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: American Journal of Physiology-Gastrointestinal and Liver Physiology, American Physiological Society, Vol. 311, No. 4 ( 2016-10-01), p. G699-G712
    Abstract: Obesity presents a significant public health concern given its association with increased cancer incidence, unfavorable prognosis, and metastasis. However, there is very little literature on the effects of weight loss, following obesity, on risk for colon cancer or liver cancer. Therefore, we sought to study whether intentional weight loss through diet manipulation was capable of mitigating colon and liver cancer in mice. We fed mice with a high-fat diet (HFD) comprised of 47% carbohydrates, 40% fat, and 13% protein for 20 wk to mimic human obesity. Subsequently, azoxymethane (AOM) was used to promote colon and liver carcinogenesis. A subset of obese mice was then switched to a low-fat diet (LFD) containing 67.5% carbohydrate, 12.2% fat, and 20% protein to promote intentional weight loss. Body weight loss and excess fat reduction did not protect mice from colon cancer progression and liver dysplastic lesion in the AOM-chemical-cancer model even though these mice had improved blood glucose and leptin levels. Intentional weight loss in AOM-treated mice actually produced histological changes that resemble dysplastic alterations in the liver and presented a higher percentage of F4/80 + CD206 + macrophages and activated T cells (CD4 + CD69 + ) in the spleen and lymph nodes, respectively. In addition, the liver of AOM-treated mice exposed to a HFD during the entire period of the experiment exhibited a marked increase in proliferation and pNF-κB activation. Altogether, these data suggest that intentional weight loss following chemical-induced carcinogenesis does not affect colon tumorigenesis but may in fact negatively impact liver repair mechanisms.
    Type of Medium: Online Resource
    ISSN: 0193-1857 , 1522-1547
    Language: English
    Publisher: American Physiological Society
    Publication Date: 2016
    detail.hit.zdb_id: 1477329-6
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: American Journal of Physiology-Gastrointestinal and Liver Physiology, American Physiological Society, Vol. 314, No. 1 ( 2018-01-01), p. G22-G31
    Abstract: We examined the role of macrophages in inflammation associated with colorectal cancer (CRC). Given the emerging evidence on immune-microbiota interactions in CRC, we also sought to examine the interaction between macrophages and gut microbiota. To induce CRC, male C57BL/6 mice ( n = 32) received a single injection of azoxymethane (AOM), followed by three cycles of dextran sodium sulfate (DSS)-supplemented water in weeks 1, 4, and 7. Prior to the final DSS cycle ( week 7) and twice weekly until euthanasia, mice ( n = 16/group) received either 200 μl ip of clodronate-filled liposomes (CLD) or phosphate-buffered saline (PBS) encapsulated liposomes to deplete macrophages. Colon tissue was analyzed for polyp burden, macrophage markers, transcription factors, and inflammatory mediators. Stool samples were collected, and DNA was isolated and subsequently sequenced for 16S rRNA. Clodronate liposomes decreased tumor number by ∼36% and specifically large (≥1 mm) tumors by ∼36% ( P 〈 0.05). This was consistent with a decrease in gene expression of EMR1 in the colon tissue and polyp tissue as well as expression of select markers associated with M1 (IL-6) and M2 macrophages (IL-13, IL-10, TGFβ, CCL17) in the colon tissue ( P 〈 0.05). Similarly, there was a decrease in STAT3 and p38 MAPK and ERK signaling in colon tissue. Clodronate liposomes increased the relative abundance of the Firmicutes phylum ( P 〈 0.05) and specifically Lactobacillaceae and Clostridiaceae families, which have been associated with reduced CRC risk. Overall, these data support the development of therapeutic strategies to target macrophages in CRC and provide support for further evaluation of immune-microbiota interactions in CRC. NEW & NOTEWORTHY We found that macrophage depletion during late-stage tumorigenesis is effective at reducing tumor growth. This was associated with a decrease in macrophage markers and chemokines in the colon tissue and a decrease in transcription factors that are linked to colorectal cancer. The macrophage-depleted group was found to have an increased abundance of Firmicutes, a phylum with documented anti-tumorigenic effects. Overall, these data support the development of therapeutic strategies to target macrophages in colorectal cancer.
    Type of Medium: Online Resource
    ISSN: 0193-1857 , 1522-1547
    Language: English
    Publisher: American Physiological Society
    Publication Date: 2018
    detail.hit.zdb_id: 1477329-6
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: World Journal of Hepatology, Baishideng Publishing Group Inc., Vol. 11, No. 8 ( 2019-8-27), p. 619-637
    Type of Medium: Online Resource
    ISSN: 1948-5182
    Language: Unknown
    Publisher: Baishideng Publishing Group Inc.
    Publication Date: 2019
    detail.hit.zdb_id: 2573703-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Physiological Reports, Wiley, Vol. 5, No. 18 ( 2017-09), p. e13412-
    Type of Medium: Online Resource
    ISSN: 2051-817X
    Language: English
    Publisher: Wiley
    Publication Date: 2017
    detail.hit.zdb_id: 2724325-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of Animal Ecology, Wiley, Vol. 89, No. 9 ( 2020-09), p. 1997-2012
    Abstract: Camera trap technology has galvanized the study of predator–prey ecology in wild animal communities by expanding the scale and diversity of predator–prey interactions that can be analysed. While observational data from systematic camera arrays have informed inferences on the spatiotemporal outcomes of predator–prey interactions, the capacity for observational studies to identify mechanistic drivers of species interactions is limited. Experimental study designs that utilize camera traps uniquely allow for testing hypothesized mechanisms that drive predator and prey behaviour, incorporating environmental realism not possible in the laboratory while benefiting from the distinct capacity of camera traps to generate large datasets from multiple species with minimal observer interference. However, such pairings of camera traps with experimental methods remain underutilized. We review recent advances in the experimental application of camera traps to investigate fundamental mechanisms underlying predator–prey ecology and present a conceptual guide for designing experimental camera trap studies. Only 9% of camera trap studies on predator–prey ecology in our review use experimental methods, but the application of experimental approaches is increasing. To illustrate the utility of camera trap‐based experiments using a case study, we propose a study design that integrates observational and experimental techniques to test a perennial question in predator–prey ecology: how prey balance foraging and safety, as formalized by the risk allocation hypothesis. We discuss applications of camera trap‐based experiments to evaluate the diversity of anthropogenic influences on wildlife communities globally. Finally, we review challenges to conducting experimental camera trap studies. Experimental camera trap studies have already begun to play an important role in understanding the predator–prey ecology of free‐living animals, and such methods will become increasingly critical to quantifying drivers of community interactions in a rapidly changing world. We recommend increased application of experimental methods in the study of predator and prey responses to humans, synanthropic and invasive species, and other anthropogenic disturbances.
    Type of Medium: Online Resource
    ISSN: 0021-8790 , 1365-2656
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2020
    detail.hit.zdb_id: 2006616-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 200, No. 1_Supplement ( 2018-05-01), p. 42.20-42.20
    Abstract: Low-grade, chronic inflammation is thought to play a role in obesity-associated metabolic dysfunction. In obesity, macrophages account for approximately 50% of adipose tissue cells and can secrete a variety of proinflammatory cytokines. In this study, we sought to deplete macrophages in order to decrease macrophage-mediated inflammation and rescue metabolic dysfunction. Following 16 weeks of high-fat diet (HFD) (40% of total kcal from fat) or AIN-76A control diet consumption (n=30), male C57BL/6J mice within each diet cohort were assigned to either a clodronate (CLD)-liposome or PBS-liposome-treatment (n=15/group). Mice received 200 μl (1mg) i.p. injections of CLD or PBS-encapsulated liposomes (control group) twice weekly for 4 weeks to deplete macrophages. Metabolic function was assessed via the HOMA-IR, glucose and insulin tolerance tests, and serum free fatty acids. Adipose tissue, liver, and blood were analyzed for macrophage infiltration, inflammatory mediators and circulating cell populations. HFD-fed mice exhibited an obese phenotype compared to the control diet; however, macrophage depletion was unable to rescue metabolic dysfunction. Interestingly, macrophage-depleted mice had & gt;35% increase in circulating neutrophils and adipose tissue Ly6G content. The increase in neutrophils, likely a compensation for the depletion of macrophages, was linked to an increase in the proinflammatory cytokines, IL-6 and IL1β, in the adipose tissue. Additionally, the decrease in macrophages resulted in iron-deficient anemia. Our study suggests that depleting macrophages in an obese setting is not an effective therapy for rescuing metabolic dysfunction and may increase the risk for anemia and adipose tissue inflammation.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2018
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Frontiers in Oncology, Frontiers Media SA, Vol. 12 ( 2023-1-4)
    Abstract: Advanced age and obesity are independent risk and progression factors for triple negative breast cancer (TNBC), which presents significant public health concerns for the aging population and its increasing burden of obesity. Due to parallels between advanced age- and obesityrelated biology, particularly adipose inflammation, we hypothesized that advanced age and obesity each accelerate mammary tumor growth through convergent, and likely interactive, mechanisms. Methods To test this hypothesis, we orthotopically transplanted murine syngeneic TNBC cells into the mammary glands of young normoweight control (7 months), young diet-induced obese (DIO), aged normoweight control (17 months), and aged DIO female C57BL/6J mice. Results Here we report accelerated tumor growth in aged control and young DIO mice, compared with young controls. Transcriptional analyses revealed, with a few exceptions, overlapping patterns of mammary tumor inflammation and tumor immunosuppression in aged control mice and young DIO mice, relative to young controls. Moreover, aged control and young DIO tumors, compared with young controls, had reduced abundance ofcytotoxic CD8 T cells. Finally, DIO in advanced age exacerbated mammary tumor growth, inflammation and tumor immunosuppression. Discussion These findings demonstrate commonalities in the mechanisms driving TNBC in aged and obese mice, relative to young normoweight controls. Moreover, we found that advanced age and DIO interact to accelerate mammary tumor progression. Given the US population is getting older and more obese, age- and obesity-related biological differences will need to be considered when developing mechanism-based strategies for preventing or controlling breast cancer.
    Type of Medium: Online Resource
    ISSN: 2234-943X
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2023
    detail.hit.zdb_id: 2649216-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2022
    In:  Cancer and Metastasis Reviews Vol. 41, No. 3 ( 2022-09), p. 607-625
    In: Cancer and Metastasis Reviews, Springer Science and Business Media LLC, Vol. 41, No. 3 ( 2022-09), p. 607-625
    Abstract: Obesity, exceptionally prevalent in the USA, promotes the incidence and progression of numerous cancer types including breast cancer. Complex, interacting metabolic and immune dysregulation marks the development of both breast cancer and obesity. Obesity promotes chronic low-grade inflammation, particularly in white adipose tissue, which drives immune dysfunction marked by increased pro-inflammatory cytokine production, alternative macrophage activation, and reduced T cell function. Breast tissue is predominantly composed of white adipose, and developing breast cancer readily and directly interacts with cells and signals from adipose remodeled by obesity. This review discusses the biological mechanisms through which obesity promotes breast cancer, the role of obesity in breast cancer health disparities, and dietary interventions to mitigate the adverse effects of obesity on breast cancer. We detail the intersection of obesity and breast cancer, with an emphasis on the shared and unique patterns of immune dysregulation in these disease processes. We have highlighted key areas of breast cancer biology exacerbated by obesity, including incidence, progression, and therapeutic response. We posit that interception of obesity-driven breast cancer will require interventions that limit protumor signaling from obese adipose tissue and that consider genetic, structural, and social determinants of the obesity–breast cancer link. Finally, we detail the evidence for various dietary interventions to offset obesity effects in clinical and preclinical studies of breast cancer. In light of the strong associations between obesity and breast cancer and the rising rates of obesity in many parts of the world, the development of effective, safe, well-tolerated, and equitable interventions to limit the burden of obesity on breast cancer are urgently needed.
    Type of Medium: Online Resource
    ISSN: 0167-7659 , 1573-7233
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2004180-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 612-612
    Abstract: Introduction: Breast cancer (BC) is the second leading cause of cancer-related mortality among American women and is exacerbated by obesity. The basal-like (BL) subtype accounts for 10-15% of all BCs and is a particularly aggressive disease due to its extreme intrinsic diversity compared with other subtypes. Paradoxically, obesity promotes immune checkpoint inhibition (ICI) response in some cancers. Further, obesity promotes altered intestinal microbial composition, and recent clinical studies of fecal microbiota transplants suggest causal links between the microbiome and immunotherapy responses. Hence, we evaluated the correlation of tumoral and cecal microbiota with anti-Programmed Cell Death Protein 1 (PD1) immunotherapy response in a murine model of BLBC and obesity. Methods: In an initial study, normoweight female C57BL/6J mice (n=8/group) received orthotopic injection of one of two novel C3TAg BLBC cell lines (1.02 and 2.51) syngeneic to C57BL/6J derived from a spontaneous tumor excised from B6-C3TAg transgenic mice. Gene expression profiling (Affymetrix arrays) with Gene Set Enrichment Analysis (GSEA) was performed on the resulting transplanted tumors. In a second study, female C57BL/6J mice were divided into a diet-induced obesity (DIO) group (n=30), promoted by high-fat diet feeding, and a lean control group (n=30). After 24 weeks, 2.51 cells were orthotopically injected into the fourth mammary fat pad of all mice, followed by treatment with anti-PD1 or IgG control antibodies (n=15/diet group). Tumor and cecal DNA was extracted and subjected to 16S rRNA amplicon sequencing. Results: GSEA from the initial study revealed that orthotopically transplanted line 2.51 tumors, relative to line 1.02 tumors, had lower levels of stromal remodeling as indicated by significant enrichment of gene sets related to extracellular matrix proteoglycans, collagen chain trimerization, and collagen biosynthesis and modification. Gene sets related to insulin and IGF-1 signaling were also lower in line 2.51 tumors than line 1.02 tumors. The in-life portion of the second study revealed a heterogeneous response to ICI therapies among lean and obese mice. To understand this diversity of response, 16S rRNA sequencing analysis of tumoral and cecal microbiota in response to DIO and ICI therapy is underway. Discussion: These data reveal striking differences in tumor microenvironment and nutrient sensing as being potentially important determinants of differential tumor growth in BLBC. Ultimately, this research may contribute to the development of precision nutrition approaches using pro-/pre-biotics to promote microbial communities, targeting the tumor microenvironment, which may improve immunotherapy response and disrupt the obesity-associated exacerbation of breast cancer. Financial Support: This work was supported by R35CA197627 to SDH. Citation Format: Tori L. McFarlane, Meredith S. Carson, Elaine M. Glenny, Violet A. Kiesel, Ashlee Taylor, Daniel Roth, Jody Albright, Melissa VerHague, John E. French, Michael F. Coleman, Stephen D. Hursting. Microbial determinants of immune checkpoint inhibition response in a murine model of obesity and metastatic basal-like breast cancer [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 612.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...