You are on page 1of 45

RANGKUMAN IMMUNOLOGY 7

Immunity to Microbes and Immunodeficiency

Immunity to Microbes
Overview of Immune Responses to Microbes

● Disease
- short lived: host response effective
- Chronic: response not adequate
● Defense against microbes→ mediated by effector mechanism of innate
and adaptive immunity
- Innate: early defense
- Adaptive: lebih kuat dan berkelanjutan → protection pada
paparan ulang
● Many pathogen→ resist innate immunity → protection dependent on
adaptive immunity
● Immune system→ responds in specialized ways to different type of
microbe
● Survival and pathogenicity→ bergantung pada ability to evade or
resist the effector mechanism of immunity
● Latent or persistent→ immune response controls but does not
eliminate. Ex: DNA virus→ herpes and poxvirus, bacteria→ M
tuberculosis (survive in phagocytic vesicles) → become activated when
immune system weakened
● Analysis of immune response → uji klinis infeksi
Helpful for
- Microbe cannot be cultured
- Not detectable in blood
- Present in tissues that are not accessible
Contoh
- IgM→ recent infection. IgG→ past infection
- Test for skin reaction → assay for T cell responses. Ex: tuberculin
test

Immunity to Extracellular Bacteria

● Extracellular bacteria → bisa replikasi di luar host cells. Ex: di blood, connective tissue, lumen
● Disease → caused by 2 principal mechanism
1. Bacteria induce inflammation → injury in site of infection
2. Bacteria produce toxin → ex:
- endotoxin (LPS in cell wall of gram -) → stimulate production of cytokine
- exotoxin (secreted) → cytotoxic and kill host cell.
a. diphtheria toxin → shut down protein synthesis in infected cells.
b. Cholera toxin→ interferes w ion and water transport
c. tetanus toxin→ inhibit neuromuscular transmission.
d. Anthrax toxin: disrupt biochemical signaling pathway

Innate Immunity to Extracellular Bacteria Adaptive Immunity to Extracellular Bacteria


a. Activation of phagocytes and inflammation → major defense
EB efficiently killed by phagocytes→ karena EB tidak bisa bertahan di
dalam sel
1. tissue-resident DC and macrophage→ activated by microbes→
secrete cytokine → promote leukocyte infiltration
2. Neutrophil and monocyte-derived macrophage→ use receptor
to recognize EB
a. Mannose and scavenger receptor→ promote phagocytosis
b. TLR→ promote microbicidal activities Humoral immunity→ major protective immune response
c. Fc and complement receptor→ promote phagocytosis to EB
b. Complement activation ● Antibody
- bacteria express mannose – mannose binding lectin/ N-acetyl – - polysaccharide→ T-independent → Ex: S,
ficolin → activate complement pathway pneumoniae, neisseria
- Opsonization→ enhance phagocytosis of bacteria - Spleen: production of antibody and
- MAC complex → lyses bacteria. Ex: Neisseria (thin cell walls) phagocytic clearance of opsonized
bacteria→ pada indiv splenectomy ↑ infection
by encapsulated bacteria
Mekanisme antibodi
- Neutralization; high affinity IgG, IgM, IgA
- Opsonization: IgG1 and IgG3
- Complement activation: IgM, IgG1, IgG3
● Protein antigen of EB→ activate CD4+ Thelper
- Produce cytokine

Injurious Effect of Immune Responses to Extracellular Bacteria

● Inflammation and Sepsis


1. Sepsis ● Superantigen → bind to TCR and MHC II (not on
- Consequence of severe infection by bacteria and fungi peptide binding cleft) → activate many more clones
- Septic shock: most severe and fatal → circulatory collapse of T cells → activate many T cells → large amount
(shock) of cytokine → sepsis-like systemic inflammatory
- Early phase: response syndrome
a. Caused by cytokine (macrophage→ LPS and
peptidoglycan)
b. TNF, IL-6, IL-1→ sitokin utama
c. Early burst of large amounts of cytokine→ cytokine
storm
Late complication of humoral immune response
● Poststreptococcal glomerulonephritis→ antibody-antigen complexes
deposited in kidney glomeruli

Immune Evasion by Extracellular Bacteria


● Bacteria w polysaccharide-rich capsules→ resist phagocytosis
● Sialic acid in capsules→ inhibit complement activation by alternative
pathway
● Variation of surface antigen
- Gonococci and E. coli→ pili→ major antigen: protein pilin → pilin
genes undergo extensive gene conversion → evade attack by
pilin-specific antibodies
- chemical alteration in surface oligosaccharides→ sialylation
- Rilis surface antigen→ buat membrane blebs→ antibodies
dibuang dari tubuh mikroba

Immunity to Intracellular Bacteria

Innate Immunity to Intracellular Bacteria Adaptive Immunity to Intracellular Bacteria

● Mainly by phagocytes and NK cells ● Mainly by Cell-mediated Immunity


Phagocyte ● HIV→ deficient in cell-mediated immunity→
● Phagocyte→ early: neutrophil, late: macrophage→ ingest these extremely susceptible to IB
microbes → but IB resistant to degradation within phagocytes ● 2 tipe reaksi sel T
● Reseptor a. CD4+Th1→ activate phagocytes (CD40L and
- TLR and NLR → recognize product of bacteria → activation of IFN-y) → secrete IFN-y (produce NO, ROS),
phagocytes TNF (recruit other leukocyte)
- CDS (cystosolic DNA receptor) --> STING pathway→ IFN type 1 b. CD8+T cells→ eliminating microbes that
responses escape phagocytes (ex: Listeria creates holes
NK cell in phagosomal membrane→ bacteria escape
● Intracellular bacteria→ activate NK by induce expression of NK into cytosol)
cell-activating ligand + stimulate production IL-12 dan IL-15 (NK
cell-activating cytokines)
● NK cell→ produce IFN-y → activate phagocytes →killing of
phagocytosed bacteria

Injurious Effect

● Macrophage activating→ DTH (delayed type hypersensitivity) → injury


● IB→ resisten pada phagocyte→ bertahan lama→ chronic T cell and
macrophage activation → formation granuloma surrounding microbe
(histologic hallmark for IB infection)--> prevent spread of microbe →
severe functional impairment bcs of tissue necrosis and fibrosis

Immune Evasion by Intracellular Bacteria


● Inhibit phagolysosome fusion
● Escaping into the cytosol
● Directly inactivating microbicidal substances. Such as ROS

Immunity to Fungi

Innate Immunity

● Major: phagocytes and the complement system


Phagocyte
- Recognize by: TLR & dectin and mannose receptor
- Neutrophil→ extracellular fungi
- Neutropenia→ disseminated fungal infection
- Neutrophil→ fungicidal substances→ ROS and lysosomal enzymes
Complement
- Complement activation → defense fungi that enter the bloodstream.
Ex: candida
- Fungi→ activate alternative/ lectin complement pathway
- Complement→ opsonize fungi → but not form MAC (bcs fungi have
thick walls)

Adaptive Immunity to Fungi Immune Evasion by Fungi

● Peran penting: Th17 ● Little is known


1. Fungi→ recognize by CLR (C- type lectin receptor) and TLR ● Virulent strain of C. neoformans→ inhibit
2. Stimulate DCs and macrophage to produce cytokine→ promote production of cytokine (TNF and IL-12 by
Th17 development (IL-1, IL-6, IL23) macrophage) and ↑ IL-10→ inhibit macrophage
● Th1-mediated cellular immunity→ against intracellular fungi infection activation
-Histoplasma capsulatum (facultative intracellular) → lives in
macrophage
● Th2→ ineffective at clearing fungi and may cause harm
- Aspergillus infection of airways→ Th2 reaction→ reaksi alergi →
bronchopulmonary aspergillosis
Immunity to Viruses

Innate Immunity to Viruses

● Associated with production of IFN type 1 by infected cells,


plasmacytoid DCs, and macrophages
● Pathways trigger IFN production :
a. recognition of viral RNA by endosomal TLRs → activation of
cytoplasmic RIG-like receptors
b. recognition of viral DNA by endosomal TLRs → activation of
STING pathway
c. activation of protein kinases → activate IRF transcription
factors → stimulate IFN gene transcription → inhibit viral
replication in both infected and uninfected cells & production
of restriction factors
NK cells kill virus-infected cells early in the course of infection
before adaptive immune responses have developed
● viral infection inhibits class I MHC expression on infected cells
+ induces expression of additional activating ligands → NK cell
inhibitory receptor is not engaged & activating receptor trigger
responses of NK cells

Adaptive Immunity to Viruses

Mediated by :
● CTLs → killing infected cells
● Antibodies → block virus binding and entry

Antibodies
● Effective only during the extracellular stage
● Antiviral antibodies bind to viral envelope / capsid antigens →
neutralizing antibodies → prevent virus attachment and entry
into host cells
● Antibodies opsonize viral particles → promote clearance by
phagocytes
CTLs
● Most virus-specific CTLs are CD8+ T cells that recognize
cytosolic, viral peptides presented by class I MHC molecules
● Programmed cell death
● Activation of nucleases within infected cells → degrade viral
genomes
● Cytokines (IFN-γ) → activates phagocytes

There are some conditions in adaptive immune response : ● CTLs caused tissue injury
● Latent infections → viral DNA exist in cell (host immune e.g. Hepatitis B virus, lymphocytic choriomeningitis virus (LCMV)
response controls but does not eliminate the infection) →
recurrent infection during immunocompromised
e.g. shingles – latent varicella, cold sores – latent HSV

Immune Evasion by Viruses

Antigenic Variation Inhibit the ability to induce antiviral state


e.g. SARS-CoV, MERS-CoV, and SARS-CoV-2
● Affected most commonly surface glycoproteins
● Principal mechanisms : point mutations & reassortment of ● shut off the host type I IFN response
RNA genomes (in RNA viruses) → antigenic drift and antigenic ● modify viral RNA (by adding 7-methyl guanosine cap & 2’O-methy
shift group to the ribose in the next nucleotide) → prevents recognition
a. Antigenic drift → mutations in the genes that encode by RIG-I and MDA-5
surface proteins, then the variation occurs
b. Antigenic shift → reassortment of viral genes results in
major changes in antigenic structure which creates distinct
viruses
Inhibition of MCH class I

● Viruses make a variety of proteins that


block different steps in antigen
processing, transport, and presentation
→ blocks the assembly and expression
of class I MHC → cannot be recognized
or killed by CD8+ CTLs.

Inactivation of CTL Cause “exhaustion” of CTL


e.g. Poxviruses
● encode molecules secreted by infected ● chronic viral infections → persistent
cells → bind to several cytokines (IFN-γ, antigen stimulation → upregulation of
TNF, IL-1, IL-18, and chemokines) → may PD-1 transcription factor
function as competitive antagonists of
the cytokines
e.g. Epstein-Barr virus Inactivation of immunocompetent cells
● production of IL-10 like protein → e.g HIV → infecting and eliminating CD4+ T
inhibits activation of macrophages & cells
DCs → suppress cell-mediated
immunity
Immunity to Parasites
Parasitic infection refers to infection with animal parasites such as protozoa, helminth, and ectoparasites (e.g ticks and mites)

Innate Immunity to Parasites

● Principal to protozoa → phagocytosis


● Some protozoa express surface molecules that are recognized by TLRs and activate phagocytes
e.g. Plasmodium species, Toxoplasma gondii, and Cryptosporidium species express glycolipids → activate TLR2 and TLR4

● Principal to helminths → eosinophils, alternative pathway of complement

Adaptive Immunity to Parasites

● Principal to protozoa that survive within macrophages →


cell-mediated immunity (macrophage activation by Th1
cell–derived cytokines)

● Principal to helminthic infections → mediated by the activation


of Th2 cells
● Helminths → naive CD4+ T cells to Th2 cells → secrete :
a. IL-4 → production of IgE → binds to the Fcε receptor of mast
cells
b. IL-5 → activates eosinophils
Immune Evasion by Parasites

● Parasites evade by reducing immunogenicity & inhibiting host Protozoan conceal themselves from the immune system
immune responses ● either by living inside host cells or developing cysts

Change surface antigens during life cycle in vertebrate hosts


● stage-specific change in antigen expression → mature tissue Parasites inhibit host immune responses by multiple mechanisms
stages produce antigens different from the infective stages ● T cell anergy (severe schistosomiasis & filarial infections)
● e.g. infective sporozoite stage of Plasmodium is antigenically ● lymphatic filariasis in lymph nodes → deficient immunity
distinct from merozoites that reside in the host and responsible ● Leishmania → stimulate the development of Tregs → suppress the
for chronic infection, Trypanosoma brucei and Trypanosoma immune response
rhodesiense ● Production of immune suppressive cytokines

Become resistant to immune effector mechanisms during


residence in vertebrate hosts
● e.g. schistosome larvae (develop a tegument) → resistant to
complement and CTLs
Strategies for Vaccine Development

● Fundamental principle → administer a killed or attenuated ● Most vaccines in use today work by inducing humoral immunity →
form of an infectious agent, or a component of a microbe, that the best vaccines are stimulate development of long-lived plasma
does not cause disease but elicits an immune response that cells that produce high-affinity antibodies and memory B cells
provides protection against infection ● There are major challenges in developing effective vaccines → the
● Vaccines most effective if the infectious agent does not immunologic correlates of protection are often poorly defined and
establish latency, undergo antigenic variation, interfere with fundamental questions about how to maximally stimulate durable
host immune response, and limited to human hosts memory, effective T cells, and long-lived plasma cells remain
unresolved

Attenuated and Inactivated Bacterial and Viral Vaccines

● Some of the earliest (first generation) and most effective


● Advantage → elicit many of innate and adaptive immune responses (both humoral and cell-mediated) → the ideal way of inducing
protective immunity
● The attenuated or killed bacterial vaccines currently in use generally induce limited protection and are effective for only short periods
● Live, attenuated viral vaccines are usually more effective → polio, measles, yellow fever
● Viral vaccines often induce long-lasting specific immunity
● The major concern is safety → vaccine-induced disease

Purified Antigen (Subunit) Vaccines

● Composed of antigens purified from microbes / inactivated ● Purified protein vaccines stimulate helper T cells and antibody
toxins and usually administered with an adjuvant → responses, but they do not generate potent CTLs → exogenous
prevention of diseases caused by bacterial toxins proteins (and peptides) usually enter the class II MHC pathway of
● Diphtheria and tetanus are two infections whose antigen presentation → vaccines are not recognized efficiently by
life-threatening consequences have been largely controlled class I MHC–restricted CD8+ T cells (except in the special situation
because of immunization of children with toxoid preparations of cross-presentation)
● Vaccines composed of bacterial polysaccharide antigens are
used against Pneumococcus and Haemophilus influenzae
● Polysaccharides (T-independent antigens) → tend to elicit
low-affinity antibody responses
● High-affinity generated by coupling to proteins to form
conjugate vaccines → elicit helper T cells to simulate germinal
center reactions, which would not occur with simple
polysaccharide vaccines
e.g. H. influenzae, pneumococcal, and meningococcal vaccines
Synthetic Antigen Vaccines

● A goal of vaccine research has been to identify the most ● It is possible to deduce the protein sequences of microbial antigens
immunogenic microbial antigens or epitopes, to synthesize from nucleotide sequence data and to prepare large quantities of
these in the laboratory, and to use the synthetic antigens as proteins by recombinant DNA
vaccines. ● Vaccines made of recombinant DNA–derived antigens are now in
use for hepatitis B virus and HPV

Live Viral Vaccines Involving Recombinant Viruses

● Introduce genes encoding microbial antigens into a ● A potential problem with recombinant viruses is that the viruses
noncytopathic virus → virus serves as a source of the antigen in may infect host cells, and even though they are not pathogenic,
an inoculated individual they may produce antigens that stimulate CTL responses that kill
● Advantage of viral vectors : like other live viruses, induce the the infected host cells
full complement of immune responses, including strong CTL
responses

DNA Vaccines mRNA Vaccines

● Inoculation of a plasmid containing complementary DNA ● Uses messenger RNA (mRNA) encoding microbial antigens
(cDNA) encoding a protein antigen leads to humoral and ● The main advantages : can be rapidly developed, the ability to
cell-mediated immune responses to the antigen bypass the need for the large-scale manufacture and purification of
● Bacterial plasmids are rich in unmethylated CpG nucleotides protein antigens, the ability to combine mRNAs encoding many
→ recognized by TLR9 in DCs and other cells → could be different protein antigens from a pathogen into a single vaccine
effective even when administered without adjuvants

Adjuvants and Immunomodulators


● The initiation of T cell–dependent immune responses against ● Approved for patients :
protein antigens requires adjuvants → elicit innate immune a. Aluminum hydroxide gel (promote mostly B cell responses)
responses → increased expression of costimulators and b. Bacterial product, monophosphoryl lipid A, alone or with
cytokines, such as IL-12, that stimulate T cell growth and aluminum salt
differentiation c. Lipid formulation called squalene (activate phagocytes)
● Heat-killed bacteria are powerful adjuvants that are commonly d. CG-rich oligonucleotides (CpG DNA) (for hepatitis B vaccines)
used in experimental animals → activating TLR9 → elicit potent innate immune reactions

Passive Immunization

● Most commonly used for rapid treatment of potentially fatal


diseases caused by toxins, such as tetanus, and for protection
from rabies, hepatitis, and SARS-CoV-2
● Convalescent plasma has been used in cases of Ebola and
COVID-19.
● Recombinant monoclonal neutralizing antibodies are now
utilized as a therapy for COVID-19
● Short-lived
● Does not induce memory
Primary and Acquired Immunodeficiencies
● The primary immunodeficiencies are genetic defects that ● Secondary, or acquired, immunodeficiencies are not inherited
result in an increased susceptibility to infection that is diseases but develop as a consequence of malnutrition, disseminated
frequently manifested in infancy and early childhood but cancer, treatment with immunosuppressive drugs, or infection of cells
is sometimes first clinically detected later in life of the immune system

Overview of Immunodeficiency Diseases

● The principal consequence of immunodeficiency is


increased susceptibility to infection → opportunistic
microorganism
● Deficient humoral immunity → infection by encapsulated,
pus-forming bacteria and some viruses
● Defects in cell-mediated immunity → infection by viruses,
other intracellular microbes / reactivation of latent
infections
● Patients with immunodeficiencies are also susceptible to
certain types of cancer
Primary (Congenital) Immunodeficiencies

● Primary immunodeficiencies → monogenic disease, caused ● Expression, clinical manifestation from same mutation may
by germline mutations in genes be variable
● Earliest → X-linked recessive disease, majority → autosomal ❖ Contributed by phenotypic variability → coinheritance of
recessive modifier genes, environmental factos, epigenetic
● Often seen in consanguineous families (same mutation modifications
inherited from both parents) ● Diagnosis of primary immunodeficiencies made from:
● Other cases in nonconsanguineous → one defective allele of measurement of serum Ig levels, flow cytometry of immune
a specific gene from parent A, and different defective cells, assessment of neutrophil function in vitro
mutation in the same gene from parent B → compound ● In the US → required for newborns to be screened → looking
heterozygotes for DNA that is deleted during TCR gene rearrangement →
● Some primary immunodeficiency from autosomal dominant, failure to detect these DNA: absence of T cell development
include gain-of-function mutations & loss-of function ❖ Early diagnosis → early correction of defect by
mutations (impaired function but not complete loss of hematopoietic stem cell transplantation
protein) ● Defects in one mechanism → individuals susceptible to only
Example: some infections
❖ Activating mutations in PIK3CD (encodes hyperactive PI3 ● Certain immunodeficiencies → associated with autoimmunity
kinase δ (gain-of-function) ❖ Mutation → changes in regulatory mechanism, or
❖ Mutation in CTLA4 → haploinsufficiency ❖ Persistent infection → tissue injury → activation of
(loss-of-function) autoreactive lymphocytes

Defects in Innate Immunity

● Innate immunity → first line of defense → congenital disorders leads to recurrent infections
● Includes: NK cell deficiencies, defects in TLR signaling & in IL-12/IFN-y pathway
● Phagocyte defects → infections of the skin & respiratory tract with bacteria & fungi, e.g. Aspergillus & Candida; deep-seated
abscess & oral stomatitis
● TLR & type 1 IFN signaling defects → recurrent pyogenic infections, severe viral infections
● IL-12 & IFN-y pathway defects→ intracellular pathogens (mycobacterial)

Defective Microbicidal Activity of Phagocytes: Chronic Granulomatous Disease (CGD)

● ⅔ (mostly) → X-linked recessive, remainder autosomal ● Leading cause of death → invasive Aspergillus infections
recessive ● Organism produce catalase (destroys the hydrogen peroxide
● CGD cause by mutations in phagocyte oxidase (PHOX) produced by host cells) → in CGD, neutrophils cannot
enzyme complex accommodate this situation → chronic cell-mediated immune
❖ X linked form → mutation in gene encoding 91-kD α response → T cell-mediated macrophage activation →
subunit of cytochrome b558/PHOX-91 granulomas from activated macrophages
= defective production of superoxide anion (ROS ● Cytokine IFN-y → ↑ transcription of gene encoding PHOX-91
species), major microbicidal mechanism of phagocytes, → ↑ production of superoxide → therapy to CGD
esp neutrophils → failure to kill phagocytosed microbes
● CGD → recurrent infections with fungi & bacteria, e.g.
Staphylococcus

Leukocyte Adhesion Deficiencies

● A group of autosomal recessive disorders → defects in Leukocyte adhesion deficiency type 1 (LAD-1)
leukocyte & endothelial adhesion molecules
● Failure of leukocyte (particularly neutrophil) recruitment to ● Cause: defect in adhesion-dependent functions of
site of infection = severe periodontitis, recurrent bacterial leukocytes (adherence to endothelium, neutrophils
infections, inability to make pus aggregation & chemotaxis, phagocytosis, cytotoxicity by
neutrophils, NK cells, T lymphocytes)
● Defect: mutation in CD18 gene → absent/reduced
expression of β2 integrins (leukocyte function-associated
antigen 1, LFA-1/CD11aCD18; MAC-1/CD11bCD18;
p150,95/CD11cCD18) → for adhesion of leukocytes to other
cells, binding of T lymphocytes to APCs
● Manifestation:
❖ Recurrent bacterial & fungal infection
❖ Impaired wound healing
❖ Delayed umbilical cord separation (normally occurs
because inflammation and neutrophil infiltration)
❖ Leukocytosis

Leukocyte adhesion deficiency type 2 (LAD-2)

● Cause: defective attachment of leukocytes to endothelium,


absence of leukocyte rolling → defective recruitment
● Defect:
❖ Mutation in guanosine diphosphate (GDP)-fucose
transporter (responsible of fucose transport into Golgi)
→ absence of sialyl Lewis X (carbohydrate ligand on
leukocytes, required for binding to E-selectin & P-selectin
on cytokine-activated endothelium)
● Manifestation:
❖ Recurrent infections
❖ Leukocytosis
❖ Development abnormalities (nonimmunological
manifestations related to defective fucose metabolism →
“congenital disorder of glycosylations”) (CDG)
*abnormality in fucosylation → contributes to Bombay blood
group phenotype
❖ Lack of fucosylated H glycan that forms the core of A, B,
O blood group antigens

Leukocyte adhesion deficiency type 3 (LAD-3)

● Cause: defect in adhesion-dependent functions of


leukocytes, platelet aggregation
● Defect:
❖ inside-out signaling pathway that mediates
chemokine-induced integrin activation → required for
leukocytes-endothelium binding & platelet aggregation;
❖ mutations in gene KINDLIN-3 (protein that binds to
cytoplasmic tail of integrin → involved in signaling)
● Manifestation:
❖ Repeated bacterial infections
❖ Delayed umbilical cord separation
❖ Bleeding disorder, life-threatening (defective platelet
aggregation

Defects in NK Cells and Phagocytes

Autosomal dominant mutations Subject: Gene encoding GATA2 transcription factor (key roles in hematopoietic development)
→ diminished precursor populations in bone marrow
Impact: loss of NK, ↓ monocytes, DC, B cells

Autosomal recessive mutations Subject: DNA Helicase MCM4 (minichromosome maintenance complex component 4)
Impact: loss of NK (subset CD56), adrenal insufficiency & growth retardation

Subject: CD16 (FCyRIIIA) → Fc receptor that mediates antibody-dependent cell-mediated


cytotoxicity
Impact: loss of NK function = severe infections from viruses, mainly herpesvirus & papillomavirus

Chediak-Higashi syndrome

Subject: Gene encoding protein LYST → regulates intracellular trafficking of lysosomes


Manifestation: formation of giant lysosomes in neutrophils, monocytes, and lymphocytes;
defect in chemotaxis & phagocytosis, impaired NK function (abnormality in cytoplasmic granules)
Impact:
❖ Defective phagosome-lyososome fusion in neutrophils & macrophages = reduced
resistance to infection
❖ Defective melanosome formation in melanocytes = albinism
❖ Lyososmal abnormalities in nervous system cells = nerve defects & platelets = bleeding
disorders
❖ Recurrent infections by pyogenic bacteria
❖ Infiltration of various organ by nonneoplastic lymphocytes
❖ Leukopenia

Inherited Defects in TLR Pathways, NF-kB Signaling, and Type 1 IFNs

● TLR mutation → herpes simplex encephalitis Mutations in MyD88 & IRAK4 → severe invasive bacterial
● Almost all virus → generate double-stranded RNA (dsRNA) infections in early life, esp. pneumococcal pneumonia
transcript, recognized by TLR3
● Major signaling pathway for TLRs & IL-1 receptor → involves
MyD88 adaptor & IRAK-4 and IRAK-1 kinases → result in
NF-kB dependent induction of inflammatory cytokines Autosomal recessive mutations in TRIF, autosomal dominant
● TLR signaling → use TRIF adaptor protein & TBK1 mutations in TRAF3 E3 ligase, autosomal dominant mutations
(serine-threonine kinase, functions in downstream of TRIF to in TBK1 → susceptibility to herpes simplex encephalitis
activate IRF3 & NF-kB)
● TLR 3, 7, 8, 9 in endosome → recognize nucleic acids → Homozygous mutations in UNC93B → reduced type 1 IFN →
require UNC93B protein (endoplasmic reticulum membrane susceptibility to herpes simplex enxephalitis
protein, deliver newly synthesized TLRs to endosome)
● Signaling downstream of endosomal TLRs → synthesis & Loss-of function STAT1 mutations → severe viral infections
secretion of type 1 IFN → activate STAT1 (signal transducer &
activator of transcription 1) transcription factor

Point mutations in IKKy (inhibitor of kB kinase y)/NEMO (NF-kB


essential modulator) → X-linked recessive disease → anhidrotic
ectodermal dysplasia with immunodeficiency (EDA-ID)
→abnormal differentiation of ectoderm-derived structure,
impaired immune function → infections from encapsulated
pyogenic bacteria & intracellular bacterial such as mycobacteria,
viruses, fungi e.g. P. jiroveci

Defects in the IL-12/IFN-y Pathway

● IL-12 secreted by DCs & macrophages, IL-12R signaling → synthesis of IFN-y by helper T cells, cytotoxic T cells, NK cells
● Mutations in genes encoding IL-12p40, IL-12Rβ1 chain, IFN-y receptor, and mutations in STAT1 & IKKy/NEMO → susceptibility
to environmental Mycobacterium species (atypical mycobacteria): Mycobacterium avium, M. kansasii, M. fortuitum
● Disorder → Mendelian susceptibility to mycobacterial (MSMD) → severe disease caused by weakly virulent mycobacteria, as
well as other intracellular pathogens such as Salmonella

Defects in Splenic Development


● Autosomal dominant “isolated congenital asplenia”
Caused by:
❖ Heterozygous missense mutations in NBX2.5 (encodes transcription factor)
❖ Genes controlling left-right laterality
● Suffer from severe infections with encapsulated bacteria, esp Streptococcus pneumoniae
Severe Combined Immunodeficiency
● Immunodeficiency that affect both humoral & cell-mediated immunity
● Usually results from impaired T lymphocyte → defective cell mediated immunity, with or without defects in B cell maturation
● Severe infections include pneumonia, meningitis, bacteremia
● Most dangerous organism in SCID → P. jiroveci = severe pneumonia
● Chickenpox (varicella) → usually only in skin & mucous membrane, in SCID could affect lungs, liver, brain
● CMV may be easily transmitted from mothers with SCID to their offspring → fatal pneumonia in newborn
● Children with SCID:
❖ GI infections caused by rotavirus, CMV, protozoa Cryptosporidium & Giardia lamblia = persistent diarrhea, malabsorption
❖ Develop infections from live-attenuated vaccine (chickenpox, measles, mumps, rubella, rotavirus)
● 50% autosomal recessive, rest are X-linked

X-Linked SCID

● Caused by mutations in gene encoding common y (yc) chain for cytokine receptors (IL-2, IL-4, IL-7, IL-9, IL-15, IL-21)
● Inability of lymphopoietic cytokine IL-7 → impaired maturation of T & NK cells
● Failure of IL-15 → impaired NK cell development
● Heterozygous females → phenotypically normal carriers, males inherit abnormal X chromosome manifest this disease

ADA Deficiency & Other Forms of SCID Caused by Defects in Nucleotide Metabolism

● Most common of autosomal recessive SCID → deficiency in ● Rarer autosomal recessive form → deficiency of purine
adenosine deaminase (ADA) enzyme nucleoside phosphorylase (PNP), catalyze the conversion of:
● ADA function → salvage pathway of purine synthesis, inosine → hypoxanthine & guanosine → guanine
catalyzes deamination of: adenosine → inosine & ● Deficiency of PNP → accumulation of deoxyguanosine &
2’-deoxyadenosine → 2’deoxyinosine deoxyguanosine triphosphate → toxic effect to immature
● Deficiency of ADA → accumulation of deoxydenosine and its lymphocytes (mainly T cells)
precursors, S-adenosylhomocysteine & deoxyadenosine ● Manifestation → neurologic deterioration, autoimmune
triphosphate (dATP) → toxic effect → inhibition of DNA hemolytic anemia
synthesis ● Severe form of SCID → “reticular dysgenesis”
● Developing lymphocytes → less efficient in degrading dATP ❖ Cause: defect in development of lymphoid & myeloid
→ more sensitive progenitors
● Manifestation → deafness, costochondral abnormalities, liver ❖ Defect: mutation in adenylate kinase 2 (AK2) gene
damage, behavioral problems, reduced number of B & T cells (regulation the level of adenosine diphosphate) →
increased apoptosis of lymphoid & myeloid precursors
❖ Manifestations: absence of T & B lymphocytes, most
myeloid cells

Autosomal Recessive Mutations in Cytokine Signaling Components

● Clinically identical to X-linked SCID


● Mutations affecting IL-17 receptor α chain or the JAK3 kinase (IL-17 associated with γc chain → signaling) → defect in T cell
development
● IL-15 not affected → normal NK cell

Severe Combined Immunodeficiency Caused by Defects in V(D)J Recombination & Pre-TCR Checkpoint Signaling

● Absence of V(D)J recombination → failure to express pre-TCR ❖ CD3 δ or ε chain or in the CD3-associated ζ chain
& pre-B cell receptor ❖ Gene encoding T cell receptor α chain (TCRα) constant
● Mutations in: region → defect in αβ T cell development → chronic
❖ RAG1/RAG2 genes → encode protein mediate cleavage varicella zoster, EBV infections, autoimmunity & atopy
step during recombination because absence of regulatory T cells (eosinophilia,
❖ ARTEMIS gene → encode nuclease, resolve coding-end vitiligo, eczema, alopecia areata, etc)
hairpins during V(D)J recombination ❖ Autosomal recessive mutations in LCK (encodes tyrosine
❖ Gene encoding catalytic subunit of the DNA-dependent kinase for pre-TCR & TCR signaling)
protein kinase (DNA-PK), DNA ligase 4 → proteins ❖ Hypomorphic mutations (partially reduce function) in
involved in double-stranded break repair/nonhomologous RAG genes/ARTEMIS → “Omenn syndrome” → reduced
end-joining of DNA generation of T & B cells
❖ Gene encoding CD45 phosphatase (positive regulator of ● Causing autosomal recessive forms of SCID
SRC family kinases, such as FYN, LCK, LYN)

DiGeorge Syndome and Other Forms of SCID Due to Defective Thymic Epithelial Development

● DiGeorge Syndrome → defective development of the thymus ● Autosomal recessive FOXN1 (transcription factor of
& parathyroid glands (structure from third and fourth Forkhead family, required for development of thymic anlage
pharyngeal pouches) & ectodermal structure) mutations → SCID, alopecia, nail
Manifestation: immunodeficiency because dystrophy
hypoplasia/agenesis of timus → deficient T cell maturation → ● Mutation in CORONIN1A (encodes protein that regulates
absence peripheral blood T lymphocytes, abnormal calcium actin cytoskeleton) → defective egress of mature T cell from
homeostasis, tetany, abnormal development of the great the thymus
vessels, facial deformities ● Homozygous mutations in MST1 gene (encodes
Mutations in: deletion in the chromosomal region 22q11, serine/threonine protein kinase) → failure of T cells to
mutation in gene encoding transcription factor T-Box1 (TBX1) emigrate from the thymus, loss of native T cells in the
Susceptible to: mycobacterial, viral, fungal infections circulation → recurrent bacterial & viral infections, some
Treatment: fetal thymic/bone marrow transplantation → develop EBV-driven lymphomas & epidermodysplasia
usually not necessary because tends to improve with age verruciformis (HPV-infected warts & skin carcinoma)

Bare Lymphocyte Syndrome and Other Defects in T Cell Positive Selection

● Generation of single-positive CD4+ & CD8+ → depends on ● Autosomal recessive class I MHC deficiencies → decreased
positive selection & lineage commitment CD8+ T cell
● Bare lymphocyte syndrome → autosomal recessive disease Cause: mutations in TAP-1 or TAP-2 genes → encore the
of deficiency of MHC class II → express little or no HLA-DP, subunits of the TAP (transports peptides from the cytosol
DQ, DR on B lymphocytes, macrophages, and DCs into the ER → loaded into MHC class I)
Cause: mutations in genes encoding proteins that regulate TAP deficiency → peptides cannot be bound with MHC →
class II MHC gene transcription MHC degraded → level of MHC class I reduced
For example → mutations on transcription factor RFX5, CIITA Manifestation: necrotizing granulomatous skin lesions,
→ defective positive selection of T cells → reduction of mature respiratory tract bacterial infections
CD4+ T cells ● Another deficiency of class I MHC → mutations in TAPASIN
Treatment: hematopoietic stem cell transplantation protein (required for loading the peptides to MHC class I)
● ZAP70 deficiency → defect in lineage commitment →
reduced CD8+, but normal numbers of CD4+ T cells

SCID Caused by Defective T Cell Activation

● Mutation: gene encoding ORAI1 (component of CRAC, ● Similar phenotype in mutations of STIM1 → encodes ER
calcium release-activated calcium channel) protein (senses the depletion of calcium stores) →
Antigen receptor signaling → activation of γ isoform of contributes to opening of CRAC channel
phospholipase C (PLCγ) & inositol triphosphate ● Manifestation → not exhibit defect in T cell development,
(IP3)-dependent release of calcium ions → replenished by but their T cells cannot be properly activated
CRAC channels → influx of extracellular calcium → this
process crucial for lymphocyte activation
Antibodies Deficiencies: Defects in B Cell Development and Activation

X-Linked Agammaglobulinemia: An X-Linked Pre-BCR Signaling Defect

● Also called Bruton’s agammaglobulinemia ● Common immunodeficiency


● Mutations/deletions: in gene encoding enzyme called Bruton ● BTK → involved in signals from pre-BCR, associates with
tyrosine kinase (BTK) → failure of B cells to mature beyond survival & differentiation of pre-B cell
pre-B cell stage in bone marrow ● Treatment → periodic injections of IgG from donors
● Characterized by: absence of antibodies in the blood → ● Autosomal recessive forms → mutant in genes encoding the
undetectable serum Ig, reduced/absent B cells in peripheral µ (IgM) heavy chain, λ5 surrogate light chain, Igα (signaling
blood & lymphoid tissues, no germinal centers, no plasma cells component of pre-BCR and BCR), p85α subunit of PI3
in tissue kinase, BLNK (adaptor protein downstream of pre-BCR &
BCR)

Selective Immunoglobulin Isotype Deficiencies

● Most common → selective IgA deficiencies ● Selective IgG subclass deficiencies


❖ Many of patients are normal, other have occasional ❖ Total serum IgG levels normal but concentration of
respiratory infections & diarrhea, rarely severe recurrent subclasses are not
infections ❖ Most common → IgG3
❖ Heterogenous ❖ Recurrent bacterial infections in some individuals
❖ Defect in IgA class switching ❖ Cause → homozygous deletions of constant region (Cγ)
❖ Cause → mutations in TACI (transmembrane activator and genes → abnormal B cell differentiation
calcium modulator and cyclophilin ligand interactor) gene,
one of receptors for BAFF (B-cell activating factor) & APRIL
(a proliferation-inducing ligand)

Defects in B Cell Differentiation: Common Variable Immunodeficiencies (CVID)

● Group of heterogeneous disorders → reduced levels of serum ● Mature B lymphocytes present, but memory B cells reduced
Ig → impaired antibody response in blood & plasma → abnormal B cell differentiation
● Most common immunodeficiency in adolescents & young ● Mutations:
adults ❖ TACI
● Diagnosis → exclusion, made based on very low serum IgG ❖ PIK3CD (gene encoding the catalytic subunit of PI3
levels, decreased IgM and/or IgA, poor antibody response to Kinase δ → hyperactivation of T cells, block of B cell
vaccine development)
● Manifestation → major infection: H. influenzae & S. ❖ Loss-of-function mutations of CTLA-4 → ↓ Treg, block of
pneumoniae; autoimmune: pernicious anemia, hemolytic B
anemia, IBD, RA; tumors: lymphoma ❖ ICOS (Inducible T cell costimulator) → ↓ Tfh
● Mostly sporadic ❖ CD19 gene → signaling of CR2(CD21) coreceptor
● Monogenic forms → mainly autosomal dominant inheritance complex, essential for B cell activation by complement

Defects in T Cell-Dependent B Cell Activation: Hyper-IgM Syndrome

● X-linked hyper IgM Syndrome ● Small subset → autosomal recessive


Caused by → mutations in the gene encoding T cell effector ❖ Defects may be in CD40/enzyme activation-induced
molecule CD40L (CD154) → B cells do not undergo heavy deaminase (AID), involved in isotype switching & affinity
chain isotope switching maturation
Manifestation → defective switching of B cells to IgG & IgA ❖ Mutations in the gene encoding uracil N-glycosylase
isotype, major antibodies: IgM, impaired production of (UNG), enzyme which removes U residues from Ig genes
high-affinity antibodies, defects in cell-mediated immunity during class witching & somatic mutation
Disease → susceptibility to intracellular fungi such as P. ❖ Hypomorphic NEMO mutations → EDA-ID disorder →
jiroveci hyper-IgM state
● Abscence of AID → defect in switching & hypermutation
Abscence of UNG → defect in isotype switching only
AID & UNG → do not compromise cell mediated immunity

Defects in T Lymphocyte Activation and Function

Defects in TCR Signal Transduction

● Mutations in genes encoding T cell proteins


Examples:
❖ Impaired TCR complex expression or function caused by mutations in CD3 ε or γ genes
❖ Defective TCR-mediated signaling caused by mutations in ZAP70
❖ Mutations affecting CD3 complex, LCK
Hyper Ig-E Syndromes

● Also known as Job syndrome ❖ In autosomal recessive: mutation in gene DOCK8


● Cause: (guanine nucleotide exchange factor) → ↓ T cells, B cells,
❖ In autosomal dominant: heterozygous dominant-negative NK cells, defect in lymphocytes signaling & cytoskeletal
mutations affecting STAT3 (transcription factor for IL-6, rearrangements
IL-10, IL-17, IL-21, IL-22) *DOCK8 → participates in actin polymerization &
Diminished acute-phase IL-6 downstream maintenance of STAT3
Defective innate immunity ● Manifestation: eczema, eosinophilia, recurrent pulmonary
Defective Th-17 responses infections, staphylococcal & fungal skin abcess
Defective inhibitory responses by IL-10 ● Defective JAK-STAT signaling → ↑ Tfh cells → ↑ IL-4 & L-13
→ drive IgE response

X-Linked Lymphoproliferative Disease (XLP)

● XLP → inability to eliminate EBV → mononucleosis, ● Manifestation: ↓ NK & T cell activation


development of B cell lymphoma ● SAP → required for Tfh cell → inability to make germinal
● Cause: mutations in gene encoding SLAM (signaling centers & high affinity antibodies
lymphocyte activation molecule)-associated protein (SAP), ● Other variation → mutation in gene encoding XIAP (X-linked
involves in activation of NK cells, T & B lymphocytes inhibitor of apoptosis) → ↑ apoptosis of T & NK cells
● SAP → links the membrane proteins SLAM & 2B4 to SRC
family kinase FYN

X-Linked Immunodeficiency-Magnesium Defects-EBV Infection-Neoplasia Syndrome

● MAGT1 → transporter of magnesium intracellular → activation ● Treatment → magnesium diet supplements


of PLCγ1 → calcium signaling in T & NK Cell activation ● B cells → have high PLCγ2 → not affected
● Mutations in X chromosome encoding MAGT1 (magnesium
transporter protein 1) → defect in NK cells, CTL, CD4+ T cell
lymphopenia

Defective CTL & NK Cell Function: Familial Hemophagocytic Lymphohistiocytosis (HLH)

● Familial HLH → defective function of NK & CTL in killing infected cell → viral infection not held in check → compensatory
excessive macrophage activity (macrophage activation syndrome)
● Cause: mutations in genes encoding granule exocytosis
● Mutations:
❖ RAB27A → involved in vesicular fusion
❖ MUNC13-4 → participates in granule exocytosis
❖ Gene for component AP3 protein → disrupt intracellular transport
● Compensatory → excessive IFN-y-mediated macrophage → hemophagocytosis & lymphadenopathy
● Treatment → antibody to IFN-y

Multisystem Disorders With Immunodeficiency

Wiskott-Aldrich Syndrome

Definition Manifestation Mutation

X-linked disease characterized ● Initial stage → ● Gene encodes cytoplasmic ● In autosomal recessive form
by Eczema, thrombocytopenia, lymphocyte numbers are protein, WASP → caused by mutations in
susceptibility to bacterial normal (Wiskott-Aldrich gene encoding WIP
infections ● Principal defect → syndrome protein) → (WASP-interacting protein)
inability to produce expressed on → binds to WASP and
antibodies in T bone-marrow derived cells stabilizes it
cell-independent ● WASP interacts with
polysaccharide antigens → adaptor molecules
susceptibility to downstream of antigen
encapsulated bacteria receptor (GRB2), ARP2/3
● Lymphocytes & platelets complex (actin
smaller than normal polymerization), G
proteins of RHO family
(actin cytoskeletal
rearrangement) →
defective formation of
immune synapses,
impaired mobility of
leukocytes

Ataxia-Telangiectasia

Definition Manifestation Mutation

Autosomal recessive disorder ● Most common humoral ● Gene encodes protein kinase ATM (ataxia-telangiectasia
characterized by abnormal gait immune defects → IgA & mutated) → required for dsDNA break repair, relevant to
(ataxia), vascular malformations IgG2 deficiency V(D)J & class switching recombination → reduced levels of
(telangiectases), neurologic ● Thymic hypoplasia → T IgG, IgA, IgE
deficits, increased risk of tumors, cell defects ● Clinical → URT & LRT infections, autoimmune
and immunodeficiencies phenomena, increasingly cancers occurence

Therapeutic Approaches for Primary Treatment Disease


Immunodeficiencies Passive immunization with X-linked agammaglobulinemia
Two aims: minimize & control infections, replace the gamma globulin
defective/absent components of immune system
Hematopoietic stem cell SCID, Wiskott-Aldrich, bare
transplantation → treatment lymphocyte, LADs
of choice

Enzyme replacement ADA deficiency

Defective gene replacement ADA deficiency, X-linked SCID


with self-renewing stem cells
Secondary (Acquired) Immunodeficiencies

● Not genetic, acquired ● Chronic infections → M. tuberculosis, fungi = anergy to many


● Caused by: antigens
❖ Immunosuppression as biologic complication of disease ● Iatrogenic immunosuppersion → drug therapies that inactives
❖ Iatrogenic immunodeficiencies (complications of therapy) lymphocytes or block chemokines
❖ Infection that target immune cells ❖ Treatment for inflammatory disease
● Disease in which immunodeficiency is a frequent complication ❖ Prevent rejection of organ allograft
→ malnutrition, cancer, infections ❖ Chemotherapeutic drugs → cytotoxic to proliferating cells,
● Cancer → bone marrow tumors (metastatic, leukimias arising including mature & developing lymphocytes
in bone marrow) → produce substance that affect ● Abscence of spleen: surgical removal, treatment to
lymphocytes development hematologic disease such as autoimmune hemolytic anemia,
● Viruses: HIV, measles, HTLV-1 (human T cell lymphotropic thrombocytopenia (red cells & platelets destroyed by
virus 1) → transforms CD4+ T cells → produces malignant phacoytes in spleen) → susceptible to pneumococci &
neoplasm = T cell leukimia/lymphoma (ATL) meningococci bacteria

Human Immunodeficiency Virus & Acquired Immunodeficiency Syndrome

● AIDS → caused by infection with HIV


● HIV can infects activated CD4+ helper T cells, also macrophages & DCs (less efficiently)
● Currently there is no vaccine or permanent cure

An Overview of HIV Virology


HIV Structure and Genes

● Infectious HIV particle: two identical ● Env sequence → encode HIV Envelope
strands of RNA packaged within a core of protein, exist as trimer of glycoprotein,
viral proteins, surrounded by a gp120, g41 (inserted in viral lipid bilayer)
phospholipid bilayer envelope derived ● Pol sequence → encode reverse
from the host cell membrane transcriptase, integrase, viral protease
● Long terminal repeats (LTRs) at each end enzyme
of genome regulate viral gene ● HIV-1 genome → additional regulatory
expression, viral integration into host genes: tat, rev, vif, nef, vpr, vpu
genome, and viral replication
● Gag sequence → encode code structural
proteins

Viral Life Cycle

● HIV infection → begins when envelope ❖ After virus completes life cycle in infected
glycoprotein gp120 binds CD4 & cell → free viral particles released from
chemokine receptor (coreceptor) infected cell → bind to an uninfected cell
● 2 components of Env → transmembrane ❖ Gp120 & gp41 expressed on plasma
gp41 subunit & external gp120 subunit membrane of infected cell → mediates
→ produced by proteolytic cleavage of cell-cell fusion with uninfected cell
gp160 precursor expressing CD41 & coreceptor
● Env complex mediates fusion of virion ● Coreceptors:
envelope with membrane target cell ❖ CCR5 & CXCR4 (most important)
❖ Gp 120 subunit binding with CD4 → ❖ Several transmembrane-spanning G
conformational change → secondary protein-coupled receptor (e.g.
gp120 binding to chemokine receptor leukotriene B4 receptor)
(CCR5/CXCR4 as coreceptor) → ● Main target of HIV → CD4+, some strain
conformational change in gp41 → with very high affinity with CD4+ could
gp41 refolding into six-helix bundle, infect macrophages (M-tropic strains)
exposes a hydrophobic region (fusion ● Only 1 viral strain act as disease
peptide) → inserts into cell membrane transmitter → founder strain (T-tropic
→ viral membrane fuse with target cell strain) → low affinity of CD4, mostly use
membrane CCR5

● HIV virion enters the cell → viral reproductive cycle begins ● Tat protein → required for HIV gene expression. Without tat
❖ Nucleoprotein core of virus disrupted → RNA genome of HIV HIV genes could not be transcripted completely because
reverse-transcripted into dsDNA → viral DNA enter nucleus polymerase complex stop before mRNA completed
❖ Viral integrase enters nucleus → catalyze the DNA ● mRNA encoding various HIV protein → derived from single
integration full-genome-length transcript by differential splicing events
❖ Integrated HIV DNA → provirus ● HIV gene expression:
❖ Provirus may remain transcriptionally inactive for years → ❖ Early → expressing of regulatory genes
latent phase ❖ Late → expressing of structural genes, full-length viral
● Transcription of provirus → regulated by LTR upstream genomes are packaged
● LTR: polyadenylation signal sequence, TATA box promotor ● Rev, Tat, Nef → early gene products encoded by fully spliced
sequence, binding site of transcription factors (NF-kB & SP1) mRNA, exported from nucleus & translated into proteins in
● Initiation of transcription → activation of T cells by antigen & cytoplasm
cytokines → activation of NF-kB → binding to LTR ● Env, gag, pol → late genes, encode structural components of
Example: IL-2, TNF, lymphotoxin → T cell, IFN-y & the virus
granulocyte-macrophage colony-stimulating factor (GM-CSF) →
macrophage

● Rev → promoting export of incompletely spliced late gene RNA ● Viral protein synthesized in cytoplasm
out of the nucleus → initiates switch from early to late expression ● Assembly → packaging proviral genome within
● Pol → precursor protein, cleaved to form reverse transcriptase, nucleoprotein complex → buds from the plasma membrane,
protease, ribonuclease, integrase enzymes capturing Env & host glycoproteins as part of its envelope
● Gag → encodes protein that cleaved by protease into p24 capsid
protein, p17 matrix protein, p7 nucleocapsid protein, p6 domain, 2
spacer peptides (sp1 & sp2)
● Env → gp160, cleaved by cellular protease in ER to gp120 &
gp41

Pathogenesis of HIV Infection and AIDS

Acute (early) infection Transition From Acute Phase-Chronic Phase

● Infection of activated CD4+ T cell in mucosal lymphoid tissue & ● Viremia & development of adaptive response
death of many infected CD4+ cells ● DCs in epithelia capture the virus → migrate to lymph nodes
→ pass the virus to CD4+ T cells
● DCs express protein with mannose-binding lectin domain
(DC-SIGN), binds with HIV envelope
● Replication in lymph nodes → viremia (large number of hIV
present in blood) → acute HIV syndrome → control by
adaptive response → drop of viremia

Chronic Phase

● Lymph nodes, spleen, GI tract → sites of continuous HIV replication


● Immune system competent at handling infection with opportunistic microbes → clinical latency period
● Destruction of CD4 T cells steadily progresses
● Early in course, host may continue to replace CD4+ T cell → period of years, cycle leads to loss of CD4+ T cells

Mechanisms of Immunodeficiency Caused by HIV


● Major cause of the loss of CD4+ T cells → direct effect of ❖ Gp120 - newly synthesized intracellular CD4 → interfere
infection normal protein processing → block expression of CD4 →
❖ Budding → ↑ plasma membrane permeability, influx of lethal incapable responding to antigen
amounts of calcium/water → apoptosis/osmotic lysis
❖ Viral production → interfere cellular protein → cell death
● Additional mechanism:
❖ Chronic activation of uninfected cells with infection common
in AIDS → apoptotic death
❖ HIV-spesific CTLs → kills infected CD4+
❖ HIV envelope protein antibodies → target infected cells

● Depletion of T cell → functional defects ● DCs → also can be infected, not directly
❖ Decrease in T cell response to antigens injured by HIV but intimate contact with
❖ Nonneutralizing humoral immune naive T cells could infect these cells
response ● FDCs in germinal center of lymph nodes
● Bound HIV on CD4 → can't interact with → trap HIV
MHC & signals→ antigen response ❖ May be a reservoir and infect
inhibited, downregulate function macrophages & CD4+ cells
● Macrophages: resistant to cytophatic ❖ Normal functions are impaired
effects of HIV, may infected from ● Latent virus in Tfh cells in light zone of
phagocytosis of the virus & Fc germinal center → protected from CTLs
receptor-mediated endocytosis →
infected but not killed with HIV →
reservoir

Clinical Features of HIV

Acute phase Chronic phase/clinical latency Final phase/AIDS


● Acute HIV syndrome, period of viremia ● Last for many years, virus ● Blood CD4+ T cell count drops <200
with nonspecific symptoms contained within lymphoid tissue cells/mm3
● Spike of plasma virus, modest reduction ● Loss of CD4 → replenished from ● Viral replication accelerates
of CD4 T cell counts progenitors ● Combination of opportunistic infections,
● Asymptomatic or minor infection neoplasms, cachexia (wasting), kidney
● Predictors of progression → level failure (neprophathy), CNS (AIDS
of virus & CD4+ blood counts encelopathy or HIV-associated
● Disease progress → susceptible to neurocognitive disorder, HAND)
other infections, immune response
to this → stimulate HIV production

Immune Responses to HIV

Innate Immunity to HIV & Host Restriction Factors

● HIV → sensed by TLR, RIG-1


● Key sensors that recognize viral reverse transcription products early in infection:
❖ IFI16 (interferon inducible protein 16) → bind to HIV-derived cDNA & signals via the STING (stimulator of IFN genes) adaptor,
TBK1 protein kinase, IRF3 & IR7 transcription factors → induces type 1 IFN & host restriction factors: APOBEC3, TRIM5α,
SAMHD1, tetherin
❖ cGAS (cyclic GMP-AMP synthase)

Tetherin APOBEC3 (apolipoprotein B mRNA editing enzyme catalytic


polypeptide like 3)

● Inhibition of budding process → prevents virion release ● Cytidine deaminase that interferes with viral replication
● Antagonized by HIV protein Vpu ● HIV Vif protein target APOBEC3 for ubiquitination &
proteasomal degradation
TRIM5α SAMHD1 (SAM domain & HD domain 1)

● Interacts with HIV capsid protein causing premature uncoating ● Hydrolyzes & depletes intracellular deoxynucleoside
of virus & proteasomal degradation of viral reverse triphosphate → prevent synthesis of viral DNA by reverse
transcriptase, block nuclear translocation of viral complex transcription
● HIV-2 produces Vpx → antagonize depleting activity

Adaptive Immune Responses to HIV

● Provide limited protection ● Antibody response → detectable 6-9 weeks postinfection


● Immune response may be detrimental to the host (stimulating ● Most immunogenic HIV → envelope glycoproteins → high
uptake of opsonized virus into uninfected cells) anti-gp120& gp-40
● Initial adaptive response → CTLs specific HIV ● Early antibodies → not neutralizing
❖ Control infection in early phase ● Neutralizing antibodies cannot cope with rapidly change
❖ Could not produce memory CD8+ because needs effective epitope
CD4+ response ● 10-15% neutralizing Ig bind to a site on viral protein that
❖ Evolution of virus → lost of original CTL & CD4+ epitope the virus cannot afford to mutate, such as CD4 binding of
gp140

Mechanisms of Immune Evasion by HIV

● High mutation rate ● Epitopes of the virus shielded by HIV-glycan shield


● Most antigenically variable part of the virus → V3 loop region ● HIV Nef protein → inhibits expression of MHC Class 1
of gp120

Treatment and Prevention of AIDS & Vaccine Development

Nucleoside analogues Nonnucleoside reverse transcriptase

● Bind to & inhibits viral reverse transcriptase ● Directly bind to enzyme & inhibit its function
● Deoxythymidine nucleoside analogues: AZT, deoxycytidine ● Viral protease inhibitors → block processing of precursor
nucleoside analogues, deoxyadenosine analogues proteins into mature viral capsid & core proteins
● Often effective in reducing plasma HIV RNA, do not halt ● Used alone → mutant virus resistant → used together with
progression of HIV-induced disease 2 different reverse transcriptase inhibitor (highly active
● Mutated forms of reverse transcriptase → resistant to drugs antiretroviral therapy, HAART or antiretroviral therapy,
ART)

Integrase Inhibitors Entry inhibitors

Fusion inhibitors ● Prevent viral entry, targeting CD4/CCR5

● Target gp41 → prevent fusion of viral envelope with host cell plasma membrane

Pre-exposure prophylaxis (PrEP)

● Reverse transcriptase inhibitors, protect uninfected individuals with high risk

Vaccine

● Complicated by ability of virus to mutate ● Recombinant subunit vaccine with HIV trimer → purpose
● Involved infection of macaques with simian immunodeficiency for neutralizing antibodies to gp140 (highly conserved)
virus (SIV) → vaccine to SIV has been developed ● Use of passive immunity of broadly neutralizing antibodies
● Nonvirulent recombinant hybrid virus (part SIV, part HIV) → ● Vectored immunoprophylaxis → protein that can mediate
attenuated by deletions of viral genome, e.g. nef gene immune synthesized in the host after injection of specific
● Use of live recombinant non-HIV viral vector carrying HIV DNA into skeletal muscle
genes →structural/regulatory genes of HIV + mammalian DNA ❖ Ig heavy & light chain genes that encode broadly
expression vector neutralizing antibody → cloned into adenovirus vector →
DNA injected

You might also like