycobacterial infections, TNF plays an essential role to maintain granuloma formation. Several effector mechanisms have been implicated according to the interactions of the two active forms, soluble TNF (solTNF) and transmembrane TNF (tmTNF), with their receptors TNFR1 and TNFR2. We review the impact of these interactions in the context of mycobacterial infections. TNF is tightly regulated by binding to receptors, however, during mycobacterial infections, upstream activation signalling pathways may be influenced by key regulatory factors either at the membrane or cytosol level. Detailing the structure and activation pathways used by TNF and its receptors, such as its interaction with solTNF/TNFRs versus tmTNF/TNFRs, may bring a better understanding of the molecular mechanisms involved in activation pathways which can be helpful for the development of new therapies aimed at being more efficient against mycobacterial infections.
Collapse
|
10
|
Gallo KA, Ellsworth E, Stoub H, Conrad SE. Therapeutic potential of targeting mixed lineage kinases in cancer and inflammation. Pharmacol Ther 2019; 207:107457. [PMID: 31863814 DOI: 10.1016/j.pharmthera.2019.107457] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022]
Abstract
Dysregulation of intracellular signaling pathways is a key attribute of diseases associated with chronic inflammation, including cancer. Mitogen activated protein kinases have emerged as critical conduits of intracellular signal transmission, yet due to their ubiquitous roles in cellular processes, their direct inhibition may lead to undesired effects, thus limiting their usefulness as therapeutic targets. Mixed lineage kinases (MLKs) are mitogen-activated protein kinase kinase kinases (MAP3Ks) that interact with scaffolding proteins and function upstream of p38, JNK, ERK, and NF-kappaB to mediate diverse cellular signals. Studies involving gene silencing, genetically engineered mouse models, and small molecule inhibitors suggest that MLKs are critical in tumor progression as well as in inflammatory processes. Recent advances indicate that they may be useful targets in some types of cancer and in diseases driven by chronic inflammation including neurodegenerative diseases and metabolic diseases such as nonalcoholic steatohepatitis. This review describes existing MLK inhibitors, the roles of MLKs in various aspects of tumor progression and in the control of inflammatory processes, and the potential for therapeutic targeting of MLKs.
Collapse
Affiliation(s)
- Kathleen A Gallo
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA.
| | - Edmund Ellsworth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Hayden Stoub
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Susan E Conrad
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
11
|
He X, Liu J, Liang C, Badshah SA, Zheng K, Dang L, Guo B, Li D, Lu C, Guo Q, Fan D, Bian Y, Feng H, Xiao L, Pan X, Xiao C, Zhang B, Zhang G, Lu A. Osteoblastic PLEKHO1 contributes to joint inflammation in rheumatoid arthritis. EBioMedicine 2019; 41:538-555. [PMID: 30824383 PMCID: PMC6442230 DOI: 10.1016/j.ebiom.2019.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/30/2019] [Accepted: 02/05/2019] [Indexed: 12/29/2022] Open
Abstract
Background Osteoblasts participating in the inflammation regulation gradually obtain concerns. However, its role in joint inflammation of rheumatoid arthritis (RA) is largely unknown. Here, we investigated the role of osteoblastic pleckstrin homology domain-containing family O member 1 (PLEKHO1), a negative regulator of osteogenic lineage activity, in regulating joint inflammation in RA. Methods The level of osteoblastic PLEKHO1 in RA patients and collagen-induced arthritis (CIA) mice was examined. The role of osteoblastic PLEKHO1 in joint inflammation was evaluated by a CIA model and a K/BxN serum-transfer arthritis (STA) model which were induced in osteoblast-specific Plekho1 conditional knockout mice and mice expressing high Plekho1 exclusively in osteoblasts, respectively. The effect of osteoblastic PLEKHO1 inhibition was explored in a CIA mice model and a non-human primate arthritis model. The mechanism of osteoblastic PLEKHO1 in regulating joint inflammation were performed by a series of in vitro studies. Results PLEKHO1 was highly expressed in osteoblasts from RA patients and CIA mice. Osteoblastic Plekho1 deletion ameliorated joint inflammation, whereas overexpressing Plekho1 only within osteoblasts exacerbated local inflammation in CIA mice and STA mice. PLEKHO1 was required for TRAF2-mediated RIP1 ubiquitination to activate NF-κB for inducing inflammatory cytokines production in osteoblasts. Moreover, osteoblastic PLEKHO1 inhibition diminished joint inflammation and promoted bone formation in CIA mice and non-human primate arthritis model. Conclusions These data strongly suggest that the highly expressed PLEKHO1 in osteoblasts contributes to joint inflammation in RA. Targeting osteoblastic PLEKHO1 may exert dual therapeutic action of alleviating joint inflammation and promoting bone formation in RA.
Collapse
Affiliation(s)
- Xiaojuan He
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Precision Medicine and Innovative Drug Discovery, Hong Kong Baptist University, Hong Kong, China; Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Precision Medicine and Innovative Drug Discovery, Hong Kong Baptist University, Hong Kong, China
| | - Chao Liang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Precision Medicine and Innovative Drug Discovery, Hong Kong Baptist University, Hong Kong, China
| | - Shaikh Atik Badshah
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Precision Medicine and Innovative Drug Discovery, Hong Kong Baptist University, Hong Kong, China
| | - Kang Zheng
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lei Dang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Precision Medicine and Innovative Drug Discovery, Hong Kong Baptist University, Hong Kong, China
| | - Baosheng Guo
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Precision Medicine and Innovative Drug Discovery, Hong Kong Baptist University, Hong Kong, China
| | - Defang Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Precision Medicine and Innovative Drug Discovery, Hong Kong Baptist University, Hong Kong, China
| | - Cheng Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingqing Guo
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Danping Fan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanqin Bian
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Guanghua Integrative Medicine Hospital/Shanghai University of TCM, Shanghai, China
| | - Hui Feng
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Guanghua Integrative Medicine Hospital/Shanghai University of TCM, Shanghai, China
| | - Lianbo Xiao
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Guanghua Integrative Medicine Hospital/Shanghai University of TCM, Shanghai, China
| | - Xiaohua Pan
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Department of Orthopaedics and Traumatology, Bao-an Hospital Affiliated to Southern Medical University & Shenzhen 8th People Hospital, Shenzhen, China
| | - Cheng Xiao
- Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - BaoTing Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Precision Medicine and Innovative Drug Discovery, Hong Kong Baptist University, Hong Kong, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Institute of Precision Medicine and Innovative Drug Discovery, Hong Kong Baptist University, Hong Kong, China; School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
12
|
Prinz E, Aviram S, Aronheim A. WDR62 mediates TNFα-dependent JNK activation via TRAF2-MLK3 axis. Mol Biol Cell 2018; 29:2470-2480. [PMID: 30091641 PMCID: PMC6233063 DOI: 10.1091/mbc.e17-08-0504] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 11/11/2022] Open
Abstract
The mitogen-activated protein kinases (MAPKs) regulate a variety of cellular processes. The three main MAPK cascades are the extracellular signal-regulated kinases (ERK), c-Jun N-terminal kinase (JNK), and p38 kinases. A typical MAPK cascade is composed of MAP3K-MAP2K-MAPK kinases that are held by scaffold proteins. Scaffolds function to assemble the protein tier and contribute to the specificity and efficacy of signal transmission. WD repeat domain 62 (WDR62) is a JNK scaffold protein, interacting with JNK, MKK7, and several MAP3Ks. The loss of WDR62 in human leads to microcephaly and pachygyria. Yet the role of WDR62 in cellular function is not fully studied. We used the CRISPR/Cas9 and short hairpin RNA approaches to establish a human breast cancer cell line MDA-MB-231 with WDR62 loss of function and studied the consequence to JNK signaling. In growing cells, WDR62 is responsible for the basal expression of c-Jun. In stressed cells, WDR62 specifically mediates TNFα-dependent JNK activation through the association with both the adaptor protein, TNF receptor-associated factor 2 (TRAF2), and the MAP3K protein, mixed lineage kinase 3. TNFα-dependent JNK activation is mediated by WDR62 in HCT116 and HeLa cell lines as well. MDA-MB-231 WDR62-knockout cells display increased resistance to TNFα-induced cell death. Collectively, WDR62 coordinates the TNFα receptor signaling pathway to JNK activation through association with multiple kinases and the adaptor protein TRAF2.
Collapse
Affiliation(s)
- Elad Prinz
- Department of Cell Biology and Cancer Science, B. Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 31096, Israel
| | - Sharon Aviram
- Department of Cell Biology and Cancer Science, B. Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 31096, Israel
| | - Ami Aronheim
- Department of Cell Biology and Cancer Science, B. Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
13
|
Moon J, Ha J, Park SH. Identification of PTPN1 as a novel negative regulator of the JNK MAPK pathway using a synthetic screening for pathway-specific phosphatases. Sci Rep 2017; 7:12974. [PMID: 29021559 PMCID: PMC5636874 DOI: 10.1038/s41598-017-13494-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/25/2017] [Indexed: 12/19/2022] Open
Abstract
The mitogen activated protein kinase (MAPK) signaling cascades transmit extracellular stimulations to generate various cellular responses via the sequential and reversible phosphorylation of kinases. Since the strength and duration of kinase phosphorylation within the pathway determine the cellular response, both kinases and phosphatases play an essential role in the precise control of MAPK pathway activation and attenuation. Thus, the identification of pathway-specific phosphatases is critical for understanding the functional mechanisms by which the MAPK pathway is regulated. To identify phosphatases specific to the c-Jun N-terminal kinase (JNK) MAPK pathway, a synthetic screening approach was utilized in which phosphatases were individually tethered to the JNK pathway specific-JIP1 scaffold protein. Of 77 mammalian phosphatases tested, PTPN1 led to the inhibition of JNK pathway activation. Further analyses revealed that of three pathway member kinases, PTPN1 directly dephosphorylates JNK, the terminal kinase of the pathway, and negatively regulates the JNK MAPK pathway. Specifically, PTPN1 appears to regulate the overall signaling magnitude, rather than the adaptation timing, suggesting that PTPN1 might be involved in the control and maintenance of signaling noise. Finally, the negative regulation of the JNK MAPK pathway by PTPN1 was found to reduce the tumor necrosis factor α (TNFα)-dependent cell death response.
Collapse
Affiliation(s)
- Jiyoung Moon
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Korea
| | - Jain Ha
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Korea
| | - Sang-Hyun Park
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
14
|
Rattanasinchai C, Gallo KA. MLK3 Signaling in Cancer Invasion. Cancers (Basel) 2016; 8:cancers8050051. [PMID: 27213454 PMCID: PMC4880868 DOI: 10.3390/cancers8050051] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/05/2016] [Accepted: 05/10/2016] [Indexed: 02/07/2023] Open
Abstract
Mixed-lineage kinase 3 (MLK3) was first cloned in 1994; however, only in the past decade has MLK3 become recognized as a player in oncogenic signaling. MLK3 is a mitogen-activated protein kinase kinase kinase (MAP3K) that mediates signals from several cell surface receptors including receptor tyrosine kinases (RTKs), chemokine receptors, and cytokine receptors. Once activated, MLK3 transduces signals to multiple downstream pathways, primarily to c-Jun terminal kinase (JNK) MAPK, as well as to extracellular-signal-regulated kinase (ERK) MAPK, P38 MAPK, and NF-κB, resulting in both transcriptional and post-translational regulation of multiple effector proteins. In several types of cancer, MLK3 signaling is implicated in promoting cell proliferation, as well as driving cell migration, invasion and metastasis.
Collapse
Affiliation(s)
| | - Kathleen A Gallo
- Cell and Molecular Biology program, Michigan State University, East Lansing, MI 48824, USA.
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
15
|
Ceccarelli A, Di Venere A, Nicolai E, De Luca A, Minicozzi V, Rosato N, Caccuri AM, Mei G. TNFR-Associated Factor-2 (TRAF2): Not Only a Trimer. Biochemistry 2015; 54:6153-61. [PMID: 26390021 DOI: 10.1021/acs.biochem.5b00674] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
TNF receptor-associated factors (TRAFs) are characterized by an oligomeric structure that plays a fundamental role in the binding process with membrane receptors. In this work, we studied the trimer-to-monomer (T ↔ 3M) equilibrium transition of the TRAF2 C-terminal domain using both chemical (dilution/guanidinium hydrochloride) and mechanical stress (high pressure) to induce the dissociation of the native protein into subunits. The experimental results and computer simulations indicate that stable monomers exist and that their population accounts for 15% of the total TRAF2 molecules already at a physiological intracellular concentration (≈1 μM), being instead the predominant species in the nanomolar concentration range. Because the total amount of TRAF2 changes during a cell cycle, the monomer-trimer equilibrium can be crucial for regulating the activities of TRAF2 in vivo.
Collapse
Affiliation(s)
- Arianna Ceccarelli
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata , Via Montpellier 1, 00133 Rome, Italy
| | - Almerinda Di Venere
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata , Via Montpellier 1, 00133 Rome, Italy.,The NAST Centre for Nanoscience & Nanotechnology & Innovative Instrumentation, University of Rome Tor Vergata , Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Eleonora Nicolai
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata , Via Montpellier 1, 00133 Rome, Italy.,The NAST Centre for Nanoscience & Nanotechnology & Innovative Instrumentation, University of Rome Tor Vergata , Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Anastasia De Luca
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata , Via Montpellier 1, 00133 Rome, Italy.,The NAST Centre for Nanoscience & Nanotechnology & Innovative Instrumentation, University of Rome Tor Vergata , Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Velia Minicozzi
- Physics Department, INFN University of Rome Tor Vergata , Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Nicola Rosato
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata , Via Montpellier 1, 00133 Rome, Italy.,The NAST Centre for Nanoscience & Nanotechnology & Innovative Instrumentation, University of Rome Tor Vergata , Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Anna Maria Caccuri
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata , Via Montpellier 1, 00133 Rome, Italy.,The NAST Centre for Nanoscience & Nanotechnology & Innovative Instrumentation, University of Rome Tor Vergata , Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Giampiero Mei
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata , Via Montpellier 1, 00133 Rome, Italy.,The NAST Centre for Nanoscience & Nanotechnology & Innovative Instrumentation, University of Rome Tor Vergata , Via della Ricerca Scientifica 1, 00133 Rome, Italy
| |
Collapse
|
16
|
Petersen SL, Chen TT, Lawrence DA, Marsters SA, Gonzalvez F, Ashkenazi A. TRAF2 is a biologically important necroptosis suppressor. Cell Death Differ 2015; 22:1846-57. [PMID: 25882049 DOI: 10.1038/cdd.2015.35] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/11/2015] [Accepted: 02/24/2015] [Indexed: 12/26/2022] Open
Abstract
Tumor necrosis factor α (TNFα) triggers necroptotic cell death through an intracellular signaling complex containing receptor-interacting protein kinase (RIPK) 1 and RIPK3, called the necrosome. RIPK1 phosphorylates RIPK3, which phosphorylates the pseudokinase mixed lineage kinase-domain-like (MLKL)-driving its oligomerization and membrane-disrupting necroptotic activity. Here, we show that TNF receptor-associated factor 2 (TRAF2)-previously implicated in apoptosis suppression-also inhibits necroptotic signaling by TNFα. TRAF2 disruption in mouse fibroblasts augmented TNFα-driven necrosome formation and RIPK3-MLKL association, promoting necroptosis. TRAF2 constitutively associated with MLKL, whereas TNFα reversed this via cylindromatosis-dependent TRAF2 deubiquitination. Ectopic interaction of TRAF2 and MLKL required the C-terminal portion but not the N-terminal, RING, or CIM region of TRAF2. Induced TRAF2 knockout (KO) in adult mice caused rapid lethality, in conjunction with increased hepatic necrosome assembly. By contrast, TRAF2 KO on a RIPK3 KO background caused delayed mortality, in concert with elevated intestinal caspase-8 protein and activity. Combined injection of TNFR1-Fc, Fas-Fc and DR5-Fc decoys prevented death upon TRAF2 KO. However, Fas-Fc and DR5-Fc were ineffective, whereas TNFR1-Fc and interferon α receptor (IFNAR1)-Fc were partially protective against lethality upon combined TRAF2 and RIPK3 KO. These results identify TRAF2 as an important biological suppressor of necroptosis in vitro and in vivo.
Collapse
Affiliation(s)
- S L Petersen
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - T T Chen
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - D A Lawrence
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - S A Marsters
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - F Gonzalvez
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - A Ashkenazi
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
17
|
Humphrey RK, Ray A, Gonuguntla S, Hao E, Jhala US. Loss of TRB3 alters dynamics of MLK3-JNK signaling and inhibits cytokine-activated pancreatic beta cell death. J Biol Chem 2014; 289:29994-30004. [PMID: 25204656 PMCID: PMC4208007 DOI: 10.1074/jbc.m114.575613] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/11/2014] [Indexed: 01/13/2023] Open
Abstract
Disabling cellular defense mechanisms is essential for induction of apoptosis. We have previously shown that cytokine-mediated activation of the MAP3K MLK3 stabilizes TRB3 protein levels to inhibit AKT and compromise beta cell survival. Here, we show that genetic deletion of TRB3 results in basal activation of AKT, preserves mitochondrial integrity, and confers resistance against cytokine-induced pancreatic beta cell death. Mechanistically, we find that TRB3 stabilizes MLK3, most likely by suppressing AKT-directed phosphorylation, ubiquitination, and proteasomal degradation of MLK3. Accordingly, TRB3(-/-) islets show a decrease in both the amplitude and duration of cytokine-stimulated MLK3 induction and JNK activation. It is well known that JNK signaling is facilitated by a feed forward loop of sequential kinase phosphorylation and is reinforced by a mutual stabilization of the module components. The failure of TRB3(-/-) islets to mount an optimal JNK activation response, coupled with the ability of TRB3 to engage and maintain steady state levels of MLK3, recasts TRB3 as an integral functional component of the JNK module in pancreatic beta cells.
Collapse
Affiliation(s)
- Rohan K Humphrey
- From the Pediatric Diabetes Research Center, University of California, San Diego School of Medicine, La Jolla, California 92037
| | - Anamika Ray
- From the Pediatric Diabetes Research Center, University of California, San Diego School of Medicine, La Jolla, California 92037
| | - Sumati Gonuguntla
- From the Pediatric Diabetes Research Center, University of California, San Diego School of Medicine, La Jolla, California 92037
| | - Ergeng Hao
- From the Pediatric Diabetes Research Center, University of California, San Diego School of Medicine, La Jolla, California 92037
| | - Ulupi S Jhala
- From the Pediatric Diabetes Research Center, University of California, San Diego School of Medicine, La Jolla, California 92037
| |
Collapse
|
18
|
Humphrey RK, Ray A, Gonuguntla S, Hao E, Jhala US. Loss of TRB3 alters dynamics of MLK3-JNK signaling and inhibits cytokine-activated pancreatic beta cell death. J Biol Chem 2014. [PMID: 25204656 DOI: 10.1074/jbc.m114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Disabling cellular defense mechanisms is essential for induction of apoptosis. We have previously shown that cytokine-mediated activation of the MAP3K MLK3 stabilizes TRB3 protein levels to inhibit AKT and compromise beta cell survival. Here, we show that genetic deletion of TRB3 results in basal activation of AKT, preserves mitochondrial integrity, and confers resistance against cytokine-induced pancreatic beta cell death. Mechanistically, we find that TRB3 stabilizes MLK3, most likely by suppressing AKT-directed phosphorylation, ubiquitination, and proteasomal degradation of MLK3. Accordingly, TRB3(-/-) islets show a decrease in both the amplitude and duration of cytokine-stimulated MLK3 induction and JNK activation. It is well known that JNK signaling is facilitated by a feed forward loop of sequential kinase phosphorylation and is reinforced by a mutual stabilization of the module components. The failure of TRB3(-/-) islets to mount an optimal JNK activation response, coupled with the ability of TRB3 to engage and maintain steady state levels of MLK3, recasts TRB3 as an integral functional component of the JNK module in pancreatic beta cells.
Collapse
Affiliation(s)
- Rohan K Humphrey
- From the Pediatric Diabetes Research Center, University of California, San Diego School of Medicine, La Jolla, California 92037
| | - Anamika Ray
- From the Pediatric Diabetes Research Center, University of California, San Diego School of Medicine, La Jolla, California 92037
| | - Sumati Gonuguntla
- From the Pediatric Diabetes Research Center, University of California, San Diego School of Medicine, La Jolla, California 92037
| | - Ergeng Hao
- From the Pediatric Diabetes Research Center, University of California, San Diego School of Medicine, La Jolla, California 92037
| | - Ulupi S Jhala
- From the Pediatric Diabetes Research Center, University of California, San Diego School of Medicine, La Jolla, California 92037
| |
Collapse
|
19
|
Blessing NA, Brockman AL, Chadee DN. The E3 ligase CHIP mediates ubiquitination and degradation of mixed-lineage kinase 3. Mol Cell Biol 2014; 34:3132-43. [PMID: 24912674 PMCID: PMC4135596 DOI: 10.1128/mcb.00296-14] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 03/30/2014] [Accepted: 05/28/2014] [Indexed: 01/30/2023] Open
Abstract
Mixed-lineage kinase 3 (MLK3) activates mitogen-activated protein kinase (MAPK) signaling pathways and has important functions in migration, invasion, proliferation, tumorigenesis, and apoptosis. We investigated the role of the E3 ligase carboxyl terminus of Hsc70-interacting protein (CHIP) in the regulation of MLK3 protein levels. We show that CHIP interacts with MLK3 and, together with the E2 ubiquitin-conjugating enzyme UbcH5 (UbcH5a, -b, -c, or -d), ubiquitinates MLK3 in vitro. CHIP or Hsp70 overexpression promoted endogenous MLK3 ubiquitination and induced a decline in MLK3 protein levels in cells with Hsp90 inhibition. Furthermore, CHIP overexpression caused a proteasome-dependent reduction in exogenous MLK3 protein. Geldanamycin (GA), heat shock, and osmotic shock treatments also reduced the level of MLK3 protein via a CHIP-dependent mechanism. In addition, CHIP depletion in ovarian cancer SKOV3 cells increased cell invasion, and the enhancement of invasiveness was abrogated by small interfering RNA (siRNA)-mediated knockdown of MLK3. Thus, CHIP modulates MLK3 protein levels in response to GA and stress stimuli, and CHIP-dependent regulation of MLK3 is required for suppression of SKOV3 ovarian cancer cell invasion.
Collapse
Affiliation(s)
- Natalya A Blessing
- Department of Biological Sciences, The University of Toledo, Toledo, Ohio, USA
| | - April L Brockman
- Department of Biological Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Deborah N Chadee
- Department of Biological Sciences, The University of Toledo, Toledo, Ohio, USA
| |
Collapse
|
20
|
Su X, Min S, Cao S, Yan H, Zhao Y, Li H, Chai L, Mei S, Yang J, Zhang Y, Zhang Z, Liu F, Sun W, Che Y, Yang R. LRRC19 expressed in the kidney induces TRAF2/6-mediated signals to prevent infection by uropathogenic bacteria. Nat Commun 2014; 5:4434. [PMID: 25026888 DOI: 10.1038/ncomms5434] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/18/2014] [Indexed: 02/06/2023] Open
Abstract
The innate immune-dependent bactericidal effects are critical for preventing microbial colonization in the urinary system. However, the mechanisms involved in establishing innate immune responses in kidney are not completely understood. Here we describe the role of a novel member of the LRR (leucine-rich repeat) class of transmembrane proteins, LRRC19 (LRR-containing 19) in eliminating uropathogenic bacteria. LRRC19 is predominantly expressed in human and mouse kidney tubular epithelial cells and LRRC19-deficient mice are more susceptible to uropathogenic Escherichia coli (UPEC) infection than wild-type or TLR4 knockout mice. Recognition of UPEC by LRRC19 induces the production of cytokines, chemokines and antimicrobial substances through TRAF2- and TRAF6-mediated NF-κB and MAPK signalling pathways. Thus, LRRC19 may be a critical pathogen-recognition receptor in kidney mediating the elimination of UPEC infection.
Collapse
Affiliation(s)
- Xiaomin Su
- 1] Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China [2]
| | - Siping Min
- 1] Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China [2]
| | - Shuisong Cao
- 1] Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China [2]
| | - Hui Yan
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yining Zhao
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Hui Li
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Limin Chai
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Shiyue Mei
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Jia Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yuan Zhang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Zhujun Zhang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Feifei Liu
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Wei Sun
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yongzhe Che
- 1] Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China [2] Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China
| | - Rongcun Yang
- 1] Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China [2] Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China [3] State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
21
|
Rana A, Rana B, Mishra R, Sondarva G, Rangasamy V, Das S, Viswakarma N, Kanthasamy A. Mixed Lineage Kinase-c-Jun N-Terminal Kinase Axis: A Potential Therapeutic Target in Cancer. Genes Cancer 2014; 4:334-41. [PMID: 24349631 DOI: 10.1177/1947601913485415] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mixed lineage kinases (MLKs) are members of the mitogen-activated protein kinase kinase kinase (MAP3K) family and are reported to activate MAP kinase pathways. There have been at least 9 members of the MLK family identified to date, although the physiological functions of all the family members are yet unknown. However, MLKs in general have been implicated in neurodegenerative diseases, including Parkinson and Alzheimer diseases. Recent reports suggest that some of the MLK members could play a role in cancer via modulating cell migration, invasion, cell cycle, and apoptosis. This review article will first describe the biology of MLK members and then discuss the current progress that relates to their functions in cancer.
Collapse
Affiliation(s)
- Ajay Rana
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, IL, USA ; Hines Veterans Affairs Medical Center, Hines, IL, USA
| | - Basabi Rana
- Hines Veterans Affairs Medical Center, Hines, IL, USA ; Division of Gastroenterology, Department of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Rajakishore Mishra
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, IL, USA ; Centre for Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ranchi, India
| | - Gautam Sondarva
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | - Velusamy Rangasamy
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, IL, USA ; Department of Veterinary Parasitology, Veterinary College and Research Institute, Namakkal, India
| | - Subhasis Das
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | - Navin Viswakarma
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| |
Collapse
|
22
|
Abstract
The binding of tumour necrosis factor α (TNFα) to cell surface receptors engages multiple signal transduction pathways, including three groups of mitogen-activated protein (MAP) kinases: extracellular-signal-regulated kinases (ERKs); the cJun NH2-terminal kinases (JNKs); and the p38 MAP kinases. These MAP kinase signalling pathways induce a secondary response by increasing the expression of several inflammatory cytokines (including TNFα) that contribute to the biological activity of TNFα. MAP kinases therefore function both upstream and down-stream of signalling by TNFα receptors. Here we review mechanisms that mediate these actions of MAP kinases during the response to TNFα.
Collapse
Affiliation(s)
- Guadalupe Sabio
- Department of Vascular Biology and Inflammation, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | - Roger J Davis
- Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
23
|
Abstract
Mitogen-activated protein kinase (MAPK) signaling pathways are composed of a phosphorelay signaling module where an activated MAP kinase kinase kinase (MAP3K) phosphorylates and activates a MAPK kinase (MAP2K) that in turn phosphorylates and activates a MAPK. The biological outcome of MAPK signaling is the regulation of cellular responses such as proliferation, differentiation, migration, and apoptosis. The MAP3K mixed lineage kinase 3 (MLK3) phosphorylates MAP2Ks to activate multiple MAPK signaling pathways, and MLK3 also has functions in cell signaling that are independent of its kinase activity. The recent elucidation of essential functions for MLK3 in tumour cell proliferation, migration, and invasion has drawn attention to the MLKs as potential therapeutic targets for cancer treatments. The mounting evidence that suggests a role for MLK3 in tumourigenesis and establishment of the malignant phenotype is the focus of this review.
Collapse
Affiliation(s)
- Deborah N Chadee
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA.
| |
Collapse
|
24
|
Kant S, Swat W, Zhang S, Zhang ZY, Neel BG, Flavell RA, Davis RJ. TNF-stimulated MAP kinase activation mediated by a Rho family GTPase signaling pathway. Genes Dev 2011; 25:2069-78. [PMID: 21979919 DOI: 10.1101/gad.17224711] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The biological response to tumor necrosis factor (TNF) involves activation of MAP kinases. Here we report a mechanism of MAP kinase activation by TNF that is mediated by the Rho GTPase family members Rac/Cdc42. This signaling pathway requires Src-dependent activation of the guanosine nucleotide exchange factor Vav, activation of Rac/Cdc42, and the engagement of the Rac/Cdc42 interaction site (CRIB motif) on mixed-lineage protein kinases (MLKs). We show that this pathway is essential for full MAP kinase activation during the response to TNF. Moreover, this MLK pathway contributes to inflammation in vivo.
Collapse
Affiliation(s)
- Shashi Kant
- Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Thylur RP, Senthivinayagam S, Campbell EM, Rangasamy V, Thorenoor N, Sondarva G, Mehrotra S, Mishra P, Zook E, Le PT, Rana A, Rana B. Mixed lineage kinase 3 modulates β-catenin signaling in cancer cells. J Biol Chem 2011; 286:37470-82. [PMID: 21880738 PMCID: PMC3199493 DOI: 10.1074/jbc.m111.298943] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Indexed: 12/21/2022] Open
Abstract
Expression of β-catenin is strictly regulated in normal cells via the glycogen synthase kinase 3β (GSK3β)- adenomatous polyposis coli-axin-mediated degradation pathway. Mechanisms leading to inactivation of this pathway (example: activation of Wnt/β-catenin signaling or mutations of members of the degradation complex) can result in β-catenin stabilization and activation of β-catenin/T-cell factor (TCF) signaling. β-Catenin-mediated cellular events are diverse and complex. A better understanding of the cellular signaling networks that control β-catenin pathway is important for designing effective therapeutic strategies targeting this axis. To gain more insight, we focused on determining any possible cross-talk between β-catenin and mixed lineage kinase 3 (MLK3), a MAPK kinase kinase member. Our studies indicated that MLK3 can induce β-catenin expression via post-translational stabilization in various cancer cells, including prostate cancer. This function of MLK3 was dependent on its kinase activity. MLK3 can interact with β-catenin and phosphorylate it in vitro. Overexpression of GSK3β-WT or the S9A mutant was unable to antagonize MLK3-induced stabilization, suggesting this to be independent of GSK3β pathway. Surprisingly, despite stabilizing β-catenin, MLK3 inhibited TCF transcriptional activity in the presence of both WT and S37A β-catenin. These resulted in reduced expression of β-catenin/TCF downstream targets Survivin and myc. Immunoprecipitation studies indicated that MLK3 did not decrease β-catenin/TCF interaction but promoted interaction between β-catenin and KLF4, a known repressor of β-catenin/TCF transcriptional activity. In addition, co-expression of MLK3 and β-catenin resulted in significant G(2)/M arrest. These studies provide a novel insight toward the regulation of β-catenin pathway, which can be targeted to control cancer cell proliferation, particularly those with aberrant activation of β-catenin signaling.
Collapse
Affiliation(s)
- Ramesh P. Thylur
- From the Departments of Medicine, Division of Gastroenterology, Hepatology, and Nutrition
| | | | | | | | - Nithyananda Thorenoor
- From the Departments of Medicine, Division of Gastroenterology, Hepatology, and Nutrition
| | | | | | - Prajna Mishra
- From the Departments of Medicine, Division of Gastroenterology, Hepatology, and Nutrition
| | | | | | - Ajay Rana
- Molecular Pharmacology and Therapeutics
- the Hines Veterans Affairs Medical Center, Hines, Illinois 60141
| | - Basabi Rana
- From the Departments of Medicine, Division of Gastroenterology, Hepatology, and Nutrition
- Cell and Molecular Physiology, Loyola University Chicago, Maywood, Ilinois 60153 and
- the Hines Veterans Affairs Medical Center, Hines, Illinois 60141
| |
Collapse
|
26
|
Pindel A, Sadler A. The Role of Protein Kinase R in the Interferon Response. J Interferon Cytokine Res 2011; 31:59-70. [DOI: 10.1089/jir.2010.0099] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Agnieszka Pindel
- Centre for Cancer Research, Monash Institute of Medical Research, Monash University, Melbourne, Australia
| | - Anthony Sadler
- Centre for Cancer Research, Monash Institute of Medical Research, Monash University, Melbourne, Australia
| |
Collapse
|
27
|
Zhang Y, Yang Z, Yang Y, Wang S, Shi L, Xie W, Sun K, Zou K, Wang L, Xiong J, Xiang J, Wu J. Production of transgenic mice by random recombination of targeted genes in female germline stem cells. J Mol Cell Biol 2010; 3:132-41. [DOI: 10.1093/jmcb/mjq043] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|