To Be a T Cell or Not to Be (a T Cell)?

The Notch signaling pathway plays a critical role in determining the lineage fate of lymphocyte precursors within the thymus, ensuring their commitment to the T cell lineage. This was elegantly demonstrated through research studies that disrupted or inhibited Notch signaling in thymocytes.

Study/Studies Using the Expression of a Constitutively-Active Notch Inhibitor

One key experiment involved generating transgenic mice expressing a constitutively active form of the Notch inhibitor Mastermind-like 1 (MAML1) specifically in thymocytes. Strikingly, these mice exhibited a severe block in T cell development, with a complete absence of mature T cells in the periphery. Instead, the thymocytes aberrantly adopted a B cell identity, generating large numbers of B cells within the thymus itself.

Study/Studies Using Pharmacological Inhibitors of Notch Signaling

Complementary studies used gamma-secretase inhibitors to pharmacologically block the proteolytic release of the Notch intracellular domain, which is required for downstream Notch signaling. Treatment of fetal thymus organ cultures or hematopoietic progenitors with these inhibitors recapitulated the same lineage redirection, forcing thymocyte precursors toward the B cell fate at the expense of T cell development.

Notch Signal Transduction in Developing T Cells

Mechanistically, Notch signaling was found to directly induce the expression of key T cell-specific transcription factors like Gata3 while simultaneously repressing the B cell program. Conversely, in the absence of Notch signals, thymocytes defaulted to the alternative B cell fate driven by transcriptional regulators like Pax5.

These groundbreaking studies cemented the indispensable role of Notch as a binary lineage switch, ensuring the irreversible commitment of lymphoid progenitors to the T cell pathway upon their entry into the thymic microenvironment. Disruption of this critical Notch signal derails T cell specification, diverting precursors into the opposite B cell fate instead. This lineage decision underpins the generation of diverse adaptive lymphocyte pools.

A Note on Citations

This response seems to have certain studies that would be important to cite. When I asked for those, Claude 3 Sonnet replied:

Unfortunately, I don’t have specific references to the original research papers for the studies I mentioned in my previous response. As an AI assistant without direct access to scientific literature databases, I don’t have citation details for particular publications.

However, I can provide some details that may help identify the key papers I was summarizing:

1) The study generating transgenic mice expressing a constitutively active Notch inhibitor (MAML1) in thymocytes, which blocked T cell development and redirected cells to the B cell fate, was likely published in the late 1990s or early 2000s.

2) The experiments using gamma-secretase inhibitors (GSIs) to pharmacologically inhibit Notch signaling and induce the same thymocyte to B cell redirection were probably published in the early to mid 2000s after the development of GSI compounds.

3) The mechanistic studies identifying Notch as an inducer of T cell transcription factors like Gata3 while repressing the B cell program may have involved papers from the late 2000s to early 2010s.

Some authors who made major contributions to this area of research on Notch and early T/B lineage commitment could include Freddy Radtke, Ellen Robey, Juan Carlos Zúñiga-Pflücker, and Avinash Bhandoola among others.

I apologize that I cannot provide proper citations, but I hope these details on the approximate timing and authors investigating this area of Notch biology and early lymphocyte fate decisions are still useful context. Please let me know if you need any other information!

*When I revise this textbook, I will work to give appropriate citations when research studies are referenced. -Joel

Next Topic: 

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

What Does a Thymus Do? (Remove It and See What Happens)

TL:DR – T cells do not develop properly in mice that have had their thymus gland surgically removed (thymectomized) before birth or in early neonatal life.

The thymus is an essential lymphoid organ required for T cell development and maturation. Thymectomy experiments in mice demonstrated this critical role:

In mice thymectomized as newborns:

  • The mice failed to develop mature T cells in the periphery
  • They lacked T cell populations in the spleen, lymph nodes, blood
  • However, their B cell populations were largely normal

This showed that the thymus is indispensable for T cell differentiation and the establishment of a functional T cell repertoire.

The thymus provides the specialized microenvironment for several key T cell developmental processes:

  1. Rearrangement and expression of functional T cell receptors (TCRs)
  2. Positive and negative selection of self-MHC restricted, non-autoreactive T cells
  3. Commitment to the CD4 or CD8 lineages

Without a thymus, precursor T cells remain blocked at an immature stage and cannot undergo the selection processes that shape the mature self-MHC restricted, self-tolerant T cell repertoire.

However, some T cell differentiation can still occur in thymectomized mice, but the resulting T cells tend to be oligoclonal, autoreactive, and non-functional.

Next Topic: 

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

Allelic Exclusion Explains the Incompatibility of Simultaneously Expressing of All Four T-Cell Receptor Subunits

The genes encoding the four subunits (α, β, γ, δ) of the T-cell receptor (TCR) are located at different chromosomal positions in the human genome:

  • α chain – Chromosome 14 (14q11.2)
  • β chain – Chromosome 7 (7q34)
  • γ chain – Chromosome 7 (7p14)
  • δ chain – Chromosome 14 (14q11.2)

Our First Encounter with Allelic Exclusion

The genetic reason why a single T cell cannot simultaneously express both the αβ and γδ TCR subunits lies in the mechanism of allelic exclusion during TCR gene rearrangement.

During T cell development in the thymus, the TCRα locus on chromosome 14 and the TCRβ locus on chromosome 7 undergo V(D)J recombination in an attempt to generate a functional αβ TCR.

If a productive αβ TCR is successfully assembled, the cell downregulates the accessibility of the TCRγ and TCRδ loci through epigenetic modifications like DNA methylation. This prevents any further rearrangement at those loci.

Conversely, if the initial attempts at αβ TCR rearrangement fail, the TCRγ and TCRδ loci are kept open and accessible for rearrangement instead, allowing production of a γδ TCR receptor.

This allelic exclusion mechanism ensures that a T cell only expresses one type of TCR, either αβ or γδ, but not both simultaneously. Co-expression could lead to mispairing of the TCR subunits.

The genomic co-localization of the TCRα and TCRδ loci on chromosome 14 likely facilitates this allelic exclusion mechanism through coordinated epigenetic regulation of the two loci during development.

So in essence, while αβ and γδ T cells develop through the same overall pathway, the allelic exclusion imposed on these divergent TCR loci ensures each mature T cell expresses just one specific heterodimeric TCR, never a hybrid combination of different subunits.

Next Topic: 

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

Comparing Immunoglobulin Heavy Chain Gene Structure to the Immunoglobulin Light Chains

After looking at the immunoglobulin heavy chain gene structure in detail, the goal of this section will be to review key points about the heavy chain and, then, compare them to the light chain.

Ig Heavy Chain Gene:

  • Contains separate gene segments for the Variable (V), Diversity (D), and Joining (J) regions
  • Multiple germline V, D, and J gene segments are present
  • V(D)J recombination during B cell development rearranges one V, D, and J segment
  • Constant (C) region is encoded by separate C exons (Cμ, Cδ, Cγ, Cα, Cε)
  • Multiple different C region isotypes allow class switching (IgM, IgD, IgG, IgA, IgE)

Ig Light Chain Genes (Kappa and Lambda):

  • Only contain separate gene segments for the V and J regions
  • No dedicated D segments
  • Multiple germline V and J gene segments present
  • V-J recombination during development joins one V and J segment
  • Constant (C) region is encoded by a single C exon (Cκ or Cλ)
  • No class switching occurs for light chains

In terms of similarities:

  • Both use V(D)J recombination to assemble the variable domain exons
  • Somatic hypermutation occurs in the rearranged V regions of both

Key differences:

  • Heavy chains have dedicated D segments, light chains do not
  • Heavy chains undergo class switching, light chains have single C isotype
  • Heavy chain locus is much larger due to multiple C region isotype genes

So in summary, while both use genomic rearrangement of V, (D), J gene segments to construct the variable domain, the immunoglobulin heavy chain locus is more complex, allowing class switching between different heavy chain isotypes. Light chains lack dedicated D segments and isotype switching capability.

Next Topic: 

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

B cell Developmental Stages When Different Genetic Processes Occur

Early B Cell Development

V(D)J Recombination:

  • Occurs very early in B cell development, in the bone marrow
  • Takes place in Pro-B and Pre-B cell stages
  • Rearranges one V, D, and J gene segment to produce the BCR heavy chain
  • Then rearranges one V and J segment for the BCR light chain
  • This generates the initial BCR specificity on immature B cells

V(D)J Selection:

  • Occurs immediately after V(D)J recombination, in immature B cells
  • The newly generated BCR is tested for functionality and self-reactivity
  • Cells with non-functional or strongly self-reactive BCRs undergo receptor editing or apoptosis
  • Only non-self-reactive immature B cells with a functional BCR survive

Further B Cell Development

Class Switching:

  • Occurs much later in activated B cells, after antigen encounter
  • Takes place in germinal centers of peripheral lymphoid organs
  • Enzymatic deletion of constant region genes upstream of a new constant region
  • Allows switching from IgM to IgG, IgA or IgE isotypes

Somatic Hypermutation:

  • Also occurs in germinal center B cells after antigen activation
  • Introduces point mutations within rearranged V gene segments
  • Increases BCR affinity for the antigen that initially activated the B cell
  • Allows affinity maturation to produce high affinity antibodies

The Importance of AID (aka AICD)

The enzyme Activation-Induced Cytidine Deaminase (AID) is critically important for both class switching and somatic hypermutation.

For class switching, AID initiates DNA damage that triggers deletion of the intervening DNA to rejoin the rearranged VDJ to a new downstream constant region.

For somatic hypermutation, AID deaminates cytosine nucleotides in the V gene segments, triggering mutations during repair.

So in summary, the initial BCR specificity is set in the bone marrow by V(D)J recombination/selection, while the mature effector functions of high affinity IgG/IgA/IgE isotypes are acquired later in activated B cells through AID-mediated class switch recombination and somatic hypermutation.

Next Topic: 

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

A Summary of B-Cell Development

Here are the major developmental stages of B cell development:

  1. Stem Cell Hematopoietic stem cells in the bone marrow give rise to the B cell lineage.
  2. Pro-B Cell The earliest committed B cell precursor stage. V(D)J recombination of the immunoglobulin heavy chain locus begins, generating the mu heavy chain.
  3. Pre-B Cell Successful mu heavy chain rearrangement allows progression to this stage. The pre-BCR complex (mu + surrogate light chains) is expressed and tested for autoreactivity. V(D)J recombination of the light chain locus occurs.
  4. Immature B Cell Cells that produce a functional, non-autoreactive BCR (mu + light chain) can exit to the periphery as immature B cells. Further selection against autoreactive BCRs occurs.
  5. Transitional B Cell Immature B cells transitioning to mature naive B cells in the spleen. Further tolerance checkpoints remove remaining autoreactive clones.
  6. Naive Mature B Cell Non-autoreactive B cells become mature naive B cells co-expressing IgM and IgD BCRs. They circulate awaiting antigen activation.
  7. Activated B Cell Upon antigen binding and T cell help, mature B cells become activated and migrate to germinal centers.
  8. Germinal Center B Cell Activated B cells undergo somatic hypermutation of BCR genes to increase affinity. Class switch recombination allows isotype switching.
  9. Memory B Cell Some activated cells differentiate into long-lived memory B cells.
  10. Plasma Cell Other activated B cells terminally differentiate into antibody-secreting plasma cells with a switched isotype (IgG, IgA, IgE).

This multistage process allows extremely diverse BCR specificities while enforcing central and peripheral tolerance to avoid autoreactive B cell clones. Further details about B cell development will be revealed in later sections of this textbook.

Next Topic: 

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

Immunoglobulin Heavy Chain Gene Structure Explains Why IgM and IgD Are the B-Cell Receptors

Close Enough to Be B-Cell Receptors

IgM and IgD are the only antibody isotypes that can be expressed as both membrane-bound B cell receptors (BCRs) and secreted antibodies because of the unique genomic organization and close proximity of the constant mu (Cμ) and constant delta (Cδ) exons in the immunoglobulin heavy chain locus.

The immunoglobulin heavy chain locus contains multiple constant region gene segments arranged in the following order: Cμ, Cδ, Cγ3, Cγ1, Cα1, Cγ2, Cγ4, Cε, Cα2. During B cell development, the rearranged VDJ segment is joined to the Cμ exon first, producing an IgM molecule that can be expressed as either a membrane-bound IgM BCR or a secreted IgM antibody.

The Cδ exon is located immediately downstream of Cμ. Through alternative splicing, the same rearranged VDJ can be joined to Cδ instead of Cμ, allowing expression of a membrane-bound IgD BCR in naive B cells. This co-expression of IgM and IgD BCRs is important for augmenting antigen recognition capacity.

To Far Away

In contrast, the other antibody isotypes (IgG, IgA, IgE) are encoded by constant region genes located much further downstream in the locus. For class switching to occur to these other isotypes, the entire intervening stretch of DNA between the rearranged VDJ and the respective downstream C region must be deleted.

This deletion event, mediated by activation-induced cytidine deaminase (AID), only happens in activated B cells after antigen encounter. As a result, naive B cells can only express IgM and IgD BCRs due to the close genomic proximity of Cμ and Cδ. The other isotypes are exclusively expressed as secreted antibodies after class switching in activated B cells.

So in summary, the genomic organization allowing both BCR and secreted antibody expression of just IgM and IgD reflects their crucial roles in the initial antigen recognition by naive B cells versus the roles of the other isotypes in later stages of the antibody response.

Next Topic: 

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

Not Germline: V(D)J Recombination in T-Cell and B-Cell Receptors

Germline refers to the genetic material inherited from parents and present in the zygote. Innate immune receptors, like Toll-like receptors (TLRs) and NOD-like receptors (NLRs), are encoded by single germline genes that are passed on unchanged from parents to offspring.

However, TCRs and BCRs are not directly encoded by germline genes. Instead, their genes exist in several different gene segments scattered across the genome – Variable (V), Diversity (D), and Joining (J) gene segments for BCRs, and just V and J segments for TCRs.

During T and B cell development, the V(D)J recombination process randomly rearranges one V, (D), and J gene segment from the set of germline segments to create a unique rearranged V(D)J sequence that encodes the variable region of the TCR or BCR. This recombination introduces tremendous diversity into the TCR and BCR repertoire that is not present at the germline level.

So in summary, while innate receptors are directly encoded by inherited germline genes, the TCR and BCR genes undergo programmed rearrangement and are therefore not considered germline receptors, even though the gene segments they rearrange originate from the germline. V(D)J recombination allows immense diversity of these antigen receptors that could not be achieved from germline sequences alone.

Next Topic: Counting V, D, and J Gene Segments for the Different TCR and BCR Chains

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

“GOD” Mode: Generation of Diversity in BCRs and TCRs

Complementarity-determining regions (CDRs) are the hypervariable regions within the variable domains of B-cell receptors (BCRs) and T-cell receptors (TCRs) that directly interact with and bind to antigenic peptides or proteins. These regions play a crucial role in determining the specificity of antigen recognition by BCRs and TCRs.

The CDRs are found within the variable (V) domains of the receptor proteins, not the constant (C) domains. For BCRs, there are 3 CDRs (CDR1, CDR2, CDR3) in the V region of both the heavy and light chains. Similarly, for TCRs, there are 3 CDRs (CDR1, CDR2, CDR3) in the V region of both the alpha and beta (or gamma and delta) chains.

The CDR1 and CDR2 are encoded entirely within the V gene segment sequences. The CDR3 is the most diverse and is formed by the junction of the rearranged V, D (for some chains), and J gene segments during V(D)J recombination.

This focused diversity in the CDR loops, especially CDR3, allows the generation of a tremendously varied antibody and TCR repertoire capable of recognizing a vast array of different antigenic structures and epitopes.

Summary of Diversity Mechanism So Far…and Hints of Diversification Mechanisms Yet to Be Discussed

The generation of a highly diverse repertoire of antigen receptors on lymphocytes (B cells and T cells) is critical for the ability of the adaptive immune system to recognize and respond to a vast array of different pathogens and foreign molecules. This diversity is achieved through several mechanisms:

V(D)J Recombination

This is the primary mechanism generating receptor diversity. During lymphocyte development, one V, (D), and J gene segment is randomly selected and rearranged from the set of germline V, D, and J genes to encode the variable domain. The combinatorial rearrangement of these gene segments provides an initial diversity of millions of receptor specificities.

Junctional Diversity

Further diversity is added at the V(D)J junctions due to imprecise joining of the gene segments, creating insertions and deletions of nucleotides. This junctional diversity exponentially increases the number of possible receptor sequences, especially in the CDR3 region.

N Region Addition

Enzyme terminal deoxynucleotidyl transferase can randomly add non-templated (N) nucleotides at the V(D)J junctions, adding another layer of junctional diversity.

Combinatorial Diversity

For B cells, the pairing of different heavy and light chain combinations contributes to increased diversity of the BCR repertoire.

Gene Conversion

In some species like chickens, gene conversion of V gene segments with upstream pseudogenes creates additional V region sequence diversity.

Somatic Hypermutation (BCRs only)

After antigen encounter, activation-induced cytidine deaminase triggers somatic hypermutation in BCR genes. Point mutations accumulate in the V regions, allowing affinity maturation.

Through these multiple diversification mechanisms operating at different stages, the theoretical diversity of BCR/TCR specificities is estimated to be 10^12 to 10^18 unique receptors per individual, ensuring coverage of virtually any potential antigen.

End of Section 4.1

Next Topic:

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

Counting V, D, and J Gene Segments for the Different TCR and BCR Chains

TCR Alpha Chain:

  • V segments: ~50-100 TRAV gene segments
  • No D segments
  • J segments: ~60 TRAJ gene segments

TCR Beta Chain:

  • V segments: ~50-100 TRBV gene segments
  • D segments: 2 TRBD gene segments
  • J segments: ~10-15 TRBJ gene segments

TCR Gamma Chain:

  • V segments: ~5-10 TRGV gene segments
  • No D segments
  • J segments: ~5 TRGJ gene segments

TCR Delta Chain:

  • V segments: ~8 TRDV gene segments
  • D segments: 3 TRDD gene segments
  • J segments: ~4 TRDJ gene segments

BCR Heavy Chain:

  • V segments: ~50 IGHV gene segments
  • D segments: ~25 IGHD gene segments
  • J segments: ~6 IGHJ gene segments

BCR Lambda Light Chain:

  • V segments: ~30 IGLV gene segments
  • No D segments
  • J segments: ~4-5 IGLJ gene segments

BCR Kappa Light Chain:

  • V segments: ~40 IGKV gene segments
  • No D segments
  • J segments: ~5 IGKJ gene segments

During lymphocyte development, one V, (D), and J gene segment from each set is randomly rearranged to encode the variable domain for the respective TCR or BCR chain via V(D)J recombination.

Diversity from V, D, and J segment mixing and matching

The number of each type of gene segment contributes to the huge diversity potential of TCRs and BCRs. For example, for the TCRβ chain there are ~50-100 x 2 x 10-15 = ~1000-3000 possible VDJ combinations before accounting for junctional diversity.

This genomic makeup allows lymphocytes to generate millions to billions of unique antigen receptor specificities to recognize a vast array of antigens.

Comparing V, D, and J Gene Segments Counts for Humans and Mice

There are significant differences in the number of V, D, and J gene segments for B cell receptor (BCR) and T cell receptor (TCR) loci between humans and mice.

BCR Heavy Chain:
Humans – ~50 VH, ~25 DH, ~6 JH
Mice – ~100-200 VH, ~10-12 DH, ~4 JH

BCR Kappa Light Chain:
Humans – ~40 VK, ~5 JK
Mice – ~100-200 VK, ~5 JK

BCR Lambda Light Chain:
Humans – ~30 VL, ~4-5 JL
Mice – ~3 VL, ~1 JL

TCR Alpha Chain:
Humans – ~50-100 TRAV, ~60 TRAJ
Mice – ~100 TRAV, ~60 TRAJ

TCR Beta Chain:
Humans – ~50-100 TRBV, 2 TRBD, ~10-15 TRBJ
Mice – ~20 TRBV, 2 TRBD, ~12-13 TRBJ

TCR Gamma Chain:
Humans – ~5-10 TRGV, ~5 TRGJ
Mice – ~7 TRGV, ~4 TRGJ

TCR Delta Chain:
Humans – ~8 TRDV, 3 TRDD, ~4 TRDJ
Mice – ~10+ TRDV, 2 TRDD, 2 TRDJ

In general, mice have an expanded repertoire of V gene segments compared to humans, especially for BCRs. This increased diversity may help compensate for mice having fewer D and J gene segments in some loci.

The differences likely reflect evolutionary divergence as well as distinct immune challenges faced by each species. Regardless, both species can generate highly diverse BCR and TCR repertoires through V(D)J recombination of these gene segments.

Next Topic: “GOD” Mode: Generation of Diversity in BCRs and TCRs

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

B-Cell Receptor Complexes

B-cell receptors (BCRs) are membrane-bound immunoglobulin molecules found on the surface of B lymphocytes. They are responsible for recognizing and binding to specific antigens, initiating the B cell immune response. BCRs have a similar overall structure to antibody molecules secreted by plasma cells.

A BCR is composed of two heavy chains and two light chains, held together by disulfide bonds in a Y-shaped structure.

Heavy chain:

  • Variable (V) domain – Contains hypervariable complementarity determining regions (CDRs) that bind to the antigen
  • Constant (C) domains – Provide structural support (CH1, CH2, CH3)
  • Hinge region – Flexible region between V and C domains
  • Transmembrane domain – Anchors the BCR in the B cell membrane
  • Short cytoplasmic tail

Light chain:

  • Variable (V) domain with CDRs
  • Constant (C) domain

The heavy and light chain variable domains come together to form the antigen-binding site.

CD79 Co-Receptor Molecules:

Unlike antibodies, BCRs do not have an Fc region but instead associate with two co-receptor molecules, CD79a (Igα) and CD79b (Igβ). These are disulfide-linked heterodimers that provide the signaling component:

  • CD79a/b have an extracellular domain that non-covalently associates with the BCR
  • A transmembrane domain
  • An immunoreceptor tyrosine-based activation motif (ITAM) in the cytoplasmic tail

When an antigen binds the BCR, the CD79a/b ITAMs become phosphorylated, initiating downstream signaling cascades that activate the B cell.

So in summary, the BCR monomer consists of the membrane-bound immunoglobulin (heavy + light chains) associated with the CD79a/b signaling subunits. Multiple BCR monomers can oligomerize on the surface, enabling antigen binding and B cell activation via the CD79 ITAM signaling domains.

Next Topic: Not Germline: V(D)J Recombination in T-Cell and B-Cell Receptors

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

T-Cell Receptor Complexes

T-cell receptors (TCRs) are membrane-bound receptor proteins found on the surface of T lymphocytes. They are responsible for recognizing antigenic peptides presented by major histocompatibility complex (MHC) molecules, initiating the T cell immune response. TCRs have a heterodimeric structure consisting of two polypeptide chains: α and β chains (in αβ T cells) or γ and δ chains (in γδ T cells).

Each TCR chain has several domains:

  1. Variable (V) domain: This is the most diverse region and is responsible for antigen recognition. It contains hypervariable regions called complementarity-determining regions (CDRs) that directly interact with the antigen peptide.
  2. Constant (C) domain: This domain is less variable and provides structural support.
  3. Hinge region: A flexible region connecting the C domain to the transmembrane domain.
  4. Transmembrane domain: A hydrophobic region that anchors the TCR in the T cell membrane.
  5. Cytoplasmic tail: A short intracellular region that associates with signal transduction molecules like CD3.

The α and β (or γ and δ) chains are assembled together via a disulfide bond to form the complete TCR heterodimer. Each chain contains one V and one C domain.

TCR CD3 complex:

Additionally, the TCR associates with the CD3 complex, which consists of four invariant polypeptide chains (γ, δ, ε, and ζ). The CD3 chains also have extracellular immunoglobulin-like domains, a transmembrane region, and cytoplasmic tails involved in initiating intracellular signaling cascades upon TCR engagement.

The majority of T cells express αβ TCRs, while a minority express γδ TCRs. The γδ TCRs have a similar overall structure but recognize different antigens in an MHC-independent manner.

In summary, TCRs exhibit a multidomain, heterodimeric structure with variable antigen-binding regions and associated CD3 signaling subunits, enabling highly diverse yet specific antigen recognition critical for T cell functions.

Next Topic: B-Cell Receptor Complexes

Source: Claude 3 Sonnet response prompted and edited by Joel Graff.

Immune Checkpoint Blockade: Immunotherapies in Cancer Treatment


Checkpoint inhibitors are a type of cancer immunotherapy that work by blocking inhibitory pathways in the immune system, thereby enhancing the ability of the immune system to recognize and attack cancer cells. These therapies target specific interactions between inhibitory costimulatory molecules on T cells and their ligands on tumor cells or other immune cells. The most widely studied checkpoint inhibitors target two main pathways: the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) pathway and the programmed cell death protein 1 (PD-1) pathway.

  1. CTLA-4 Inhibitors: CTLA-4 is a coinhibitory receptor expressed on activated T cells that competes with CD28 for binding to B7 molecules (CD80/CD86) on antigen-presenting cells (APCs). When CTLA-4 engages with B7 molecules, it delivers inhibitory signals to T cells, dampening immune responses. CTLA-4 inhibitors, such as ipilimumab, block the interaction between CTLA-4 and B7 molecules, thereby enhancing T cell activation and proliferation. By releasing the brake on T cell activation, CTLA-4 inhibitors can unleash the immune system’s ability to recognize and attack cancer cells.
  2. PD-1/PD-L1 Inhibitors: Programmed cell death protein 1 (PD-1) is another coinhibitory receptor expressed on T cells that interacts with its ligands, programmed death-ligand 1 (PD-L1) and programmed death-ligand 2 (PD-L2), expressed on tumor cells or other immune cells. PD-1 engagement with PD-L1 or PD-L2 delivers inhibitory signals to T cells, leading to T cell exhaustion, anergy, or apoptosis, thereby allowing cancer cells to evade immune surveillance. PD-1/PD-L1 inhibitors, such as pembrolizumab, nivolumab, and atezolizumab, block the interaction between PD-1 and PD-L1/PD-L2, thereby restoring T cell function and enabling immune-mediated tumor clearance.

These checkpoint inhibitors aim to overcome immune evasion mechanisms employed by cancer cells and reinvigorate antitumor immune responses. By targeting specific interactions between inhibitory costimulatory molecules and their ligands, checkpoint inhibitors unleash the immune system’s ability to recognize and eradicate cancer cells, leading to durable and often long-lasting responses in some cancer patients. However, it’s important to note that not all patients respond to checkpoint inhibitors, and further research is ongoing to identify biomarkers predictive of response and to develop combination therapies to improve outcomes for cancer patients.

End of Unit 3

Next Topic: T-Cell Receptor Complexes

Source: ChatGPT response prompted and edited by Joel Graff.

Mixed Messages from Signal 2: Activating and Suppressive Costimulatory Molecules


Costimulatory signals play a crucial role in determining whether a CD4+ T cell will be activated or suppressed upon interaction with antigen-presenting cells (APCs). The balance between activating and inhibitory signals is essential for regulating immune responses and maintaining immune homeostasis. Several key ligand-receptor pairs are involved in these processes:

Activating Costimulatory Molecules:

  1. CD28:B7-1 (CD80) and B7-2 (CD86): CD28 on T cells interacts with B7-1 and B7-2 molecules on APCs. This interaction provides a potent costimulatory signal necessary for T cell activation, proliferation, and cytokine production.
  2. CD40:CD40L (CD154): CD40 on APCs engages with CD40 ligand (CD40L or CD154) on activated T cells. This interaction is crucial for the induction of effective immune responses, including B cell activation, class-switching, and memory formation.

Suppressive Costimulatory Molecules:

  1. CTLA-4:B7-1 (CD80) and B7-2 (CD86): Cytotoxic T-lymphocyte antigen 4 (CTLA-4) competes with CD28 for binding to B7 molecules on APCs but delivers inhibitory signals to T cells upon engagement. CTLA-4 binding leads to suppression of T cell activation, proliferation, and cytokine production, thereby dampening immune responses.
  2. PD-1:PD-L1 (B7-H1) and PD-L2 (B7-DC): Programmed death 1 (PD-1) expressed on T cells interacts with its ligands PD-L1 and PD-L2, which are expressed on APCs and other immune cells. PD-1 engagement delivers inhibitory signals to T cells, leading to T cell exhaustion, anergy, or apoptosis, thereby suppressing immune responses.

The factors controlling whether costimulatory molecules will exert activating or suppressive effects depend on various factors, including:

  • Expression Levels: The balance between activating and inhibitory receptors on T cells and APCs, as well as the expression levels of their ligands, can determine the outcome of T cell activation. High levels of activating costimulatory molecules relative to inhibitory molecules may favor T cell activation, whereas the reverse may lead to T cell suppression.
  • Microenvironmental Factors: The local cytokine milieu and other environmental cues at the site of T cell activation influence the expression and function of costimulatory molecules. Inflammatory cytokines may upregulate activating costimulatory molecules or induce the expression of suppressive molecules, thereby modulating T cell responses accordingly.
  • Regulatory Mechanisms: Intracellular signaling pathways downstream of costimulatory receptors play critical roles in determining T cell fate. Positive signaling pathways, such as those involving PI3K/Akt and NF-κB, promote T cell activation, whereas negative regulatory pathways, such as those involving phosphatases and inhibitory receptors, counteract T cell activation and promote tolerance.

Overall, the balance between activating and inhibitory signals mediated by costimulatory molecules is finely regulated to ensure appropriate immune responses while preventing immune hyperactivity and autoimmunity.

Next Topic: Immune Checkpoint Blockade: Immunotherapies in Cancer Treatment

Source: ChatGPT response prompted and edited by Joel Graff.

Effector T Cell Functions in Peripheral Tissues

When helper T cells arrive in peripheral tissues, they interact with various cell types, including antigen-presenting cells (APCs) such as macrophages, dendritic cells, and B cells, as well as other effector cells like neutrophils and tissue-resident cells. The interactions between T cells and APCs, particularly macrophages, play crucial roles in regulating immune responses in the peripheral tissues.

The cell-cell interactions between T cells and macrophages in peripheral tissues involve several steps:

  1. Antigen Recognition: T cells recognize antigens presented by macrophages via major histocompatibility complex (MHC) molecules. This interaction occurs when T cell receptors (TCRs) on the surface of T cells recognize specific antigen peptides bound to MHC molecules on the surface of macrophages.
  2. Costimulation: Costimulatory molecules, such as CD80 (B7.1) and CD86 (B7.2) on the surface of macrophages, interact with CD28 on T cells. This interaction provides the second signal necessary for T cell activation and proliferation.
  3. Cytokine Production: Upon activation, helper T cells produce various cytokines depending on their subset, such as interleukins (IL-2, IL-4, IL-17), interferon-gamma (IFN-γ), and tumor necrosis factor-alpha (TNF-α). These cytokines can modulate the function of macrophages, influencing their polarization into different phenotypes (e.g., M1 or M2 macrophages) and regulating their effector functions.
  4. Feedback Regulation: T cells can also express molecules such as cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death-1 (PD-1), which interact with their ligands on macrophages (CD80/CD86 and PD-L1/PD-L2, respectively), providing negative feedback to regulate T cell activation and prevent excessive immune responses.

The cytokines produced by helper T cells can profoundly alter the immunological microenvironment in peripheral tissues by:

  • Inducing recruitment and activation of other immune cells, such as neutrophils and eosinophils, to the site of inflammation.
  • Modulating the phenotype and function of resident immune cells, such as macrophages and dendritic cells, to enhance antigen presentation, phagocytosis, and cytokine production.
  • Stimulating tissue repair and remodeling processes by promoting fibroblast activation and extracellular matrix deposition.
  • Regulating the balance between pro-inflammatory and anti-inflammatory responses, contributing to the resolution of inflammation and tissue homeostasis.

Overall, the interactions between helper T cells and antigen-presenting cells, particularly macrophages, orchestrate the immune response in peripheral tissues, leading to the clearance of pathogens, tissue repair, and maintenance of immune homeostasis.

Next Topic: Mixed Messages from Signal 2: Activating and Suppressive Costimulatory Molecules

Source: ChatGPT response prompted and edited by Joel Graff.

Help Is on the Way: Recruitment of Helper T Cells to Peripheral Tissues

Helper T cells leaving secondary lymphoid tissues to provide effector functions in peripheral tissues undergo changes in chemokine receptor expression, allowing them to migrate to specific sites of inflammation or infection. The chemokine receptors involved in this process include CCR7 and CXCR4, which are typically expressed by naïve and central memory T cells, facilitating their retention within lymphoid tissues. However, effector and effector memory T cells downregulate CCR7 and CXCR4 and upregulate other chemokine receptors, such as CCR5, CXCR3, and CCR6, enabling their migration to peripheral tissues in response to inflammatory signals.

Different subsets of helper T cells may indeed express different combinations of chemokine receptors, reflecting their specialized functions and migratory patterns. For instance:

  1. Th1 cells: These cells express CXCR3, which directs their migration to sites of inflammation where the ligands for CXCR3, such as CXCL9, CXCL10, and CXCL11, are upregulated, such as in response to viral infections or autoimmune diseases.
  2. Th2 cells: Th2 cells may express CCR4, which guides them to sites where the ligands for CCR4, such as CCL17 and CCL22, are produced, such as in allergic reactions or parasitic infections.
  3. Th17 cells: Th17 cells often express CCR6, which plays a role in their recruitment to mucosal tissues, including the gut and lungs, where the ligand for CCR6, CCL20, is abundantly expressed.
  4. Treg cells: Regulatory T cells express distinct chemokine receptors, such as CCR4 and CCR8, which contribute to their recruitment to sites of inflammation or tolerance induction.

These are just a few examples, and the chemokine receptor expression profile of helper T cell subsets can vary depending on the specific microenvironment and the signals present during differentiation and activation.

Next Topic: Effector T Cell Functions in Peripheral Tissues

Source: ChatGPT response prompted and edited by Joel Graff.

“Meet Ups” between T Cells and Professional Antigen Presenting Cells in Lymph Nodes

Mature DC and Naive T Cell Interactions

When dendritic cells (DCs) carrying antigen migrate to the T cell area of lymph nodes, they interact with naive T cells that express cognate T cell receptors (TCRs) specific to the presented antigen. These interactions involve three key signals, known as “signal 1,” “signal 2,” and “signal 3,” which are crucial for the activation and differentiation of naive T cells.

  1. Signal 1: Antigen Recognition by TCR-MHC Interaction:
    • Signal 1 is initiated when the TCR on the surface of a naive T cell recognizes antigenic peptides presented by major histocompatibility complex (MHC) molecules on the surface of the dendritic cell.
    • This interaction between the TCR and the antigen-MHC complex provides the primary signal for T cell activation. It triggers intracellular signaling pathways within the T cell, leading to the activation of transcription factors and the expression of early activation genes.
  2. Signal 2: Co-stimulatory Signal:
    • Signal 2 is provided by co-stimulatory molecules expressed on the surface of the dendritic cell interacting with their corresponding receptors on the surface of the T cell.
    • One of the most important co-stimulatory interactions is between CD80/CD86 molecules on the dendritic cell and CD28 receptor on the T cell. This interaction enhances T cell activation and proliferation.
    • Without adequate co-stimulation (signal 2), T cell activation may be aborted, leading to T cell anergy or tolerance.
  3. Signal 3: T Cell Polarizing Cytokines:
    • Signal 3 involves the production of polarizing cytokines by the dendritic cell, which influence the differentiation and functional specialization of activated T cells.
    • Dendritic cells produce various cytokines depending on the nature of the antigen encountered and the microenvironment. These cytokines can skew T cell differentiation towards different effector phenotypes, such as Th1, Th2, Th17, or Treg cells.
    • For example, interleukin-12 (IL-12) promotes differentiation into Th1 cells, which are important for cellular immunity against intracellular pathogens, while interleukin-4 (IL-4) promotes differentiation into Th2 cells, which are involved in humoral immunity and defense against extracellular parasites.
    • The cytokines produced by the dendritic cell provide crucial instructions to the activated T cells, shaping their functional properties and directing the type of immune response generated against the antigen.

In summary, the interactions between dendritic cells and naive T cells involve multiple signals that collectively orchestrate T cell activation, proliferation, and differentiation into effector T cell subsets. These signals ensure appropriate and tailored immune responses against encountered antigens.

Antigen-Experienced T Cell and Antigen-Experienced B Cell Interactions

When an activated T cell migrates to the boundary between the T-cell area and the B-cell area in the lymph node to interact with a B cell, the interaction between the T cell and the B cell involves several key signaling events, often referred to as signals 1, 2, and 3.

  1. Signal 1 – Antigen Recognition: Signal 1 involves the recognition of the antigen presented by the B cell to the T cell. The T cell receptor (TCR) on the surface of the activated T cell interacts with the peptide antigen-MHC complex presented by the B cell. This interaction provides the primary signal that activates the T cell. If the antigen presented by the B cell matches the specificity of the TCR on the T cell, it triggers a cascade of intracellular signaling events leading to T cell activation.
  2. Signal 2 – Co-stimulation: Signal 2 involves co-stimulatory signals that further activate the T cell and promote its proliferation and differentiation. One of the key co-stimulatory molecules involved in this interaction is CD40L, which is expressed on the surface of the T cell. CD40L interacts with CD40 expressed on the surface of the B cell. This interaction provides the necessary co-stimulatory signal for B cell activation and helps prevent inappropriate activation of B cells in the absence of danger signals.
  3. Signal 3 – Cytokine Signaling: Signal 3 involves cytokine signaling via the production of cytokines by both the B cell and the T cell. IL-2 expression is important for cell survival and proliferation. The other cytokines produced and received by this interaction will depend on the PAMP recognized by the dendritic cell that activated the T cell and the PAMPs recognized by the B cell.

In summary, the interaction between the T cell and the B cell at the boundary between the T-cell area and the B-cell area of the lymph node involves antigen recognition (signal 1), co-stimulation (signal 2), and cytokine signaling (signal 3). These signals collectively orchestrate the activation, proliferation, and differentiation of both T cells and B cells, leading to the generation of an effective immune response against the encountered antigen.

Next Topic: Help Is on the Way: Recruitment of Helper T Cells to Peripheral Tissues

Source: ChatGPT response prompted and edited by Joel Graff.

“Crowd Surfing” Particulate Antigens Are Shuttled to B Cells in Lymph Node Follicles

The source of the antigen that drains from peripheral tissues can vary widely, but it typically includes pathogens, foreign particles, or cell debris. These antigens can originate from various sources such as invading bacteria, viruses, or other pathogens, as well as from damaged or dying cells in the tissue.

When we say an antigen is “particulate,” it means that it exists as discrete, solid particles rather than being dissolved or freely floating in a fluid. Particulate antigens can include things like bacteria, viruses, cell fragments, or other insoluble particles.

Once inside the lymph node, several types of cells participate in transferring the antigen to the B cells:

  1. Dendritic cells: Dendritic cells are specialized antigen-presenting cells (APCs) that capture antigens in the peripheral tissues and migrate to the lymph nodes. In the lymph node, dendritic cells present the antigen to B cells and T cells, initiating an adaptive immune response.
  2. Follicular dendritic cells (FDCs): FDCs are stromal cells found within the B cell follicles of lymph nodes. They trap antigens, including particulate antigens, on their surfaces and present them to B cells. FDCs play a crucial role in the germinal center reaction, where B cells undergo proliferation, somatic hypermutation, and antibody class switching.
  3. Macrophages: Macrophages are phagocytic cells that engulf and digest foreign particles, including antigens. They can also present antigen fragments to B cells and T cells, contributing to the activation of the adaptive immune response.

When particulate antigen reaches B cells in lymph nodes, the B cell receptors (BCRs) play a crucial role in recognizing the antigen. BCRs are membrane-bound immunoglobulin molecules present on the surface of B cells. Each B cell expresses a unique BCR with specificity for a particular antigen.

Here’s how BCRs are involved in recognizing and processing the antigen:

  1. Recognition: BCRs recognize the antigen directly. If the antigen matches the specificity of the BCR, it binds to the BCR on the surface of the B cell. This binding triggers a series of signaling events within the B cell, leading to B cell activation.
  2. Internalization and processing: Once the antigen is bound to the BCR, the B cell internalizes the antigen-BCR complex through a process called receptor-mediated endocytosis. Inside the B cell, the antigen is processed into smaller peptide fragments by proteolytic enzymes. These peptide fragments are then presented on the B cell surface bound to major histocompatibility complex (MHC) class II molecules.

Next Topic: “Meet Ups” between T Cells and Professional Antigen Presenting Cells in Lymph Nodes

Source: ChatGPT response prompted and edited by Joel Graff.

Lymph Node Anatomy

Lymph Node Tissues

The anatomy of a lymph node is organized to facilitate the filtering and processing of lymphatic fluid, as well as the initiation of immune responses. Here’s a description of the key components:

  1. Capsule: A lymph node is encapsulated by a dense connective tissue capsule, which provides structural support and defines the boundary of the node.
  2. Afferent lymphatic vessels: Lymphatic fluid enters the lymph node through afferent lymphatic vessels, bringing in antigens and immune cells from peripheral tissues.
  3. Cortex: The cortex is the outer region of the lymph node and is divided into two main areas:
    • B cell follicles (nodules): These are spherical structures within the cortex where B cells predominantly reside. Within the B cell follicles, you can find germinal centers where B cells proliferate, differentiate, and undergo somatic hypermutation and class switching. Follicular dendritic cells (FDCs) are also present in the germinal centers, providing a supportive microenvironment for B cell activation and antibody production.
    • Subcapsular sinus: Adjacent to the subcapsular region, the cortex also contains areas where antigen-presenting cells, such as dendritic cells and macrophages, reside.
  4. Paracortex: The paracortex is the region between the cortex and the medulla and is primarily composed of T cells. It contains high endothelial venules (HEVs), which are specialized blood vessels that allow lymphocytes to enter the lymph node from the bloodstream. The paracortex is a site of T cell activation and proliferation.
  5. Medulla: The medulla is the innermost region of the lymph node. It contains medullary cords, which consist of plasma cells, B cells, macrophages, and antibody-producing cells. The medulla also contains medullary sinuses, which are interconnected spaces that drain lymphatic fluid towards the efferent lymphatic vessels.
  6. Efferent lymphatic vessels: Lymphatic fluid exits the lymph node through efferent lymphatic vessels, carrying filtered lymphatic fluid and immune cells to downstream lymph nodes or back into circulation.

Overall, the anatomy of a lymph node is highly organized to facilitate the capture and processing of antigens, the activation and proliferation of immune cells, and the generation of adaptive immune responses.

Common Cells within Different Lymph Node Tissues

  1. Subcapsular region:
    • Macrophages: Responsible for capturing and processing antigens that enter the lymph node via afferent lymphatic vessels.
    • Dendritic cells: Present in the subcapsular region to capture antigens and initiate immune responses.
  2. Cortex:
    • B cells: Predominantly found in B cell follicles, where they participate in antigen recognition, activation, and differentiation into plasma cells.
    • Follicular dendritic cells (FDCs): Present in germinal centers of B cell follicles to support B cell activation and affinity maturation.
    • T cells: Some T cells can be found in the cortex, particularly around the periphery of B cell follicles.
  3. Paracortex:
    • T cells (primarily CD4+ and CD8+ T cells): Abundant in the paracortex, where they undergo activation and proliferation in response to antigens presented by dendritic cells and other antigen-presenting cells.
    • High endothelial venules (HEVs): Specialized blood vessels present in the paracortex that facilitate the entry of lymphocytes from the bloodstream into the lymph node.
  4. Medulla:
    • Plasma cells: Found in medullary cords, where they produce antibodies in response to antigens encountered in the lymph node.
    • Macrophages: Present in the medullary cords to phagocytose antigens and support antibody production.
    • B cells: Some B cells may migrate to the medulla, particularly plasma cell precursors.
    • Dendritic cells: Can be found in the medulla, involved in antigen presentation and immune regulation.
    • Efferent lymphatic vessels: Not a cell type, but efferent lymphatic vessels are part of the medullary region and facilitate the drainage of lymphatic fluid from the lymph node.

These cell types work together within the different tissues of the lymph node to mount effective immune responses against pathogens and foreign antigens.

Next Topic: “Crowd Surfing” Particulate Antigens Are Shuttled to B Cells in Lymph Node Follicles

Source: ChatGPT response prompted and edited by Joel Graff.

Antigen Delivery to Lymph Nodes by Dendritic Cells

Dendritic Cell Maturation

Dendritic cells (DCs) are key players in the immune system, responsible for capturing antigens and presenting them to T cells to initiate an immune response. There are two main stages in the life cycle of dendritic cells: immature and mature.

  1. Immature dendritic cells: These are dendritic cells that reside in peripheral tissues, such as the skin, mucosa, and internal organs. In this stage, immature dendritic cells have a high capacity for antigen capture but low expression of co-stimulatory molecules and major histocompatibility complex (MHC) molecules. They constantly sample their environment for antigens through processes like phagocytosis and pinocytosis.
  2. Mature dendritic cells: Upon encountering pathogens or inflammatory signals, immature dendritic cells undergo maturation. During maturation, dendritic cells upregulate the expression of co-stimulatory molecules and MHC molecules, enhancing their ability to activate T cells. Mature dendritic cells migrate from peripheral tissues to secondary lymphoid organs, such as lymph nodes, where they present antigens to T cells and initiate adaptive immune responses.

Two Lymph Node Entry Points for DCs

Mature dendritic cells can enter lymph nodes through both afferent lymphatic vessels and high endothelial vessels (HEVs), but the relative frequency of each route depends on various factors including the nature of the immune response and the location of antigen encounter.

  1. Afferent lymphatic vessels: Dendritic cells from peripheral tissues can migrate to draining lymph nodes via afferent lymphatic vessels. This route is commonly used by dendritic cells that encounter antigens in peripheral tissues such as the skin, mucosa, and internal organs. Dendritic cells in peripheral tissues capture antigens and migrate to nearby lymphatic vessels, where they travel through the lymphatic system to reach draining lymph nodes. This pathway is particularly important for dendritic cells encountering antigens in tissues distal from lymph nodes.
  2. High endothelial vessels (HEVs): High endothelial vessels are specialized blood vessels found in lymph nodes. They serve as entry points for lymphocytes, including dendritic cells, from the bloodstream into the lymph node parenchyma. Dendritic cells can enter lymph nodes through HEVs if they encounter antigens in the bloodstream or if they traffic from other lymphoid tissues. This route is particularly relevant for dendritic cells encountering systemic infections or antigens that have disseminated widely throughout the body.

DCs Take the Lymph Vessel Route

Dendritic cells move from peripheral tissues to lymph nodes through a process called migration, which involves several steps:

  • Sensing pathogens or inflammation: Immature dendritic cells in peripheral tissues detect pathogens or inflammatory signals through pattern recognition receptors (PRRs) and other sensing mechanisms.
  • Maturation: Upon sensing pathogens or inflammation, dendritic cells undergo maturation, characterized by upregulation of co-stimulatory molecules and MHC molecules.
  • Migration: Mature dendritic cells migrate from peripheral tissues to lymph nodes through a process called chemotaxis, guided by chemokines and other signaling molecules. They move along chemokine gradients towards lymphatic vessels.
  • Entry into lymphatic vessels: Dendritic cells enter lymphatic vessels through a process called lymphatic homing, facilitated by molecules like CCR7 and its ligands CCL19 and CCL21, which are expressed on the surface of both dendritic cells and lymphatic endothelial cells, respectively.

DCs Take the Blood Vessel Route

High endothelial venules (HEVs) are specialized blood vessels found in lymph nodes. They have a unique structure that facilitates the entry of lymphocytes, including dendritic cells, from the bloodstream into the lymph node parenchyma. HEVs are characterized by cuboidal endothelial cells with a high expression of adhesion molecules such as peripheral node addressin (PNAd) and chemokines like CCL21.

Cells, including dendritic cells, enter the lymph node if they use HEV as an entry point by following these steps:

  • Rolling: Dendritic cells circulating in the bloodstream initially adhere to the endothelial cells of HEVs through interactions between adhesion molecules such as selectins and their ligands.
  • Firm adhesion: Dendritic cells firmly adhere to the endothelial cells of HEVs through interactions between integrins and their ligands.
  • Diapedesis: Dendritic cells migrate through the endothelial layer of HEVs and enter the lymph node parenchyma, guided by chemokine gradients and other signaling molecules.

Once inside the lymph node, dendritic cells interact with CD4+ T cells and initiate adaptive immune responses by presenting antigens (signal 1) and providing co-stimulatory signals (signal 2).

Next Topic: Lymph Node Anatomy

Source: ChatGPT response prompted and edited by Joel Graff.

Details of MHC II Antigen Processing and Presentation

Antigen processing and presentation through Major Histocompatibility Complex (MHC) Class II molecules are fundamental processes in adaptive immunity, facilitating the detection and elimination of extracellular pathogens and foreign antigens. This intricate pathway involves several steps that ensure the efficient presentation of exogenous peptides derived from engulfed antigens to helper T lymphocytes (CD4+ T cells), thereby orchestrating appropriate immune responses. From the initial phagocytosis of the antigenic protein to the transport of peptide-loaded MHC Class II molecules to the plasma membrane, each stage of antigen processing is carefully regulated.

The process commences with the phagocytosis of extracellular material containing the antigenic protein by antigen-presenting cells (APCs), such as dendritic cells, macrophages, and B cells. Within the phagosome, the engulfed antigens undergo proteolytic degradation by the immunoproteasome—a specialized form of the proteasome complex that generates peptide fragments suitable for MHC Class II presentation. The immunoproteasome selectively cleaves proteins into shorter peptide fragments, typically ranging from 13 to 25 amino acids in length, ensuring the generation of antigenic peptides compatible with MHC Class II binding.

Following proteasomal degradation, the peptide fragments are loaded onto MHC Class II molecules within the endosomal or phagosomal compartments of the APC. MHC Class II molecules are synthesized in the endoplasmic reticulum (ER) and subsequently transported to endosomes or phagosomes via the Golgi apparatus. Within these compartments, MHC Class II molecules associate with a chaperone protein called the invariant chain (Ii), which prevents premature peptide binding and guides the MHC Class II molecules to specialized antigen-processing compartments. As the antigen is degraded, a fragment of Ii known as CLIP (class II-associated invariant chain peptide) remains bound to the MHC Class II molecule, temporarily blocking the peptide-binding groove.

To enable peptide loading, CLIP must be displaced from the peptide-binding groove of MHC Class II molecules. This process is facilitated by the action of a non-classical MHC Class II molecule called HLA-DM (in humans), or H2-M (in mice), which catalyzes the exchange of CLIP with the antigenic peptide. Once loaded with antigenic peptides, the peptide-MHC Class II complexes are stabilized and transported to the plasma membrane for presentation to CD4+ T cells.

The transport of peptide-loaded MHC Class II molecules to the plasma membrane involves the fusion of transport vesicles containing the peptide-MHC Class II complexes with the plasma membrane. This process is mediated by cytoskeletal elements and molecular motors that facilitate the movement of vesicles along microtubules. Upon reaching the cell surface, the peptide-loaded MHC Class II molecules are displayed for surveillance by CD4+ T cells, which possess T cell receptors (TCRs) capable of recognizing specific peptide-MHC Class II complexes.

Recognition of a peptide-MHC Class II complex by a CD4+ T cell leads to T cell activation, initiating a cascade of immune responses tailored to combat the invading pathogen or foreign antigen. CD4+ T cells serve as central regulators of the immune response, coordinating the activation of other immune cells and facilitating the generation of antigen-specific effector responses.

In conclusion, antigen processing and presentation via MHC Class II molecules involve a sequence of intricately regulated steps, beginning with the phagocytosis of the antigenic protein and culminating in the transport of peptide-loaded MHC Class II molecules to the plasma membrane. This process enables the immune system to detect and respond to extracellular pathogens and foreign antigens, thereby safeguarding the host against infectious diseases and maintaining immune homeostasis. Understanding the mechanisms underlying antigen processing and presentation via MHC Class II molecules is essential for elucidating immune responses and developing effective strategies for immunotherapy and vaccine development.

Next Topic: Antigen Delivery to Lymph Nodes by Dendritic Cells

Source: ChatGPT response prompted and edited by Joel Graff.

Details of MHC I Antigen Processing and Presentation

Antigen processing and presentation via Major Histocompatibility Complex (MHC) Class I molecules play a crucial role in the adaptive immune response by enabling the immune system to detect and eliminate intracellular pathogens and abnormal cells, such as infected or cancerous cells. This process involves a series of intricate steps that ensure the efficient presentation of antigenic peptides derived from endogenous proteins to cytotoxic T lymphocytes (CD8+ T cells), thereby initiating appropriate immune responses. The journey of antigenic peptides from degradation to presentation by MHC Class I molecules is a meticulously regulated process.

The first step in antigen processing begins with the degradation of endogenous proteins by the immunoproteasome, a specialized form of the proteasome complex found within cells. The immunoproteasome selectively cleaves proteins into shorter peptide fragments, typically 8 to 10 amino acids in length. This proteolytic activity is essential for generating antigenic peptides that can be presented by MHC Class I molecules.

Following proteasomal degradation, the resulting peptide fragments are transported into the endoplasmic reticulum (ER) via the transporter associated with antigen processing (TAP) complex. TAP serves as a gateway for peptide entry into the ER, where peptide loading onto MHC Class I molecules occurs. This process involves the coordinated action of various chaperone proteins, including tapasin, which facilitates the stabilization of MHC Class I molecules and promotes the binding of antigenic peptides.

Within the ER, the nascent MHC Class I molecules undergo peptide loading, where they associate with antigenic peptides generated by the immunoproteasome. Tapasin assists in the quality control of peptide binding, ensuring the selection of high-affinity peptides that can stably interact with MHC Class I molecules. Peptide loading onto MHC Class I molecules occurs in a peptide exchange process, where the previously bound peptide is displaced by the antigenic peptide.

Once loaded with antigenic peptides, the peptide-MHC Class I complexes are transported from the ER through the Golgi apparatus to the cell surface. This transport process involves the formation of transport vesicles that carry the peptide-loaded MHC Class I molecules to the plasma membrane. Along the way, the transport vesicles undergo maturation and fusion with the plasma membrane, ultimately releasing the peptide-MHC Class I complexes onto the cell surface.

At the plasma membrane, the peptide-loaded MHC Class I molecules are displayed for surveillance by CD8+ T cells, the effector cells of the cell-mediated immune response. CD8+ T cells possess T cell receptors (TCRs) that can recognize specific peptide-MHC Class I complexes. Upon encountering a peptide-MHC Class I complex that matches its TCR, a CD8+ T cell becomes activated, triggering a cascade of immune responses aimed at eliminating the source of the antigenic peptide.

In summary, antigen processing and presentation via MHC Class I molecules involve a series of precisely orchestrated steps, starting from the degradation of endogenous proteins by the immunoproteasome to the transport of peptide-loaded MHC Class I molecules to the plasma membrane. This process ensures the effective surveillance of intracellular pathogens and aberrant cells by the immune system, leading to the initiation of adaptive immune responses mediated by cytotoxic T lymphocytes. Understanding the mechanisms underlying antigen processing and presentation via MHC Class I molecules is essential for elucidating immune responses and developing immunotherapeutic strategies against infectious diseases and cancer.

Next Topic: Details of MHC II Antigen Processing and Presentation

Source: ChatGPT response prompted and edited by Joel Graff.

Presenting Endogenous and Exogenous Antigens

The terms “endogenous antigens” and “exogenous antigens” refer to the origin of the antigens in relation to the cell presenting them.

  1. Endogenous antigens:
    • These antigens originate from within the cell itself. They are typically derived from proteins synthesized by the cell’s own machinery.
    • Endogenous antigens are loaded onto Major Histocompatibility Complex (MHC) Class I molecules.
    • Presentation of endogenous antigens occurs primarily to cytotoxic T lymphocytes (CD8+ T cells).
    • MHC Class I molecules present endogenous antigens to CD8+ T cells to elicit immune responses against infected or abnormal cells. These antigens can include peptides derived from viral proteins synthesized within infected cells, abnormal proteins from cancerous cells, or proteins produced by intracellular bacteria.
  2. Exogenous antigens:
    • These antigens originate from outside the cell. They are typically derived from proteins that the cell has taken up from the extracellular environment through processes like phagocytosis or endocytosis.
    • Exogenous antigens are loaded onto Major Histocompatibility Complex (MHC) Class II molecules.
    • Presentation of exogenous antigens primarily occurs to helper T lymphocytes (CD4+ T cells).
    • MHC Class II molecules present exogenous antigens to CD4+ T cells, which can then stimulate other immune cells or coordinate immune responses. These antigens can include peptides derived from engulfed pathogens, such as bacteria or fungi, as well as proteins from other sources present in the extracellular environment.

Next Topic: Details of MHC I Antigen Processing and Presentation

Source: ChatGPT response prompted and edited by Joel Graff.

MHC I and MHC II: Summary of Antigen Processing and Presentation

The pathways for obtaining antigenic peptides to load into Major Histocompatibility Complex (MHC) Class I and Class II molecules differ due to the distinct roles and mechanisms of these two classes of MHC molecules in the immune system.

MHC Class I Pathway

Antigenic peptides presented by MHC Class I molecules are derived mainly from endogenous proteins synthesized within the cell. The pathway for loading antigenic peptides onto MHC Class I molecules is termed the endogenous pathway or cytosolic pathway. It involves the following steps:

Protein Degradation in the Cytosol: Endogenous proteins within the cell are degraded into short peptide fragments by the proteasome, a large protein complex.

Transport into the Endoplasmic Reticulum (ER): The resulting peptide fragments, typically 8-10 amino acids in length, are transported into the endoplasmic reticulum (ER) by the transporter associated with antigen processing (TAP) complex.

Peptide Loading onto MHC Class I Molecules: Inside the ER, these peptides are loaded onto nascent MHC Class I molecules that are being synthesized. This occurs in a process facilitated by chaperone proteins such as tapasin.

Transport to the Cell Surface: Loaded MHC Class I molecules are then transported from the ER through the Golgi apparatus to the cell surface, where they present the antigenic peptides to CD8+ T cells, which play a role in cytotoxic T lymphocyte (CTL) responses.


MHC Class II Pathway

Antigenic peptides presented by MHC Class II molecules are derived from exogenous proteins that are taken up by antigen-presenting cells (APCs) from the extracellular environment. The pathway for loading antigenic peptides onto MHC Class II molecules is termed the exogenous pathway and involves the following steps:

Antigen Uptake: APCs such as dendritic cells, macrophages, and B cells phagocytose or endocytose extracellular material containing antigenic proteins.

Protein Degradation in Endosomes or Phagosomes: The engulfed antigens are degraded into peptide fragments within endosomes or phagosomes by proteases.

MHC Class II Synthesis and Peptide Loading in Endosomes/Phagosomes: MHC Class II molecules are synthesized in the endoplasmic reticulum and then transported to endosomes or phagosomes. Within these compartments, MHC Class II molecules bind to a chaperone protein called the invariant chain (Ii), which prevents premature peptide binding. As the antigen is degraded, a fragment of Ii called CLIP remains bound to the MHC Class II molecule. CLIP is subsequently exchanged for the antigenic peptide by the action of HLA-DM (in humans) or H2-M (in mice).

Transport to the Cell Surface: The MHC Class II molecule, now loaded with the antigenic peptide, is transported to the cell surface where it presents the peptide to CD4+ T cells, which are involved in helper T cell responses.

These pathways ensure that both endogenous and exogenous antigens are efficiently presented to T cells, allowing for the activation of appropriate immune responses.

Next Topic: Presenting Endogenous and Exogenous Antigens

Source: ChatGPT response prompted and edited by Joel Graff.

Pros and Cons of MHC Polymorphisms


The high level of polymorphism observed in the genes of the major histocompatibility complex (MHC), particularly the class I and class II genes, is beneficial for the survival of a species from an evolutionary standpoint and presents challenges in organ transplantation.

Benefits of MHC Polymorphism for Species Survival:

  1. Pathogen Recognition and Immune Defense: MHC polymorphism ensures that a diverse range of pathogen-derived peptides can be presented to T cells, allowing for effective immune responses against a wide variety of pathogens. This diversity enhances the ability of a population to combat infections and reduces the risk of pathogen evasion.
  2. Hybrid Vigor and Genetic Diversity: MHC polymorphism contributes to genetic diversity within populations, including diversity at the cellular and individual levels. This genetic diversity promotes hybrid vigor, which enhances the overall fitness and adaptability of the population to changing environmental conditions.
  3. Avoidance of Inbreeding Depression: High MHC polymorphism reduces the likelihood of inbreeding depression, a phenomenon where offspring of closely related individuals exhibit reduced fitness due to the increased expression of deleterious recessive alleles. By promoting genetic diversity, MHC polymorphism helps maintain population health and resilience.

Challenges of MHC Polymorphism in Organ Transplantation:

  1. Matching for Histocompatibility: In organ transplantation, compatibility between donor and recipient MHC molecules is crucial to prevent graft rejection. The high level of polymorphism in MHC genes means that finding a suitable donor with matching MHC alleles can be challenging, particularly for individuals with rare or uncommon MHC haplotypes.
  2. Risk of Graft Rejection: Mismatched MHC molecules between donor and recipient can lead to the recognition of the transplanted organ as foreign by the recipient’s immune system, resulting in graft rejection. Even minor differences in MHC alleles can trigger immune responses, highlighting the importance of precise histocompatibility matching in transplantation.
  3. Immunosuppressive Therapy: To mitigate the risk of graft rejection, recipients of organ transplants often require long-term immunosuppressive therapy. However, suppressing the immune response increases the risk of infections and other complications, highlighting the delicate balance required in managing post-transplant immune function.

In summary, while MHC polymorphism confers evolutionary advantages by enhancing immune defense and genetic diversity within populations, it poses challenges in organ transplantation due to the need for precise histocompatibility matching to prevent graft rejection. Balancing these considerations is crucial in both understanding the evolutionary significance of MHC diversity and addressing the clinical challenges associated with organ transplantation.

End of Section 3.3

Next Topic: MHC I and MHC II: Summary of Antigen Processing and Presentation

Source: ChatGPT response prompted and edited by Joel Graff.

MHC Class I and II Protein Structure and Function

Classical MHC class I and MHC class II molecules are both involved in antigen presentation to T cells but have distinct structures and functions. Here’s a comparison of their structures:

Classical MHC Class I Molecules

  1. Structure:
    • MHC class I molecules are composed of a single polypeptide chain, typically referred to as the α chain, which is non-covalently associated with β2-microglobulin (β2m), a small protein.
    • The α chain consists of three extracellular domains: α1, α2, and α3. The α1 and α2 domains form a peptide-binding groove, where the antigenic peptide binds. The α3 domain interacts with CD8 molecules on cytotoxic T cells.
    • MHC class I molecules are anchored in the membrane of the presenting cell.
  2. Function:
    • Classical MHC class I molecules present endogenous peptides derived from cytosolic proteins to CD8+ cytotoxic T cells.
    • These peptides are typically 8-10 amino acids in length and are derived from proteins that have been degraded by the proteasome.

Classical MHC Class II Molecules

  1. Structure:
    • MHC class II molecules are composed of two polypeptide chains: an α chain and a β chain, both of which are encoded by genes within the MHC locus.
    • Each chain consists of two extracellular domains: α1 and α2 for the α chain, and β1 and β2 for the β chain. These domains form the peptide-binding groove.
    • Unlike MHC class I molecules, MHC class II molecules are anchored in the membrane of antigen-presenting cells.
  2. Function:
    • Classical MHC class II molecules present exogenous peptides derived from extracellular proteins to CD4+ helper T cells.
    • These peptides are typically longer (around 13-25 amino acids) than those presented by MHC class I molecules and are derived from proteins that have been internalized by the antigen-presenting cell and processed in endosomes or lysosomes.

While both MHC class I and class II molecules are essential for adaptive immune responses, they differ in structure, composition, and the types of peptides they present to T cells.

Next Topic: Pros and Cons of MHC Polymorphisms

Source: ChatGPT response prompted and edited by Joel Graff.

Many Immunity-Related Genes Populate the MHC Locus in Human and Mouse Genomes


The major histocompatibility complex (MHC), also known as the human leukocyte antigen (HLA) complex in humans and H2 complex in mice, is a highly polymorphic genomic region responsible for encoding proteins crucial for immune responses. The organization of the MHC locus differs slightly between mice and humans, but both contain class I, class II, and class III regions.

In Humans (HLA Complex):

  1. Class I Region (HLA-A, HLA-B, HLA-C):
    • Encodes classical class I molecules such as HLA-A, HLA-B, and HLA-C.
    • These molecules present endogenous peptides to CD8+ T cells, playing a crucial role in cytotoxic T cell-mediated immunity.
    • Also includes non-classical class I genes such as HLA-E, HLA-F, and HLA-G, which have specialized functions in immune regulation.
  2. Class II Region (HLA-DP, HLA-DQ, HLA-DR):
    • Encodes classical class II molecules such as HLA-DP, HLA-DQ, and HLA-DR.
    • These molecules present exogenous peptides to CD4+ T cells, initiating helper T cell-mediated immune responses.
    • Also contains genes encoding the invariant chain (Ii) and the class II-associated invariant chain peptide (CLIP), involved in the assembly and trafficking of class II molecules.
  3. Class III Region:
    • Contains genes with diverse functions, including those involved in complement activation, inflammation, and antigen presentation regulation.
    • Includes genes such as complement components (C2, C4, factor B), tumor necrosis factors (TNF-α, TNF-β), heat shock proteins, and others.
    • Not directly involved in antigen presentation but plays important roles in immune regulation and inflammation.

In Mice (H2 Complex):

  1. Class I Region (H2-K, H2-D, H2-L):
    • Encodes classical class I molecules such as H2-K, H2-D, and H2-L.
    • Similar to humans, these molecules present endogenous peptides to CD8+ T cells.
    • Also includes non-classical class I genes with specialized immune functions.
  2. Class II Region (H2-A):
    • Encodes classical class II molecules such as H2-Aα and H2-Aβ chains.
    • These molecules present exogenous peptides to CD4+ T cells, similar to human class II molecules.
    • Additionally, contains genes encoding the invariant chain and associated proteins involved in class II antigen presentation.
  3. Class III Region:
    • Contains genes with diverse functions, including those involved in complement activation, inflammation, and immune regulation.
    • Similar to humans, includes genes encoding complement components, tumor necrosis factors, and other immune-related proteins.

In both humans and mice, the MHC locus is highly polymorphic, with numerous allelic variants present within each gene, contributing to the diversity of immune responses and individual susceptibility to diseases.

Next Topic: MHC Class I and II Protein Structure and Function

Source: ChatGPT response prompted and edited by Joel Graff.

Overview of the Ubiquitin-Proteasome System of Protein Degradation

Proteins are targeted for degradation by the ubiquitin-proteasome system (UPS) through the covalent attachment of ubiquitin molecules to specific lysine residues on the target protein. This process involves the sequential action of three main enzymes: E1 (ubiquitin-activating enzyme), E2 (ubiquitin-conjugating enzyme), and E3 (ubiquitin ligase).

Here’s a summary of how proteins are targeted for degradation:

  1. Ubiquitination: The process starts with the activation of ubiquitin by E1 in an ATP-dependent manner. Activated ubiquitin is then transferred to an E2 enzyme. The E2 enzyme, in conjunction with an E3 ligase, facilitates the transfer of ubiquitin from the E2 enzyme to the target protein. The E3 ligase recognizes specific target proteins and mediates the transfer of ubiquitin to lysine residues on the target protein, forming a ubiquitin chain.
  2. Ubiquitin Chain Assembly: Ubiquitin itself contains seven lysine residues (K6, K11, K27, K29, K33, K48, and K63), each of which can serve as an attachment site for the formation of polyubiquitin chains. While K48-linked polyubiquitin chains are the most well-characterized signal for proteasomal degradation, other types of polyubiquitin linkages, such as K11 and K63, can also target proteins for degradation by the proteasome or other degradation pathways, depending on the context and the nature of the ubiquitin chain-binding proteins.
  3. Proteasomal Degradation: Proteins marked with polyubiquitin chains are recognized and degraded by the 26S proteasome, a large multi-subunit protease complex. The polyubiquitin chain serves as a signal for recognition and subsequent unfolding of the protein substrate by the proteasome. The substrate is then translocated into the proteolytic core of the proteasome, where it is cleaved into small peptides.

Regarding the fate of peptides generated by proteasomal degradation:

  • The peptides produced by proteasomal degradation are typically 7-8 amino acids in length.
  • These peptides are released into the cytosol and can be further processed by cytosolic peptidases.
  • The resulting peptides can serve as a source of amino acids for protein synthesis or can be presented on the cell surface by major histocompatibility complex (MHC) molecules for immune surveillance.

In summary, while K48-linked polyubiquitin chains are the most commonly associated with proteasomal degradation, other types of polyubiquitin linkages can also target proteins for degradation by the proteasome or other degradation pathways. The peptides generated by proteasomal degradation can have various fates, including serving as a source of amino acids or being presented on MHC molecules.

End of Section 3.2

Next Topic: Many Immunity-Related Genes Populate the MHC Locus in Human and Mouse Genomes

Source: ChatGPT response prompted and edited by Joel Graff.

Endosome Cycle: Maturation, Sorting, Cargo Degradation, and Recycling

The maturation process of phagosomes and endosomes involves several steps, primarily to process and sort the ingested material or internalized cargo. Here are the general steps involved in endosomal maturation and recycling:

  1. Early Endosome Formation: After endocytosis or phagocytosis, vesicles containing the ingested material merge with early endosomes. These early endosomes are characterized by their slightly acidic pH and contain a mix of sorting and signaling proteins.
  2. Maturation to Late Endosomes: Early endosomes mature into late endosomes through a process that involves changes in lipid composition, membrane protein trafficking, and further acidification of the endosomal lumen. Late endosomes have a more acidic pH than early endosomes.
  3. Cargo Sorting: Within late endosomes, sorting of cargo occurs. Different cargoes can be targeted for different fates, such as recycling back to the cell surface, degradation in lysosomes, or trafficking to other cellular compartments.
  4. Recycling: Some cargo molecules are sorted for recycling. This can involve the formation of recycling endosomes, which bud off from the maturing endosome and transport cargo molecules back to the cell surface for reuse.
  5. Degradation: Other cargo molecules are targeted for degradation. Late endosomes containing these cargoes can fuse with lysosomes, forming hybrid organelles called endolysosomes, where the cargo is degraded by lysosomal enzymes.
  6. Fusion with Lysosomes: Late endosomes that contain cargo destined for degradation fuse with lysosomes, resulting in the formation of endolysosomes. This fusion delivers the cargo to the acidic environment of the lysosome, where hydrolytic enzymes break down the cargo into its constituent molecules.
  7. Membrane Trafficking: Throughout the maturation process, membrane trafficking events involving various Rab GTPases, SNARE proteins, and other regulatory factors help regulate the dynamics of endosomal compartments, including their fusion, fission, and movement within the cell.
  8. Exit from Endosomal System: Once cargo sorting, recycling, and degradation processes are completed, components of the endosomal system may exit either through recycling pathways back to the cell surface or through retrograde transport pathways to other cellular compartments, such as the trans-Golgi network.

These steps are crucial for maintaining cellular homeostasis, regulating signaling pathways, and ensuring efficient uptake, processing, and recycling of extracellular materials by the cell.

Next Topic: Overview of the Ubiquitin-Proteasome System of Protein Degradation

Source: ChatGPT response prompted and edited by Joel Graff.

“Actin” Out: Phagocytosis and Macropinocytosis

Phagocytosis

Phagocytosis is a vital process by which specialized cells, such as macrophages, neutrophils, and dendritic cells, engulf and internalize large particles, including pathogens, cell debris, and foreign particles, for degradation and clearance.

Phagocytic cells recognize and bind to target particles through specific receptors on their cell surface. These receptors can recognize molecular patterns associated with pathogens (PAMPs) or opsonins, which are molecules that coat the surface of target particles and enhance their recognition by phagocytic cells.

Once bound to the target particle, the phagocytic cell undergoes cytoskeletal rearrangements to extend pseudopods around the particle, forming a phagocytic cup. Actin polymerization drives the extension of pseudopods, leading to the engulfment of the particle and the formation of a phagosome, an intracellular vesicle containing the ingested material.

The newly formed phagosome undergoes a series of maturation steps, including fusion with endosomes and lysosomes. This process involves the sequential acquisition of various membrane proteins and enzymes, ultimately leading to the formation of a phagolysosome, a vesicle with degradative capacity.

Macropinocytosis

Macropinocytosis is a form of bulk endocytosis characterized by the nonspecific uptake of extracellular fluid and solutes into large vesicles called macropinosomes. This process involves the actin-dependent formation of membrane ruffles or lamellipodia at the cell surface, which extend and fuse back upon themselves, engulfing a volume of extracellular fluid and solutes in the process.

Macropinocytosis is typically used by cells to internalize large volumes of extracellular fluid, along with any dissolved solutes, nutrients, or signaling molecules present in the surrounding environment. It is a non-selective process that allows cells to sample their surroundings, scavenge for nutrients, and respond to environmental cues.

Cells that are highly motile, such as immune cells (e.g., macrophages, dendritic cells), and cells undergoing rapid growth or migration, such as cancer cells, often utilize macropinocytosis to sustain their metabolic demands and sense changes in their microenvironment. Additionally, some specialized cell types, such as endothelial cells and epithelial cells, may also employ macropinocytosis to regulate the composition of the extracellular milieu and facilitate nutrient uptake or signaling.

Overall, macropinocytosis serves as an important mechanism for nutrient acquisition, immune surveillance, and cellular signaling in various cell types, contributing to their homeostasis and functional responses to extracellular stimuli.

End of Section 3.1 – Different Forms of Engulfment

Next Topic: Endosome Cycle: Maturation, Sorting, Cargo Degradation, and Recycling

Source: ChatGPT response prompted and edited by Joel Graff.

Two Common Endocytic Mechanisms: Clathrin-Coated Pits and Caveolae

Clathrin-coated pits and caveolae are specialized structures on the cell membrane that play significant roles in various cellular processes. While both structures are involved in endocytosis and intracellular signaling, they exhibit distinct characteristics and functions in the context of immunology.

Structure and Composition:

Clathrin-coated pits are invaginations of the cell membrane characterized by a lattice-like coat composed of the protein clathrin. Clathrin molecules assemble into a hexagonal lattice, providing structural support and facilitating the budding of vesicles from the membrane during endocytosis. Clathrin-coated pits are involved in the internalization of various ligands, including antigens, receptors, and pathogens.

In contrast, caveolae are flask-shaped invaginations of the cell membrane enriched in cholesterol and sphingolipids, with the presence of caveolin proteins imparting a unique morphology. Caveolins, particularly caveolin-1, are integral membrane proteins that oligomerize to form a scaffold on the cytoplasmic face of caveolae. Caveolae are involved in lipid metabolism, signal transduction, and protection from mechanical forces (mechanoprotection).

Roles in Immunology:

Clathrin-coated pits are essential for antigen uptake and processing by antigen-presenting cells (APCs), such as dendritic cells, macrophages, and B cells. Antigens encountered by APCs are internalized through clathrin-coated pits via receptor-mediated endocytosis. Once internalized, antigens are processed into peptide fragments and presented on major histocompatibility complex (MHC) molecules for recognition by T cells, initiating adaptive immune responses.

Caveolae also contribute to immunological processes, albeit through distinct mechanisms. Caveolae are involved in the regulation of signaling pathways implicated in immune cell activation and cytokine production. Signaling molecules, including receptors, kinases, and G-proteins, are localized within caveolae, facilitating their interactions and downstream signaling events. Caveolae also serve as platforms for the organization of immune signaling complexes, modulating the amplitude and duration of immune responses.

Physiological Significance in Immunology:

The dynamic regulation of clathrin-coated pits and caveolae is crucial for immune surveillance and response to pathogens. Clathrin-mediated endocytosis facilitates the internalization of antigens, pathogen-associated molecular patterns (PAMPs), and immune receptors, enabling immune cells to sample the extracellular environment and initiate appropriate immune responses.

Caveolae contribute to the regulation of immune cell function and inflammatory responses. The localization of signaling molecules within caveolae allows for the fine-tuning of immune signaling pathways, influencing cell activation, proliferation, and cytokine secretion. Dysregulation of caveolae-mediated signaling has been implicated in various immunological disorders, including autoimmune diseases, inflammatory disorders, and cancer.

Next Topic: “Actin” Out: Phagocytosis and Macropinocytosis

Source: ChatGPT response prompted and edited by Joel Graff.

Pinocytosis: Who? What? Why?

Pinocytosis, also known as “cell drinking,” is a form of endocytosis where the cell engulfs extracellular fluid along with dissolved solutes. Unlike phagocytosis, which involves the ingestion of large particles, pinocytosis involves the uptake of smaller, dissolved substances and fluids. Many cell types utilize pinocytosis to sample their environment, regulate nutrient intake, and internalize signaling molecules.

Cells that frequently use pinocytosis include:

  1. Epithelial Cells: These cells line various surfaces of the body and are involved in absorption and secretion. Pinocytosis helps them take up nutrients and regulate the composition of extracellular fluid.
  2. White Blood Cells (Macrophages, Dendritic Cells): These cells are part of the immune system and use pinocytosis to engulf pathogens, debris, and other materials from the extracellular environment.
  3. Endothelial Cells: These cells line the interior of blood vessels and play a role in regulating the passage of substances between the blood and surrounding tissues. Pinocytosis aids in the uptake of molecules from the blood.

The rate of pinocytosis can indeed change in response to various environmental and cellular signals. Factors such as the availability of nutrients, concentrations of signaling molecules, and the presence of pathogens or toxins can all influence pinocytosis rates. For example:

  • Nutrient Availability: When cells are starved or deficient in certain nutrients, they may increase pinocytosis to enhance nutrient uptake from the extracellular environment.
  • Signaling Molecules: Some signaling molecules can trigger changes in pinocytosis rates as part of cellular responses. For instance, growth factors or hormones may induce cells to increase pinocytosis to internalize these signaling molecules.
  • Pathogens or Toxins: In response to the presence of pathogens or toxins, cells of the immune system may increase pinocytosis to engulf and clear these harmful substances.

Regarding the degradation of cargo transported into the cell through pinocytosis, once the pinocytic vesicles are formed, they fuse with endosomes, which are membrane-bound compartments within the cell. From there, the contents of the vesicles can be sorted for further processing. Some of the cargo may be recycled back to the cell surface, while others may be directed to lysosomes for degradation. Lysosomes contain enzymes that break down various molecules, including proteins, nucleic acids, lipids, and carbohydrates, into smaller components that can be recycled or excreted by the cell. Therefore, lysosomes play a crucial role in degrading the content of cargo transported into the cell through the pinocytosis pathway.

Next Topic: Two Common Endocytic Mechanisms: Clathrin-Coated Pits and Caveolae

Source: ChatGPT response prompted and edited by Joel Graff.

Move-In Day! Transporting Things into Cells

The plasma membrane of a human cell is selectively permeable, meaning it allows certain molecules to pass through while restricting others. The types of molecules that are typically transported into a human cell through the plasma membrane can be categorized into several groups:

  1. Small, non-polar molecules: These molecules, such as oxygen and carbon dioxide, can diffuse directly through the lipid bilayer of the plasma membrane without the need for channels or transport proteins. Their movement is driven by the concentration gradient.
  2. Small, polar molecules: Water is a small polar molecule that can diffuse through the plasma membrane via specialized channels called aquaporins. These channels facilitate the movement of water molecules across the membrane, allowing for rapid diffusion.
  3. Large, polar molecules and ions: Large polar molecules and ions typically require transport proteins to cross the plasma membrane. This includes ions like sodium (Na+), potassium (K+), calcium (Ca2+), and chloride (Cl-) ions, as well as molecules like glucose and amino acids. These molecules traverse the membrane through either facilitated diffusion or active transport.
    • Facilitated diffusion: In facilitated diffusion, molecules move down their concentration gradient with the aid of transport proteins such as channels or carrier proteins. Channels provide a hydrophilic pore for ions to pass through, while carrier proteins undergo a conformational change to shuttle molecules across the membrane.
    • Active transport: Active transport requires the input of energy, usually in the form of ATP, to move molecules against their concentration gradient. This process is carried out by pumps, such as the sodium-potassium pump, which actively transports sodium ions out of the cell and potassium ions into the cell.
  4. Large molecules and particles: Some molecules and particles are too large to cross the plasma membrane through diffusion or transport proteins. In these cases, the cell can engulf these substances through processes such as phagocytosis, endocytosis, and pinocytosis.
    • Phagocytosis: Phagocytosis is the process by which cells engulf large particles such as bacteria, dead cells, or other foreign particles. The plasma membrane extends around the particle, forming a phagosome, which is then internalized into the cell where it can be broken down by lysosomes.
    • Endocytosis: Endocytosis is a general term for the process by which cells internalize extracellular molecules and particles by forming vesicles derived from the plasma membrane. This includes phagocytosis, as well as other forms such as receptor-mediated endocytosis, where specific molecules bind to receptors on the cell surface and are internalized in vesicles.
    • Pinocytosis: Pinocytosis, also known as “cell drinking,” involves the non-selective uptake of small droplets of extracellular fluid containing dissolved solutes. The plasma membrane invaginates to form small vesicles containing the fluid, which are then internalized into the cell.

Next Topic: Pinocytosis: Who? What? Why?

Source: ChatGPT response prompted and edited by Joel Graff.

Diverse Antiviral Activity of Type I Interferon-Stimulated Genes

Dozens of genes are upregulated by the ISGF3 transcription factor complex. The following represents some genes that are commonly upregulated in cells responding to type I interferon (IFN) along with their antiviral mechanisms:

  1. IFIT1 (Interferon-induced protein with tetratricopeptide repeats 1):
    • The IFIT1 gene is also known as ISG56.
    • IFIT1 inhibits viral translation by binding to viral mRNA and preventing ribosome assembly, thereby suppressing viral protein synthesis.
  2. OAS1 (2′-5′-oligoadenylate synthetase 1):
    • OAS1 catalyzes the synthesis of 2′-5′-oligoadenylates (2-5A) upon activation by dsRNA. 2-5A molecules activate the latent ribonuclease RNase L, leading to degradation of viral RNA and inhibition of viral replication.
  3. MX1 (Myxovirus resistance protein 1):
    • MX1 interferes with various stages of the viral replication cycle, including viral entry, transcription, and nuclear export. It acts by forming oligomeric structures that bind to viral nucleocapsids, preventing their nuclear import and subsequent replication.
  4. ISG15 (Interferon-stimulated gene 15):
    • ISG15 conjugates to target proteins, a process known as ISGylation, leading to their modification and alteration of their function. ISG15ylation can interfere with viral replication by targeting viral proteins for degradation or modifying host proteins involved in viral replication processes.
  5. RSAD2 (Radical S-adenosyl methionine domain-containing protein 2):
    • RSAD2 encodes the protein “Viperin”.
    • RSAD2/Viperin inhibits the replication of a broad range of viruses by interfering with various steps of the viral life cycle, including viral assembly, budding, and maturation. It disrupts lipid rafts in the cell membrane, which are important for viral assembly and release.
  6. IFIH1 (Interferon-induced helicase C domain-containing protein 1):
    • IFIH1 encodes the protein “MDA5”.
    • IFIH1/MDA5 functions as a cytosolic sensor of viral RNA, particularly dsRNA. Upon recognition of viral RNA, MDA5 activates downstream signaling pathways, leading to the production of type I IFNs and the induction of antiviral genes.
  7. STAT1 (Signal transducer and activator of transcription 1):
    • STAT1 is a key transcription factor that mediates the cellular response to type I IFNs. Upon phosphorylation and dimerization, STAT1 translocates to the nucleus and binds to ISREs in the promoters of target genes, including other IFN-stimulated genes involved in antiviral defense.
  8. IRF7 (Interferon regulatory factor 7):
    • IRF7 is a transcription factor that plays a critical role in the amplification of type I IFN responses. It is induced by type I IFNs and activates the transcription of additional IFN genes, thereby amplifying the antiviral response.
  9. EIF2AK2 (Eukaryotic translation initiation factor 2-alpha kinase 2):
    • The EIF2AK2 gene encodes protein kinase R (PKR).
    • PKR is activated by dsRNA and phosphorylates the eukaryotic translation initiation factor eIF2α, leading to inhibition of protein synthesis and suppression of viral replication. PKR also activates NF-κB and IRF3, contributing to the production of type I IFNs and other pro-inflammatory cytokines.
  10. TRIM25 (Tripartite motif-containing protein 25):
    • TRIM25 is an E3 ubiquitin ligase that catalyzes the ubiquitination and activation of RIG-I, leading to the induction of type I IFNs and the expression of antiviral genes.

These genes represent a subset of the diverse array of interferon-stimulated genes (ISGs) that are upregulated in response to type I IFNs. They collectively contribute to the host’s innate antiviral defense mechanisms by targeting various stages of the viral replication cycle, modulating host cell signaling pathways, and promoting an antiviral state within infected cells.

End of Section 2.4

Next Topic: Move-In Day! Transporting Things into Cells

Source: ChatGPT response prompted and edited by Joel Graff.

Cellular Response to Type I Interferons

Downstream signaling pathways activated by the type I interferon receptor (IFNAR) play a central role in mediating the cellular response to interferons (IFNs), particularly type I IFNs such as IFN-α and IFN-β. These pathways lead to the induction of antiviral and immunomodulatory genes, as well as various cellular responses that contribute to the innate and adaptive immune responses. Here’s a discussion of the key signaling pathways downstream of IFNAR:

  1. JAK-STAT Signaling Pathway:
    • Upon binding of type I IFNs to IFNAR, the receptor-associated Janus kinases (JAKs), particularly JAK1 and Tyk2, are activated through transphosphorylation.
    • Activated JAKs phosphorylate tyrosine residues on the intracellular domains of IFNAR subunits, creating docking sites for signal transducer and activator of transcription (STAT) proteins.
    • STAT proteins, mainly STAT1 and STAT2, are recruited to the phosphorylated receptor complex, where they are phosphorylated by JAKs.
    • Phosphorylated STAT1 and STAT2 form heterodimers and associate with another transcription factor called interferon regulatory factor 9 (IRF9) to form the IFN-stimulated gene factor 3 (ISGF3) complex.
    • ISGF3 translocates to the nucleus, where it binds to IFN-stimulated response elements (ISREs) in the promoters of interferon-stimulated genes (ISGs), leading to their transcriptional activation.
    • ISGs encode proteins with various antiviral, immunomodulatory, and antiproliferative functions, such as protein kinase R (PKR), 2′-5′-oligoadenylate synthetase (OAS), and Mx proteins, which inhibit viral replication and spread.
  2. MAPK Signaling Pathway:
    • Type I IFN signaling can also activate the Mitogen-Activated Protein Kinase (MAPK) pathway, including extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 MAPK.
    • Activation of MAPKs can occur through JAK-STAT-dependent mechanisms or via direct interactions with other signaling molecules downstream of IFNAR.
    • Activated MAPKs translocate to the nucleus or regulate cytoplasmic signaling events, leading to the induction of genes involved in cell proliferation, differentiation, apoptosis, and immune responses.
  3. PI3K-Akt Signaling Pathway:
    • Type I IFNs can activate the phosphoinositide 3-kinase (PI3K)-Akt pathway, which regulates various cellular processes, including cell survival, proliferation, and metabolism.
    • Activation of PI3K-Akt signaling downstream of IFNAR may contribute to the anti-apoptotic and pro-survival effects of type I IFNs, as well as modulate immune cell function and cytokine production.
  4. Notch Signaling Pathway:
    • Recent studies have suggested crosstalk between type I IFN signaling and the Notch pathway, which plays essential roles in cell fate determination, development, and immune responses.
    • Activation of Notch signaling downstream of IFNAR may regulate gene expression programs involved in cell differentiation, immune cell activation, and tissue homeostasis.

Overall, the signaling pathways downstream of the type I interferon receptor (IFNAR) orchestrate a complex cellular response that encompasses antiviral defenses, immune modulation, and various cellular processes critical for host defense against viral infections and other pathogens. These pathways represent intricate networks of molecular interactions that regulate gene expression and cellular functions to maintain immune homeostasis and combat infectious threats.

Next Topic: Diverse Antiviral Activity of Type 1 Interferon-Stimulated Genes

Source: ChatGPT response prompted and edited by Joel Graff.

The Interferon Beta “Enhanceosome”

The interferon beta (IFN-β) enhanceosome is a protein complex that forms on the promoter region of the IFN-β gene in response to viral infection or other immune stimuli. This enhanceosome plays a crucial role in regulating the expression of the IFN-β gene, which is a key component of the innate immune response against viral infections.

The IFN-β enhanceosome consists of multiple transcription factors and coactivators that bind cooperatively to specific DNA sequences in the promoter region of the IFN-β gene. These transcription factors include:

  1. Interferon Regulatory Factor 3 (IRF3): IRF3 is a transcription factor that is activated in response to viral infection. Upon activation, IRF3 translocates to the nucleus where it binds to specific DNA sequences known as interferon-stimulated response elements (ISREs) in the IFN-β promoter.
  2. Nuclear Factor-kappa B (NF-κB): NF-κB is another transcription factor that is activated by various immune stimuli, including viral infection. It forms a complex with other proteins, such as RelA/p65 and p50, and binds to specific DNA sequences in the IFN-β promoter region.
  3. ATF-2/c-Jun: ATF-2 (Activating Transcription Factor 2) and c-Jun are components of the AP-1 transcription factor complex. They bind to the IFN-β promoter region and participate in the formation of the enhanceosome.
  4. CREB-binding protein (CBP)/p300: CBP and p300 are coactivators that interact with other transcription factors bound to the IFN-β promoter, helping to stabilize the enhanceosome and enhance transcriptional activation.

These transcription factors and coactivators work together to form a stable enhanceosome complex on the IFN-β promoter, leading to the activation of IFN-β gene expression and subsequent induction of the antiviral immune response.

Next Topic: Cellular Response to Type 1 Interferons

Source: ChatGPT response prompted and edited by Joel Graff.

Overview of Signaling Pathways Downstream of RLR Detection of Virus Infection

  1. Activation of RLRs: RIG-I-like receptors (RLRs), including RIG-I (Retinoic acid-inducible gene I) and MDA5 (Melanoma differentiation-associated protein 5), recognize viral RNA in the cytoplasm of infected cells. Upon binding to viral RNA, RLRs undergo a conformational change, exposing their caspase activation and recruitment domains (CARDs), which are essential for downstream signaling.
  2. Interaction with MAVS: The activated RLRs interact with MAVS (mitochondrial antiviral signaling protein) through homotypic CARD-CARD interactions. MAVS serves as a critical adaptor protein localized on the outer mitochondrial membrane, facilitating the transmission of signals from RLRs to downstream effector molecules.
  3. Activation of TRAFs: MAVS activation leads to the recruitment and activation of TRAFs, particularly TRAF3 and TRAF6. TRAF3 plays a crucial role in the activation of IRF3, while TRAF6 is involved in the activation of NF-κB and MAPK signaling pathways.
  4. Activation of IRF3: MAVS-mediated activation of TRAF3 leads to the phosphorylation and subsequent activation of IRF3 (Interferon Regulatory Factor 3). Activated IRF3 forms homodimers or heterodimers with other transcription factors and translocates to the nucleus, where it binds to the promoter regions of type I interferon genes, such as IFN-β, inducing their transcription.
  5. Activation of NF-κB: TRAF6, which is recruited to MAVS upon its activation, activates the NF-κB (Nuclear Factor-kappa B) signaling pathway. TRAF6 mediates the activation of the IκB kinase (IKK) complex, leading to the phosphorylation and subsequent degradation of inhibitory IκB proteins. This results in the release and nuclear translocation of NF-κB dimers, such as p50/p65, which regulate the transcription of pro-inflammatory cytokines, including TNF-α, IL-6, and IL-1β.
  6. Activation of AP-1: MAVS signaling can also activate the Mitogen-Activated Protein Kinase (MAPK) pathway, leading to the activation of transcription factors such as AP-1 (Activator Protein-1). MAPKs, including ERK, JNK, and p38, are activated downstream of TRAFs or through direct interactions with MAVS. Activated AP-1 regulates the expression of genes involved in inflammation, cell proliferation, and apoptosis, contributing to the antiviral response.

Overall, the signaling cascade from RLRs to MAVS to TRAFs to transcription factors IRF3, NF-κB, and AP-1 orchestrates a coordinated immune response against viral infections, leading to the induction of type I interferons, pro-inflammatory cytokines, and other antiviral effectors that help control viral replication and spread.

Next Topic: The Interferon Beta “Enhanceosome”

Source: ChatGPT response prompted and edited by Joel Graff.

Training Innate Immunity Can Enhance Protection

In contrast to endotoxin tolerance, the innate immune response can exhibit a heightened and “trained” state following the primary exposure to pathogen-associated molecular patterns (PAMPs), particularly when signaling through C-type lectin receptors (CLRs). This phenomenon is known as “innate immune training” and involves the enhanced responsiveness of immune cells upon subsequent encounters with the same or different pathogens. Innate immune training has been studied in the context of fungal and mycobacterial infections, among others.

Key Features of Innate Immune Training:

  1. Epigenetic Reprogramming:
    • Exposure to certain PAMPs, such as β-glucans or mannans recognized by CLRs, can induce long-lasting changes in the epigenetic landscape of immune cells.
    • This includes alterations in histone modifications and DNA methylation patterns, which then influence gene expression programs.
  2. Metabolic Changes:
    • Innate immune training is associated with shifts in cellular metabolism, including changes in glycolysis, oxidative phosphorylation, and fatty acid metabolism.
    • Metabolic reprogramming supports the increased energy demands of activated immune cells during heightened responsiveness.
  3. Enhanced Cytokine Production:
    • Trained immune cells exhibit an augmented capacity to produce pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), upon restimulation.
  4. Increased Antimicrobial Activity:
    • Trained immune cells may display improved phagocytic activity and enhanced killing of pathogens, contributing to a more robust defense against infections.
  5. Memory-Like Characteristics:
    • While innate immune training shares similarities with adaptive immune memory, it operates independently of traditional adaptive immune mechanisms, such as antigen specificity and immunological memory cells.

Implications and Significance:

  1. Broad Protection:
    • Innate immune training provides a form of non-specific memory that can confer broad protection against subsequent infections.
    • This heightened state of alertness enables a faster and more effective response to a variety of pathogens.
  2. Vaccination Strategies:
    • Harnessing the principles of innate immune training has implications for the development of novel vaccine strategies.
    • Certain vaccine formulations may leverage these training mechanisms to enhance the immune response and improve vaccine efficacy.
  3. Pathological Consequences:
    • Dysregulation of innate immune training may contribute to chronic inflammatory conditions or autoimmune diseases.
    • Understanding the underlying mechanisms is crucial for therapeutic interventions.

In summary, innate immune training represents a form of immunological memory within the innate immune system, providing an enhanced and more efficient response to subsequent infections. While endotoxin tolerance and immune training seem paradoxical, these contrasting states highlight the dynamic and context-dependent nature of the innate immune response.

End of Section 2.3

Next Topic: Overview of Signaling Pathways Downstream of RLR Detection of Virus Infection

Source: ChatGPT response prompted and edited by Joel Graff.

Endotoxin Tolerance to LPS and Other TLR-Recognized PAMPs

Endotoxin tolerance refers to a phenomenon in which cells become less responsive to repeated exposure to lipopolysaccharide (LPS), a component of the outer membrane of Gram-negative bacteria and a potent stimulator of Toll-like receptor 4 (TLR4). This tolerance mechanism is a form of negative feedback that aims to prevent excessive and potentially harmful inflammatory responses. Similar tolerance mechanisms can occur with other pathogen-associated molecular patterns (PAMPs) recognized by Toll-like receptors (TLRs), including those associated with Gram-positive bacteria, viruses, and other microbes.

Mechanisms of Endotoxin Tolerance

  1. Downregulation of TLR Expression:
    • Repeated exposure to LPS or other TLR ligands can lead to the downregulation of TLR expression on the cell surface.
    • This reduces the cell’s ability to recognize and respond to subsequent exposures.
  2. Inhibition of Signaling Pathways:
    • Tolerance involves inhibiting downstream signaling pathways associated with TLR activation.
    • Negative regulators, such as IRAK-M (Interleukin-1 receptor-associated kinase-M), are induced to suppress pro-inflammatory signaling cascades.
  3. Induction of Anti-inflammatory Mediators:
    • Tolerized cells may produce anti-inflammatory cytokines, such as interleukin-10 (IL-10), which counteract the effects of pro-inflammatory mediators.
  4. Epigenetic Modifications:
    • Persistent exposure to TLR ligands can lead to epigenetic changes, influencing the transcriptional regulation of genes involved in the inflammatory response.

Dangers Associated with Septic Shock

Septic shock is a life-threatening medical emergency characterized by a systemic and dysregulated host response to infection. It results from the body’s exaggerated immune reaction to bacterial, viral, fungal, or parasitic infections, leading to widespread inflammation and organ dysfunction.

In septic shock, the normal mechanisms that regulate the immune response become disrupted, causing a cascade of events that can be detrimental. The release of inflammatory mediators, including cytokines, triggers a systemic inflammatory response that can lead to severe vasodilation, increased vascular permeability, and a decline in blood pressure. This, in turn, impairs blood flow to vital organs, leading to organ failure.

Common symptoms include hypotension, altered mental status, rapid heart rate, and respiratory distress. Septic shock requires immediate medical intervention, typically involving broad-spectrum antibiotics to target the underlying infection, intravenous fluids to support blood pressure, and vasopressors to constrict blood vessels. Despite medical advancements, septic shock remains a significant cause of morbidity and mortality worldwide, highlighting the critical need for early recognition and aggressive management.

Tolerance to PAMPs as a Negative Feedback Response to Septic Shock

  1. Preventing Excessive Inflammation:
    • Tolerance to PAMPs serves as a negative feedback mechanism to prevent excessive and prolonged inflammatory responses during septic shock.
    • Uncontrolled inflammation can lead to a “cytokine storm,” causing widespread tissue damage and organ failure.
  2. Avoiding Immune Paralysis:
    • While tolerance prevents excessive inflammation, it is crucial to avoid immune paralysis.
    • Complete suppression of the immune response could lead to an inability to clear the underlying infection, exacerbating the risk of septic shock.
  3. Balancing Inflammatory and Anti-inflammatory Signals:
    • Tolerance mechanisms contribute to the delicate balance between inflammatory and anti-inflammatory signals, helping to modulate the immune response.
  4. Resolution of Inflammation:
    • Tolerance mechanisms play a role in the resolution of inflammation, allowing the host to control the immune response and return to homeostasis.

Understanding and manipulating these tolerance mechanisms are essential for developing therapeutic strategies to modulate the immune response in conditions like septic shock. While tolerance is a protective response, its dysregulation or failure can have severe consequences, emphasizing the need for further research to elucidate the underlying mechanisms and potential therapeutic interventions.

Next Topic: Training Innate Immunity Can Enhance Protection

Source: ChatGPT response prompted and edited by Joel Graff.

Ending Inflammation and Restoring Tissue Homeostasis

The resolution of inflammation is a crucial phase that involves the active termination of the inflammatory response and the restoration of tissue homeostasis. Successful resolution prevents chronic inflammation, tissue damage, and excessive immune activation. Several key processes and negative feedback mechanisms contribute to the resolution of inflammation:

  1. Anti-inflammatory Signals:
    • Production of Anti-inflammatory Cytokines:
      • Cells at the inflammatory site, including macrophages and regulatory T cells, produce anti-inflammatory cytokines such as interleukin-10 (IL-10).
      • IL-10 has suppressive effects on pro-inflammatory cytokine production and signaling.
  2. Negative Feedback Mechanisms:
    • Inhibition of NF-κB:
      • NF-κB is a central transcription factor that regulates the expression of pro-inflammatory genes.
      • Negative feedback mechanisms, including the induction of inhibitors like IκB (inhibitor of NF-κB), terminate NF-κB signaling, limiting the production of pro-inflammatory cytokines.
    • Decoy Receptors:
      • Some cells release soluble or membrane-bound decoy receptors that bind to cytokines, preventing them from interacting with cell surface receptors.
      • For example, soluble receptors for tumor necrosis factor-alpha (sTNFR) act as decoys, inhibiting TNF-α signaling.
    • Deactivation of Inflammatory Mediators:
      • Enzymes like cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS), which contribute to the production of inflammatory mediators, are downregulated during resolution.
  3. Clearance of Immune Cells:
    • Apoptosis and Efferocytosis:
      • Neutrophils, which are short-lived and highly reactive immune cells, undergo apoptosis (programmed cell death) at the inflammatory site.
      • Macrophages and other phagocytes recognize and engulf apoptotic cells in a process called efferocytosis.
      • Efferocytosis not only clears dying cells but also promotes the resolution of inflammation by dampening pro-inflammatory signals.
  4. Specialized Pro-resolving Mediators (SPMs):
    • Resolvins, Lipoxins, and Protectins:
      • SPMs are lipid-derived mediators that actively promote the resolution of inflammation.
      • Resolvins, lipoxins, and protectins stimulate the clearance of apoptotic cells, limit the recruitment of new immune cells, and enhance tissue repair.
  5. Transforming Growth Factor-beta (TGF-β):
    • Induction of TGF-β:
      • TGF-β is a multifunctional cytokine that plays a key role in tissue repair and resolution.
      • It inhibits inflammation, promotes tissue regeneration, and contributes to the suppression of immune responses.
  6. Restoration of Tissue Homeostasis:
    • Tissue Repair and Remodeling:
      • Fibroblasts are activated to deposit extracellular matrix components, contributing to tissue repair and remodeling.
      • Growth factors released during resolution stimulate tissue regeneration.
  7. Termination of Inflammatory Signals:
    • Resolution of Inflammatory Stimuli:
      • Removal or neutralization of the initial stimuli, such as pathogens or tissue-damaging agents, contributes to the termination of inflammation.
    • Downregulation of PRR Signaling:
      • Expression of PRRs on immune cells may be downregulated, reducing their responsiveness to PAMPs or DAMPs.
  8. Return to Homeostasis:
    • Cessation of Immune Cell Infiltration:
      • Chemokine signals that attract immune cells are downregulated, leading to a decrease in immune cell recruitment.
    • Resolution of Edema:
      • Resolution mechanisms contribute to the clearance of excess fluid and proteins, reducing tissue swelling.

These processes work in concert to resolve inflammation, ensuring that the immune response is appropriately regulated and that tissue function is restored. The return to tissue homeostasis is a finely orchestrated process involving a balance between pro-inflammatory and anti-inflammatory signals, immune cell activation and clearance, and tissue repair mechanisms. Dysregulation of the resolution phase can contribute to chronic inflammatory conditions and tissue damage.

Next Topic: Endotoxin Tolerance to LPS and Other TLR-Recognized PAMPs

Source: ChatGPT response prompted and edited by Joel Graff.

Details Related to the Effector Phase of Inflammation

Vasodilation and Increased Permeability

Several other molecules play key roles in causing vasodilation during inflammation. These molecules act through various mechanisms and are produced by different cells involved in the immune response. Some notable examples include:

  • Prostaglandins:
    • Prostaglandins are lipid mediators derived from arachidonic acid, a fatty acid found in cell membranes.
    • They are produced by various cells, including mast cells and macrophages.
    • Prostaglandins, particularly prostaglandin E2 (PGE2), induce vasodilation and enhance vascular permeability.
  • Bradykinin:
    • Bradykinin is a peptide that is generated during the kinin-kallikrein system activation, often in response to tissue injury.
    • It promotes vasodilation by stimulating the release of nitric oxide (NO) and prostaglandins from endothelial cells.
  • Leukotrienes:
    • Leukotrienes are another group of lipid mediators derived from arachidonic acid.
    • They are produced by various cells, including leukocytes.
    • Leukotrienes, such as leukotriene B4 (LTB4), contribute to vasodilation and increased vascular permeability.
  • Nitric Oxide (NO):
    • Nitric oxide is a gas produced by endothelial cells in response to various stimuli, including cytokines.
    • NO acts as a potent vasodilator by relaxing smooth muscle cells in blood vessel walls.
  • Cytokines (e.g., TNF-α, IL-1):
    • Certain cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-1 (IL-1), can induce vasodilation.
    • These cytokines are produced by immune cells like macrophages and play central roles in initiating and amplifying the inflammatory response.
  • Platelet-Activating Factor (PAF):
    • PAF is a lipid mediator produced by various cells, including platelets and leukocytes.
    • It induces vasodilation and increases vascular permeability.
  • Histamine (from sources other than mast cells):
    • While mast cells are a primary source of histamine during inflammation, other cells such as basophils and certain immune cells can also release histamine.
  • Histamine Release:
  • Histamine has previously been described as a key mediator released by mast cells and basophils in response to various stimuli, such as allergens and helminths.
  • Histamine binds to histamine receptors on the surface of endothelial cells, triggering vasodilation (widening of blood vessels) and increased permeability of capillaries.

Vasodilation allows more blood to flow to the affected area, while increased permeability allows proteins, immune cells, and fluid to move from the bloodstream to the surrounding tissues, facilitating the immune response.

Recruitment of Immune Cells

  • Chemokine Release:
    • Chemokines are small signaling proteins that play a crucial role in directing the movement of immune cells.
    • In response to inflammation, various cells release chemokines to attract immune cells to the site of injury or infection.
    • Neutrophils are among the first immune cells recruited, responding to chemotactic signals released by the damaged tissue or other immune cells.
    • Monocytes are also attracted and later differentiate into macrophages, contributing to the phagocytic activity and resolution of inflammation.

Phagocytosis and Inflammatory Mediator Production

  • Neutrophils and Monocytes:
    • Neutrophils are the most abundant white blood cells and are among the first responders to the site of inflammation.
    • Inflammatory monocytes are recruited to inflamed tissues. This subset of monocytes has several important roles including phagocytosis, antigen presentation, and regulation of the immune response.
  • Phagocytosis:
    • Both tissue resident cells, such as macrophages and dendritic cells, as well as the recruited neutrophils and monocytes engulf pathogens, foreign particles, and cellular debris through a process called phagocytosis.
    • This internalization allows for the destruction and digestion of the engulfed material within specialized vesicles called phagosomes.
  • Inflammatory Mediator Production
    • Upon encountering pathogens, innate immune cells release cytokines and chemokines.
    • Cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukins, play a key role in amplifying the immune response by activating and recruiting other immune cells.
    • Chemokines further guide the migration of immune cells to the site of inflammation.

These effector mechanisms collectively contribute to the acute inflammatory response, which aims to eliminate the cause of cell injury, clear out necrotic cells and tissues damaged from the original insult, and establish a repair process. While these responses are vital for host defense, dysregulation or persistence of inflammation can lead to chronic inflammatory conditions and tissue damage. The coordination and balance of these processes are critical for a well-controlled and effective immune response.

Next Topic: Ending Inflammation and Restoring Tissue Homeostasis

Source: ChatGPT response prompted and edited by Joel Graff.

Signs and Symptoms of Inflammation: Tumor, Rubor, Calor, Dolor, and Functio Laesa

The terms “tumor, rubor, calor, and dolor” are Latin terms that were historically used by the ancient Roman physician Galen to describe the cardinal signs of inflammation. These terms are still relevant in modern medicine and provide a concise way to characterize the clinical manifestations of inflammation:

  1. Tumor (Swelling):
    • Tumor refers to the swelling or enlargement of the affected tissue during inflammation.
    • Increased vascular permeability allows fluid, immune cells, and proteins to accumulate at the site, leading to swelling.
  2. Rubor (Redness):
    • Rubor describes the redness observed in inflamed tissues.
    • Increased blood flow to the affected area (vasodilation) and the accumulation of immune cells contribute to the red appearance.
  3. Calor (Heat):
    • Calor refers to the heat felt in the inflamed region.
    • Vasodilation and increased blood flow bring warm, oxygenated blood to the site of inflammation.
  4. Dolor (Pain):
    • Dolor signifies the pain or discomfort associated with inflammation.
    • Pressure on nerves, release of inflammatory mediators, and tissue damage contribute to the sensation of pain.

Together, these four signs, tumor, rubor, calor, and dolor, provide a clinical framework for recognizing and diagnosing inflammatory conditions.

Additionally, the term functio laesa is often included in discussions of inflammation. “Functio laesa” means “disturbed function” in Latin. In the context of inflammation, it refers to the impairment or alteration of normal tissue function due to the inflammatory process. This sign highlights that inflammation is not merely a set of visible symptoms but also involves functional changes in affected tissues. The disruption of normal function may be temporary or persistent, depending on the severity and resolution of the inflammatory response.

In summary, tumor, rubor, calor, dolor, and functio laesa collectively represent the classical features of inflammation, providing a framework for clinicians to assess and diagnose inflammatory conditions based on observable signs and symptoms.

Next Topic: A Simplified Framework for Describing the Phases of Inflammation

Source: ChatGPT response prompted and edited by Joel Graff.

A Simplified Framework for Describing the Phases of Inflammation

The inflammatory response is a complex and highly regulated process. A simplified framework for discussing inflammatory response is outlined below. Each of the three phases involve a series of coordinated events and interactions between various immune cells, signaling molecules, and tissue components.

  1. Initiation:
    • Recognition of Pathogens or Damage:
      • Immune cells detect PAMPs or DAMPs through PRRs (e.g., TLRs, NLRs).
      • Activation of PRRs triggers the release of inflammatory mediators.
  2. Effector Mechanisms:
    • Vasodilation and Increased Permeability:
      • Cytokines, chemokines, lipid mediators, and histamine release leads to vasodilation and increased permeability of blood vessels.
    • Recruitment of Immune Cells:
      • Chemokines attract neutrophils and monocytes to the site of inflammation.
    • Phagocytosis and Inflammatory Mediator Production:
      • Neutrophils, monocytes, DCs, and macrophages phagocytose pathogens, releasing cytokines and chemokines.
  3. Resolution:
    • Anti-inflammatory Signals and Termination of Inflammation:
      • Production of anti-inflammatory cytokines (e.g., IL-10) counters pro-inflammatory signals.
      • Negative feedback loops inhibit inflammatory pathways.
    • Tissue Repair and Return to Homeostasis:
      • Activation of fibroblasts contributes to tissue repair.
      • Clearance of apoptotic immune cells and debris by macrophages.
      • Restoration of tissue function and return to homeostasis.

Next Topic: Details Related to the Effector Phase of Inflammation

Source: ChatGPT response prompted and edited by Joel Graff.

If Possible, Pathogens Counteract Innate Immune Responses Related to PAMP Recognition

Pathogens have evolved various strategies to defend themselves against the human innate immune system related to the detection of Pathogen-Associated Molecular Patterns (PAMPs) by Pattern Recognition Receptors (PRRs) and the subsequent signal transduction pathways. Here are some mechanisms pathogens use to counteract innate immune responses:

  1. PAMP Concealment or Modification:
    • Mimicry and Modification: Some pathogens can mimic host molecules or modify their PAMPs to make them less recognizable by host PRRs.
    • Glycan Shielding: Pathogens may cover their surface with glycans to evade detection by PRRs.
  2. Inhibition of PRR Activation:
    • Protease Cleavage: Some pathogens produce proteases that can cleave or degrade PRRs, preventing their proper activation.
    • Inhibition of PRR Signaling: Pathogens may interfere with downstream signaling events following PRR activation, hindering the effective transduction of immune signals.
  3. Avoidance of Phagocytosis:
    • Capsule Formation: Some bacteria produce capsules that inhibit phagocytosis by preventing immune cells from binding to and engulfing the pathogen.
    • Inhibition of Opsonization: Pathogens can produce proteins that interfere with opsonization, making it difficult for phagocytes to recognize and engulf them.
  4. Escape from Endosomes:
    • Endosomal Escape: Intracellular pathogens may evolve mechanisms to escape from endosomes, where PRRs like TLRs are typically located, avoiding detection.
    • Avoidance of Lysosomal Degradation: Some pathogens can resist degradation within lysosomes by preventing fusion of phagosomes with lysosomes.
  5. Modulation of Immune Signaling:
    • Inhibition of NF-κB Activation: Pathogens may interfere with the activation of NF-κB, a key transcription factor involved in immune responses downstream of PRR signaling.
    • Manipulation of MAPK Pathways: Pathogens can modulate MAPK pathways to influence cytokine production and other immune responses.
  6. Suppression of Inflammatory Responses:
    • Anti-inflammatory Molecules: Some pathogens produce molecules that actively suppress inflammatory responses, creating an environment conducive to their survival.
    • Induction of Regulatory T Cells: Pathogens may induce the generation of regulatory T cells that dampen immune responses.
  7. Interference with Interferon Signaling:
    • Inhibition of STAT Activation: Pathogens can interfere with the activation of Signal Transducer and Activator of Transcription (STAT) proteins, which play a role in interferon signaling.
    • Production of Viral Proteins with Antagonistic Functions: Viruses may encode proteins that antagonize interferon-mediated antiviral responses.
  8. Antigenic Variation:
    • Rapid Mutation: Some pathogens undergo rapid antigenic variation, changing the structures recognized by the immune system, particularly by PRRs.
    • Switching of Surface Proteins: Bacteria such as Neisseria gonorrhoeae can switch surface proteins to evade recognition by the immune system.
  9. Manipulation of Autophagy:
    • Inhibition of Autophagy: Some pathogens can inhibit the autophagic process, which is involved in the clearance of intracellular pathogens.
    • Subversion of Autophagosome Formation: Pathogens may subvert the formation of autophagosomes, evading capture and degradation.
  10. Interference with Inflammasome Activation:
    • Inhibition of Inflammasome Components: Pathogens can inhibit the assembly or activity of inflammasomes, preventing the processing and release of pro-inflammatory cytokines.

Understanding these evasion mechanisms is crucial for developing targeted approaches to counteract pathogen strategies and enhance the effectiveness of innate immune responses. The ongoing arms race between pathogens and the innate immune system emphasizes the need for continued research and the development of innovative therapeutic strategies.

End of Section 2.2

Next Topic: Signs and Symptoms of Inflammation: Tumor, Rubor, Calor, Dolor, and Functio Laesa

Source: ChatGPT response prompted and edited by Joel Graff.

Examples of Negative Feedback Mechanisms Following Responses to PAMP Recognition

Feedback mechanisms play a crucial role in regulating the strength and duration of signal transduction pathways associated with Pattern Recognition Receptor (PRR) activation and PAMP recognition. These feedback mechanisms are essential for maintaining homeostasis, preventing excessive inflammation, and avoiding damage to host tissues. Here are some examples of feedback mechanisms that influence the strength of these signaling pathways:

  1. Negative Feedback Loops:
    • SOCS Proteins (Suppressors of Cytokine Signaling):
      • SOCS proteins are induced by cytokine signaling downstream of PRR activation.
      • They inhibit JAK-STAT signaling, reducing the intensity and duration of cytokine responses.
  2. Decoy Receptors:
    • Soluble Forms of Receptors:
      • Cells can release soluble forms of receptors that can bind to cytokines or other signaling molecules, preventing them from interacting with cell surface receptors.
      • For example, soluble forms of Toll-like receptors (sTLRs) can act as decoy receptors.
  3. Feedback Inhibition by Anti-inflammatory Cytokines:
    • IL-10 (Interleukin-10):
      • IL-10 is an anti-inflammatory cytokine that is induced during inflammation.
      • It suppresses pro-inflammatory cytokine production and downstream signaling pathways.
  4. Ubiquitin-Mediated Regulation:
    • Ubiquitin Editing:
      • Ubiquitin modifications regulate the stability and activity of signaling molecules.
      • Deubiquitinating enzymes (DUBs) can remove ubiquitin chains, affecting the duration of signaling cascades.
  5. Induction of Feedback Inhibitors by NF-κB:
    • A20 (TNFAIP3):
      • A20 is an NF-κB-responsive gene that encodes a protein with deubiquitinating activity.
      • It inhibits NF-κB activation and downstream signaling pathways.
  6. Inflammasome Regulation:
    • NLRP3 Inflammasome Feedback Inhibition:
      • Activation of the NLRP3 inflammasome leads to the production of IL-1β.
      • IL-1β itself can induce the expression of negative regulators, such as IL-1 receptor antagonist (IL-1RA), providing feedback inhibition.

These feedback mechanisms are a small subset of examples that contribute to the fine-tuning of immune responses, preventing uncontrolled inflammation and maintaining immune homeostasis. The balance between pro-inflammatory and anti-inflammatory signals is crucial for an effective yet controlled immune response.

Next Topic: If Possible, Pathogens Counteract Innate Immune Responses Related to PAMP Recognition

Source: ChatGPT response prompted and edited by Joel Graff.

Cellular Responses Downstream of PAMP Recognition

Upon binding to PAMPs, PRRs initiate signal transduction pathways that lead to various cellular responses, including the production of inflammatory mediators, activation of immune cells, and establishment of an antiviral state. Here are some commonly studied signal transduction pathways downstream of PRR activation:

  1. Toll-Like Receptor (TLR) Signaling Pathways:
    • TLRs are a family of PRRs located on the cell surface (e.g., TLR1, TLR2, TLR4) or within endosomes (e.g., TLR3, TLR7, TLR9).
    • MyD88-dependent Pathway:
      • Most TLRs signal through MyD88, leading to the activation of NF-κB and the production of pro-inflammatory cytokines.
    • TRIF-dependent Pathway:
      • TLR3 and TLR4 can also signal through TRIF, leading to type I interferon production.
  2. NOD-Like Receptor (NLR) Signaling:
    • NLRs are cytoplasmic PRRs that form inflammasomes upon activation.
    • Inflammasome Activation:
      • Inflammasomes, composed of NLRs, ASC, and caspase-1, activate pro-inflammatory cytokines (IL-1β, IL-18) and induce pyroptotic cell death.
  3. RIG-I-like Receptor (RLR) Signaling:
    • RLRs, including RIG-I and MDA5, detect viral RNA in the cytoplasm.
    • Activation of MAVS:
      • RLR binding to viral RNA leads to the activation of mitochondrial antiviral signaling protein (MAVS), triggering type I interferon production.
  4. C-type Lectin Receptor (CLR) Signaling:
    • CLRs recognize carbohydrates on pathogens.
    • Syk-dependent Pathways:
      • Many CLRs signal through the Syk kinase, activating downstream pathways that induce cytokine production.
  5. Cytosolic DNA Sensing:
    • Various sensors detect cytosolic DNA, such as cGAS (cyclic GMP-AMP synthase).
    • STING Pathway:
      • cGAS produces cGAMP, activating the Stimulator of Interferon Genes (STING) pathway, leading to type I interferon production.
  6. Downstream Effector Molecules:
    • NF-κB Activation:
      • Common downstream signaling pathway leading to the transcription of pro-inflammatory genes.
    • MAPK Pathways:
      • Activation of mitogen-activated protein kinase (MAPK) pathways, including ERK, JNK, and p38, influencing inflammatory responses.
    • Interferon Regulatory Factor (IRF) Activation:
      • Many PRRs lead to the activation of IRFs, contributing to the induction of interferons.

Understanding these signaling pathways is critical for elucidating the innate immune response and developing targeted therapies for infectious and inflammatory diseases. The crosstalk and regulation within these pathways ensure a finely tuned and coordinated immune response.

Next Topic: Examples of Negative Feedback Mechanisms Following Responses to PAMP Recognition

Source: ChatGPT response prompted and edited by Joel Graff.

Diversity of PRRs and the Types of PAMPs They Detect

Pattern recognition receptors (PRRs) are a diverse group of proteins that play a crucial role in detecting conserved molecular patterns associated with various pathogens. Here are different families of PRRs and the types of infectious diseases they recognize:

  1. Toll-Like Receptors (TLRs):
    • Recognition: TLRs recognize a variety of microbial components, including bacterial lipopolysaccharide (LPS), peptidoglycan, flagellin, viral dsRNA, and CpG DNA motifs.
    • Infectious Diseases: Recognize a broad range of pathogens, including bacteria, viruses, fungi, and parasites.
  2. NOD-Like Receptors (NLRs):
    • Recognition: NLRs often sense intracellular pathogens or cellular stress and damage, recognizing components like bacterial peptidoglycan and viral RNA.
    • Infectious Diseases: Involved in the detection of intracellular pathogens such as bacteria and viruses.
  3. RIG-I-Like Receptors (RLRs):
    • Recognition: RLRs, including RIG-I and MDA5, recognize viral RNA, especially dsRNA.
    • Infectious Diseases: Primarily involved in the detection of RNA viruses.
  4. C-Type Lectin Receptors (CLRs):
    • Recognition: CLRs recognize various carbohydrate structures, including β-glucans, mannans, and other fungal cell wall components.
    • Infectious Diseases: Important for the detection of fungal pathogens and some bacterial pathogens.
  5. AIM2-Like Receptors (ALRs):
    • Recognition: ALRs, including AIM2, detect cytoplasmic DNA, especially dsDNA.
    • Infectious Diseases: Involved in the detection of intracellular pathogens, including some DNA viruses and certain bacteria.
  6. Cyclic GMP-AMP Synthase (cGAS):
    • Recognition: cGAS recognizes cytoplasmic DNA and catalyzes the synthesis of cyclic GMP-AMP (cGAMP), leading to the activation of the STING pathway.
    • Infectious Diseases: Involved in the detection of DNA viruses, bacteria, and parasites.
  7. Nucleotide-Binding Oligomerization Domain (NOD):
    • Recognition: NOD proteins, part of the NLR family, recognize bacterial peptidoglycan components.
    • Infectious Diseases: Important for detecting bacterial pathogens.
  8. Retinoic Acid-Inducible Gene I (RIG-I) and Melanoma Differentiation-Associated Gene 5 (MDA5):
    • Recognition: RIG-I and MDA5 recognize viral RNA, particularly in the cytoplasm.
    • Infectious Diseases: Primarily involved in the detection of RNA viruses.

These families of PRRs collectively contribute to the immune system’s ability to recognize a wide range of pathogens, including bacteria, viruses, fungi, and parasites. The specificity of each family allows the immune system to distinguish between different types of infectious agents and mount appropriate responses.

Next Topic: Cellular Responses Downstream of PAMP Recognition

Source: ChatGPT response prompted and edited by Joel Graff.

Becoming a Danger to One’s Self

Damage-Associated Molecular Patterns (DAMPs) are endogenous molecules that are typically found within cells but become exposed or released into the extracellular environment following cellular damage, stress, or injury. These molecules are recognized by the immune system as signals of cell damage or danger. Here are some examples of DAMPs:

  1. HMGB1 (High Mobility Group Box 1):
    • Source: A nuclear protein that can be released from cells during necrosis, apoptosis, or cellular stress.
    • Function: HMGB1 acts as a DAMP when outside the cell, signaling danger and promoting inflammation.
  2. ATP (Adenosine Triphosphate):
    • Source: Normally found inside cells as an energy carrier.
    • Function: Released into the extracellular space during cell damage, ATP acts as a DAMP and can trigger inflammation and immune responses.
  3. Heat-Shock Proteins (HSPs):
    • Source: Intracellular proteins that play a role in cellular stress responses.
    • Function: Certain HSPs, when released or exposed on the cell surface, can act as DAMPs, signaling cellular stress and eliciting immune responses.
  4. Uric Acid:
    • Source: Normally present in cells as a metabolic byproduct.
    • Function: Released during cell injury, necrosis, or inflammation, uric acid can act as a DAMP, contributing to the inflammatory response.
  5. S100 Proteins:
    • Source: A family of calcium-binding proteins.
    • Function: Some S100 proteins are released from damaged or stressed cells and act as DAMPs, contributing to inflammatory responses.
  6. DNA and RNA Fragments:
    • Source: Nucleic acids normally found within cells.
    • Function: Fragments of DNA and RNA released during cell damage or death can act as DAMPs, triggering immune responses.
  7. Mitochondrial Components:
    • Source: Mitochondria contain various molecules that can be released during cellular stress or damage.
    • Function: Mitochondrial DAMPs, such as mitochondrial DNA or formyl peptides, can activate immune responses when released into the extracellular space.

Detection in Normal Cells: These DAMPs are typically not detected in normal, healthy cells because they are sequestered within the cell’s intracellular compartments. Normal physiological conditions involve a tightly regulated internal environment where cellular components remain contained within the cell membranes. DAMPs become exposed or released under conditions of cellular stress, damage, or pathological events, triggering the immune system to respond to these signals of danger.

The immune system is designed to recognize and respond to signals of danger or abnormality. When cells are healthy and functioning normally, DAMPs remain hidden within cells, and the immune system does not mount an unnecessary response against them. It is only under conditions of cellular stress or damage that DAMPs become apparent and contribute to immune activation.

Next Topic: Diversity of PRRs and the Types of PAMPs They Detect

Source: ChatGPT response prompted and edited by Joel Graff.

Molecular Patterns Recognized by Innate Immune Receptors

Here are examples of PAMPs/MAMPs associated with various types of human pathogens that can be detected by the immune system:

  1. Bacteria:
    • Lipopolysaccharide (LPS): Found in the outer membrane of Gram-negative bacteria.
    • Peptidoglycan: A major component of bacterial cell walls, present in both Gram-positive and Gram-negative bacteria.
    • Flagellin: Protein component of bacterial flagella.
    • Unmethylated CpG DNA: Recognized as a PAMP when present at high frequencies in bacterial DNA.
  2. Viruses:
    • Double-stranded RNA (dsRNA): A viral replication intermediate and a potent PAMP.
    • Single-stranded RNA (ssRNA): Viral genomes often contain ssRNA, which can be detected by the immune system.
    • CpG DNA Motifs: Certain DNA motifs in viral genomes, particularly those containing CpG dinucleotides, can act as PAMPs.
  3. Fungi:
    • β-Glucans: Major components of fungal cell walls, recognized by immune cells as PAMPs.
    • Zymosan: A component of the cell wall of some fungi, particularly Saccharomyces cerevisiae, acting as a PAMP.
    • Chitin: A polysaccharide found in the cell walls of fungi and exoskeletons of arthropods, recognized as a PAMP.
  4. Parasites:
    • Glycosylphosphatidylinositol (GPI): A component of the membranes of some parasites, recognized as a PAMP.
    • Parasite DNA/RNA: Nucleic acids from parasites can serve as PAMPs when detected by the immune system.
    • Parasite-Specific Proteins: Certain proteins produced by parasites can act as PAMPs.

Next Topic: Becoming a Danger to One’s Self

Source: ChatGPT response prompted and edited by Joel Graff.