Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Proc Natl Acad Sci U S A ; 119(21): e2202016119, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35537042

ABSTRACT

Autophagy defects are a risk factor for inflammatory bowel diseases (IBDs) through unknown mechanisms. Whole-body conditional deletion of autophagy-related gene (Atg) Atg7 in adult mice (Atg7Δ/Δ) causes tissue damage and death within 3 mo due to neurodegeneration without substantial effect on intestine. In contrast, we report here that whole-body conditional deletion of other essential Atg genes Atg5 or Fip200/Atg17 in adult mice (Atg5Δ/Δ or Fip200Δ/Δ) caused death within 5 d due to rapid autophagy inhibition, elimination of ileum stem cells, and loss of barrier function. Atg5Δ/Δ mice lost PDGFRα+ mesenchymal cells (PMCs) and Wnt signaling essential for stem cell renewal, which were partially rescued by exogenous Wnt. Matrix-assisted laser desorption ionization coupled to mass spectrometry imaging (MALDI-MSI) of Atg5Δ/Δ ileum revealed depletion of aspartate and nucleotides, consistent with metabolic insufficiency underlying PMC loss. The difference in the autophagy gene knockout phenotypes is likely due to distinct kinetics of autophagy loss, as deletion of Atg5 more gradually extended lifespan phenocopying deletion of Atg7 or Atg12. Thus, autophagy is required for PMC metabolism and ileum stem cell and mammalian survival. Failure to maintain PMCs through autophagy may therefore contribute to IBD.


Subject(s)
Autophagy , Intestines , Receptor, Platelet-Derived Growth Factor alpha , Stem Cells , Animals , Autophagy/genetics , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Autophagy-Related Proteins , Cell Survival , Mice , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Stem Cells/metabolism
2.
J Cell Biol ; 221(6)2022 06 06.
Article in English | MEDLINE | ID: mdl-35446347

ABSTRACT

The endolysosome system plays central roles in both autophagic degradation and secretory pathways, including the release of extracellular vesicles and particles (EVPs). Although previous work reveals important interconnections between autophagy and EVP-mediated secretion, our understanding of these secretory events during endolysosome inhibition remains incomplete. Here, we delineate a secretory autophagy pathway upregulated in response to endolysosomal inhibition, which mediates EVP-associated release of autophagic cargo receptors, including p62/SQSTM1. This secretion is highly regulated and dependent on multiple ATGs required for autophagosome formation, as well as the small GTPase Rab27a. Furthermore, disrupting autophagosome maturation, either via genetic inhibition of autophagosome-to-autolysosome fusion or expression of SARS-CoV-2 ORF3a, is sufficient to induce EVP secretion of autophagy cargo receptors. Finally, ATG-dependent EVP secretion buffers against the intracellular accumulation of autophagy cargo receptors when classical autophagic degradation is impaired. Thus, we propose secretory autophagy via EVPs functions as an alternate route to clear sequestered material and maintain proteostasis during endolysosomal dysfunction or impaired autophagosome maturation.


Subject(s)
Autophagy , Extracellular Vesicles , Lysosomes , Proteostasis , Autophagosomes/metabolism , Extracellular Vesicles/metabolism , Humans , Lysosomes/metabolism , SARS-CoV-2 , Sequestosome-1 Protein , Viroporin Proteins , rab27 GTP-Binding Proteins
3.
J Cell Sci ; 134(2)2021 01 22.
Article in English | MEDLINE | ID: mdl-33483365

ABSTRACT

Autophagy is deregulated in many cancers and represents an attractive target for therapeutic intervention. However, the precise contributions of autophagy to metastatic progression, the principle cause of cancer-related mortality, is only now being uncovered. While autophagy promotes primary tumor growth, metabolic adaptation and resistance to therapy, recent studies have unexpectedly revealed that autophagy suppresses the proliferative outgrowth of disseminated tumor cells into overt and lethal macrometastases. These studies suggest autophagy plays unexpected and complex roles in the initiation and progression of metastases, which will undoubtedly impact therapeutic approaches for cancer treatment. Here, we discuss the intricacies of autophagy in metastatic progression, highlighting and integrating the pleiotropic roles of autophagy on diverse cell biological processes involved in metastasis.


Subject(s)
Autophagy , Neoplasms , Humans , Neoplasm Metastasis , Neoplasms/genetics
4.
Commun Biol ; 3(1): 388, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32681145

ABSTRACT

Autophagy promotes protein degradation, and therefore has been proposed to maintain amino acid pools to sustain protein synthesis during metabolic stress. To date, how autophagy influences the protein synthesis landscape in mammalian cells remains unclear. Here, we utilize ribosome profiling to delineate the effects of genetic ablation of the autophagy regulator, ATG12, on translational control. In mammalian cells, genetic loss of autophagy does not impact global rates of cap dependent translation, even under starvation conditions. Instead, autophagy supports the translation of a subset of mRNAs enriched for cell cycle control and DNA damage repair. In particular, we demonstrate that autophagy enables the translation of the DNA damage repair protein BRCA2, which is functionally required to attenuate DNA damage and promote cell survival in response to PARP inhibition. Overall, our findings illuminate that autophagy impacts protein translation and shapes the protein landscape.


Subject(s)
Autophagy , Gene Expression Regulation , Protein Biosynthesis , RNA, Messenger/metabolism , Ribosomes/metabolism , Animals , Autophagy/physiology , Autophagy-Related Protein 12/metabolism , BRCA2 Protein/metabolism , DNA Damage , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/physiology , Ribosomes/physiology
5.
Autophagy ; 16(6): 1164-1165, 2020 06.
Article in English | MEDLINE | ID: mdl-32267786

ABSTRACT

Macroautophagy/autophagy plays complex, context-dependent roles in cancer. How autophagy governs the emergence of metastatic disease has been incompletely understood. We recently uncovered that genetic autophagy inhibition strongly attenuates primary tumor growth in mammary cancer models, yet paradoxically promotes spontaneous metastasis to the lung and enables the outgrowth of disseminated tumor cells (DTCs) into overt macro-metastases. Furthermore, at both primary and metastatic sites, genetic autophagy inhibition leads to the marked expansion of tumor cells exhibiting aggressive and pro-metastatic basal epithelial differentiation. These pro-metastatic effects of autophagy inhibition are due to the cytosolic accumulation of the autophagy cargo receptor NBR1 in autophagy-deficient tumor cells.


Subject(s)
Autophagy , Breast Neoplasms , Carrier Proteins , Humans , Intracellular Signaling Peptides and Proteins , Macroautophagy
6.
Dev Cell ; 52(5): 591-604.e6, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32084360

ABSTRACT

Although autophagy is being pursued as a therapeutic target in clinical oncology trials, its effects on metastasis, the principal cause of cancer mortality, remain unclear. Here, we utilize mammary cancer models to temporally delete essential autophagy regulators during carcinoma progression. Though genetic ablation of autophagy strongly attenuates primary mammary tumor growth, impaired autophagy promotes spontaneous metastasis and enables the outgrowth of disseminated tumor cells into overt macro-metastases. Transcriptomic analysis reveals that autophagy deficiency elicits a subpopulation of otherwise luminal tumor cells exhibiting basal differentiation traits, which is reversed upon preventing accumulation of the autophagy cargo receptor, Neighbor to BRCA1 (NBR1). Furthermore, pharmacological and genetic induction of autophagy suppresses pro-metastatic differentiation and metastatic outgrowth. Analysis of human breast cancer data reveal that autophagy gene expression inversely correlates with pro-metastatic differentiation signatures and predicts overall and distant metastasis-free survival. Overall, these findings highlight autophagy-dependent control of NBR1 as a key determinant of metastatic progression.


Subject(s)
Autophagy , Intracellular Signaling Peptides and Proteins/metabolism , Mammary Neoplasms, Experimental/metabolism , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , MCF-7 Cells , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Transcriptome
7.
Nat Cell Biol ; 22(2): 187-199, 2020 02.
Article in English | MEDLINE | ID: mdl-31932738

ABSTRACT

Traditionally viewed as an autodigestive pathway, autophagy also facilitates cellular secretion; however, the mechanisms underlying these processes remain unclear. Here, we demonstrate that components of the autophagy machinery specify secretion within extracellular vesicles (EVs). Using a proximity-dependent biotinylation proteomics strategy, we identify 200 putative targets of LC3-dependent secretion. This secretome consists of a highly interconnected network enriched in RNA-binding proteins (RBPs) and EV cargoes. Proteomic and RNA profiling of EVs identifies diverse RBPs and small non-coding RNAs requiring the LC3-conjugation machinery for packaging and secretion. Focusing on two RBPs, heterogeneous nuclear ribonucleoprotein K (HNRNPK) and scaffold-attachment factor B (SAFB), we demonstrate that these proteins interact with LC3 and are secreted within EVs enriched with lipidated LC3. Furthermore, their secretion requires the LC3-conjugation machinery, neutral sphingomyelinase 2 (nSMase2) and LC3-dependent recruitment of factor associated with nSMase2 activity (FAN). Hence, the LC3-conjugation pathway controls EV cargo loading and secretion.


Subject(s)
Autophagosomes/metabolism , Autophagy/genetics , Extracellular Vesicles/metabolism , Microtubule-Associated Proteins/genetics , RNA-Binding Proteins/genetics , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Animals , Autophagosomes/chemistry , Autophagy-Related Protein 7/deficiency , Autophagy-Related Protein 7/genetics , Autophagy-Related Proteins/deficiency , Autophagy-Related Proteins/genetics , Biological Transport , Biotinylation , Extracellular Vesicles/chemistry , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/chemistry , Lysosomes/metabolism , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Proteomics/methods , RAW 264.7 Cells , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , RNA-Binding Proteins/classification , RNA-Binding Proteins/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism
8.
Elife ; 82019 05 28.
Article in English | MEDLINE | ID: mdl-31134896

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is a heterogeneous disease comprised of a basal-like subtype with mesenchymal gene signatures, undifferentiated histopathology and worse prognosis compared to the classical subtype. Despite their prognostic and therapeutic value, the key drivers that establish and control subtype identity remain unknown. Here, we demonstrate that PDA subtypes are not permanently encoded, and identify the GLI2 transcription factor as a master regulator of subtype inter-conversion. GLI2 is elevated in basal-like PDA lines and patient specimens, and forced GLI2 activation is sufficient to convert classical PDA cells to basal-like. Mechanistically, GLI2 upregulates expression of the pro-tumorigenic secreted protein, Osteopontin (OPN), which is especially critical for metastatic growth in vivo and adaptation to oncogenic KRAS ablation. Accordingly, elevated GLI2 and OPN levels predict shortened overall survival of PDA patients. Thus, the GLI2-OPN circuit is a driver of PDA cell plasticity that establishes and maintains an aggressive variant of this disease.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cell Plasticity , Gene Expression Regulation , Nuclear Proteins/metabolism , Osteopontin/metabolism , Pancreatic Neoplasms/pathology , Transcription, Genetic , Zinc Finger Protein Gli2/metabolism , Animals , Cell Line , Disease Models, Animal , Humans , Mice , Models, Theoretical , Neoplasm Transplantation , Transplantation, Heterologous
9.
Mol Cell ; 72(2): 203-204, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30340019

ABSTRACT

In this issue of Molecular Cell, Wan et al. (2018) uncover WIPI2 as a critical rheostat in the control of autophagic degradation by mTORC1 and demonstrate the physiological utility of this signaling axis in promoting the clearance of hepatic lipids.


Subject(s)
Autophagy , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Signal Transduction , TOR Serine-Threonine Kinases
10.
J Clin Invest ; 126(12): 4417-4429, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27775547

ABSTRACT

The rising success of cancer immunotherapy has produced immense interest in defining the clinical contexts that may benefit from this therapeutic approach. To this end, there is a need to ascertain how the therapeutic modulation of intrinsic cancer cell programs influences the anticancer immune response. For example, the role of autophagy as a tumor cell survival and metabolic fitness pathway is being therapeutically targeted in ongoing clinical trials that combine cancer therapies with antimalarial drugs for the treatment of a broad spectrum of cancers, many of which will likely benefit from immunotherapy. However, our current understanding of the interplay between autophagy and the immune response remains incomplete. Here, we have evaluated how autophagy inhibition impacts the antitumor immune response in immune-competent mouse models of melanoma and mammary cancer. We observed equivalent levels of T cell infiltration and function within autophagy-competent and -deficient tumors, even upon treatment with the anthracycline chemotherapeutic doxorubicin. Similarly, we found equivalent T cell responses upon systemic treatment of tumor-bearing mice with antimalarial drugs. Our findings demonstrate that antitumor adaptive immunity is not adversely impaired by autophagy inhibition in these models, allowing for the future possibility of combining autophagy inhibitors with immunotherapy in certain clinical contexts.


Subject(s)
Antimalarials/pharmacology , Autophagy/drug effects , Immunity, Cellular/drug effects , Mammary Neoplasms, Experimental , Melanoma , T-Lymphocytes/immunology , Animals , Autophagy/immunology , Cell Line, Tumor , Female , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Melanoma/drug therapy , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Transgenic , T-Lymphocytes/pathology
11.
Cancer Res ; 76(10): 2932-43, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27197230

ABSTRACT

Triple-negative breast cancer (TNBC) is considered an early onset subtype of breast cancer that carries with it a poorer prognosis in young rather than older women for reasons that remain poorly understood. Hematopoiesis in the bone marrow becomes altered with age and may therefore affect the composition of tumor-infiltrating hematopoietic cells and subsequent tumor progression. In this study, we investigated how age- and tumor-dependent changes to bone marrow-derived hematopoietic cells impact TNBC progression. Using multiple mouse models of TNBC tumorigenesis and metastasis, we found that a specific population of bone marrow cells (BMC) upregulated CSF-1R and secreted the growth factor granulin to support stromal activation and robust tumor growth in young mice. However, the same cell population in old mice expressed low levels of CSF1R and granulin and failed to promote tumor outgrowth, suggesting that age influences the tumorigenic capacity of BMCs in response to tumor-associated signals. Importantly, BMCs from young mice were sufficient to activate a tumor-supportive microenvironment and induce tumor progression in old mice. These results indicate that hematopoietic age is an important determinant of TNBC aggressiveness and provide rationale for investigating age-stratified therapies designed to prevent the protumorigenic effects of activated BMCs. Cancer Res; 76(10); 2932-43. ©2016 AACR.


Subject(s)
Bone Marrow Cells/pathology , Hematopoiesis/physiology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment , Age Factors , Age of Onset , Animals , Apoptosis , Blotting, Western , Bone Marrow Cells/metabolism , Cell Proliferation , Disease Progression , Female , Flow Cytometry , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Progranulins , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Nat Cell Biol ; 17(1): 1-3, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25679028
13.
Cancer Discov ; 4(6): 716-29, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24670723

ABSTRACT

UNLABELLED: Neoplastic cells rely on the tumor microenvironment (TME) for survival and progression factors. Indeed, senescent and cancer-associated fibroblasts (CAF) express factors that promote tumorigenesis that are collectively referred to as the senescence-associated secretory phenotype (SASP). Despite their importance in tumorigenesis, the mechanisms that control TME-derived factor expression remain poorly understood. Here, we address a key unanswered question: how the SASP is sustained in senescent fibroblasts and CAFs. We find that the mitogen-activated protein kinase p38 (p38MAPK) controls AUF1 occupancy on SASP mRNAs and thus controls their stability. The importance of this regulatory mechanism is underscored by our findings that stromal-specific p38MAPK inhibition abrogates the tumor-promoting activities of CAFs and senescent fibroblasts. Our data suggest that targeting SASP mRNA stability through inhibition of p38MAPK will significantly aid the development of clinical strategies to target the TME. SIGNIFICANCE: The TME plays a key role in tumorigenesis. We demonstrate that p38MAPK governs a posttranscriptional mechanism that sustains the protumorigenic SASP. Inhibition of p38MAPK abrogates the tumor-promoting activities of CAFs and senescent fibroblasts. Thus, p38MAPK is a TME-specific Achilles' heel that may be exploited as a new therapeutic target.


Subject(s)
Fibroblasts/metabolism , Neoplasms/metabolism , Tumor Microenvironment , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Cells, Cultured , Cellular Senescence , Female , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Humans , Imidazoles/pharmacology , Lipopolysaccharides , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Tumor Necrosis Factor-alpha/blood , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
14.
Cancer Discov ; 3(8): 922-35, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23689072

ABSTRACT

The causes for malignant progression of disseminated tumors and the reasons recurrence rates differ in women with different breast cancer subtypes are unknown. Here, we report novel mechanisms of tumor plasticity that are mandated by microenvironmental factors and show that recurrence rates are not strictly due to cell-intrinsic properties. Specifically, outgrowth of the same population of incipient tumors is accelerated in mice with triple-negative breast cancer (TNBC) relative to those with luminal breast cancer. Systemic signals provided by overt TNBCs cause the formation of a tumor-supportive microenvironment enriched for EGF and insulin-like growth factor-I (IGF-I) at distant indolent tumor sites. Bioavailability of EGF and IGF-I enhances the expression of transcription factors associated with pluripotency, proliferation, and epithelial-mesenchymal transition. Combinatorial therapy with EGF receptor and IGF-I receptor inhibitors prevents malignant progression. These results suggest that plasticity and recurrence rates can be dictated by host systemic factors and offer novel therapeutic potential for patients with TNBC.


Subject(s)
Epidermal Growth Factor/metabolism , Insulin-Like Growth Factor I/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation , Disease Progression , Epidermal Growth Factor/genetics , Epithelial-Mesenchymal Transition , ErbB Receptors/antagonists & inhibitors , Female , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor I/genetics , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Recurrence, Local , Neoplasm Transplantation , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Stromal Cells/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Tumor Microenvironment/physiology
15.
Biochim Biophys Acta ; 1832(7): 1070-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23123598

ABSTRACT

Studies of epithelial cancers (i.e., carcinomas) traditionally focused on transformation of the epithelium (i.e., the cancer cells) and how aberrant signaling within the cancer cells modulates the surrounding tissue of origin. In more recent decades, the normal cells, blood vessels, molecules, and extracellular components that surround the tumor cells, collectively known as the "tumor microenvironment" or "stroma", have received increasing attention and are now thought to be key regulators of tumor initiation and progression. Of particular relevance to the work reviewed herein are the fibroblasts, which make up the major cell type within the microenvironment of most carcinomas. Due to their inherent heterogeneity, plasticity, and function, it is perhaps not surprising that fibroblasts are ideal modulators of normal and cancerous epithelium; however, these aspects also present challenges if we are to interrupt their tumor-supportive functions. Here, we review the current body of knowledge and the many questions that still remain about the special entity known as the cancer-associated fibroblast. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.


Subject(s)
Fibroblasts , Neoplasms , Cell Transformation, Neoplastic , Disease Progression , Fibroblasts/metabolism , Humans , Neoplasms/metabolism , Signal Transduction , Tumor Microenvironment
16.
Cancer Discov ; 2(12): 1150-65, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22896036

ABSTRACT

UNLABELLED: Breast cancer recurrence rates vary following treatment, suggesting that tumor cells disseminate early from primary sites but remain indolent indefinitely before progressing to symptomatic disease. The reasons why some indolent disseminated tumors erupt into overt disease are unknown. We discovered a novel process by which certain luminal breast cancer (LBC) cells and patient tumor specimens (LBC "instigators") establish a systemic macroenvironment that supports outgrowth of otherwise-indolent disseminated tumors ("responders"). Instigating LBCs secrete cytokines that are absorbed by platelets, which are recruited to responding tumor sites where they aid vessel formation. Instigator-activated bone marrow cells enrich responding tumor cell expression of CD24, an adhesion molecule for platelets, and provide a source of VEGF receptor 2(+) tumor vessel cells. This cascade results in growth of responder adenocarcinomas and is abolished when platelet activation is inhibited by aspirin. These findings highlight the macroenvironment as an important component of disease progression that can be exploited therapeutically. SIGNIFICANCE: Currently, processes that mediate progression of otherwise indolent tumors are not well understood, making it difficult to accurately predict which cancer patients are likely to relapse. Our findings highlight the macroenvironment as an important component of disease progression that can be exploited to more accurately identify patients who would benefit from adjuvant therapy.


Subject(s)
Blood Platelets/pathology , Bone Marrow Cells/pathology , Breast Neoplasms/pathology , Animals , Blood Platelets/metabolism , Bone Marrow Cells/metabolism , Breast Neoplasms/blood , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , CD24 Antigen/metabolism , Cell Communication/physiology , Disease Progression , Female , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/pathology , Prognosis , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
J Relig Health ; 50(3): 575-91, 2011 Sep.
Article in English | MEDLINE | ID: mdl-19830564

ABSTRACT

This study investigated the relationships between negative attitudes towards homosexuals and two traditional ideologies: religiosity and nationalism, and explored the link with attachment style. An Internet survey yielded 290 participants, of highly diverse ages, nationalities, and religious backgrounds. The participants provided demographic details, and completed measures of adult attachment, nationalism, religiosity, and both explicit and implicit measures of homonegativity. The results indicated that both nationalism and religiosity were highly significant predictors of homonegativity. In the religious group, homonegativity and religiosity were positively related. This finding was greater for less securely attached individuals. Avoidance moderated the relationship in religious females, while anxiety moderated the relationship in religious males. No significant attachment moderation was found between the nationalism-homonegativity relationships.


Subject(s)
Homosexuality/psychology , Object Attachment , Religion , Adolescent , Adult , Aged , Data Collection , Female , Humans , Male , Middle Aged , United States , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...