LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 9 of total 9

Search options

  1. Article ; Online: The PD-L1/PD-1 Axis Blocks Neutrophil Cytotoxicity in Cancer.

    Yajuk, Olga / Baron, Maya / Toker, Sapir / Zelter, Tamir / Fainsod-Levi, Tanya / Granot, Zvi

    Cells

    2021  Volume 10, Issue 6

    Abstract: The PD-L1/PD-1 axis mediates immune tolerance and promotes tumor growth and progression via the inhibition of anti-tumor immunity. Blocking the interaction between PD-L1 and PD-1 was clinically shown to be beneficial in maintaining the anti-tumor ... ...

    Abstract The PD-L1/PD-1 axis mediates immune tolerance and promotes tumor growth and progression via the inhibition of anti-tumor immunity. Blocking the interaction between PD-L1 and PD-1 was clinically shown to be beneficial in maintaining the anti-tumor functions of the adaptive immune system. Still, the consequences of blocking the PD-L1/PD-1 axis on innate immune responses remain largely unexplored. In this context, neutrophils were shown to consist of distinct subpopulations, which possess either pro- or anti-tumor properties. PD-L1-expressing neutrophils are considered pro-tumor as they are able to suppress cytotoxic T cells and are propagated with disease progression. That said, we found that PD-L1 expression is not limited to tumor promoting neutrophils, but is also evident in anti-tumor neutrophils. We show that neutrophil cytotoxicity is effectively and efficiently blocked by tumor cell-expressed PD-1. Furthermore, the blocking of either neutrophil PD-L1 or tumor cell PD-1 maintains neutrophil cytotoxicity. Importantly, we show that tumor cell PD-1 blocks neutrophil cytotoxicity and promotes tumor growth via a mechanism independent of adaptive immunity. Taken together, these findings highlight the therapeutic potential of enhancing anti-tumor innate immune responses via blocking of the PD-L1/PD-1 axis.
    MeSH term(s) Adaptive Immunity ; Animals ; B7-H1 Antigen/metabolism ; B7-H1 Antigen/physiology ; Cell Line, Tumor ; Female ; Humans ; Immune Tolerance ; Immunity, Innate ; Male ; Mice ; Mice, Inbred BALB C ; Mice, Inbred C57BL ; Mice, Inbred NOD ; Neoplasms/metabolism ; Neoplasms/pathology ; Neutrophils/metabolism ; Neutrophils/physiology ; Programmed Cell Death 1 Receptor/metabolism ; Programmed Cell Death 1 Receptor/physiology
    Chemical Substances B7-H1 Antigen ; Cd274 protein, mouse ; Pdcd1 protein, mouse ; Programmed Cell Death 1 Receptor
    Language English
    Publishing date 2021-06-15
    Publishing country Switzerland
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2661518-6
    ISSN 2073-4409 ; 2073-4409
    ISSN (online) 2073-4409
    ISSN 2073-4409
    DOI 10.3390/cells10061510
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article: Neutrophil Cathepsin G and Tumor Cell RAGE Facilitate Neutrophil Anti-Tumor Cytotoxicity.

    Sionov, Ronit Vogt / Fainsod-Levi, Tanya / Zelter, Tamir / Polyansky, Lola / Pham, Christine T / Granot, Zvi

    Oncoimmunology

    2019  Volume 8, Issue 9, Page(s) e1624129

    Abstract: Neutrophils are a heterogeneous population of myeloid cells which may either promote or hinder tumor growth and progression. Anti-tumor neutrophils have the capacity to kill tumor cells in a contact-dependent manner. However, the molecular mechanisms ... ...

    Abstract Neutrophils are a heterogeneous population of myeloid cells which may either promote or hinder tumor growth and progression. Anti-tumor neutrophils have the capacity to kill tumor cells in a contact-dependent manner. However, the molecular mechanisms underlying tumor cell recognition by neutrophils remained unexplored. Tumor cells were shown to express aberrant glycosylation patterns and neutrophils are equipped with receptors capable of recognizing such glycosylations. Accordingly, we hypothesized that the receptor for advanced glycation end products (RAGE) may facilitate neutrophil recognition of tumor cells. Indeed, RAGE decoy receptors and RAGE-specific blocking antibodies dramatically reduce tumor cell susceptibility to neutrophil cytotoxicity. Unexpectedly, we found that tumor cell RAGE rather than neutrophil RAGE is important for the killing process. We further identified neutrophil Cathepsin G as the neutrophil component interacting with tumor cell RAGE. Cathepsin G-deficient neutrophils show impaired ability to kill tumor cells, suggesting that RAGE-Cathepsin G interaction is required for neutrophil cytotoxicity. These data unravel new aspects of neutrophil anti-tumor activity and identify a novel role for RAGE and Cathepsin G in neutrophil-mediated cytotoxicity.
    Language English
    Publishing date 2019-06-11
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2645309-5
    ISSN 2162-402X ; 2162-4011
    ISSN (online) 2162-402X
    ISSN 2162-4011
    DOI 10.1080/2162402X.2019.1624129
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: TRPM2 modulates neutrophil attraction to murine tumor cells by regulating CXCL2 expression.

    Gershkovitz, Maya / Fainsod-Levi, Tanya / Zelter, Tamir / Sionov, Ronit V / Granot, Zvi

    Cancer immunology, immunotherapy : CII

    2018  Volume 68, Issue 1, Page(s) 33–43

    Abstract: In recent years, immune cells were shown to play critical roles in tumor growth and metastatic progression. In this context, neutrophils were shown to possess both pro- and anti-tumor properties. To exert their anti-tumor effect, neutrophils need to ... ...

    Abstract In recent years, immune cells were shown to play critical roles in tumor growth and metastatic progression. In this context, neutrophils were shown to possess both pro- and anti-tumor properties. To exert their anti-tumor effect, neutrophils need to migrate towards, and form physical contact with tumor cells. Neutrophils secrete H
    MeSH term(s) Animals ; Cell Line, Tumor ; Cell Movement/immunology ; Chemokine CXCL2/genetics ; Chemokine CXCL2/immunology ; Chemokine CXCL2/metabolism ; Chemotaxis, Leukocyte/immunology ; Coculture Techniques ; Cytotoxicity, Immunologic/immunology ; Female ; Gene Expression Regulation, Neoplastic/immunology ; Mice, Inbred BALB C ; Neoplasms/immunology ; Neoplasms/metabolism ; Neoplasms/pathology ; Neutrophils/cytology ; Neutrophils/immunology ; RNA Interference/immunology ; TRPM Cation Channels/genetics ; TRPM Cation Channels/immunology ; TRPM Cation Channels/metabolism
    Chemical Substances Chemokine CXCL2 ; Cxcl2 protein, mouse ; TRPM Cation Channels ; TRPM2 protein, mouse
    Language English
    Publishing date 2018-09-24
    Publishing country Germany
    Document type Journal Article
    ZDB-ID 195342-4
    ISSN 1432-0851 ; 0340-7004
    ISSN (online) 1432-0851
    ISSN 0340-7004
    DOI 10.1007/s00262-018-2249-2
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: IRE1 endoribonuclease signaling promotes myeloid cell infiltration in glioblastoma.

    Obacz, Joanna / Archambeau, Jérôme / Lafont, Elodie / Nivet, Manon / Martin, Sophie / Aubry, Marc / Voutetakis, Konstantinos / Pineau, Raphael / Boniface, Rachel / Sicari, Daria / Pelizzari-Raymundo, Diana / Ghukasyan, Gevorg / McGrath, Eoghan / Vlachavas, Efstathios-Iason / Le Gallo, Matthieu / Le Reste, Pierre Jean / Barroso, Kim / Fainsod-Levi, Tanya / Obiedat, Akram /
    Granot, Zvi / Tirosh, Boaz / Samal, Juhi / Pandit, Abhay / Négroni, Luc / Soriano, Nicolas / Monnier, Annabelle / Mosser, Jean / Chatziioannou, Aristotelis / Quillien, Véronique / Chevet, Eric / Avril, Tony

    Neuro-oncology

    2023  Volume 26, Issue 5, Page(s) 858–871

    Abstract: Background: Intrinsic or environmental stresses trigger the accumulation of improperly folded proteins in the endoplasmic reticulum (ER), leading to ER stress. To cope with this, cells have evolved an adaptive mechanism named the unfolded protein ... ...

    Abstract Background: Intrinsic or environmental stresses trigger the accumulation of improperly folded proteins in the endoplasmic reticulum (ER), leading to ER stress. To cope with this, cells have evolved an adaptive mechanism named the unfolded protein response (UPR) which is hijacked by tumor cells to develop malignant features. Glioblastoma (GB), the most aggressive and lethal primary brain tumor, relies on UPR to sustain growth. We recently showed that IRE1 alpha (referred to IRE1 hereafter), 1 of the UPR transducers, promotes GB invasion, angiogenesis, and infiltration by macrophage. Hence, high tumor IRE1 activity in tumor cells predicts a worse outcome. Herein, we characterized the IRE1-dependent signaling that shapes the immune microenvironment toward monocytes/macrophages and neutrophils.
    Methods: We used human and mouse cellular models in which IRE1 was genetically or pharmacologically invalidated and which were tested in vivo. Publicly available datasets from GB patients were also analyzed to confirm our findings.
    Results: We showed that IRE1 signaling, through both the transcription factor XBP1s and the regulated IRE1-dependent decay controls the expression of the ubiquitin-conjugating E2 enzyme UBE2D3. In turn, UBE2D3 activates the NFκB pathway, resulting in chemokine production and myeloid infiltration in tumors.
    Conclusions: Our work identifies a novel IRE1/UBE2D3 proinflammatory axis that plays an instrumental role in GB immune regulation.
    MeSH term(s) Glioblastoma/pathology ; Glioblastoma/metabolism ; Humans ; Mice ; Endoribonucleases/metabolism ; Endoribonucleases/genetics ; Animals ; Protein Serine-Threonine Kinases/metabolism ; Protein Serine-Threonine Kinases/genetics ; Signal Transduction ; Brain Neoplasms/pathology ; Brain Neoplasms/metabolism ; Myeloid Cells/metabolism ; Myeloid Cells/pathology ; Unfolded Protein Response ; Tumor Microenvironment ; Tumor Cells, Cultured ; Endoplasmic Reticulum Stress
    Chemical Substances Endoribonucleases (EC 3.1.-) ; Protein Serine-Threonine Kinases (EC 2.7.11.1) ; ERN1 protein, human (EC 2.7.11.1)
    Language English
    Publishing date 2023-12-27
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2028601-6
    ISSN 1523-5866 ; 1522-8517
    ISSN (online) 1523-5866
    ISSN 1522-8517
    DOI 10.1093/neuonc/noad256
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression.

    Parhi, Lishay / Alon-Maimon, Tamar / Sol, Asaf / Nejman, Deborah / Shhadeh, Amjad / Fainsod-Levi, Tanya / Yajuk, Olga / Isaacson, Batya / Abed, Jawad / Maalouf, Naseem / Nissan, Aviram / Sandbank, Judith / Yehuda-Shnaidman, Einav / Ponath, Falk / Vogel, Jörg / Mandelboim, Ofer / Granot, Zvi / Straussman, Ravid / Bachrach, Gilad

    Nature communications

    2020  Volume 11, Issue 1, Page(s) 3259

    Abstract: Fusobacterium nucleatum is an oral anaerobe recently found to be prevalent in human colorectal cancer (CRC) where it is associated with poor treatment outcome. In mice, hematogenous F. nucleatum can colonize CRC tissue using its lectin Fap2, which ... ...

    Abstract Fusobacterium nucleatum is an oral anaerobe recently found to be prevalent in human colorectal cancer (CRC) where it is associated with poor treatment outcome. In mice, hematogenous F. nucleatum can colonize CRC tissue using its lectin Fap2, which attaches to tumor-displayed Gal-GalNAc. Here, we show that Gal-GalNAc levels increase as human breast cancer progresses, and that occurrence of F. nucleatum gDNA in breast cancer samples correlates with high Gal-GalNAc levels. We demonstrate Fap2-dependent binding of the bacterium to breast cancer samples, which is inhibited by GalNAc. Intravascularly inoculated Fap2-expressing F. nucleatum ATCC 23726 specifically colonize mice mammary tumors, whereas Fap2-deficient bacteria are impaired in tumor colonization. Inoculation with F. nucleatum suppresses accumulation of tumor infiltrating T cells and promotes tumor growth and metastatic progression, the latter two of which can be counteracted by antibiotic treatment. Thus, targeting F. nucleatum or Fap2 might be beneficial during treatment of breast cancer.
    MeSH term(s) Animals ; Anti-Bacterial Agents/pharmacology ; Bacterial Proteins/metabolism ; Breast Neoplasms/diagnostic imaging ; Breast Neoplasms/immunology ; Breast Neoplasms/microbiology ; Breast Neoplasms/pathology ; Cell Line, Tumor ; Cell Proliferation/drug effects ; Colony Count, Microbial ; DNA, Bacterial/genetics ; Disease Models, Animal ; Disease Progression ; Female ; Fusobacterium nucleatum/drug effects ; Fusobacterium nucleatum/genetics ; Fusobacterium nucleatum/growth & development ; Galactosamine/metabolism ; Galactose/metabolism ; Genome, Bacterial/genetics ; Humans ; Immunity/drug effects ; Lung Neoplasms/secondary ; Mice, Inbred BALB C ; Neoplasm Metastasis
    Chemical Substances Anti-Bacterial Agents ; Bacterial Proteins ; DNA, Bacterial ; Galactosamine (7535-00-4) ; Galactose (X2RN3Q8DNE)
    Language English
    Publishing date 2020-06-26
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-020-16967-2
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Microenvironmental Cues Determine Tumor Cell Susceptibility to Neutrophil Cytotoxicity.

    Gershkovitz, Maya / Fainsod-Levi, Tanya / Khawaled, Saleh / Shaul, Merav E / Sionov, Ronit V / Cohen-Daniel, Leonor / Aqeilan, Rami I / Shaul, Yoav D / Fridlender, Zvi G / Granot, Zvi

    Cancer research

    2018  Volume 78, Issue 17, Page(s) 5050–5059

    Abstract: We have recently shown that neutrophil antitumor cytotoxicity is ... ...

    Abstract We have recently shown that neutrophil antitumor cytotoxicity is Ca
    MeSH term(s) Animals ; Cell Line, Tumor ; Cell Proliferation/genetics ; Disease Models, Animal ; Epithelial-Mesenchymal Transition/genetics ; Gene Expression Regulation, Neoplastic/genetics ; Humans ; Hydrogen Peroxide/metabolism ; Lung Neoplasms/genetics ; Lung Neoplasms/pathology ; Mice ; Neoplasm Metastasis ; Neutrophil Activation/genetics ; Neutrophil Activation/immunology ; Neutrophils/metabolism ; Neutrophils/pathology ; TRPM Cation Channels/genetics ; Tumor Microenvironment/genetics
    Chemical Substances TRPM Cation Channels ; TRPM2 protein, mouse ; Hydrogen Peroxide (BBX060AN9V)
    Language English
    Publishing date 2018-07-02
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 1432-1
    ISSN 1538-7445 ; 0008-5472
    ISSN (online) 1538-7445
    ISSN 0008-5472
    DOI 10.1158/0008-5472.CAN-18-0540
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Hyperglycemia Impairs Neutrophil Mobilization Leading to Enhanced Metastatic Seeding.

    Fainsod-Levi, Tanya / Gershkovitz, Maya / Völs, Sandra / Kumar, Saran / Khawaled, Saleh / Sagiv, Jitka Y / Sionov, Ronit V / Grunewald, Myriam / Keshet, Eli / Granot, Zvi

    Cell reports

    2017  Volume 21, Issue 9, Page(s) 2384–2392

    Abstract: Preexisting diabetes is a risk factor for the development of multiple types of cancer. Additionally, diabetic patients face a poorer prognosis when diagnosed with cancer. To gain insight into the effects of hyperglycemia, a hallmark of diabetes, on tumor ...

    Abstract Preexisting diabetes is a risk factor for the development of multiple types of cancer. Additionally, diabetic patients face a poorer prognosis when diagnosed with cancer. To gain insight into the effects of hyperglycemia, a hallmark of diabetes, on tumor growth and metastatic progression, we combined mouse models of cancer and hyperglycemia. We show that while hyperglycemia attenuates primary tumor growth, it concomitantly increases metastatic seeding in a distant organ. We further show that the increase in metastatic seeding is due to impaired secretion of granulocyte colony-stimulating factor (G-CSF) and impaired neutrophil mobilization. Normalizing blood glucose levels using insulin rescues neutrophil recruitment and tumor growth and concomitantly reduces metastatic seeding. These results provide links among hyperglycemia-induced changes in neutrophil mobilization, primary tumor growth, and metastatic progression. Furthermore, our observations highlight the importance of normalizing blood glucose levels in hyperglycemic cancer patients.
    MeSH term(s) Animals ; Cell Line, Tumor ; Enzyme-Linked Immunosorbent Assay ; Female ; Granulocyte Colony-Stimulating Factor/immunology ; Granulocyte Colony-Stimulating Factor/metabolism ; Hyperglycemia/immunology ; Hyperglycemia/metabolism ; Leukocytes/immunology ; Leukocytes/metabolism ; Mice ; Mice, Inbred BALB C ; Neoplasm Metastasis/immunology ; Neutrophils/physiology
    Chemical Substances Granulocyte Colony-Stimulating Factor (143011-72-7)
    Language English
    Publishing date 2017-11-28
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2649101-1
    ISSN 2211-1247 ; 2211-1247
    ISSN (online) 2211-1247
    ISSN 2211-1247
    DOI 10.1016/j.celrep.2017.11.010
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression.

    Parhi, Lishay / Alon-Maimon, Tamar / Sol, Asaf / Nejman, Deborah / Shhadeh, Amjad / Fainsod-Levi, Tanya / Yajuk, Olga / Isaacson, Batya / Abed, Jawad / Maalouf, Naseem / Nissan, Aviram / Sandbank, Judith / Yehuda-Shnaidman, Einav / Ponath, Falk / Vogel, Jörg / Mandelboim, Ofer / Granot, Zvi / Straussman, Ravid / Bachrach, Gilad

    11 ; 1 ; 3259 ; Nature communications ; England

    2020  

    Abstract: Fusobacterium nucleatum is an oral anaerobe recently found to be prevalent in human colorectal cancer (CRC) where it is associated with poor treatment outcome. In mice, hematogenous F. nucleatum can colonize CRC tissue using its lectin Fap2, which ... ...

    Abstract Fusobacterium nucleatum is an oral anaerobe recently found to be prevalent in human colorectal cancer (CRC) where it is associated with poor treatment outcome. In mice, hematogenous F. nucleatum can colonize CRC tissue using its lectin Fap2, which attaches to tumor-displayed Gal-GalNAc. Here, we show that Gal-GalNAc levels increase as human breast cancer progresses, and that occurrence of F. nucleatum gDNA in breast cancer samples correlates with high Gal-GalNAc levels. We demonstrate Fap2-dependent binding of the bacterium to breast cancer samples, which is inhibited by GalNAc. Intravascularly inoculated Fap2-expressing F. nucleatum ATCC 23726 specifically colonize mice mammary tumors, whereas Fap2-deficient bacteria are impaired in tumor colonization. Inoculation with F. nucleatum suppresses accumulation of tumor infiltrating T cells and promotes tumor growth and metastatic progression, the latter two of which can be counteracted by antibiotic treatment. Thus, targeting F. nucleatum or Fap2 might be beneficial during treatment of breast cancer.
    Language English
    Publishing date 2020-06-26
    Publisher Nature Research
    Publishing country de
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

  9. Article ; Online: TRPM2 Mediates Neutrophil Killing of Disseminated Tumor Cells.

    Gershkovitz, Maya / Caspi, Yaki / Fainsod-Levi, Tanya / Katz, Ben / Michaeli, Janna / Khawaled, Saleh / Lev, Shaya / Polyansky, Lola / Shaul, Merav E / Sionov, Ronit V / Cohen-Daniel, Leonor / Aqeilan, Rami I / Shaul, Yoav D / Mori, Yasuo / Karni, Rotem / Fridlender, Zvi G / Binshtok, Alexander M / Granot, Zvi

    Cancer research

    2018  Volume 78, Issue 10, Page(s) 2680–2690

    Abstract: Neutrophils play a critical role in cancer, with both protumor and antitumor neutrophil subpopulations reported. The antitumor neutrophil subpopulation has the capacity to kill tumor cells and limit metastatic spread, yet not all tumor cells are equally ... ...

    Abstract Neutrophils play a critical role in cancer, with both protumor and antitumor neutrophil subpopulations reported. The antitumor neutrophil subpopulation has the capacity to kill tumor cells and limit metastatic spread, yet not all tumor cells are equally susceptible to neutrophil cytotoxicity. Because cells that evade neutrophils have greater chances of forming metastases, we explored the mechanism neutrophils use to kill tumor cells. Neutrophil cytotoxicity was previously shown to be mediated by secretion of H
    MeSH term(s) Animals ; Breast Neoplasms/pathology ; CRISPR-Cas Systems/genetics ; Calcium/metabolism ; Calcium Channels/metabolism ; Cell Line, Tumor ; Cell Proliferation/genetics ; Female ; Humans ; Hydrogen Peroxide/metabolism ; Mice ; Mice, Inbred BALB C ; Neoplastic Cells, Circulating/immunology ; Neoplastic Cells, Circulating/pathology ; Neutrophils/immunology ; Neutrophils/metabolism ; TRPM Cation Channels/genetics ; TRPM Cation Channels/metabolism
    Chemical Substances Calcium Channels ; TRPM Cation Channels ; TRPM2 protein, mouse ; Hydrogen Peroxide (BBX060AN9V) ; Calcium (SY7Q814VUP)
    Language English
    Publishing date 2018-02-28
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 1432-1
    ISSN 1538-7445 ; 0008-5472
    ISSN (online) 1538-7445
    ISSN 0008-5472
    DOI 10.1158/0008-5472.CAN-17-3614
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top