Podcast
Questions and Answers
Within the framework of T cell development and selection, what critical mechanism prevents autoimmunity by inducing apoptosis in T cells that exhibit high-affinity binding to self-antigens presented on thymic epithelial cells, while simultaneously ensuring the survival of T cells with moderate affinity for self-MHC molecules?
Within the framework of T cell development and selection, what critical mechanism prevents autoimmunity by inducing apoptosis in T cells that exhibit high-affinity binding to self-antigens presented on thymic epithelial cells, while simultaneously ensuring the survival of T cells with moderate affinity for self-MHC molecules?
- Receptor editing in immature T cells, where the T cell receptor (TCR) undergoes rearrangement to alter its affinity for self-antigens, preventing strong autoreactive responses.
- Peripheral tolerance induced by regulatory T cells (Tregs) that suppress autoreactive T cells in the periphery through the expression of CTLA-4 and secretion of immunosuppressive cytokines such as IL-10 and TGF-β.
- Negative selection in the bone marrow, where autoreactive B cells are eliminated via receptor editing or clonal deletion, preventing their migration to the thymus for further T cell interactions.
- Positive selection mediated by cortical thymic epithelial cells (cTECs), followed by negative selection mediated by medullary thymic epithelial cells (mTECs) expressing tissue-restricted antigens (TRAs) under the control of the autoimmune regulator (AIRE) gene. (correct)
In the intricate interplay of innate and adaptive immune responses, which cells are responsible for the critical transition of linking those responses by internalizing pathogens or antigens, processing them into smaller peptides, and presenting those peptides to T cells via MHC class II molecules, thereby initiating T cell activation and cytokine production?
In the intricate interplay of innate and adaptive immune responses, which cells are responsible for the critical transition of linking those responses by internalizing pathogens or antigens, processing them into smaller peptides, and presenting those peptides to T cells via MHC class II molecules, thereby initiating T cell activation and cytokine production?
- Dendritic cells, which are specialized antigen-presenting cells that capture and process antigens, migrate to lymph nodes, and activate T cells, thereby bridging the innate and adaptive immune systems. (correct)
- Neutrophils, which are the first responders in the innate immune system, engulfing and destroying pathogens through phagocytosis and degranulation, but lacking the ability to present antigens to T cells.
- B cells, which recognize and bind to specific antigens, undergo receptor-mediated endocytosis, and present antigens to T cells, but primarily function in antibody production rather than initiating innate immune responses.
- Macrophages, which reside in tissues and perform phagocytosis, clean up cellular debris, and present antigens to T cells, but do not migrate to lymph nodes to initiate adaptive immune responses.
Imagine that during a clinical trial B cells are rendered unable to undergo somatic hypermutation (SHM). How is this specifically going to impact an individual?
Imagine that during a clinical trial B cells are rendered unable to undergo somatic hypermutation (SHM). How is this specifically going to impact an individual?
- Impaired plasma cell differentiation, leading to a complete absence of antibody production and subsequent susceptibility to opportunistic infections.
- Diminished affinity maturation, resulting in decreased antibody specificity and reduced ability to neutralize rapidly evolving pathogens. (correct)
- Compromised isotype switching leading to unchecked Th1 polarisation owing to a lack of IgG production.
- Disrupted central tolerance, causing systemic autoimmunity due to the uncontrolled survival of self-reactive B cells.
Consider an innovative therapeutic strategy that promotes immune tolerance by selectively upregulating the expression of FoxP3 in autoreactive CD4+ T cells that successfully evaded central tolerance mechanisms. What implications does this strategy have regarding control of peripheral tolerance?
Consider an innovative therapeutic strategy that promotes immune tolerance by selectively upregulating the expression of FoxP3 in autoreactive CD4+ T cells that successfully evaded central tolerance mechanisms. What implications does this strategy have regarding control of peripheral tolerance?
Under circumstances of chronic viral infection, what phenomenon critically impairs T cell function and how does programmed cell death protein 1 (PD-1) directly contribute?
Under circumstances of chronic viral infection, what phenomenon critically impairs T cell function and how does programmed cell death protein 1 (PD-1) directly contribute?
How does HLA class I downregulation by malignant cells specifically influence the balance between innate and adaptive immunity?
How does HLA class I downregulation by malignant cells specifically influence the balance between innate and adaptive immunity?
When considering the complexity of post-translational modifications of MHC molecules, how do alterations such as citrullination or glycosylation affect antigen presentation and the subsequent T cell response?
When considering the complexity of post-translational modifications of MHC molecules, how do alterations such as citrullination or glycosylation affect antigen presentation and the subsequent T cell response?
Supposing you're developing a novel class of adjuvants that aim to harness the power of innate immune sensors, how would triggering intracellular NOD-like receptors (NLRs) and specifically aim at amplifying adaptive immunity to a co-administered vaccine?
Supposing you're developing a novel class of adjuvants that aim to harness the power of innate immune sensors, how would triggering intracellular NOD-like receptors (NLRs) and specifically aim at amplifying adaptive immunity to a co-administered vaccine?
How does chronic HIV infection lead to the progressive depletion of CD4+ T cells, and what are the specific mechanisms leading to CD4+ T cell death?
How does chronic HIV infection lead to the progressive depletion of CD4+ T cells, and what are the specific mechanisms leading to CD4+ T cell death?
In the pathogenesis of autoimmune diseases like rheumatoid arthritis (RA), what is the role of citrullinated proteins and how do they contribute to chronic inflammation and joint destruction?
In the pathogenesis of autoimmune diseases like rheumatoid arthritis (RA), what is the role of citrullinated proteins and how do they contribute to chronic inflammation and joint destruction?
How does dysregulation in the signaling pathways of pattern recognition receptors (PRRs) on innate immune cells potentiate the development of inflammatory and autoimmune diseases?
How does dysregulation in the signaling pathways of pattern recognition receptors (PRRs) on innate immune cells potentiate the development of inflammatory and autoimmune diseases?
How do defects in the complement regulatory proteins (e.g., C1 inhibitor, factor H) contribute to the pathogenesis of autoimmune diseases such as systemic lupus erythematosus (SLE) and atypical hemolytic uremic syndrome (aHUS)?
How do defects in the complement regulatory proteins (e.g., C1 inhibitor, factor H) contribute to the pathogenesis of autoimmune diseases such as systemic lupus erythematosus (SLE) and atypical hemolytic uremic syndrome (aHUS)?
How can a breakdown in peripheral tolerance mechanisms, specifically those involving regulatory T cells (Tregs), lead to the development of autoimmune diseases?
How can a breakdown in peripheral tolerance mechanisms, specifically those involving regulatory T cells (Tregs), lead to the development of autoimmune diseases?
What features of the cytokine milieu within the synovial microenvironment perpetuate chronic inflammation and joint destruction in rheumatoid arthritis (RA)?
What features of the cytokine milieu within the synovial microenvironment perpetuate chronic inflammation and joint destruction in rheumatoid arthritis (RA)?
Considering the pathogenesis of systemic lupus erythematosus (SLE), how do plasmacytoid dendritic cells (pDCs) contribute to systemic autoimmunity and inflammation?
Considering the pathogenesis of systemic lupus erythematosus (SLE), how do plasmacytoid dendritic cells (pDCs) contribute to systemic autoimmunity and inflammation?
In the development of type 1 diabetes (T1D), how do autoreactive T cells specifically recognize and destroy pancreatic beta cells, leading to insulin deficiency?
In the development of type 1 diabetes (T1D), how do autoreactive T cells specifically recognize and destroy pancreatic beta cells, leading to insulin deficiency?
Considering the development of multiple sclerosis (MS), how does molecular mimicry contribute to autoreactive T cell activation and central nervous system (CNS) demyelination?
Considering the development of multiple sclerosis (MS), how does molecular mimicry contribute to autoreactive T cell activation and central nervous system (CNS) demyelination?
How would a genetic defect leading to impaired expression of the autoimmune regulator (AIRE) gene in thymic epithelial cells contribute to the pathogenesis of autoimmune diseases?
How would a genetic defect leading to impaired expression of the autoimmune regulator (AIRE) gene in thymic epithelial cells contribute to the pathogenesis of autoimmune diseases?
In the context of Type I hypersensitivity reactions, what is the mechanistic basis for the 'late phase' response that occurs several hours after initial allergen exposure?
In the context of Type I hypersensitivity reactions, what is the mechanistic basis for the 'late phase' response that occurs several hours after initial allergen exposure?
During antibody-mediated (Type II) hypersensitivity, how does the mechanism of antibody-dependent cell-mediated cytotoxicity (ADCC) lead to tissue damage and cellular destruction?
During antibody-mediated (Type II) hypersensitivity, how does the mechanism of antibody-dependent cell-mediated cytotoxicity (ADCC) lead to tissue damage and cellular destruction?
In the context of Type III hypersensitivity reactions, what mechanistic processes determine the extent and location of tissue damage resulting from immune complex deposition?
In the context of Type III hypersensitivity reactions, what mechanistic processes determine the extent and location of tissue damage resulting from immune complex deposition?
Within the complex landscape of Type IV hypersensitivity responses, what specific mechanisms govern the selective recruitment of immune cells to the site of antigen exposure, leading to the characteristic delayed-type hypersensitivity (DTH) reactions?
Within the complex landscape of Type IV hypersensitivity responses, what specific mechanisms govern the selective recruitment of immune cells to the site of antigen exposure, leading to the characteristic delayed-type hypersensitivity (DTH) reactions?
Characterizing a theoretical immunodeficiency, you find increased B cell numbers in circulation but a marked lack of germinal centers within lymph nodes. Furthermore, in vitro stimulation reveals impaired B cell proliferation upon BCR crosslinking. Which signaling molecule is most likely deficient in these patients?
Characterizing a theoretical immunodeficiency, you find increased B cell numbers in circulation but a marked lack of germinal centers within lymph nodes. Furthermore, in vitro stimulation reveals impaired B cell proliferation upon BCR crosslinking. Which signaling molecule is most likely deficient in these patients?
In individuals presenting with hyper IgM syndrome due to a defect in CD40L, what additional immune deficiency would these individuals MOST likely present with?
In individuals presenting with hyper IgM syndrome due to a defect in CD40L, what additional immune deficiency would these individuals MOST likely present with?
Consider two patients, one diagnosed with Severe Combined Immunodeficiency (SCID) due to a mutation in the IL2RG gene (encoding the common \gamma chain) and another with DiGeorge syndrome. What immune defects and functional capabilities will be shared in BOTH of these patients?
Consider two patients, one diagnosed with Severe Combined Immunodeficiency (SCID) due to a mutation in the IL2RG gene (encoding the common \gamma chain) and another with DiGeorge syndrome. What immune defects and functional capabilities will be shared in BOTH of these patients?
An experimental drug is designed to selectively deplete regulatory T cells (Tregs) in the tumor microenvironment to enhance anti-tumor immunity. However, clinical trials reveal severe autoimmune side effects in a subset of patients. What refinement to this drug is ideal?
An experimental drug is designed to selectively deplete regulatory T cells (Tregs) in the tumor microenvironment to enhance anti-tumor immunity. However, clinical trials reveal severe autoimmune side effects in a subset of patients. What refinement to this drug is ideal?
Bone marrow transplantation is being considered for a nine month old infant. The transplantation team are in disagreement which source of hematopoietic stem cells (HSCs) has the best chance of generating immune self-tolerance and long-term-engraftment. Who should they choose?
Bone marrow transplantation is being considered for a nine month old infant. The transplantation team are in disagreement which source of hematopoietic stem cells (HSCs) has the best chance of generating immune self-tolerance and long-term-engraftment. Who should they choose?
Following successful reconstitution of T cell populations following bone marrow transplantation for treatment of SCID, your patient develops chronic diarrhea, a maculopapular rash, and hepatosplenomegaly. Histological analysis of the involved tissues shows extensive lymphocyte infiltration and tissue destruction. What explains this constellation of symptoms?
Following successful reconstitution of T cell populations following bone marrow transplantation for treatment of SCID, your patient develops chronic diarrhea, a maculopapular rash, and hepatosplenomegaly. Histological analysis of the involved tissues shows extensive lymphocyte infiltration and tissue destruction. What explains this constellation of symptoms?
In Multiple Myeloma that is characterised by excessive production of monoclonal immunoglobulins, what would the MOST likely downstream defect be that contributes to renal damage?
In Multiple Myeloma that is characterised by excessive production of monoclonal immunoglobulins, what would the MOST likely downstream defect be that contributes to renal damage?
In HIV-infected individuals, what cellular mechanism accounts for the establishment and maintenance of viral reservoirs despite antiretroviral therapy (ART)?
In HIV-infected individuals, what cellular mechanism accounts for the establishment and maintenance of viral reservoirs despite antiretroviral therapy (ART)?
You have a patient with HIV presenting with a constellation of symptoms indicative of progressive multifocal leukoencephalopathy (PML). What statement best explains this condition given the context of HIV-mediated immunosuppression?
You have a patient with HIV presenting with a constellation of symptoms indicative of progressive multifocal leukoencephalopathy (PML). What statement best explains this condition given the context of HIV-mediated immunosuppression?
In the complex interplay of T cell activation, what specific intracellular signaling event, subsequent to T cell receptor (TCR) engagement and co-stimulation, is indispensable for the induction of gene transcription required for T cell effector function and proliferation, and how might pharmacological targeting of this event modulate autoimmune responses?
In the complex interplay of T cell activation, what specific intracellular signaling event, subsequent to T cell receptor (TCR) engagement and co-stimulation, is indispensable for the induction of gene transcription required for T cell effector function and proliferation, and how might pharmacological targeting of this event modulate autoimmune responses?
What role does the interplay between central and peripheral tolerance mechanisms play in preventing autoimmune diseases, and how might disruptions in these processes, particularly concerning the regulation of autoreactive T cells, contribute to the pathogenesis of such conditions?
What role does the interplay between central and peripheral tolerance mechanisms play in preventing autoimmune diseases, and how might disruptions in these processes, particularly concerning the regulation of autoreactive T cells, contribute to the pathogenesis of such conditions?
Considering the multifaceted roles of cytokines in regulating the immune response, what are the implications for therapeutic interventions targeting specific cytokine pathways in autoimmune diseases, taking into account potential compensatory mechanisms and off-target effects?
Considering the multifaceted roles of cytokines in regulating the immune response, what are the implications for therapeutic interventions targeting specific cytokine pathways in autoimmune diseases, taking into account potential compensatory mechanisms and off-target effects?
If a novel therapeutic approach aims to selectively inhibit the function of plasmacytoid dendritic cells (pDCs) in patients with systemic lupus erythematosus (SLE), what downstream immunological consequences would be anticipated, specifically regarding the production of type I interferons and the activation of autoreactive B cells?
If a novel therapeutic approach aims to selectively inhibit the function of plasmacytoid dendritic cells (pDCs) in patients with systemic lupus erythematosus (SLE), what downstream immunological consequences would be anticipated, specifically regarding the production of type I interferons and the activation of autoreactive B cells?
Given the complexity of HLA gene polymorphism and its role in antigen presentation, how does the diversity of HLA alleles within a population influence susceptibility and resistance to infectious diseases and autoimmune disorders, and what are the implications for personalized medicine approaches?
Given the complexity of HLA gene polymorphism and its role in antigen presentation, how does the diversity of HLA alleles within a population influence susceptibility and resistance to infectious diseases and autoimmune disorders, and what are the implications for personalized medicine approaches?
How does the chronic activation of the innate immune system, particularly through pattern recognition receptors (PRRs), contribute to the development and perpetuation of autoimmune diseases, and what are the potential therapeutic strategies to modulate this persistent activation?
How does the chronic activation of the innate immune system, particularly through pattern recognition receptors (PRRs), contribute to the development and perpetuation of autoimmune diseases, and what are the potential therapeutic strategies to modulate this persistent activation?
In the context of T cell dysfunction, what specific molecular mechanisms lead to the impaired ability of cytotoxic T lymphocytes (CTLs) to effectively eliminate tumor cells, and how can this knowledge be leveraged to improve cancer immunotherapy strategies?
In the context of T cell dysfunction, what specific molecular mechanisms lead to the impaired ability of cytotoxic T lymphocytes (CTLs) to effectively eliminate tumor cells, and how can this knowledge be leveraged to improve cancer immunotherapy strategies?
Considering the different types of hypersensitivity reactions, how does the mechanism underlying Type IV hypersensitivity, specifically the role of sensitized T cells and macrophages, contribute to the pathogenesis of chronic contact dermatitis, and what targeted therapeutic interventions could disrupt this process?
Considering the different types of hypersensitivity reactions, how does the mechanism underlying Type IV hypersensitivity, specifically the role of sensitized T cells and macrophages, contribute to the pathogenesis of chronic contact dermatitis, and what targeted therapeutic interventions could disrupt this process?
In the context of autoimmune diseases and considering the mechanisms of molecular mimicry, how might microbial infections trigger autoimmune responses and what criteria must be met to definitively establish a causative link between a specific pathogen and an autoimmune condition?
In the context of autoimmune diseases and considering the mechanisms of molecular mimicry, how might microbial infections trigger autoimmune responses and what criteria must be met to definitively establish a causative link between a specific pathogen and an autoimmune condition?
Given the complexity of the immune response in graft rejection scenarios, what specific mechanisms underlie chronic rejection of solid organ transplants, focusing on the roles of alloantibodies, T cell-mediated inflammation, and the resulting vascular damage, and what strategies could be employed to mitigate these processes?
Given the complexity of the immune response in graft rejection scenarios, what specific mechanisms underlie chronic rejection of solid organ transplants, focusing on the roles of alloantibodies, T cell-mediated inflammation, and the resulting vascular damage, and what strategies could be employed to mitigate these processes?
Within the classification of immunodeficiency disorders, what are the key distinctions between primary and secondary forms of immunodeficiency, focusing on their etiologies, typical clinical manifestations, and the implications for diagnostic strategies and therapeutic interventions?
Within the classification of immunodeficiency disorders, what are the key distinctions between primary and secondary forms of immunodeficiency, focusing on their etiologies, typical clinical manifestations, and the implications for diagnostic strategies and therapeutic interventions?
How would you differentiate between leukocyte adhesion deficiency (LAD) type 1, caused by mutations in the ITGB2 gene encoding the CD18 subunit, and LAD type 2, resulting from defects in the SLC35C1 gene affecting the expression of sialyl-Lewis X, regarding their underlying molecular mechanisms, cellular phenotypes, and consequent clinical presentations?
How would you differentiate between leukocyte adhesion deficiency (LAD) type 1, caused by mutations in the ITGB2 gene encoding the CD18 subunit, and LAD type 2, resulting from defects in the SLC35C1 gene affecting the expression of sialyl-Lewis X, regarding their underlying molecular mechanisms, cellular phenotypes, and consequent clinical presentations?
If the development of a novel cancer immunotherapy involves the ex vivo expansion and adoptive transfer of tumor-infiltrating lymphocytes (TILs) with enhanced cytotoxic activity, what specific modifications to the TILs, such as genetic engineering or pharmacological pre-conditioning, would most effectively overcome immunosuppressive mechanisms within the tumor microenvironment and improve therapeutic efficacy?
If the development of a novel cancer immunotherapy involves the ex vivo expansion and adoptive transfer of tumor-infiltrating lymphocytes (TILs) with enhanced cytotoxic activity, what specific modifications to the TILs, such as genetic engineering or pharmacological pre-conditioning, would most effectively overcome immunosuppressive mechanisms within the tumor microenvironment and improve therapeutic efficacy?
Considering the complexities of HIV pathogenesis, what specific cellular and molecular mechanisms account for the establishment and long-term maintenance of latent viral reservoirs within infected individuals despite effective antiretroviral therapy (ART), and how might these reservoirs be specifically targeted for eradication?
Considering the complexities of HIV pathogenesis, what specific cellular and molecular mechanisms account for the establishment and long-term maintenance of latent viral reservoirs within infected individuals despite effective antiretroviral therapy (ART), and how might these reservoirs be specifically targeted for eradication?
What are the distinct immunological mechanisms underlying the pathogenesis of rheumatoid arthritis (RA) and psoriatic arthritis (PsA), focusing on the specific roles of autoantibodies, T cell subsets, cytokines, and the characteristic features of synovial inflammation and joint destruction in each disease?
What are the distinct immunological mechanisms underlying the pathogenesis of rheumatoid arthritis (RA) and psoriatic arthritis (PsA), focusing on the specific roles of autoantibodies, T cell subsets, cytokines, and the characteristic features of synovial inflammation and joint destruction in each disease?
Flashcards
What is immunity?
What is immunity?
The body's defense system against harmful agents and abnormal cells.
How does the immune system work?
How does the immune system work?
Recognizing antigens on pathogens or damaged cells and responding to neutralize or destroy them
Innate immune system
Innate immune system
A fast, non-specific immune response.
Adaptive immune system
Adaptive immune system
Signup and view all the flashcards
Physical barriers
Physical barriers
Signup and view all the flashcards
Neutrophils
Neutrophils
Signup and view all the flashcards
Macrophages
Macrophages
Signup and view all the flashcards
Dendritic Cells
Dendritic Cells
Signup and view all the flashcards
Complement System/Cascade
Complement System/Cascade
Signup and view all the flashcards
Inflammation
Inflammation
Signup and view all the flashcards
Types of T cells
Types of T cells
Signup and view all the flashcards
MHC (HLA)
MHC (HLA)
Signup and view all the flashcards
B cells
B cells
Signup and view all the flashcards
Antibodies
Antibodies
Signup and view all the flashcards
Helper T Cells (CD4+)
Helper T Cells (CD4+)
Signup and view all the flashcards
Cytotoxic T Cells (CD8+)
Cytotoxic T Cells (CD8+)
Signup and view all the flashcards
Regulatory T Cells (Tregs)
Regulatory T Cells (Tregs)
Signup and view all the flashcards
MHC and HLA
MHC and HLA
Signup and view all the flashcards
Human MHC molecules
Human MHC molecules
Signup and view all the flashcards
HLA Class 1
HLA Class 1
Signup and view all the flashcards
HLA Class 2
HLA Class 2
Signup and view all the flashcards
HLA Dysfunction
HLA Dysfunction
Signup and view all the flashcards
Leukocytes
Leukocytes
Signup and view all the flashcards
Types of granulocytes
Types of granulocytes
Signup and view all the flashcards
Types of agranulocytes
Types of agranulocytes
Signup and view all the flashcards
Organs of the immune system
Organs of the immune system
Signup and view all the flashcards
What is the role of bone marrow?
What is the role of bone marrow?
Signup and view all the flashcards
Thymus
Thymus
Signup and view all the flashcards
Spleen
Spleen
Signup and view all the flashcards
Lymph nodes
Lymph nodes
Signup and view all the flashcards
Mucosal-associated lymphoid tissues (MALT)
Mucosal-associated lymphoid tissues (MALT)
Signup and view all the flashcards
Lymphatic system
Lymphatic system
Signup and view all the flashcards
Antigen Recognition
Antigen Recognition
Signup and view all the flashcards
Effector Phase
Effector Phase
Signup and view all the flashcards
Immunologic Memory
Immunologic Memory
Signup and view all the flashcards
Immune Response control
Immune Response control
Signup and view all the flashcards
Regulatory mechanisms of the immune system
Regulatory mechanisms of the immune system
Signup and view all the flashcards
Regulatory T-cells (Tregs)
Regulatory T-cells (Tregs)
Signup and view all the flashcards
Cytokine Regulation
Cytokine Regulation
Signup and view all the flashcards
Abnormal immune responses
Abnormal immune responses
Signup and view all the flashcards
Autoimmunity
Autoimmunity
Signup and view all the flashcards
AutoImmunity key factors
AutoImmunity key factors
Signup and view all the flashcards
Types of Autoimmunity
Types of Autoimmunity
Signup and view all the flashcards
Hypersensitivity
Hypersensitivity
Signup and view all the flashcards
Type 1 Hypersensitivity
Type 1 Hypersensitivity
Signup and view all the flashcards
Type 2 Hypersensitivity
Type 2 Hypersensitivity
Signup and view all the flashcards
Type 3 Hypersensitivity
Type 3 Hypersensitivity
Signup and view all the flashcards
Type 4 Hypersensitivity
Type 4 Hypersensitivity
Signup and view all the flashcards
Immunodeficiency
Immunodeficiency
Signup and view all the flashcards
Examples of immunodeficiency
Examples of immunodeficiency
Signup and view all the flashcards
Immunosuppression
Immunosuppression
Signup and view all the flashcards
Leukaemia
Leukaemia
Signup and view all the flashcards
Lymphoma
Lymphoma
Signup and view all the flashcards
Study Notes
Lecture 9: Abnormal Immunity
- Immunity is the body's defense system, protecting against harmful agents and abnormal cells
- The immune system works by recognizing antigens on pathogens or damaged host cells
- The immune system responds to neutralize or destroy antigens
Types of Immunity
- The immune system offers protection via two main systems: the innate immune system and the adaptive immune system
- These systems work together to defend against harmful substances
- Each system plays specific roles in defense
Innate Immunity
- Provides a fast and non-specific response to intruders
- Physical barriers are the first line of defense: skin, mucosal membranes, cilia, and other anatomical defenses
- Immune cells contribute to the first line of defense through neutrophils, macrophages, and dendritic cells
- Neutrophils are the first responders that phagocytose pathogens
- Macrophages engulf pathogens, clean up dead cells, and activate adaptive immunity
- Dendritic cells link innate and adaptive immunity by presenting antigens to T cells
- The complement system/cascade is a group of proteins that work together to opsonize pathogens & promote inflammation
- Inflammation is triggered by infection or injury; aims to contain infection, but chronic inflammation can lead to tissue damage/autoimmunity
Adaptive Immunity
- Provides a slower, more specific response if the innate system fails
- It discerns the cause of the issue, which takes time, but it more accurate and remembers previous battles
- T cells are responsible for cellular immunity
- Helper T cells (CD4+) assist in activating other immune cells
- Cytotoxic T cells (CD8+) destroy infected or cancerous cells
- MHC (HLA) aids in presenting antigens to T cells and recognizing foreign pathogens which signals the immune system
- B cells produce antibodies that specifically recognize & neutralize pathogens
- After an initial encounter, adaptive immunity develops immunological memory
- This enables a faster & stronger response if re-exposure to the same pathogen occurs
T Cells
- Essential components of the adaptive immune response
- They have varied types with specific functions
- Helper T cells (CD4+) aid in activation and coordination of other cells of the immune system
- Helper T cells are activated when they recognize an antigen presented by HLA Class 2 molecules on antigen presenting cells
- Cytotoxic T cells (CD8+) directly kill infected or abnormal cells, such as infected or cancerous cells
- Activation of cytotoxic T cells occurs upon recognizing antigens presented by MHC Class 1 molecules on infected cells
- Activation of Cytotoxic T cells releases perforins and granzymes
- Regulatory T cells (Tregs) assist in the maintenance of immune tolerance and prevent autoimmunity by suppressing other immune cells' activity
- Tregs limit immune responses to ensure the body's own tissues aren't attacked, balancing the immune response
T Cell Control Mechanisms
- T cells originate within bone marrow but mature within the thymus
- During maturation, T cells undergo positive and negative selection
- Activation requires two signals: T cell receptor must recognize a specific antigen presented by HLA molecules
- A costimulatory signal from antigen presenting cells occurs
- Cytokines such as interleukins influence T cells, determining differentiation of T cell subgroups
T Cell Dysfunction
- May lead to severe immunodeficiencies, autoimmune disorders, cancer, and chronic infections
- If T cells are dysfunctional, the body is unable to mount an effective immune response
- Sometimes this results from a genetic abnormality/deletion, like DiGeorge Syndrome which affects the thymus
- If Tregs can't adequately suppress immune activity, autoimmune diseases are more likely
- Some T cells mistakenly attack systems, like T cells that target the CNS with Multiple Sclerosis
- Defects in cytotoxic T cell activation/function impair the immune system's ability to detect/destroy cancerous cells and allow them to grow uncontrollably
- Downregulation of HLA Class 1 expression is another way tumor cells evade immune detection
- Defective T helper cells can lead to ineffective immune responses, allowing persistent infections to thrive
- In HIV/AIDS, the virus targets CD4+ T cells specifically, weakening the entire immune system
MHC and HLA
- MHC molecules, or Major Histocompatibility Complex, are proteins on cell surfaces that aid immune response and are crucial in antigen presentation
- They facilitate immune recognition & response to foreign pathogens
- MHC molecules are found in all vertebrates and are referred to as HLA genes in humans, or Human Leukocyte Antigen
- There are two main classes of MHC molecules: HLA Class 1 and HLA Class 2
HLA Class 1
- Found on most nucleated cells, presenting endogenous antigens to cytotoxic T cells (CD8+)
- If a cell is infected, has a virus, or is abnormal, it presents proteins on its MHC molecule like a flagging system
- This allows cytotoxic T cells to recognize these foreign antigens, eliminate infected/abnormal cells
HLA Class 2
- Found mainly on antigen presenting cells (APCs), which are specialized immune cells
- HLA class 2 present exogenous antigens to helper T cells (CD4+)
- Helper T cells help activate other immune cells, including B cells and cytotoxic T cells
HLA Control Mechanisms
- Antigen processing is where molecules must bind to a processed peptide, or antigen, before presenting it on the cell surface
- Breaking down proteins from viruses, bacteria, or tumor cells into small peptide fragments involves the process which bind to the HLA molecules
- HLA genes are highly polymorphic, so more pathogens can be recognized because there is higher genetic diversity
- The expression of HLA molecules can be regulated by cytokines and other immune signals
HLA dysfunction
- Mutations or deficiencies in HLA molecules can result in an inability to present antigens properly
- Example: Bare Lymphocyte Syndrome occurs when defects in HLA 1 or 2 result in an impaired immune response
- Triggering an autoimmune response where the immune system attacks its own tissues happens if molecules present self-antigens or mistakenly present normal proteins as foreign
- A connection between HLA alleles and autoimmune diseases is documented e.g. RhA, Ankylosing Spondylitis, Psoriasis, and Crohn's
Further components of the immune system
- White blood cells (WBCs) or leukocytes, primary cells that are involved in immune responses
- Granulocytes and agranulocytes, the main categories blood cells divide into
- Neutrophils, eosinophils, and basophils are Granulocytes
- Monocytes, lymphocytes T and B cells and natural killer cells are Agranulocytes
- Antigen presenting cells (APCs) and mast cells
- Organs of the immune system:
- Bone marrow, primary site of blood cell production, including immune cells and stem cells in bone marrow gives rise to WBCs, RBCs and platelets
- Thymus is located behind the the sternum where T cells mature which makes them capable of recognizing specific antigens
- Spleen is located in the abdomen, filtering blood and removing old/damaged RBCs and aids in trapping pathogens and presenting them to immune cells
- Lymph nodes are small, bean shaped structures found throughout the body that filter lymph and help immune cells recognize pathogens/respond
- Mucosal-associated lymphoid tissues (MALT) are structures, including tonsils, adenoids, Payer's patches in the intestines, and appendix, involved in immune responses which respond to pathogens
Lymphatic System
- A network consisting of vessels, tissues, and organs maintains fluid balance that is essential for the immune system
- Closely related to the circulatory system, transporting wastes to facilitate immune responses with lymph instead of blood
- Lymph is the clear fluid circulating through these vessels, it contains lymphocytes, other immune cells, products of waste, fats and proteins
- Lymphatic vessels carry lymph from tissues to lymph nodes, which activates immune responses
- Lymph nodes filter lymph and house immune cells which respond to pathogens
Immune Response Process
- The immune system recognizes foreign substances by antigen recognition, where immune cells encounter the foreign substance
- The surface of APCs capture, process and present antigen fragments using HLA molecules
- APCs travel to lymph nodes, interacting with naïve lymphocytes, both T and B cells
- Effector T and B cells, capable of performing their functions for Cytotoxic T cells once activated, directly attack cells through apoptosis
- Helper T cells coordinate immune responses by activating other immune cells
- B cells release antibodies into the bloodstream, After pathogen clearance, some activated lymphocytes, B and T cells, become memory cells creating immunological memory
- Memory B cells store genetic blueprints for specific antibodies against the antigens
- Memory T cells form and rapidly respond to the antigen
- Immunological memory provides long-lasting immunity and is the foundation for how vaccinations work
Control of the Immune Response
- Is tightly regulated to defend against pathogens avoiding autoimmunity/inflammation
- Various regulatory mechanisms control the immune response, including tolerance, regulatory T-cells or Tregs, and cytokine regulation
- Regulatory mechanisms control and balance the immune response through Activation, resolution, self-regulation, and prevention of overactivation
- Immune responses should be resolved appropriately once the pathogen is cleared to prevent chronic inflammation/tissue damage
- Preventing excessive immune activation is crucial for autoimmune disorders
- Tolerance is the process where the immune system learns to recognize and not attack the body's cells/tissues which prevents autoimmunity
- Central Tolerance occurs in the thymus for T-cells and in the bone marrow for B-cells, eliminating those reactive to self antigens through negative selection
- Peripheral Tolerance occurs in peripheral tissues after maturation, with extra mechanisms preventing self attacking
Regulatory T-Cells
- Specialized subset of T cells crucial in maintaining immune homeostasis
- Prevents excessive/inappropriate responses such as autoimmunity and inflammation
- CD4+ T cells express the CD25 marker and the FoxP3 transcription factor
- They suppress activity of other T cells/immune cells, for maintenance and over activation prevention
- Tregs actively suppress autoimmune responses via autoreactive T cells targeting self-antigens
- They help resolve/regulate inflammation, preventing tissue damage
- Tregs promote tolerance by ignoring harmless substances
Cytokine Regulation
- Cytokine regulation involves signaling molecules mediating/regulating immune responses
- Various immune cells produce cytokines, including T cells, macrophages, and dendritic cells
- Coordinating the immune response is one of their purposes
- Appropriate immune responses to an infection/injury using cytokine regulation
- Prevents the immune system from excessive or inappropriate activation
- Pro-inflammatory cytokines include interleukin-1 (IL-1), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6) and promote inflammation and immune activation
- Cytokines recruit immune cells to infection sites and stimulate the immune system
- Anti-inflammatory cytokines prevent excessive inflammation/tissue damage, thus reduce the immune response.
- Example: Interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β) promote the resolution of inflammation and promote tissue repair. Cytokine Imbalance
Abnormal Immune Responses
- This is when an overreaction, underreaction, or misidentification of self tissues as foreign occurs
- Autoimmunity, hypersensitivity, immunodeficiency, and immunosuppression are the mechanisms
- A condition where the immune system mistakenly attacks its own tissues is autoimmunity
- It happens when the system fails to distinguish self from non-self
- Molecular Mimicry is when the immune system confuses normal body cells with foreign invaders or antigens resembling self-antigens
- Loss of Tolerance occurs if immune cells target own tissues must be eliminated, but they are not during T cell development
- A combination of genetics and environmental factors such as stress, chemicals and infections generally result in autoimmunity
Genetic Factors
- Having certain genes is associated with an increase in susceptibility to autoimmune diseases e.g. HLA related gene complex
- Environmental factors for consideration are infection, certain medications, and smoking, they can also trigger responses in predisposed individuals
- Hormonal factors are considered because most autoimmune diseases is more common in women, hormones play a role in development
Autoimmune Disease Examples
- Rheumatoid Arthritis is when joints become inflamed/painful, and may include painful, oedematous joints in patients feet, toes and foot deformities
- Psoriatic Arthritis is common with psoriasis, with symptoms like skin lesions, toe pain, & dactylitis.
- Systemic Lupus Erythematous (SLE) can cause skin rashes & joint pain, especially in the presence of ulcerations/infections
Hypersensitivity
- An immune system that is triggered by harmless substances with these four types:
- Type 1 (immediate): IgE- mediated allergies include anaphylaxis and/or asthma, chemical mediators cause allergy symptoms, and allergic tendencies stem from genetics
- - Type 2 (cytotoxic)*: Immune system attacks its own cells, such as haemolytic anaemia, IgG and IgM are involved,
- Damage to certain cells stems from antibody response Once an antibody binds to a cell, the NK cells, phagocytes target the problem at the tissues, also known as complement activation/cell-mediated cytotoxicity
- Type 3 (immune complex): Disposition of antigen-antibody complexes causing inflammation e.g. SLE the intended target is soluble and not membrane bound but antibody-mediated
- Type 4 (Delayed): T-cell mediated responses include contact dermatitis which manifest in hours/days
- Only hypersensitive response to be triggered by antigen-specific T cells
- CD8+ cytotoxic T cells and CD4+ helper T cells recognize those with either class I or class II MHC (HLA) molecules
Type 1 Hypersensitivity, Allergic Reaction
- The body experiences dust, pollen which signals B cells to produce IgE antibodies leading to mast cells and basophils in airways and connective tissues
- Mast cells release histamine which signal the symptoms of Anaphylaxis, a life threatening condition, Bee sting, or allergy.
- Generalized reaction- causes Generalized vasodilation causing major bp issues, and possibly Bronchoconstriction causing difficulty breathing
Type 2 hypersensitivity, Exam
- Transfusion reactions (truly foreign antigens)
- Graft rejection (truly foreign antigens)
- Goodpasture's syndrome (auto-antibodies to the glomerular basement membrane)
- Grave's disease (antibodies against the thyroid stimulating hormone receptor)
- The antibody mimics the effect of TSH causing the gland to produce excessive thyroxine
Type 3 Hypersensitivity Exam
- Deposition of antibody-antigen complexes are left in skin, kidney and joints Immune complex glomerulonephritis can be caused by group A/ẞ haemolytic streptococci after a infection in throat or skin, and may affect children
- Presents with blood in the urine, the need to urinate, oliguria, with protein in the urine, Treat with antibacterials and dialysis
- The Arthus reaction is local type III hypersensitivity and with Extrinsic Allergic Alveolitis-Inhaled antigen triggers IgG with Type III rxn in the lung
Type 4 Hypersensitivity
- T cell medicated responses take hours/days, and this reaction is the only type triggered by antigen-specific T cells CD8+ cytotoxic T cells and CD4+ helper T cells recognize the antigen when complexed both with class I or class II
- Macrophages function as antigen-presenting cells to helper T cells and help them produce a proliferation of Interluekin 1 and regulate the delayed sensitivity reactions
Type 4 Hypersensitivity Processes
- Activated cytotoxic T cells destroys cells that come into contact
- Natural killer cells, without immunization, or by antibody-dependent cellular cytotoxicity ADCC can kill other cells
- Activated macrophages are more phagocytic or destroy the pathogen Sufficient exposure to M.tuberculosis, for leprosy , where the macrophages clump and produce lymphocytes form granulomas Occurs when the immune system cant clear the antigen so it walled off When Immune production declines with age the patient may have renewal in microbacteria causing reactivation Contact dermatitis occurs when exposed to salts & metals so antigens react by binding normal body protein stimulate the immune system
Immunodeficiency
- This Happens when the immune system cant mount an effective response in infections.
- Can either be primary, as from a genetic disorders, or from a secondary/external reason, like HIV, chemo etc
- With Diabetes the patients may have issues with blood and wound healing and impaired neutrophil production.
- HIV can cause issues in reduced CD4 + T cells, which lead to fungal and bacterial infections
Immunosuppression
- Suppression reduces and reduces the responsiveness of the immune
- May be from transplanted organs Use Drugs that helps transplanted patients, auto immune drugs to cause immunosuppression, Corticosteroid and biologics are used to treat autoimmune
Immune System Cancers
- Blood and bone marrow cancer-Leukemia Characterized by replacement of Bone Marrow Cells, decreasing the amount of Red blood cells,WBC and also decreasing thrombocyopaenia
Lymphoma
- Cancer of the Lymph Tissues Hodgkins Diese accounts for about 48% of such lymphoma cases and also includes Reed Sternberg Cells and Non Hodkins spreads very easily and faster and involves Viral Aetiology. Lymphoma occurs if there are issues with humor, body and cellular response
Cancer
- Multiple myeloma which is plasma cell cancer causing and affecting several areas that are active marrow sites.
- Has been linked to Bence Jones (bence johns) proteins as symptoms of anemia and bone in-pairment
- HIV attacks weaken the immune system.
- Binding CD4 molecules help, the Virus attack and reduces T cells below microliters unit of measurement
- Also CD8 kills T and harms the Immune system
HIV immunity
- As CD4 drops to 400mcl immunity declines and the pathogen can damage tissue
- The patient will get Opportunistic infections
Studying That Suits You
Use AI to generate personalized quizzes and flashcards to suit your learning preferences.