GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 2103-2103
    Abstract: In the United States, a woman has a 12% chance of developing breast cancer, and current treatments offer little relief to patients diagnosed with metastatic disease. Tumorigenesis and successful establishment of metastases depend upon tumor cell interactions with the surrounding immune microenvironment. Elevated tumor infiltration of immunosuppressive (M2) macrophages correlates with poor prognosis of breast cancer patients. The tumor microenvironment remarkably orchestrates molecular mechanisms that program these macrophages toward the M2 phenotype. Also, metabolic programming is instrumental in orchestrating the polarization of macrophages to assume an M1 (tumor-eradicating) or an M2 (tumor-promoting) phenotype. Aberrant activation of Hedgehog (Hh) signaling in breast cancer cells enables them to survive, proliferate, and metastasize, thus making it a promising target for breast cancer treatment. Hh signaling also enables a crosstalk between breast cancer cells and cells in their milieu, thus contributing to M2 macrophage polarization. We used two immunocompetent orthotopic mouse models of mammary tumors to test the effect of inhibiting Hh signaling on tumor-associated macrophages, and discovered that treatment with the pharmacologic Hh inhibitor, Vismodegib, induced a significant shift in the profile of tumor-infiltrating macrophages. We hypothesized that Hh activity calibrates the metabolism in macrophages, leading to enhanced M2 phenotype and function within the tumor microenvironment. Using a mass spectrometry-enabled untargeted metabolomics approach, we identified that inhibiting Hh signaling reduces flux through the hexosamine biosynthetic pathway, resulting in reduced cellular O-GlcNAcylation in M2 macrophages. This impinges upon diminished STAT6 O-GlcNAcylation, which consequently decreases fatty acid oxidation and ultimately enacts a metabolic cascade including lipid utilization, cellular bioenergetics, and mitochondrial dynamics. As such, inhibiting Hh activity mitigates the metabolomic and bioenergetic underpinnings of the immunosuppressive program of M2 macrophages, resulting in macrophages that are functionally and phenotypically reminiscent of inflammatory, anti-tumor macrophages. In conclusion, we discovered a novel role for Hh signaling in promoting polarization of tumor-associated macrophages to the M2 type through recalibrating their metabolic circuitries, ultimately leading to diminished M2 phenotype and function within the tumor microenvironment. This is the first evidence highlighting the relevance of Hh signaling in controlling a complex metabolic network in immune cells. This knowledge will help us to better understand how to target and diminish the pro-tumorigenic functions of tumor-infiltrating macrophages. Citation Format: Dominique C. Hinshaw, Ann Hanna, Tshering Lama-Sherpa, Brandon Metge, Sarah C. Kammerud, Gloria A. Benavides, Atul Kumar, Heba A. Alsheikh, Mateus Mota, Dongquan Chen, Scott Ballinger, Jeffrey C. Rathmell, Selvarangan Ponnazhagan, Victor Darley-Usmar, Rajeev S. Samant, Lalita A. Shevde. Hedgehog signaling regulates metabolism and polarization of mammary tumor-associated macrophages [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2103.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 11, No. 5 ( 2023-05-03), p. 687-702
    Abstract: The tumor immune microenvironment dynamically evolves to support tumor growth and progression. Immunosuppressive regulatory T cells (Treg) promote tumor growth and metastatic seeding in patients with breast cancer. Deregulation of plasticity between Treg and Th17 cells creates an immune regulatory framework that enables tumor progression. Here, we discovered a functional role for Hedgehog (Hh) signaling in promoting Treg differentiation and immunosuppressive activity, and when Hh activity was inhibited, Tregs adopted a Th17-like phenotype complemented by an enhanced inflammatory profile. Mechanistically, Hh signaling promoted O-GlcNAc modifications of critical Treg and Th17 transcription factors, Foxp3 and STAT3, respectively, that orchestrated this transition. Blocking Hh reprogramed Tregs metabolically, dampened their immunosuppressive activity, and supported their transdifferentiation into inflammatory Th17 cells that enhanced the recruitment of cytotoxic CD8+ T cells into tumors. Our results demonstrate a previously unknown role for Hh signaling in the regulation of Treg differentiation and activity and the switch between Tregs and Th17 cells in the tumor microenvironment.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 21 ( 2021-11-01), p. 5425-5437
    Abstract: Elevated infiltration of immunosuppressive alternatively polarized (M2) macrophages is associated with poor prognosis in patients with cancer. The tumor microenvironment remarkably orchestrates molecular mechanisms that program these macrophages. Here we identify a novel role for oncogenic Hedgehog (Hh) signaling in programming signature metabolic circuitries that regulate alternative polarization of tumor-associated macrophages. Two immunocompetent orthotopic mouse models of mammary tumors were used to test the effect of inhibiting Hh signaling on tumor-associated macrophages. Treatment with the pharmacologic Hh inhibitor vismodegib induced a significant shift in the profile of tumor-infiltrating macrophages. Mass spectrometry-based metabolomic analysis showed Hh inhibition induced significant alterations in metabolic processes, including metabolic sensing, mitochondrial adaptations, and lipid metabolism. In particular, inhibition of Hh in M2 macrophages reduced flux through the UDP-GlcNAc biosynthesis pathway. Consequently, O-GlcNAc-modification of STAT6 decreased, mitigating the immune-suppressive program of M2 macrophages, and the metabolically demanding M2 macrophages shifted their metabolism and bioenergetics from fatty acid oxidation to glycolysis. M2 macrophages enriched from vismodegib-treated mammary tumors showed characteristically decreased O-GlcNAcylation and altered mitochondrial dynamics. These Hh-inhibited macrophages are reminiscent of inflammatory (M1) macrophages, phenotypically characterized by fragmented mitochondria. This is the first report highlighting the relevance of Hh signaling in controlling a complex metabolic network in immune cells. These data describe a novel immunometabolic function of Hh signaling that can be clinically exploited. Significance: These findings illustrate that Hh activity regulates a metabolic and bioenergetic regulatory program in tumor-associated macrophages that promotes their immune-suppressive polarization.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Oncogenesis, Springer Science and Business Media LLC, Vol. 10, No. 6 ( 2021-06-02)
    Abstract: Molecular dynamics of developmental processes are repurposed by cancer cells to support cancer initiation and progression. Disruption of the delicate balance between cellular differentiation and plasticity during mammary development leads to breast cancer initiation and metastatic progression. STAT5A is essential for differentiation of secretory mammary alveolar epithelium. Active STAT5A characterizes breast cancer patients for favorable prognosis. N-Myc and STAT Interactor protein (NMI) was initially discovered as a protein that interacts with various STATs; however, the relevance of these interactions to normal mammary development and cancer was not known. We observe that NMI protein is expressed in the mammary ductal epithelium at the onset of puberty and is induced in pregnancy. NMI protein is decreased in 70% of patient specimens with metastatic breast cancer compared to primary tumors. Here we present our finding that NMI and STAT5A cooperatively mediate normal mammary development. Loss of NMI in vivo caused a decrease in STAT5A activity in normal mammary epithelial as well as breast cancer cells. Analysis of STAT5A mammary specific controlled genetic program in the context of NMI knockout revealed ISG20 (interferon stimulated exonuclease gene 20, a protein involved in rRNA biogenesis) as an unfailing negatively regulated target. Role of ISG20 has never been described in metastatic process of mammary tumors. We observed that overexpression of ISG20 is increased in metastases compared to matched primary breast tumor tissues. Our observations reveal that NMI-STAT5A mediated signaling keeps a check on ISG20 expression via miR-17–92 cluster. We show that uncontrolled ISG20 expression drives tumor progression and metastasis.
    Type of Medium: Online Resource
    ISSN: 2157-9024
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2674437-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 20, No. 1 ( 2022-01-01), p. 150-160
    Abstract: Metastases account for the majority of mortalities related to breast cancer. The onset and sustained presence of hypoxia strongly correlates with increased incidence of metastasis and unfavorable prognosis in patients with breast cancer. The Hedgehog (Hh) signaling pathway is dysregulated in breast cancer, and its abnormal activity enables tumor progression and metastasis. In addition to programming tumor cell behavior, Hh activity enables tumor cells to craft a metastasis-conducive microenvironment. Hypoxia is a prominent feature of growing tumors that impacts multiple signaling circuits that converge upon malignant progression. We investigated the role of Hh activity in crafting a hypoxic environment of breast cancer. We used radioactive tracer [18F]-fluoromisonidazole (FMISO) positron emission tomography (PET) to image tumor hypoxia. We show that tumors competent for Hh activity are able to establish a hypoxic milieu; pharmacologic inhibition of Hh signaling in a syngeneic mammary tumor model mitigates tumor hypoxia. Furthermore, in hypoxia, Hh activity is robustly activated in tumor cells and institutes increased HIF signaling in a VHL-dependent manner. The findings establish a novel perspective on Hh activity in crafting a hypoxic tumor landscape and molecularly navigating the tumor cells to adapt to hypoxic conditions. Implications: Importantly, we present a translational strategy of utilizing longitudinal hypoxia imaging to measure the efficacy of vismodegib in a preclinical model of triple-negative breast cancer.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Letters, Elsevier BV, Vol. 517 ( 2021-10), p. 24-34
    Type of Medium: Online Resource
    ISSN: 0304-3835
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 195674-7
    detail.hit.zdb_id: 2004212-7
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Elsevier BV ; 2021
    In:  iScience Vol. 24, No. 1 ( 2021-01), p. 102010-
    In: iScience, Elsevier BV, Vol. 24, No. 1 ( 2021-01), p. 102010-
    Type of Medium: Online Resource
    ISSN: 2589-0042
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 2927064-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Laboratory Investigation, Elsevier BV, Vol. 101, No. 11 ( 2021-11), p. 1439-1448
    Type of Medium: Online Resource
    ISSN: 0023-6837
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 2041329-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cell Death & Disease, Springer Science and Business Media LLC, Vol. 12, No. 3 ( 2021-03-04)
    Abstract: Triple-negative breast cancer (TNBC) patients with upregulated Wnt/β-catenin signaling often have poor clinical prognoses. During pathological examinations of breast cancer sections stained for β-catenin, we made the serendipitous observation that relative to non-TNBC, specimens from TNBC patients have a greater abundance of nucleoli. There was a remarkable direct relationship between nuclear β-catenin and greater numbers of nucleoli in TNBC tissues. These surprising observations spurred our investigations to decipher the differential functional relevance of the nucleolus in TNBC versus non-TNBC cells. Comparative nucleolar proteomics revealed that the majority of the nucleolar proteins in TNBC cells were potential targets of β-catenin signaling. Next, we undertook an analysis of the nucleolar proteome in TNBC cells in response to β-catenin inhibition. This effort revealed that a vital component of pre-rRNA processing, LAS1 like ribosome biogenesis factor (LAS1L) was significantly decreased in the nucleoli of β-catenin inhibited TNBC cells. Here we demonstrate that LAS1L protein expression is significantly elevated in TNBC patients, and it functionally is important for mammary tumor growth in xenograft models and enables invasive attributes. Our observations highlight a novel function for β-catenin in orchestrating nucleolar activity in TNBCs.
    Type of Medium: Online Resource
    ISSN: 2041-4889
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2541626-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 115-115
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 115-115
    Abstract: Breast cancer is the most commonly diagnosed cancer in women in the US. Metastasis accounts for the majority of breast cancer-related deaths. Rapidly proliferating primary tumors experience oxygen levels that are lower in comparison to the normal tissue. The proportions of these hypoxic tumor cells in breast cancer strongly correlate to the incidence of metastasis. Around 70% of breast cancers present with bone metastasis. Tumor cells that metastasize to the bone can lead to an imbalance in bone homeostasis, often leading to osteolysis. Our lab has previously reported that the activation of Hedgehog (Hh) signaling, one of the embryonic developmental pathways found to be abnormally activated in breast cancer, leads to increased osteolysis in vivo. The incidence of osteolytic metastasis at the tibio-femoral junction was significantly reduced in the Hh signaling-deficient mice. Since a hypoxic microenvironment is prevalent in bone, it is imperative to understand how Hh signaling drives tumor cell adaptation to hypoxia. The hypoxic tumor microenvironment in the primary tumor might select for metastatic cells that are better able to metastasize to the bone. We hypothesize that Hh signaling enables the adaptation of breast cancer cells to hypoxia and consequently their metastatic potential. Hh signaling can be canonically activated through smoothened (SMO)-dependent activation of the transcriptional regulator GLI. Cellular response to hypoxia is mediated by the hypoxia-inducible factors (HIFs). To understand the importance of Hh signaling activation in hypoxic condition, we inhibited the Hh signaling pathway using GLI and SMO inhibitors. Additionally, we knocked-down GLI1 in breast cancer cell lines. We have identified a feed-forward mechanism between Hh signaling and HIF signaling in hypoxic conditions leading to sustained hypoxia response by stabilization of HIF-1α. Future investigations will address the significance of Hh signaling inhibition in a murine model of TNBC. This study will help determine the therapeutic benefit of hedgehog inhibitors in controlling hypoxia-mediated metastasis and tumorigenesis. Citation Format: Tshering D. Lama-Sherpa, Sarah C. Kammerud, Lalita A. Shevde. Hedgehog signaling pathway promotes breast cancer adaptation to hypoxia [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 115.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...