MAFB is a biomarker for cancer severity, prognostic marker, and a potential target for cancer immunotherapy

Author(s):  
Samir Omar ◽  
Naohiro Kobayashi ◽  
Mennatullah Siyam ◽  
Manoj Yadav ◽  
Yuri Inoue ◽  
...  

Abstract MAFB is a transcription factor specifically expressed in macrophages. Using in vitro and in vivo in mouse tumor models, our previous study suggested that MAFB could be a suitable marker for tumor-associated macrophages (TAMs), besides MAFB is expressed in anti-inflammatory alternatively activated M2 macrophages in vitro. TAMs play a key role in the tumor microenvironment (TME) by inducing immunosuppression, angiogenesis, tumor invasion, and metastasis. However, finding a suitable specific biomarker and target for TAMs is challenging. Here, we demonstrated that MAFB could be used as a biomarker for TAMs and consequently, for severity in various human cancers, including lung, liver, colon, and pancreatic cancers, according to the immunohistochemical analysis of the expression of MAFB, CD68, and CD204. Moreover, In a cohort of lung adenocarcinomas patients (n = 120), increased MAFB expression was related to increased tendency towards metastasis and poor overall survival rate. Further, we showed that MAFB expression was positively correlated to the expression of CD204 and CD68 in both human hepatocarcinoma and colon cancers. Our findings indicate that MAFB as a specific biomarker can be used as prognostic marker for Metastasis potential in Lung adenocarcinomas patients and also a biomarker for the severe Liver, Colon and pancreatic cancers. In addition, we showed that MAFB was expressed in Tumor associated macrophages expressing Programmed cell death protein-1 and/or Programmed cell death ligand 1 (TAM PD-1+ and TAM PD-L1+) cells in both human lung adenocarcinomas and Lewis lung carcinoma (LLC) mouse model. These findings indicate that MAFB can be a potential target for drug development against TAM PD-1+ and TAM PD-L1+ cells. In summary, transcriptional factor MAFB can be used as a specific biomarker, prognostic marker and a potential target for cancer immunotherapy against TAMs.

2021 ◽  
Author(s):  
Omar Samir ◽  
Naohiro Kobayashi ◽  
Mennatullah Siyam ◽  
Manoj Yadav ◽  
Yuri Inoue ◽  
...  

Abstract MAFB is a transcription factor specifically expressed in macrophages. Tumor associated macrophages (TAMs) play a key role in the tumor microenvironment (TME) by inducing immunosuppression, angiogenesis, tumor invasion, and metastasis. However, finding a suitable specific biomarker and target for TAMs is challenging. Our previous study1 suggested that MAFB could be a suitable marker for tumor-associated macrophages (TAMs) besides MAFB is expressed in anti-inflammatory alternatively activated M2 macrophages in vitro. In the current study, in a cohort of patients with lung adenocarcinoma (n = 120), increased MAFB expression was related to increased metastasis and poor overall survival rate. Our findings indicate that MAFB can be used as a prognostic marker for assessing metastatic potential in patients with lung adenocarcinoma. Further, we showed that MAFB expression was positively correlated with the expression of CD204 and CD68 in hepatocarcinoma, colon and pancreatic cancers. We demonstrated that MAFB could be used as a biomarker for TAMs and consequently, for assessing severity in various human cancers, including lung, liver, colon, and pancreatic cancers, according to the immunohistochemical analysis of the expression of MAFB, CD68, and CD204. In addition, we showed that MAFB was expressed in TAMs expressing Programmed cell death protein-1 and/or Programmed cell death ligand 1 (TAM PD-1+ and TAM PD-L1+) cells in lung adenocarcinoma and Lewis lung carcinoma (LLC) mouse model. These findings indicate that MAFB can be a potential target for drug development against TAM PD-1+ and TAM PD-L1+ cells. In summary, transcriptional factor MAFB can be used as a specific biomarker, prognostic marker, and a potential target for cancer immunotherapy against TAMs.


2021 ◽  
Vol 220 ◽  
pp. 153390
Author(s):  
Negar Khoshghamat ◽  
Niloufar Jafari ◽  
Mehrdad Moetamani-Ahmadi ◽  
Ghazaleh Khalili-Tanha ◽  
Mohammad-Hossein Khajavi rad ◽  
...  

Blood ◽  
1996 ◽  
Vol 87 (11) ◽  
pp. 4746-4753 ◽  
Author(s):  
A Cayota ◽  
F Vuillier ◽  
G Gonzalez ◽  
G Dighiero

Oxidative stress has been proposed to be involved in the immunologic defeat observed in effector calls of the immune system as well as in lymphocyte cell death and viral replication in human immunodeficiency virus (HIV)-infected patients. Because thiol-containing antioxidants such as N-acetyl-L-cysteine have been shown to have beneficial effects on CD4+ lymphocyte survival and to inhibit programmed cell death and HIV-1 replication, they may play a role in therapeutic strategies of this disease. In this work we have studied the cellular thiol levels and the affect of in vitro antioxidant treatment of purified CD4+ lymphocytes from HIV-infected patients, and correlated these parameters to proliferative responses and programmed cell death. We show that CD4+ lymphocytes from HIV-infected patients display impaired proliferative responses and a significant decrease in cellular thiol levels, indicating a disturbed redox status. Interestingly, antioxidant treatment succeeded to restore defective proliferative responses to CD3- mediated activation in 8 of 11 patients (high antioxidant responders). In contrast to high responders, patients failing to respond to antioxidant treatment (low antioxidant responders), were characterized by an abnormal ratio of apoptotic cells, which was not affected by N- acetyl-L-cysteine and/or 2-beta-mercaptoethanol preincubation. These results demonstrate for the first time that antioxidant treatment is able to revert the impaired proliferative activity of CD4 cells from HIV-infected patients and could help designing therapeutic strategies with antioxidant drugs. However, this action is not observed in cells undergoing programmed cell death.


2018 ◽  
Vol 3 (29) ◽  
pp. eaat7061 ◽  
Author(s):  
Bei Wang ◽  
Wen Zhang ◽  
Vladimir Jankovic ◽  
Jacquelynn Golubov ◽  
Patrick Poon ◽  
...  

Most patients with cancer do not develop durable antitumor responses after programmed cell death protein 1 (PD-1) or programmed cell death ligand 1(PD-L1) checkpoint inhibition monotherapy because of an ephemeral reversal of T cell dysfunction and failure to promote long-lasting immunological T cell memory. Activating costimulatory pathways to induce stronger T cell activation may improve the efficacy of checkpoint inhibition and lead to durable antitumor responses. We performed single-cell RNA sequencing of more than 2000 tumor-infiltrating CD8+T cells in mice receiving both PD-1 and GITR (glucocorticoid-induced tumor necrosis factor receptor–related protein) antibodies and found that this combination synergistically enhanced the effector function of expanded CD8+T cells by restoring the balance of key homeostatic regulators CD226 and T cell immunoreceptor with Ig and ITIM domains (TIGIT), leading to a robust survival benefit. Combination therapy decreased CD8+T cell dysfunction and induced a highly proliferative precursor effector memory T cell phenotype in a CD226-dependent manner. PD-1 inhibition rescued CD226 activity by preventing PD-1–Src homology region 2 (SHP2) dephosphophorylation of the CD226 intracellular domain, whereas GITR agonism decreased TIGIT expression. Unmasking the molecular pathways driving durable antitumor responses will be essential to the development of rational approaches to optimizing cancer immunotherapy.


2012 ◽  
Vol 209 (6) ◽  
pp. 1201-1217 ◽  
Author(s):  
Tadashi Yokosuka ◽  
Masako Takamatsu ◽  
Wakana Kobayashi-Imanishi ◽  
Akiko Hashimoto-Tane ◽  
Miyuki Azuma ◽  
...  

Programmed cell death 1 (PD-1) is a negative costimulatory receptor critical for the suppression of T cell activation in vitro and in vivo. Single cell imaging elucidated a molecular mechanism of PD-1–mediated suppression. PD-1 becomes clustered with T cell receptors (TCRs) upon binding to its ligand PD-L1 and is transiently associated with the phosphatase SHP2 (Src homology 2 domain–containing tyrosine phosphatase 2). These negative costimulatory microclusters induce the dephosphorylation of the proximal TCR signaling molecules. This results in the suppression of T cell activation and blockade of the TCR-induced stop signal. In addition to PD-1 clustering, PD-1–TCR colocalization within microclusters is required for efficient PD-1–mediated suppression. This inhibitory mechanism also functions in PD-1hi T cells generated in vivo and can be overridden by a neutralizing anti–PD-L1 antibody. Therefore, PD-1 microcluster formation is important for regulation of T cell activation.


2020 ◽  
Author(s):  
Purva Karia ◽  
Keiko Yoshioka ◽  
Wolfgang Moeder

ABSTRACTThe role of mitochondria in programmed cell death (PCD) during animal growth and development is well documented, but much less is known for plants. We previously showed that the Arabidopsis thaliana triphosphate tunnel metalloenzyme (TTM) proteins TTM1 and TTM2 are tail-anchored proteins that localize in the mitochondrial outer membrane and participate in PCD during senescence and immunity, respectively. Here, we show that TTM1 is specifically involved in senescence induced by abscisic acid (ABA). Moreover, phosphorylation of TTM1 by multiple mitogen-activated protein kinases (MAPKs) regulates its function and turnover. A combination of proteomics and in vitro kinase assays revealed three major phosphorylation sites of TTM1 (S10, S437, and S490), which are phosphorylated upon perception of senescence cues such as ABA and prolonged darkness. S437 is phosphorylated by the MAP kinases MPK3 and MPK4, and S437 phosphorylation is essential for TTM1 function in senescence. These MPKs, together with three additional MAP kinases (MPK1, MPK7, and MPK6), phosphorylate S10 and S490, marking TTM1 for protein turnover, which likely prevents uncontrolled cell death. Taken together, our results show that multiple MPKs regulate the function and turnover of the mitochondrial protein TTM1 during senescence-related PCD, revealing a novel link between mitochondria and PCD.SummaryEmail addresses: [email protected]


Cells ◽  
2021 ◽  
Vol 10 (11) ◽  
pp. 3247
Author(s):  
Lingxiao Ye ◽  
Zhengxin Zhu ◽  
Xiaochuan Chen ◽  
Haoran Zhang ◽  
Jiaqi Huang ◽  
...  

Binding of programmed cell death ligand 1 (PD-L1) to its receptor programmed cell death protein 1 (PD-1) can lead to the inactivation of cytotoxic T lymphocytes, which is one of the mechanisms for immune escape of tumors. Immunotherapy based on this mechanism has been applied in clinic with some remaining issues such as drug resistance. Exosomal PD-L1 derived from tumor cells is considered to play a key role in mediating drug resistance. Here, the effects of various tumor-derived exosomes and tumor-derived exosomal PD-L1 on tumor progression are summarized and discussed. Researchers have found that high expression of exosomal PD-L1 can inhibit T cell activation in in vitro experiments, but the function of exosomal PD-L1 in vivo remains controversial. In addition, the circulating exosomal PD-L1 has high potential to act as an indicator to evaluate the clinical effect. Moreover, therapeutic strategy targeting exosomal PD-L1 is discussed, such as inhibiting the biogenesis or secretion of exosomes. Besides, some specific methods based on the strategy of inhibiting exosomes are concluded. Further study of exosomal PD-L1 may provide an effective and safe approach for tumor treatment, and targeting exosomal PD-L1 by inhibiting exosomes may be a potential method for tumor treatment.


1998 ◽  
Vol 102 (11) ◽  
pp. 2002-2010 ◽  
Author(s):  
L Guedez ◽  
W G Stetler-Stevenson ◽  
L Wolff ◽  
J Wang ◽  
P Fukushima ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document