Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Immunity ; 54(9): 1961-1975.e5, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525337

ABSTRACT

Nucleic acids are powerful triggers of innate immunity and can adopt the Z-conformation, an unusual left-handed double helix. Here, we studied the biological function(s) of Z-RNA recognition by the adenosine deaminase ADAR1, mutations in which cause Aicardi-Goutières syndrome. Adar1mZα/mZα mice, bearing two point mutations in the Z-nucleic acid binding (Zα) domain that abolish Z-RNA binding, displayed spontaneous induction of type I interferons (IFNs) in multiple organs, including in the lung, where both stromal and hematopoietic cells showed IFN-stimulated gene (ISG) induction. Lung neutrophils expressed ISGs induced by the transcription factor IRF3, indicating an initiating role for neutrophils in this IFN response. The IFN response in Adar1mZα/mZα mice required the adaptor MAVS, implicating cytosolic RNA sensing. Adenosine-to-inosine changes were enriched in transposable elements and revealed a specific requirement of ADAR1's Zα domain in editing of a subset of RNAs. Thus, endogenous RNAs in Z-conformation have immunostimulatory potential curtailed by ADAR1, with relevance to autoinflammatory disease in humans.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Adenosine Deaminase/genetics , Interferon Type I/immunology , RNA, Double-Stranded/genetics , Adenosine/genetics , Adenosine/metabolism , Animals , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Inosine/genetics , Inosine/metabolism , Interferon Type I/genetics , Mice , Mutation , Nervous System Malformations/genetics , Nervous System Malformations/immunology , RNA Editing/genetics , RNA, Double-Stranded/metabolism
2.
Bioessays ; 41(5): e1800260, 2019 05.
Article in English | MEDLINE | ID: mdl-30970156

ABSTRACT

Inflammatory mediators have an established role in inducing insulin resistance and promoting hyperglycemia. In turn, hyperglycemia has been argued to drive immune cell dysfunction as a result of mitochondrial dysfunction. Here, the authors review the evidence challenging this view. First, it is pointed out that inflammatory mediators are known to induce altered mitochondrial function. In this regard, critical care patients suffer both an elevated inflammatory tone as well as hyperglycemia, rendering it difficult to distinguish between the effects of inflammation and hyperglycemia. Second, emerging evidence indicates that a decrease in mitochondrial respiration and an increase in reactive oxygen species (ROS) production are not necessarily manifestations of pathology, but adaptations taking shape as the mitochondria is abdicating its adenosine triphosphate (ATP)-producing function (which is taken over by glycolysis) and instead becomes "retooled" for an immunological role. Collectively, these observations challenge the commonly held belief that acute hyperglycemia induces mitochondrial damage leading to immune cell dysfunction.


Subject(s)
Hyperglycemia/pathology , Inflammation/complications , Mitochondria/immunology , Humans , Hyperglycemia/immunology , Hyperglycemia/metabolism , Inflammation/metabolism , Inflammation/physiopathology , Insulin/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism
3.
Exp Cell Res ; 381(2): 280-287, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31121155

ABSTRACT

Breast cancer is frequently diagnosed in women and poses a major health problem throughout the world. Currently, the unresponsiveness of cancer cells to chemotherapeutics is a major concern. During chemotherapeutic treatment with Doxorubicin, neighbouring cells in the tumor microenvironment are also damaged. Depending on the concentration of Doxorubicin, apoptotic or senescent fibroblasts in the tumor microenvironment can then secrete a variety of bioactive molecules which promote tumor growth, metastasis and drug resistance. Mouse embryonic fibroblasts (MEFs) were treated with Doxorubicin to induce apoptosis and senescence respectively. Conditioned media was collected from the MEFs and was used to assess the paracrine effects between fibroblasts and E0771 murine breast cancer cells. Senescent fibroblasts significantly increased cell viability in E0771 cells following Doxorubicin treatment by activating Akt and ERK. Autophagy contributed to cancer cell death and not to treatment resistance in breast cancer cells. Our results highlight the complexity of the tumor microenvironment where chemotherapeutic agents such as Doxorubicin can induce significant changes fibroblasts which can affect tumor growth via the secretion of paracrine factors. Here we have demonstrated that those secreted paracrine factors enhance breast cancer growth and induce therapeutic resistance through the evasion of apoptotic cell death.


Subject(s)
Culture Media, Conditioned/pharmacology , Doxorubicin/therapeutic use , Fibroblasts/metabolism , Mammary Neoplasms, Animal/drug therapy , Paracrine Communication/physiology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cells, Cultured , Cellular Senescence/drug effects , Cellular Senescence/physiology , Coculture Techniques , Culture Media, Conditioned/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Embryo, Mammalian , Female , Fibroblasts/cytology , Fibroblasts/physiology , Mammary Neoplasms, Animal/pathology , Mice , Tumor Microenvironment/drug effects
4.
BMC Cancer ; 19(1): 757, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31370818

ABSTRACT

BACKGROUND: Doxorubicin is currently the most effective chemotherapeutic drug used to treat breast cancer. It has, however, been shown that doxorubicin can induce drug resistance resulting in poor patient prognosis and survival. Studies reported that the interaction between signalling pathways can promote drug resistance through the induction of proliferation, cell cycle progression and prevention of apoptosis. The aim of this study was therefore to determine the effects of doxorubicin on apoptosis signalling, autophagy, the mitogen-activated protein kinase (MAPK)- and phosphoinositide 3-kinase (PI3K)/Akt signalling pathway, cell cycle control, and regulators of the epithelial-mesenchymal transition (EMT) process in murine breast cancer tumours. METHODS: A tumour-bearing mouse model was established by injecting murine E0771 breast cancer cells, suspended in Hank's Balances Salt Solution and Corning® Matrigel® Basement Membrane Matrix, into female C57BL/6 mice. Fourty-seven mice were randomly divided into three groups, namely tumour control (received Hank's Balances Salt Solution), low dose doxorubicin (received total of 6 mg/ml doxorubicin) and high dose doxorubicin (received total of 15 mg/ml doxorubicin) groups. A higher tumour growth rate was, however, observed in doxorubicin-treated mice compared to the untreated controls. We therefore compared the expression levels of markers involved in cell death and survival signalling pathways, by means of western blotting and fluorescence-based immunohistochemistry. RESULTS: Doxorubicin failed to induce cell death, by means of apoptosis or autophagy, and cell cycle arrest, indicating the occurrence of drug resistance and uncontrolled proliferation. Activation of the MAPK/ extracellular-signal-regulated kinase (ERK) pathway contributed to the resistance observed in treated mice, while no significant changes were found with the PI3K/Akt pathway and other MAPK pathways. Significant changes were also observed in cell cycle p21 and DNA replication minichromosome maintenance 2 proteins. No significant changes in EMT markers were observed after doxorubicin treatment. CONCLUSIONS: Our results suggest that doxorubicin-induced drug resistance and tumour growth can occur through the adaptive role of the MAPK/ERK pathway in an effort to protect tumour cells. Previous studies have shown that the efficacy of doxorubicin can be improved by inhibition of the ERK signalling pathway and thereby treatment failure can be overcome.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Animals , Apoptosis , Autophagy , Cell Cycle , Cell Line, Tumor , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
5.
Crit Care ; 22(1): 231, 2018 09 30.
Article in English | MEDLINE | ID: mdl-30268137

ABSTRACT

Nutritional support continues to receive much attention as a possible intervention to prevent loss of lean tissue mass, promote recovery and re-establish proper immune function in critical care patients. Yet there remains much controversy regarding the clinical efficacy of such interventions. In addition to the direct effect of nutrition in terms of micro- and macronutrient content, nutritional formulations may exert an effect via the physiological response to feeding. Here, we highlight the key role of postprandial reabsorbed bile acids in attenuating both the inflammatory response and autophagy. These observations suggest that not all patients would benefit from aggressive nutritional support.


Subject(s)
Bile Acids and Salts/therapeutic use , Nutritional Support/methods , Autophagy/drug effects , Energy Intake/drug effects , Energy Intake/physiology , Humans , Nutritional Status/drug effects , Nutritional Support/statistics & numerical data
6.
Cell Biochem Funct ; 36(2): 65-79, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29399832

ABSTRACT

Autophagy plays a major role in the adaptive metabolic response of cancer cells during adverse conditions such as nutrient deprivation. However, specific data that assess metabolite profiles in context with adenosine triphosphate (ATP) availability and cell death susceptibility remain limited. Human breast cancer cells, MDAMB231, and normal breast epithelial cells, MCF12A, were subjected to short-term amino acid starvation and the cellular apoptotic and autophagic responses assessed. The role of autophagy in the control of cellular amino acid, ATP, free fatty acid, and glucose levels during amino acid starvation were compared. We demonstrate that breast cancer cells have an increased metabolic demand contributing to significant amino acid and ATP depletion in a nutrient-poor environment. Upregulation of autophagy was important for the generation of amino acids and free fatty acids and maintenance of cellular ATP levels. In contrast to normal cells, breast cancer cells were unable to maintain the response after 12 hours of amino acid starvation. Regulation of autophagic activity in these environments had indirect consequences on cell death susceptibility. Overall, our data provide support for autophagy as an important survival mechanism capable of providing metabolic substrates when cancer cells are faced with nutrient-deprived environments. SIGNIFICANCE OF STUDY: The results obtained in this study helps to expand our current knowledge on how cells respond to environmental changes; the biochemical and metabolic consequences and the physiological processes activated in response. The environmental stress applied in this study is relevant to tumour physiology, and results can be translated to cancer therapeutic and clinical research areas, ultimately assisting in the specific targeting of cancer cells while avoiding harm to normal cells.


Subject(s)
Adenosine Triphosphate/metabolism , Amino Acids/metabolism , Autophagy , Cells, Cultured , Humans
7.
Crit Care ; 21(1): 202, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28768529

ABSTRACT

There is an ongoing debate regarding the efficacy of glycaemic control in critically ill patients. Here we briefly highlight the key function of elevated glucose in critically ill patients, namely, to enable elevation of aerobic glycolysis in rapidly dividing cells. In particular, aerobic glycolysis provides metabolic intermediates necessary for expansion of biomass in immune cells and promotion of tissue repair. Furthermore, we emphasise that insulin may inhibit autophagy, a cell survival process used in the bulk degradation of cellular debris and damaged organelles. These observations provide a rational basis for tolerating elevated glucose levels in certain critically ill patients.


Subject(s)
Critical Illness/therapy , Hyperglycemia/drug therapy , Immune System Phenomena/physiology , Blood Glucose/analysis , Humans , Insulin/pharmacology , Insulin/therapeutic use , Intensive Care Units/organization & administration , Prevalence
8.
Front Oncol ; 12: 1000925, 2022.
Article in English | MEDLINE | ID: mdl-36248994

ABSTRACT

It has been established that the acute phase protein, Serum amyloid A (SAA), which is usually synthesized by the liver, is also synthesized by cancer cells and cancer-associated cells in the tumor microenvironment. SAA also activates modulators of autophagy, such as the PI3K/Akt and MAPK signaling pathways. However, the role of SAA in autophagy in breast cancer still remains to be elucidated. The aim of this study was to investigate the role of SAA in the regulation of signaling pathways and autophagy in in vitro and in vivo models of breast cancer. The MDA-MB-231 and MCF7 cell lines were transiently transfected to overexpress SAA1. A tumor-bearing SAA1/2 knockout mouse model was also utilized in this study. SAA1 overexpression activated ERK signaling in the MDA-MB-231 cells, downregulated the PI3K pathway protein, PKB/Akt, in the MCF7 cell line, while SAA1/2 knockout also inhibited Akt. Furthermore, SAA1 overexpression in vitro downregulated autophagy, while the expression of SQSTM1/p62 was increased in the MCF7 cells, and SAA1/2 knockout induced autophagy in vivo. SAA overexpression in the MDA-MB-231 and MCF7 cells resulted in an increase in cell viability and increased the expression of the proliferation marker, MCM2, in the MCF7 cells. Furthermore, knockout of SAA1/2 resulted in an altered inflammatory profile, evident in the decrease of plasma IL-1ß, IL-6 and IL-10, while increasing the plasma levels of MCP-1 and TNF-α. Lastly, SAA1/2 knockout promoted resistance to apoptosis and necrosis through the regulation of autophagy. SAA thus regulates autophagy in breast cancer cells to promote tumorigenesis.

9.
Cytokine Growth Factor Rev ; 59: 62-70, 2021 06.
Article in English | MEDLINE | ID: mdl-33144050

ABSTRACT

Breast cancer is the most frequently diagnosed cancer in women globally. Although there have been many significant advances made in the diagnosis and treatment of breast cancer, numerous unresolved challenges remain, which include prevention, early diagnosis, metastasis and recurrence. The role of inflammation in cancer development is well established and is believed to be one of the leading hallmarks of cancer progression. Recently, the role of the inflammasome, a cytosolic multiprotein complex, has received attention in different cancers. By contributing to the activation of inflammatory cytokines the inflammasome intensifies the inflammatory cascade. The inflammasome can be activated through several pathways, which include the binding of pattern associated molecular patterns (PAMPs) and damage associated molecular patterns (DAMPs) to toll-like receptors (TLRs). Serum amyloid A (SAA), a non-specific acute-phase protein, can function as an endogenous DAMP by binding to pattern recognition receptors like TLRs on both breast cancer cells and cancer associated fibroblasts (CAFs). SAA can thus stimulate the production of IL-1ß, thereby creating a favourable inflammatory environment to support tumour growth. The aim of this review is to highlight the possible role of SAA as an endogenous DAMP in the tumour microenvironment (TME) thereby promoting breast cancer growth through the activation of the NLRP3 inflammasome.


Subject(s)
Breast Neoplasms , Inflammasomes , Humans , Interleukin-1beta , NLR Family, Pyrin Domain-Containing 3 Protein , Serum Amyloid A Protein , Toll-Like Receptors , Tumor Microenvironment
10.
Cell Mol Gastroenterol Hepatol ; 12(4): 1329-1341, 2021.
Article in English | MEDLINE | ID: mdl-34217896

ABSTRACT

BACKGROUND & AIMS: Identifying new approaches to lessen inflammation, as well as the associated malignant consequences, remains crucial to improving the lives and prognosis of patients diagnosed with inflammatory bowel diseases. Although it previously has been suggested as a suitable biomarker for monitoring disease activity in patients diagnosed with Crohn's disease, the role of the acute-phase protein serum amyloid A (SAA) in inflammatory bowel disease remains unclear. In this study, we aimed to assess the role of SAA in colitis-associated cancer. METHODS: We established a model of colitis-associated cancer in wild-type and SAA double-knockout (Saa1/2-/-) mice by following the azoxymethane/dextran sulfate sodium protocol. Disease activity was monitored throughout the study while colon and tumor tissues were harvested for subsequent use in cytokine analyses, Western blot, and immunohistochemistry +experiments. RESULTS: We observed attenuated disease activity in mice deficient for Saa1/2 as evidenced by decreased weight loss, increased stool consistency, decreased rectal bleeding, and decreased colitis-associated tissue damage. Macrophage infiltration, including CD206+ M2-like macrophages, also was attenuated in SAA knockout mice, while levels of interleukin 4, interleukin 10, and tumor necrosis factor-ɑ were decreased in the distal colon. Mice deficient for SAA also showed a decreased tumor burden, and tumors were found to have increased apoptotic activity coupled with decreased expression for markers of proliferation. CONCLUSION: Based on these findings, we conclude that SAA has an active role in inflammatory bowel disease and that it could serve as a therapeutic target aimed at decreasing chronic inflammation and the associated risk of developing colitis-associated cancer.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/metabolism , Disease Susceptibility , Serum Amyloid A Protein/metabolism , Animals , Biomarkers , Cell Transformation, Neoplastic/genetics , Colitis-Associated Neoplasms/pathology , Disease Models, Animal , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Protein Isoforms , Serum Amyloid A Protein/genetics
11.
Front Cell Dev Biol ; 8: 565915, 2020.
Article in English | MEDLINE | ID: mdl-33178685

ABSTRACT

Many clinical trials are beginning to assess the effectiveness of compounds known to regulate autophagy in patients receiving anti-cancer chemotherapy. However, autophagy inhibition, through exogenous inhibitors, or activation, through starvation, has revealed conflicting roles in cancer management and chemotherapeutic outcome. This study aimed to assess the effect of amino acid starvation on doxorubicin-treated breast cancer cells by assessing the roles of autophagy and apoptosis. An in vitro breast cancer model consisting of the normal breast epithelial MCF12A and the metastatic breast cancer MDAMB231 cells was used. Apoptotic and autophagic parameters were assessed following doxorubicin treatments, alone or in combination with bafilomycin, ATG5 siRNA or amino acid starvation. Inhibition of autophagy, through ATG5 siRNA or bafilomycin treatment, increased caspase activity and intracellular doxorubicin concentrations in MCF12A and MDAMB231 cells during doxorubicin treatment. While amino acid starvation increased autophagic activity and decreased caspase activity and intracellular doxorubicin concentrations in MCF12A cells, no changes in autophagic parameters or caspase activity were observed in MDAMB231 cells. Our in vivo data showed that 24 h protein starvation during high dose doxorubicin treatment resulted in increased survival of tumor-bearing GFP-LC3 mice. Results from this study suggest that short term starvation during doxorubicin chemotherapy may be a realistic avenue for adjuvant therapy, especially with regards to the protection of non-cancerous cells. More research is however, needed to fully understand the regulation of autophagic flux during starvation.

12.
Sci Rep ; 8(1): 16798, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30429533

ABSTRACT

Many chronic diseases, including those classified as cardiovascular, neurodegenerative, or autoimmune, are characterized by persistent inflammation. The origin of this inflammation is mostly unclear, but it is typically mediated by inflammatory biomarkers, such as cytokines, and affected by both environmental and genetic factors. Recently circulating bacterial inflammagens such as lipopolysaccharide (LPS) have been implicated. We used a highly selective mouse monoclonal antibody to detect bacterial LPS in whole blood and/or platelet poor plasma of individuals with Parkinson's Disease, Alzheimer's type dementia, or Type 2 Diabetes Mellitus. Our results showed that staining is significantly enhanced (P < 0.0001) compared to healthy controls. Aberrant blood clots in these patient groups are characterized by amyloid formation as shown by the amyloid-selective stains thioflavin T and Amytracker™ 480 or 680. Correlative Light-Electron Microscopy (CLEM) illustrated that the LPS antibody staining is located in the same places as where amyloid fibrils may be observed. These data are consistent with the Iron Dysregulation and Dormant Microbes (IDDM) hypothesis in which bacterial inflammagens such as LPS are responsible for anomalous blood clotting as part of the aetiology of these chronic inflammatory diseases.


Subject(s)
Alzheimer Disease/metabolism , Diabetes Mellitus, Type 2/metabolism , Fibrin/metabolism , Lipopolysaccharides/metabolism , Microscopy, Electron/methods , Parkinson Disease/metabolism , Aged , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid/analysis , Blood Coagulation/drug effects , Blood Specimen Collection , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/pathology , Female , Humans , Inflammation/etiology , Lipopolysaccharides/adverse effects , Male , Middle Aged , Parkinson Disease/etiology , Parkinson Disease/pathology , Protein Binding
13.
Biochem Pharmacol ; 148: 174-183, 2018 02.
Article in English | MEDLINE | ID: mdl-29309757

ABSTRACT

Understanding the response of cancer cells to anti-cancer therapies is crucial to unraveling and preventing the development of therapeutic resistance. The human AHNAK protein is a giant scaffold protein implicated in several diverse cellular functions. The role of AHNAK in cancer is however unclear as the protein has previously been described as a tumor suppressor, as well as being essential for tumor metastasis and invasion, while also being implicated in selected chemotherapeutic responses. To clarify the role of AHNAK in cancer, we investigated the effect of doxorubicin treatment on AHNAK in doxorubicin-sensitive MCF-7 and doxorubicin-resistant MDA-MB-231 breast cancer cell lines, as well as in a tumor-bearing mouse model. The role of AHNAK in the cellular response of breast cancer cells to doxorubicin was also investigated. We report here, for the first time, an association between AHNAK and resistance to doxorubicin. While treatment with doxorubicin modulated AHNAK protein expression both in vitro and in vivo in a dose-dependent manner, no changes in its cellular localization were observed. AHNAK knockdown prevented doxorubicin-induced modulation of cleaved caspase 7 protein expression and cell cycle arrest, while its overexpression decreased cleaved caspase 7 and cleaved PARP levels and induced S-phase arrest, changes that were comparable to the effects of doxorubicin. This novel association was restricted to doxorubicin-resistant cells, implicating the protein in therapeutic resistance. These findings confirm that AHNAK does indeed function in the chemotherapeutic response of breast cancer cells while also emphasizing the need for further investigation into potential implications for AHNAK in terms of predicting and modulating treatment response.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics
14.
Biol Direct ; 10: 47, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26341882

ABSTRACT

BACKGROUND: The characterization of a completely novel adaptive immune system (AIS) in jawless vertebrates (hagfish and lampreys) presents an excellent opportunity for exploring similarities and differences in design principles. It also highlights a somewhat neglected question: Why did vertebrates, representing only 5 % of all animals, evolve a system as complex as an AIS twice, whereas invertebrates failed to do so? A number of theories have been presented in answer to this question. However, these theories either fail to explain why invertebrates would not similarly develop an AIS and are confounded by issues of causality, or have been challenged by more recent findings. PRESENTATION OF THE HYPOTHESIS: Instead of identifying a selective pressure that would drive the development of an AIS, we hypothesise that invertebrates failed to develop an AIS because of the evolutionary constraints imposed by these animals' physiological context. In particular, we argue that a number of vascular innovations in vertebrates allowed the effective implementation of an AIS. A lower blood volume allowed for a higher antibody titer (i.e., less 'diluted' antibody concentration), rendering these immune effectors more cost-effective. In addition, both a high circulatory velocity and the ability of endothelium to coordinate immune cell trafficking promote 'epitope sampling'. Collectively, these innovations allowed the effective implementation of AIS in vertebrates. TESTING THE HYPOTHESIS: The hypothesis posits that a number of innovations to the vascular system provided the release from constraints which allowed the implementation of an AIS. However, this hypothesis would be refuted by phylogenetic analysis demonstrating that the AIS preceded these vascular innovations. The hypothesis also suggests that vascular performance would have an impact on the efficacy of an AIS, thus predicting a correlation between the vascular parameters of a species and its relative investment in AIS. The contribution of certain vascular innovations in augmenting immune functionality of an AIS can be tested by modelling the effect of different vascular parameters on AIS efficacy. IMPLICATIONS OF THE HYPOTHESIS: The hypothesis not only explains the immunological dimorphism between vertebrates and invertebrates but also brings to attention the fact that immunity is dependent on more than just an immune system.


Subject(s)
Adaptive Immunity , Biological Evolution , Endothelium/physiology , Immune System/physiology , Invertebrates/immunology , Vertebrates/immunology , Animals , Invertebrates/genetics , Vertebrates/genetics
SELECTION OF CITATIONS
SEARCH DETAIL