ArticlePDF AvailableLiterature Review

The Immunocompromised Host: HIV Infection

Authors:

Abstract and Figures

The variety of pulmonary infections encountered in HIV-infected individuals indicates that many components of the host defense network are impaired. In addition to depletion of CD4+ T cell numbers, HIV infection results in functional deficits in CD4+ T cells, CD8+ T cells, and natural killer cells. Although some components of macrophage defense are preserved, lack of activation signals from CD4+ T cells contributes to impaired defense by macrophages. There are few data examining the functional capabilities of neutrophils in the lung, but evidence from peripheral blood neutrophils indicates that defense by these cells is also impaired. An improved understanding of these events in the lung during HIV infection could lead to specific interventions aimed at restoration of deficient function.
Content may be subject to copyright.
The Immunocompromised Host
HIV Infection
James M. Beck
Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School; and Medical Service,
Department of Veterans Affairs Medical Center, Ann Arbor, Michigan
The variety of pulmonary infections encountered in HIV-infected
individuals indicates that many components of the host defense
network are impaired. In addition to depletion of CD4Tcellnum-
bers, HIV infection results in functional de ficits in CD4T cells, CD8
T cells, and natural killer cells. Although some components of macro-
phage defense are preserved, lack of activation signals from CD4
T cells contributes to impaired defense by macrophages. There are
few data examining the functional capabilities of neutrophils in the
lung, but evidence from peripheral blood neutrophils indicates that
defense by these cells is also impaired. An improved understanding
of these events in the lung during HIV infection could lead to specific
interventions aimed at restoration of deficient function.
Keywords: HIV infections; macrophages, alveolar; neutrophils;
opportunistic infections; T-lymphocytes
The variety of pulmonary infections encountered in HIV-
infected individuals demonstrates that HIV impairs lung host
defenses significantly (1). Although most investigations focus
on HIV’s effects on systemic immunity, a significant body of
literature examines pulmonary immune mechanisms during HIV
infection. In some instances, immune defects documented in the
periphery are reflected in the lungs. In other instances, effects
of HIV on lung cells differ from those in the periphery. This
article focuses on the effects of HIV infection on pulmonary host
defenses to explain why serious pulmonary infections remain
common in this immunocompromised patient population.
The accessibility of lung cells for study by bronchoalveolar
lavage (BAL) provides an opportunity to study the effects of
HIV and opportunistic pathogens on lung host defenses. Once
impairments in pulmonary host defense are understood, novel
strategies to correct these defects may be developed for treat-
ment and prophylaxis of pulmonary infections (1).
There are several general mechanisms by which HIV infection
can impair pulmonary host defense; the literature documents
alterations in cell numbers and in cell function. First, HIV di-
rectly infects and kills cells directed against pathogens, leaving
decreased numbers of cells available to participate in host de-
fense. Second, HIV infection induces qualitative defects in the
metabolic and secretory functions of effector cells. Third, HIV-
infected cells may shift their repertoires from elaboration of
immunostimulating to immunosuppressive products. Fourth,
HIV infection could interfere with the capacity of circulating
lymphocytes, monocytes, or neutrophils to migrate into the lungs
and to clear pathogens from the alveolar spaces. Finally, coinfec-
(Received in original form July 30, 2005; accepted in final form August 17, 2005)
Supported in part by a Merit Review Award from the Department of Veterans
Affairs and R01 HL08342 from the National Institutes of Health.
Correspondence and requests for reprints should be addressed to James M. Beck,
M.D., Pulmonary Medicine (111G), Veterans Affairs Medical Center, 2215 Fuller
Road, Ann Arbor, MI 48105. E-mail: jamebeck@umich.edu
Proc Am Thorac Soc Vol 2. pp 423–427, 2005
DOI: 10.1513/pats.200507-077JS
Internet address: www.atsjournals.org
tion by a second pathogen can contribute to a breakdown in the
host defense cascade.
HIV INFECTION OF LUNG CELLS
One important mechanism by which HIV alters lung host de-
fense is by direct infection of pulmonary cells. Various HIV
strains demonstrate tropism for lymphocytes or for monocytes/
macrophages. The CD4 molecule, present on lymphocytes and
monocytes/macrophages, serves as the primary cellular receptor
for HIV-1. In the lung, CD4 is the primary receptor for HIV
on alveolar macrophages. Coreceptors are also needed for HIV
entry into cells, and cellular coreceptors determine the tropism
of HIV strains (Figure 1). Lymphocyte-tropic (T-tropic) strains
interact with the chemokine receptor CXCR4 (fusin) to control
entry into target cells. Infection with T-tropic strains can be
blocked by the CXC chemokine SDF-1, which is a CXCR4
ligand.
In contrast, monocyte-tropic (M-tropic) strains interact with
the chemokine receptor CCR5 to control entry into target cells.
HIV infection of human alveolar macrophages is preferentially
mediated by the CCR5 receptor, although alveolar macrophages
also express CXCR4. Infection with M-tropic strains can be blocked
with the CC chemokines RANTES, macrophage inflammatory
protein-1, and macrophage inflammatory protein-1,whichare
CCR5 ligands. As HIV infection progresses, T-tropic virus replaces
M-tropic virus, and this change is accompanied by more rapid
immunologic decline.
Alveolar macrophages are a primary reservoir of HIV in
the lung. HIV reverse transcriptase can be detected in alveolar
macrophages obtained by lavage from patients with AIDS, and
alveolar macrophages can be infected with HIV in vitro (2).
Reports comparing the HIV burden of alveolar macrophages
and peripheral blood monocytes are discrepant. The relative
importance of in situ HIV replication in the lung, compared with
the influx of previously infected cells from bone marrow and
blood, is unclear. The percentages of alveolar macrophages ex-
pressing HIV antigens have varied considerably in reports from
different laboratories (3). Detection of HIV by polymerase chain
reaction suggests that HIV infection of alveolar macrophages
is common (4). Alveolar macrophages become infected with
increasing frequency as HIV infection progresses (5). When di-
rect comparisons of alveolar macrophages and peripheral blood
monocytes have been made, the data suggest that viral burdens
in these two cell populations are equivalent (6).
How highly active antiretroviral therapy (HAART) modu-
lates HIV replication in the lung and pulmonary host defense
is unknown. Isolation of HIV from BAL in asymptomatic indi-
viduals decreases when CD4T cell counts are greater than
200/l, and when patients receive HAART (7), suggesting a
beneficial effect. However, HIV strains may evolve indepen-
dently in the lung and blood, as phylogenetic analysis of BAL
strains may differ from blood strains in HIV-infected individuals
(8). Zidovudine resistance may differ in peripheral blood mono-
nuclear cells and alveolar macrophages of single individuals,
suggesting that HAART may have different effects on HIV in
424 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 2 2005
Figure 1. Tropism of HIV strains for lung cells. T-tropic strains infect
T cells, using CD4 as the main receptor and CXCR4 as the coreceptor.
M-tropic strains infect alveolar macrophages, using CCR5 as the core-
ceptor, although alveolar macrophages also express CXCR4.
lung cells (9). Recent work comparing resistance patterns of
peripheral blood and BAL cells demonstrates that resistant HIV
genotypes in the lung may be most widely divergent in individu-
als exposed to combination antiretroviral therapy (10). Further
studies are needed to examine the viral load in the lung during
HAART and to determine how HAART modulates pulmonary
host defenses (11).
ALTERATIONS IN LUNG CELL NUMBERS AND
PHENOTYPES DURING HIV INFECTION
Most of the data describing alterations in lung cell numbers and
phenotypes were obtained during bronchoscopies to diagnose
pulmonary infections, making the effects of HIV and of opportu-
nistic pathogens difficult to distinguish. Additionally, these stud-
ies were conducted before the availability of HAART. These
studies revealed that lymphocyte percentages or concentrations
are increased in BAL from HIV-infected individuals compared
with uninfected individuals. Examination of BAL lymphocyte
subsets from patients with AIDS shows decreases in CD4
T cells and increases in CD8T cells. Therefore, CD4/CD8
ratios in BAL specimens may be lower than the ratios in periph-
eral blood. Some HIV-infected individuals may manifest pulmo-
nary symptoms as a result of CD8T-cell influx into the lung,
clinically diagnosed as lymphoid interstitial pneumonitis. Late
in the course of HIV infection, numbers of CD8T cells decline.
Depletion of CD8T cells may be associated with the develop-
ment of disseminated cytomegalovirus and Mycobacterium avium
infections (12). As in peripheral blood, most T cells in the lung
bear ␣␤ T-cell receptors on their surfaces, but a minority of
T cells expresses ␥␦ T-cell receptors. Numbers of ␥␦ T cells are
reported to be decreased or increased in HIV-infected patients
with opportunistic infections.
Macrophage numbers in BAL from patients with AIDS are
probably normal, but percentages are decreased by influx of
other cells. Several BAL series have reported increases in the
concentrations or in the percentages of neutrophils obtained
from patients with AIDS compared with uninfected control sub-
jects. Presumably, HAART results in the repopulation of pulmo-
nary host defense cells as in blood, but BAL studies are not
available to compare individuals receiving and not receiving
HAART.
ALTERATIONS IN LYMPHOCYTE FUNCTION
CD4T Cells
Recent evidence demonstrates that, in addition to accelerated
destruction of CD4T cells by HIV infection, production of
T cells is also impaired. Underproduction can occur from
infection-mediated death of progenitor cells and destruction of
the hematopoietic stroma. In peripheral blood, T cells from
patients with AIDS do not proliferate normally in response
to mitogens. Even in patients with AIDS who show serologic
evidence of prior infection with cytomegalovirus or herpes sim-
plex virus, T cells fail to proliferate normally in response to these
viral antigens. Failure to proliferate may be caused in part by
impaired elaboration of IL-2. Peripheral blood T cells from pa-
tients with AIDS have impaired IL-2 secretion in response to a
variety of stimuli. In vitro, recombinant IL-2 can restore some
mitogenic responses of blood T cells in patients with AIDS (13).
Clinical trials of IL-2 for HIV infection demonstrate that IL-2
increases CD4T cell counts in recipients without increasing
HIV replication, particularly when given intermittently (14).
IL-16 may prime mature CD4memory cells to respond to
IL-2, and decreased IL-16 levels correlate with HIV progression.
Therefore, IL-16 therapy may be useful in addition to IL-2 to
restore CD4T-cell responses (15).
T cells from HIV-infected individuals do not produce IFN-
normally in response to mitogens or antigens. The relative ability
of peripheral lymphocytes to elaborate IFN-correlates with
clinical status and CD4T cell count and predicts progression
to AIDS (16). Experimental work suggests that progression from
asymptomatic HIV infection to AIDS is accompanied by a switch
from Th1-like responses to Th2-like responses. In theory, pre-
vention of this switch in T-cell responses could prevent progres-
sion to AIDS. IL-12, a cytokine that favors Th1 development
and inhibits Th2 development, has been shown to restore cell-
mediated immunity in T cells obtained from HIV-infected indi-
viduals (17). Few data exist on the functional capabilities of lung
CD4cells.
CD8T Cells
As with CD4T cells, CD8T cells in blood and lung can be
infected with HIV. HIV infection also impairs the functional
capacities of CD8T cells. CD8T cells obtained from the
lungs of HIV-infected individuals do not lyse appropriate targets
in vitro (18). For example, CD8T cell–mediated cytotoxicity
for influenza virus is decreased in HIV-infected individuals (19).
Phenotypically, CD8T cells from patients with AIDS show
increased expression of activation markers, compared with unin-
fected individuals, and increased percentages of activated cells
predict progression of HIV-related disease. Peripheral CD8
T cells in HIV-infected individuals may be poor effectors because
they lack required maturation signals (20). An analogous situa-
tion likely exists in the lung.
CD8T-cell alveolitis occurs during HIV infection, but the
functional capabilities of these cells and their intended targets
require further investigation. Subpopulations of CD8T cells,
obtained from HIV-infected individuals, are cytotoxic for macro-
phages or B-cell lines expressing HIV antigens (21). The intensity
of CD8alveolitis correlates with HIV load, and the poor prog-
nosis associated with alveolitis may be a result of the elevated
viral burden (22). Unlike the periphery, local concentrations of
IL-2 and IFN-may be increased in the lung during HIV infec-
tion due to activation of CD8T cells (23). The alveolitis may
be driven by overproduction of IL-15, a cytokine with IL-2–like
effects, by alveolar macrophages. Alveolar macrophages from
HIV-infected individuals produce large quantities of IL-15,
Beck: HIV Infection 425
which enhances antigen presentation by alveolar macrophages
and causes proliferation of lung CD8T cells (24).
NK Cells
Increased numbers of NK cells in BAL have been observed in
HIV-infected individuals, but with progressive HIV disease, they
lose functional capabilities. NK cells may be nonfunctional in
HIV infection because they are dependent on signals from
CD4T cells for optimal function (25). Biologic response mod-
ifiers such as recombinant IL-2 restore lytic ability in vitro (26),
and IFN-may augment NK cell activity in early stages of HIV
infection (27).
B Cells
Analysis of peripheral B cells from HIV-infected individuals
shows elevated numbers of cells that spontaneously secrete im-
munoglobulins, and patients with AIDS have increases in serum
IgA, IgG, and IgM. The IgG increase is predominantly due to
IgG subtypes IgG1 and IgG3, with decreases in IgG2 and IgG4.
Antibody to polysaccharides in bacterial cell walls is composed
of IgG2, and patients with AIDS who have pyogenic infections
have lower serum IgG2 than patients without bacterial disease.
Concentrations of immunoglobulins in the lung may depend
on the stage of HIV infection or the presence of pulmonary
pathogens. Compared with uninfected individuals, measurement
of immunoglobulins in BAL from patients with AIDS who have
pulmonary symptoms shows increases in total amounts of IgG,
IgM, and IgA. Local immunoglobulin synthesis may occur in the
lung, as shown by increased numbers of IgG-, IgM-, and IgA-
secreting cells (28). However, BAL from asymptomatic HIV-
infected individuals contains decreased concentrations of IgG
compared with uninfected control subjects (29). During HAART,
IgG levels in asymptomatic, HIV-infected individuals are
increased compared with uninfected volunteers (30).
Antibody responses to specific antigens are impaired in HIV-
infected individuals. B-cell abnormalities begin early in HIV
infection, with failure to produce antibody in response to mito-
gen at the time of HIV seroconversion, before T-cell function
is affected. B cells from patients with AIDS show impaired
proliferation in response to mitogens and do not initiate normal
antibody synthesis in response to newly encountered antigens.
Decreased IgG concentrations in the lung may be a result of
impaired ability of alveolar macrophages to induce IgG secretion
from B cells, likely as a result of transforming growth factor-
secretion (29).
ALTERATIONS IN MACROPHAGE FUNCTION
Phagocytosis, Respiratory Burst, and Killing of Organisms
Peripheral blood monocytes from patients with AIDS are re-
ported by some investigators to be defective in chemotaxis to
several chemoattractants, but other investigators find unim-
paired chemotaxis. Alveolar macrophages from asymptomatic,
HIV-infected subjects demonstrate enhanced phagocytosis for
Staphylococcus aureus (31). BAL cells from HIV-infected individu-
als demonstrate increased fungistatic activity against Cryptococcus
neoformans compared with cells from uninfected control subjects
(32). The magnitude of the respiratory burst of alveolar macro-
phages from patients with AIDS in vitro is not different from
uninfected control subjects, and IFN-enhances the response in
cells from both groups equivalently (33). Alveolar macrophages
and monocyte-derived macrophages do not kill Toxoplasma gondii
or Chlamydia psittaci, whether obtained from patients with AIDS
or from uninfected individuals (33). When exposed in vitro to
IFN-, alveolar macrophages obtained from patients with AIDS
increase their killing of these organisms in a manner equivalent
to uninfected individuals’ cells (33). In contrast, HIV infection
may impair phagocytosis of organisms that commonly cause
pulmonary infections in susceptible individuals. For example,
alveolar macrophages from HIV-infected individuals demon-
strate decreased binding and phagocytosis of Pneumocystis
in vitro, and this defect correlates with decreased expression of
alveolar macrophage mannose receptors (34).
Antigen Presentation
During HIV infection, blood monocytes do not present antigens
to T cells normally (35). In comparison to blood monocytes,
alveolar macrophagesare considered to be relatively poor antigen-
presenting cells. However, alveolar macrophages from HIV-
infected patients have enhanced ability to present antigen com-
pared with alveolar macrophages from uninfected control subjects
(36). Dendritic cells are also important antigen-presenting cells
in the lung. HIV infection of dendritic cells is cytopathic for
these cells, and the numbers of dendritic cells are decreased
in asymptomatic HIV-infected individuals and in patients with
AIDS (37). Dendritic cells from HIV-infected individuals exhibit
defective antigen presentation and may facilitate HIV infection
of T cells (38).
Tumor Necrosis Factor Elaboration
Some patients with AIDS are reported to have elevated serum
levels of tumor necrosis factor (TNF). When peripheral blood
monocytes are examined, they are reported to have high sponta-
neous release of TNF (39) or suboptimal release after appro-
priate stimulation (40). Other investigators have found that HIV
infection of monocytes or monocyte-derived macrophages in vitro
does not induce TNF release (41). Alveolar macrophages from
asymptomatic, HIV-seropositive individuals have increased spon-
taneous TNF release, which correlates with extent of HIV expres-
sion (42). BAL cells from smokers release less TNF than BAL
cells from nonsmokers, suggesting that smoking and HIV inter-
act to suppress macrophage function (43). In the lung, TNF
decreases HIV replication in alveolar macrophages by inducing
production of RANTES and by decreasing CCR5 expression (44).
ALTERATIONS IN NEUTROPHIL FUNCTION
Although patients with AIDS may have increased numbers of
neutrophils at the time of BAL, little is known about the host
defense capabilities of these cells. In several studies, peripheral
neutrophils from some patients with AIDS who had frequent
localized infections showed decreased chemotaxisin vitro.Neutro-
phils from HIV-infected individuals have decreased expression of
CD88, the ligand for C5a, which could contribute to increased
susceptibility to bacterial infections (45). Recently, pulmonary
neutrophils from HIV-infected individuals have been shown to
express decreased immunoglobulin G Fc-receptor 1 expression
compared with neutrophils from uninfected volunteers (46).
The phagocytic capacity of neutrophils during HIV infection
is controversial and may depend on the stage and severity of
HIV infection. For example, neutrophils from individuals with
early HIV infection demonstrate enhanced phagocytosis (47).
Phagocytosis of opsonized S. aureus is decreased in the periph-
eral blood neutrophils of some, but not all, patients with AIDS
(48). The defect in phagocytosis can be corrected by in vivo
administration of granulocyte colony-stimulating factor (49).
Other investigators find decreased phagocytosis and intracellular
killing of Candida albicans by neutrophils obtained from intrave-
nous drug-using patients with AIDS, whereas cells from homo-
sexual AIDS patients function normally (50). The authors postu-
late that these defects are associated with intravenous drug use
426 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 2 2005
rather than HIV infection because cells from seronegative intra-
venous drug users were also abnormal.
The neutrophil possesses potent oxygen-dependent mecha-
nisms to kill intracellular organisms. Peripheral neutrophils from
patients with AIDS are reported to produce subnormal, normal, or
supernormal amounts of superoxidewhen stimulated with phorbol
myristate acetate. The ability of pulmonary neutrophils from HIV-
infected individuals to produce superoxide is unknown.
CONCLUSION
HIV infection impairs many aspects of host defense in the lung
and in the periphery. In addition to depletion of CD4T cell
numbers, HIV results in functional deficits in CD4T cells,
CD8T cells, and NK cells. Although some components of
macrophage defense are preserved, lack of activation signals
from CD4T cells contributes to impaired defense by macro-
phages. There are few data examining the functional capabilities
of neutrophils in the lung, but evidence from peripheral blood
neutrophils indicates that defense by these cells is also impaired.
An improved understanding of these events in the lung during
HIV infection can lead to specific interventions aimed at restora-
tion of deficient function.
Conflict of Interest Statement :J.M.B. does not have a financial relationship with
a commercial entity that has an interest in the subject of this manuscript.
References
1. Beck JM, Rosen MJ, Peavy HH. Pulmonary complications of HIV infec-
tion: report of the fourth NHLBI workshop. Am J Respir Crit Care
Med 2001;164:2120–2126.
2. Salahuddin SZ, Rose RM, Groopman JE, Markham PD, Gallo RC.
Human T lymphotropic virus type III infection of human alveolar
macrophages. Blood 1986;68:281–284.
3. Agostini C, Trentin L, Zambello R, Semenzato G. HIV-1 and the lung:
infectivity, pathogenic mechanisms and cellular immune responses
taking place in the lower respiratory tract. Am Rev Respir Dis 1993;147:
1038–1049.
4. Rose RM, Krivine A, Pinkston P, Gillis JM, Huang A, Hammer SM.
Frequent identification of HIV-1 DNA in bronchoalveolar lavage cells
obtained from individuals with the acquired immunodeficiency syn-
drome. Am Rev Respir Dis 1991;143:850–854.
5. Sierra-Madero JG, Toossi Z, Hom DL, Finegan CK, Hoenig E, Rich
EA. Relationship between load of virus in alveolar macrophages from
human immunodeficiency virus type 1-infected persons, production of
cytokines, and clinical status. J Infect Dis 1994;169:18–27.
6. Lewin SR, Kirihara J, Sonza S, Irving L, Mills J, Crowe SM. HIV-1 DNA
and mRNA concentrations are similar in peripheral blood monocytes
and alveolar macrophages in HIV-1-infected individuals. AIDS
1998;12:719–727.
7. Wood KL, Chaiyarit P, Day RB, Wang Y, Schnizlein-Bick CT, Gregory
RL, Twigg HL III. Measurements of HIV viral loads from different
levels of the respiratory tract. Chest 2003;124:536–542.
8. Itescu S, Simonelli PF, Winchester RJ, Ginsberg HS. Human immunode-
ficiency virus type 1 strains in the lungs of infected individuals evolve
independently from those in peripheral blood and are highly conserved
in the C-terminal region of the envelope V3 loop. Proc Natl Acad Sci
USA 1994;91:11378–11382.
9. Gates A, Coker RJ, Miller RF, Williamson JD, Clarke JR. Zidovudine-
resistant variants of HIV-1 in the lung. AIDS 1997;11:702–703.
10. White NC, Israe
¨l-Biet D, Coker RJ, Mitchell DM, Weber JN, Clarke
JR. Different resistance mutations can be detected simultaneously in
the blood and the lung of HIV-1 infected individuals on antiretroviral
therapy. J Med Virol 2004;72:325–357.
11. White NC, Agostini C, Israe
¨l-Biet D, Semenzato G, Clarke JR. The
growth and the control of human immunodeficiency virus in the lung:
implications for highly active antiretroviral therapy. Eur J Clin Invest
1999;29:964–972.
12. Fiala M, Herman V, Gornbein J. Role of CD8in late opportunistic
infections of patients with AIDS. Res Immunol 1992;143:903–907.
13. Sheridan JF, Aurelian L, Donnenberg AD, Quinn TC. Cell-mediated
immunity to cytomegalovirus (CMV) and herpes simplex virus (HSV)
antigens in the acquired immune deficiency syndrome: interleukin-1
and interleukin-2 modify in vitro responses. J Clin Immunol 1984;4:
304–311.
14. Sereti I, Lane HC. Immunopathogenesis of human immunodeficiency
virus: implications for immune-based therapies. Clin Infect Dis 2001;
32:1738–1755.
15. Center DM, Kornfeld H, Ryan TC, Cruikshank WW. Interleukin 16:
implications for CD4 functions and HIV-1 progression. Immunol
Today 2000;21:273–280.
16. Murray HW, Hillman JK, Rubin BY, Kelly CD, Jacobs JL, Tyler LW,
Donelly DM, Carriero SM, Godbold JH, Roberts RB. Patients at risk
for AIDS-related opportunistic infections: clinical manifestations and
impaired gamma interferon production. N Engl J Med 1985;313:1504–
1510.
17. Clerici M, Lucey DR, Berzofsky JA, Pinto LA, Wynn TA, Blatt SP,
Dolan MJ, Hendrix CW, Wolf SF, Shearer GM. Restoration of HIV-
specific cell-mediated immune responses by interleukin-12 in vitro.
Science 1993;262:1721–1724.
18. Semenzato G. Immunology of interstitial lung diseases: cellular events
taking place in the lung of sarcoidosis, hypersensitivity pneumonitis
and HIV infection. Eur Respir J 1991;4:94–102.
19. Shearer GM, Bernstein DC, Tung KSK, Via CS, Redfield R, Salahuddin
SZ, Gallo RC. A model for the selective loss of major histocompatibil-
ity complex self-restricted T cell immune response during the develop-
ment of acquired immune deficiency syndrome (AIDS). J Immunol
1986;137:2514–2521.
20. Lieberman J, Shankar P, Manjanath N, Andersson J. Dressed to kill?
A review of why antiviral CD8 T lymphocytes fail to prevent progres-
sive immunodeficiency in HIV-1 infection. Blood 2001;98:1667–1677.
21. Autran B, Plata F, Guillon JM, Joly P, Mayaud C, Debre
´P. HIV-specific
cytotoxic T lymphocytes directed against alveolar macrophages in
HIV-infected patients. Res Virol 1990;141:131–136.
22. Twigg HL, Soliman DM, Day RB, Knox KS, Anderson RJ, Wilkes DS,
Schnizlein-Bick CT. Lymphocytic alveolitis, bronchoalveolar lavage
viral load, and outcome in human immunodeficiency virus infection.
Am J Respir Crit Care Med 1999;159:1439–1444.
23. Twigg HL III, Spain BA, Soliman DM, Knox K, Sidner RA, Schnizlein-
Bick C, Wilkes DS, Iwamoto GK. Production of interferon-gamma
by lung lymphocytes in HIV-infected individuals. Am J Physiol 1999;
276:L256–L262.
24. Agostini C, Zambello R, Facco M, P erin A, Piazza F, Siviero M, Basso U,
Bortolin M, Trentin L, Semenzato G. CD8 T-cell infiltration in extra-
vascular tissues of patients with human immunodeficiency virus infec-
tion: interleukin-15 upmodulates costimulatory pathways involved in
the antigen-presenting cells-T-cell interaction. Blood 1999;93:1277–
1286.
25. AgostiniC, Poletti V, Zambello R, Trentin L, Siviero F, Spiga L, Gritti F,
Semenzato G. Phenotypical and functional analysis of bronchoalveolar
lavage lymphocytes in patients with HIV infection. Am Rev Respir
Dis 1988;138:1609–1615.
26. Agostini C, Zambello R, Trentin L, Feruglio C, Masciarelli M, Siviero F,
Poletti V, Spiga L, Gritti F, Semenzato G. Cytotoxic events taking
place in the lung of patients with HIV-1 infection: evidence of an
intrinsic defect of the major histocompatibility complex-unrestricted
killing partially restored by the incubation with rIL-2. Am Rev Respir
Dis 1990;142:516–522.
27. Poli G, Introna M, Zanaboni F, Peri G, Carbonari M, Aiuti F, Lazzarin A,
Moroni M, Mantovani A. Natural killer cells in intravenous drug
abusers with lymphadenopathy syndrome. Clin Exp Immunol 1985;
62:128–135.
28. Rankin JA, Walzer PD, Dwyer JM, Schrader CE, Enriquez RE, Merrill
WW. Immunologic alterations in bronchoalveolar lavage fluid in the
acquired immunodeficiency syndrome (AIDS). Am Rev Respir Dis
1983;128:189–194.
29. Twigg HL III, Spain BA, Soliman DM, Bowen LK, Heidler KM, Wilkes
DS. Impaired IgG production in the lungs of HIV-infected individuals.
Cell Immunol 1996;170:127–133.
30. Fahy RJ, Diaz PT, Hart J, Wewers MD. BAL and serum IgG levels in
healthy asymptomatic HIV-infected patients. Chest 2001;119:196–203.
31. Musher DM, Watson DA, Nickeson D, Gyorkey F, Lahart C, Rossen
RD. The effect of HIV infection on phagocytosis and killing of Staphy-
lococcus aureus by human pulmonary alveolar macrophages. Am J
Med 1990;299:158–163.
32. Reardon CC, Kim SJ, Wagner RP, Koziel H, Kornfeld H. Phagocytosis
and growth inhibition of Cryptococcus neoformans by human alveolar
macrophages: effects of HIV-1 infection. AIDS 1996;10:613–618.
33. Murray HW, Gellene RA, Libby DM, Rothermel CD, Rubin BY. Activa-
tion of tissue macrophages from AIDS patients: in vitro response of
Beck: HIV Infection 427
AIDS alveolar macrophages to lymphokines and interferon-.
J Immunol 1985;135:2374–2377.
34. Koziel H, Eichbaum Q, Kruskal BA, Pinkston P, Rogers RA, Armstrong
MY, Richards FF, Rose RM, Ezekowitz RA. Reduced binding and
phagocytosis of Pneumocystis carinii by alveolar macrophages from
persons infected with HIV-1 correlates with mannose receptor down-
regulation. J Clin Invest 1998;102:1332–1344.
35. Rich EA, Toossi Z, Fujiwara H, Hanigosky R, Lederman MM, Ellner JJ.
Defective accessory function of monocytes in human immunodefi-
ciency virus-related disease syndromes. J Lab Clin Med 1988;112:174–
181.
36. Twigg HL III, Lipscomb MF, Yoffe B, Barbaro DJ, Weissler JC. En-
hanced accessory cell function by alveolar macrophages from patients
infected with the human immunodeficiency virus: potential role for
depletion of CD4cells in the lung. Am J Respir Cell Mol Biol 1989;1:
391–400.
37. Macatonia SE, Lau R, Patterson S, Pinching AJ, Knight SC. Dendritic
cell infection, depletion and dysfunction in HIV-infected individuals.
Immunology 1990;71:38–45.
38. SpiegelH, Herbst H, Niedobitek G, Foss HD, Stein H. Follicular dendritic
cells are a major reservoir for human immunodeficiency virus type 1
in lymphoid tissues facilitating infection of CD4T-helper cells. Am
J Pathol 1992;140:15–22.
39. WrightSC, Jewett A, Mitsuyasu R, Bonavida B. Spontaneous cytotoxicity
and tumor necrosis factor production by peripheral blood monocytes
from AIDS patients. J Immunol 1988;141:99–104.
40. Ammann AJ, Palladino MA, Volberding P, Abrams D, Martin NL,
Conant M. Tumor necrosis factor alpha and beta in acquired immuno-
deficiency syndrome (AIDS) and AIDS-related complex. J Clin Immu-
nol 1987;7:481–485.
41. Munis JR, Richman DD, Kornbluth RS. Human immunodeficiency
virus-1 infection of macrophages in vitro neither induces tumor necro-
sis factor (TNF)/cachectin gene expression nor alters TNF/cachectin
induction by lipopolysaccharide. J Clin Invest 1990;85:591–596.
42. Agostini C, Zambello R, Trentin L, Garbisa S, Di Cella PF, Bulian P,
Onisto M, Poletti V, Spiga L, Raise E, et al. Alveolar macrophages
from patients with AIDS and AIDS-related complex constitutively
synthesize and release tumor necrosis factor alpha. Am Rev Respir
Dis 1991;144:195–201.
43. Wewers MD, Diaz PT, Wewers ME, Lowe MP, Nagaraja HN, Clanton
TL. Cigarette smoking in HIV infection induces a suppressive inflam-
matory environment in the lung. Am J Respir Crit Care Med 1998;158:
1543–1549.
44. Lane BR, Markovitz DM, Woodford NL, Rochford R, Strieter RM,
Coffey MJ. TNF-alpha inhibits HIV-1 replication in peripheral blood
monocytes and alveolar macrophages by inducing the production of
RANTES and decreasing C–C chemokine receptor 5 (CCR5) expres-
sion. J Immunol 1999;163:3653–3661.
45. Meddows-TaylorS, Pendle S, Tiemessen CT. Altered expression of CD88
and associated impairment of complement 5a-induced neutrophil re-
sponses in human immunodeficiency virus type 1-infected patients with
and without pulmonary tuberculosis. J Infect Dis 2001;183:662–665.
46. Armbruster C, Krugluger W, Huber M, Stephan K. Immunoglobulin
G Fc(gamma) receptor expression on polymorphonuclear cells in
bronchoalveolar lavage fluid of HIV-infected and HIV-seronegative
patients with bacterial pneumonia. Clin Chem Lab Med 2004;42:
192–197.
47. Bandres JC, Trial J, Musher DM, Rossen RD. Increased phagocytosis
and generation of reactive oxygen products by neutrophils and mono-
cytes of men with stage 1 human immunodeficiency virus infections.
J Infect Dis 1993;168:75–83.
48. Baldwin GC, Gasson JC, Quan SG, Fleischmann J, Weisbart R, Oette D,
Mitsuyasu RT, Golde DW. Granulocyte-macrophage colony-stimulating
factor enhances neutrophil function in acquired immunodeficiency syn-
drome patients. Proc Natl Acad Sci USA 1988;85:2763–2766.
49. Roilides E, Walsh TJ, Pizzo PA, Rubin M. Granulocyte colony-stimulating
factor enhances the phagocytic and bactericidal activity of normal and
defective human neutrophils. J Infect Dis 1991;163:579–583.
50. Lazzarin A, Uberti Foppa C, Galli M, Mantovani A, Poli G, Franzetti F,
Novati R. Impairment of polymorphonuclear leucocyte function in
patients with acquired immunodeficiency syndrome and with lymph-
adenopathy syndrome. Clin Exp Immunol 1986;65:105–111.
... Changes in mucociliary function and the presence of soluble defense molecules such defensins in respiratory secretions are examples of these modifications [2]. Innate and adaptive immune responses to infections may be compromised in the lung parenchyma [3] . For example, alveolar macrophages from HIV-infected individuals have been shown to be deficient in pathogen recognition. ...
Article
Full-text available
Introduction: Impaired pulmonary function and arterial oxygen (O2) saturation are among the symptomatic changes in people living with HIV (PLWH), associated with progression of HIV disease and anti-retroviral therapy (ART). Exercise has been considered an important adjuvant therapy for health promotion and improving the pulmonary function of PLWH. However studies are lacking on the effects circuit exercise training on o2 saturation and pulmonary function of PLWH. Objective: This study determined the effect of moderate intensity circuit exercise programme on the O2 saturation and pulmonary function of PLWH. Methods: The study design was a pretest-posttest randomized controlled design. A total of 120 participants were recruited from the HIV clinic of a teaching hospital. Fish bowl method was used to randomize the participants to control group or exercise group. Moderate intensity (50 – 75% MHR) circuit exercise programme was administered to the participants 3 times a week between 30-60 minutes per session for 8 weeks. Pulse Oximeter and hand held spirometer were used to measure the oxygen saturation and some pulmonary functions respectively. Measurement were taken at baseline and at 8-week. Descriptive statistics of mean, standard deviation and percentages were used to summarize demographic data. Paired sample t-test and Independent sample t-test determined the mean difference within the groups and between the groups respectively. Alpha level was set at P<0.05. Results: There was significant difference in the mean score of O2 saturation and pulmonary function within the exercise group after 8-week exercise intervention. There was significant difference in the mean score of O2 saturation and pulmonary function between the control group and exercise group after 8-week exercise intervention. Conclusion: An 8-week circuit exercise programme brought about improvement in O2 saturation and pulmonary function of PLWH. Circuit exercise can be effectively employed to improve lung function and ameliorate the exercise hypoxemia associated with decreased O2 saturation in PLWH.
... Human immunodeficiency virus (HIV) is known to invade the immune system thereby decreasing the CD4 ? T cells count [2]. A special attention is required for PLHIV with low CD4 cell count, high viral load, advanced stage of HIV disease and lastly those who are not taking effective ART treatment. ...
Article
Full-text available
People living with Human Immunodeficiency Virus (PLHIV) are at greater risk of developing prolonged illness due to COVID 19 leading to longer duration of virus shedding owing to their underlying immune defects. The present study compared SARS-CoV-2 infection developing at the same time among two health care workers living with and without a history of HIV and working in the same ward of a tertiary care hospital of North India. A higher viral load was reported in the SARS-CoV-2 infected worker who was immunocompromised as compared to immunocompetent patient (19,193 copies/µL vs 9.4 copies/µL). In this preliminary case report, no difference was observed in the clinical presentation of both patients at the time of diagnosis. Further studies are required to investigate the COVID-19 susceptibility and severity among HIV-infected patients.
... The higher risk associated with CKD, obesity, and immunocompromised states that are chronic stress/anxiety, T2DM and HIV/AIDS is also linked to the weakening of the immune system that characterizes these pathologies [31][32][33][34][35]. Despite a relatively higher burden of HIV/AIDS [36] and CKD [37], the African population hosts lower proportions of individuals with chronic stress/anxiety [38], obesity [39] and T2DM [40] as compared to the US and Europe. ...
Preprint
Full-text available
Introduction. Since its identification, the COVID-19 infection has caused substantial mortality and morbidity worldwide, but sub-Saharan Africa seems to defy the predictions. We aimed to verify this hypothesis using strong statistical methods. Methods. We conducted a cross-sectional study comparing the projected and actual numbers as well as population proportions of COVID-19 cases in the 46 sub-Saharan African countries on May 1st, May 29th (4 weeks later) and June 26th (8 weeks later). The source of the projected number of cases was a publication by scientists from the Center for Mathematical Modeling of Infectious Diseases of the London School of Hygiene & Tropical Medicine, whereas the actual number of cases was obtained from the WHO situation reports. We calculated the percentage difference between the projected and actual numbers of cases per country. Further, N-1 chi-square tests with Bonferroni correction were used to compare the projected and actual population proportion of COVID-19 cases, along with the 95% confidence interval of the difference between these population proportions. All statistical tests were 2-sided, with 0.05 used as threshold for statistical significance. Results. On May 1st, May 29th and June 26th, respectively 40 (86.95%), 45 (97.82%) and 41 (89.13%) of the sub-Saharan African countries reported a number of confirmed cases that was lower than the predicted number of 1000 cases for May 1st and 10000 for both May 29th and June 26th. At these dates, the population proportions of confirmed Covid-19 cases were significantly lower (p-value <0.05) than the projected proportions of cases. Across all these dates, South-Africa always exceeded the predicted number and population proportion of COVID-19 infections. Conclusion. Sub-Saharan African countries did defy the dire predictions of the COVID-19 burden. Preventive measures should be further enforced to preserve this positive outcome.
... [7][8][9][10] Even before progressing to AIDS, HIV infection leads to immune dysregulation. [11,12] This dysregulation can affect response to and efficacy of vaccines in an HIV-positive person. Reduced antibody response to influenza, [13], hepatitis B, [14] and pneumococcal polysaccharide vaccines [15] have been observed among HIV-positive persons. ...
Article
Full-text available
Human immunodeficiency virus (HIV) causes immune dysregulation, potentially affecting response to vaccines in infected persons. We investigated if unexpected adverse events (AEs) or unusual patterns of AEs after vaccination were reported among HIV-positive persons. We searched for domestic reports among HIV-positive persons to the Vaccine Adverse Event Reporting System (VAERS) during 1990–2016. We analyzed reports by age group (<19 and ≥19 years), sex, serious or non-serious status, live vaccine type (live versus inactivated), AEs reported, and CD4 counts. Of 532,235 reports received, 353 (0.07%) described HIV-positive persons, of whom 67% were aged ≥19 years, and 57% were male; most reports (75%) were non-serious. The most commonly reported inactivated vaccines were pneumococcal polysaccharide (27%) and inactivated influenza (27%); the mostly reported common live virus vaccines were combination measles, mumps, and rubella (8%) and varicella (6%). Injection site reactions were commonly reported (39%). Of 67 reports with CD4 counts available, 41 (61%) described persons immunocompromised at time of vaccination (CD4 count <500 cells/mm³), and differed from overall reports only in that varicella was the most common live virus vaccine (4 reports). Of 22 reports describing failure to protect against infection, 6 described persons immunocompromised at time of vaccination, among whom varicella vaccine was most common (3 reports). Of 66 reports describing live virus vaccines, 7 described persons with disseminated infection: 6 had disseminated varicella, 3 of whom had vaccine strain varicella-zoster virus. Of 18 reported deaths, 7 resulted from disseminated infection: 6 were among immunocompromised persons, 1 of whom had vaccine strain varicella-zoster virus. We identified no unexpected or unusual patterns of AEs among HIV-positive persons. These data reinforce current vaccine recommendations for this risk group. However, healthcare providers should know their HIV-positive patients’ immune status because immunocompromising conditions can potentially increase the risk of rare, but severe, AEs following vaccination with live virus vaccines.
... Finally, coinfection by a second pathogen can contribute to a breakdown in the host defense cascade leading to opportunistic infections. (Beck, 2005). The innate immune system plays an important role in the viral pathogen. ...
Article
Full-text available
SAMHD1 (sterile alpha motif and HD domain 1) is a protein that is found in myeloid cells, which restricts HIV1 replication. It depletes the de-oxy-nucleoside tri-phosphate (dNTPs) pool needed for a viral cDNA synthesis leading to inhibition of viral replication inside the cells. However, it does not restrict HIV2 replication in myeloid cells due to the presence of viral Vpx protein. Vpx is a virion-associated protein which augments viral infectivity and it only exists in HIV2 and it has been recently shown in Simian Immunodeficiency Virus (SIV) and which can induce degradation of SAMHD1 protein. This increases the amount of dNTPs for viral reverse transcription in cytoplasm and HIV infection. HIV2 reverse transcription is believed to be less active than HIV1 and this could be the reason for the absence of Vpx from HIV1. Protein expression and interaction between Vpx and SAMHD1 remains unclear. The interaction of SAMHD1 and HIV2-VPx patients' cells can be considered as a first step to help in the development for more effective anti-HIV drugs and possible novel intervention therapy in the future. Present review article provides comprehensive insights on the above issue. We performed a comprehensive literature search in the bibliographic database “Pubmed,” looking at studies discussing the SAMHDI and Vpx interactions.
... In the later stages, lymphocytes are attracted to the infected area. 17 T cells from HIV-infected patients do not produce IFNÀc in response to antigens, and it could be further elaborated that IFNÀc correlates with clinical status and CD4 counts and predicts progression to AIDS. It appears that lymphocytes in skin tissue could be a predictor of the immune response to an infectious agent. ...
Article
Background: Disseminated cutaneous histoplasmosis (DCH) is one of the manifestations of systemic histoplasmosis infection in HIV-positive patients. Interaction between host immune status and histoplasmosis infection is still poorly understood. It is thought that immune status, represented by CD4 counts, may be correlated with clinical and histopathological findings. Objective: To correlate the CD4 counts with the different clinical and histopathological findings in cutaneous histoplasmosis. Methods: This was a serial case report of seven HAART-treated HIV positive patients with disseminated histoplasmosis observed within the period of January 2004 through December 2014 from the Dermatology and Venereology Department, Cipto Mangunkusumo Hospital, Jakarta, Indonesia. The patients came with different complaints and clinical findings. CD4 counts were recorded prior to lesion biopsy. Results: The CD4 counts were independent from clinical morphology and distribution of lesions. Lower CD4 counts were associated with the presence of intraepidermal yeast-like cells, whereas there is the ability of forming granulomas at higher CD4 counts. Conclusion: CD4 count correlates to histopathological findings of cutaneous histoplasmosis.
... The human immunodeficiency viruses cause significant impairment in lung host defenses. Several mechanisms have been elucidated and are reviewed by Beck (36). Several mechanisms of impairment are briefly described here. ...
Article
Full-text available
Lower Respiratory Tract Infections, Page 1 of 2 Abstract Decades of advances in cancer treatments and transplantation immunology have expanded the population of severely immunocompromised patients. In addition, new therapies for the management of rheumatologic, autoimmune, and acquired immune diseases have reduced mortality among these patient groups. Pulmonary infections are the most common syndromes contributing to morbidity and mortality among immunosuppressed patients ( 1 – 3 ). Virtually any potential pathogen can result in significant illness, and pulmonary infiltrates may be caused by a variety of noninfectious syndromes as well. Management of pulmonary syndromes in these vulnerable populations is a challenge for both clinicians and microbiologists, as prompt diagnosis can prevent irreversible pulmonary complications and/or allow withdrawal of potentially toxic empiric therapies. Diagnostic approaches should consider the tempo of the pulmonary process, the extent of immunosuppression, and the radiographic patterns. In addition, the likelihood of a specific infection may be affected by recently administered prophylaxis or empiric therapies. Diagnostic Microbiology of the Immunocompromised Host, Second Edition
Article
Full-text available
Human immunodeficiency virus type 1 (HIV-1) infection causes progressive depletion of CD4/HIV-receptor-positive T helper lymphocytes, ultimately leading to AIDS. The major HIV reservoir and site of T-helper cell infection in lymphoid tissues, however, has remained poorly defined. The authors used in situ hybridization in combination with immunohistologic labeling techniques to identify the phenotype of HIV-infected cells in lymph nodes from patients at different stages of HIV-infection. The number of HIV-infected macrophages, widely considered the major site of HIV replication, was extremely low. There was no evidence for HIV-infection of endothelial and interdigitating reticulum cells. However, HIV RNA was found in small but consistent proportions of CD45RO-positive T cells and in the vast majority of follicular dendritic cells (FDC) in a pattern suggestive of active infection in addition to HIV-immunocomplex trapping on cell membranes. FDC may therefore be a major HIV reservoir and since T-helper cells travel through the FDC meshwork during their migration within lymphoreticular tissues, it appears likely that HIV-replicating T cells may infect FDC, which then infect new T cells, thus causing a gradual dissemination of the virus to all FDC and thereby a steadily increasing infection of T-helper/memory cells within germinal centers. This results in CD4+ T cell depletion, and ultimately, in immunodeficiency.
Article
The human T-cell lymphotropic virus type III (HTLV-III) is the etiologic agent of the acquired immunodeficiency syndrome (AIDS) and preferentially infects T4 lymphocytes. Other cell types, notably B lymphocytes and other nonlymphoid cells, also have been reported to be infected in vitro by HTLV-III. We now report on the susceptibility of human pulmonary macrophages to infection with HTLV-III in vitro. Alveolar macrophages infected with HTLV-III produced low levels of virus that could be transferred to allogeneic human peripheral blood mononuclear leukocytes as long as 2 weeks after initiation of infection. Unlike HTLV-III infection of T lymphocytes, macrophages appeared more resistant to viral-mediated cytopathic effects. Primary cultures of pulmonary macrophages from two of four patients with AIDS spontaneously produced low levels of virus detected as precipitable reverse transcriptase activity, suggesting that these cells were infected in vivo. Because tissue macrophages are long-lived cells, they may act as a reservoir of HTLV-III, capable of transmitting the virus to other susceptible cells such as T lymphocytes, causing periodic low- level viremia. Macrophage infection with HTLV-III may be one mechanism for the establishment of viral persistence in infected hosts.
Article
Peripheral blood mononuclear cells (PBMCs) from many asymptomatic individuals infected with human immunodeficiency virus-type 1 (HIV) are unresponsive as measured by in vitro T cell proliferation and interleukin-2 (IL-2) production to influenza virus and synthetic peptides of HIV envelope (Env). Strong influenza virus- and Env-stimulated IL-2 responses and T cell proliferation were restored when cultures were stimulated in the presence of IL-12. Interferon-gamma production by PBMCs from HIV seropositive (HIV^+) patients was also restored with IL-12. Furthermore, in vitro antigen-specific production of IL-2 and proliferation of PBMCs from HIV^- donors were suppressed by antibody to IL-12, but were not enhanced by addition of exogenous IL-12. Thus, IL-12 may be limiting in PBMCs from HIV^+ but not HIV^- individuals. These findings demonstrate that IL-12 can restore HIV-specific cell-mediated immunity in vitro in HIV-infected individuals and suggest a potential use of IL-12 in augmenting the diminished immunologic functions associated with HIV infection.
Article
We studied the relationship of CD4+ and CD8+ depletion to initial and late opportunistic infections in 62 patients with AIDS. The mean interval between initial and late infections was 12.2 months. Geometric mean (and 95% confidence intervals) of T-cell counts at the diagnosis of each infection were: (Pneumocystis carinii pneumonia) CD4+ 0.051 (0.044-0.058) x 10(9)/l, CD8+ 0.561 (0.476-0.646) x 10(9)/l; (cytomegalovirus retinitis) CD4+ 0.025 (0.019-0.031) x 10(9)/l, CD8+ 0.333 (0.183-0.483) x 10(9)/l. Mycobacterium avium-intracellulare bacteraemia closely followed cytomegalovirus dissemination. Most patients were free from late opportunistic infections caused by disseminated cytomegalovirus and M. avium-intracellulare until CD8+ declined below 0.500 x 10(9)/l. Zidovudine improved CD4+, but less so CD8+, and similarly enhanced the survival of patients treated in 1985-1990 and 1991.
Article
Granulocyte colony-stimulating factor (G-CSF) stimulates proliferation of myeloid cells and maybe a valuable adjunct in prevention and treatment of neutropenia-associated infections. Neutrophil (PMNL)phagocytic and microbicidal functions against Staphylococcus aureus and Candida albicans blastoconidia were therefore evaluated. Bacterial phagocytosis and bactericidal activity were significantly enhanced by ∼ 50%–70% after preincubation of normal PMNLw ith G-CSF in concentrations of 1000–4000 units/ml for 10 min at 37°C. G-CSF in similar concentrations enhanced the defective bactericidal activity of PMNL from HIV-1-infected patients by ∼70%–150% and reached the baseline control PMNL killing. However, G-CSF enhanced neither phagocytosis nor fungicidal activity of normal PMNL against C. albicans blastoconidia. These data demonstrate that G-CSF enhances the antibacterial but not the antifungal activities of human PMNL in vitro and also improves the defective PMNLbactericidal activity of HIV-1-infected patients.
Article
Tissue macrophages are recognized as a cellular target for infection with the human immunodeficiency virus type 1 (HIV-1). To characterize the nature of this cell-retrovirus interaction within the lower respiratory tract we analyzed fluid and cells obtained by bronchoalveolar lavage (BAL) of eight individuals with acquired immunodeficiency syndrome (AIDS) who were undergoing diagnostic fiberoptic bronchoscopy. Of these eight individuals, seven had active infection with Pneumocystis carinii; one had suspected cytomegalovirus pneumonitis. At the time of study two were receiving the antiretroviral drug zidovudine (azidothymidine [AZT]). HIV-1 could not be isolated from any of the eight samples of BAL fluid concentrated by ultracentrifugation through 20% sucrose. HIV-1 antigen (p24) was detected in one of eight samples of concentrated BAL fluid but could not be found in eight samples of media conditioned by overnight incubation with adherent BAL cells. Despite the infrequent detection of HIV-1 antigen it was possible to identify HIV-1 genomic sequences by the use of a DNA amplification technique, the polymerase chain reaction, in all eight BAL cell preparations. In BAL cells adherent for up to 5 days in culture this method detected retroviral DNA that hybridized to a complementary pair of primers located in the env and gag gene regions of HIV-1. These studies demonstrate the uniform presence of HIV-1 harboring cells within the airways of the lung in individuals with AIDS and active respiratory infection and may have implications for local organ defense.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
This paper summarizes our research and the results obtained on the topic of immunology of interstitial lung disorders. Areas of investigation mainly included sarcoidosis, hypersensitivity pneumonitis (HP), and more recently the pulmonary involvement in acquired immunodeficiency syndrome (AIDS). In sarcoidosis patients two major mechanisms account for the alveolitis, i.e. an in situ cellular proliferation and a cellular redistribution from the peripheral blood to the sites of disease activity, including the lung. These findings involve both lymphocytes (CD4 helper-related cells) and macrophages, and lead to the formation and provide maintenance of sarcoid granuloma. In patients with hypersensitivity pneumonitis the lung infiltrates are characterized by cells bearing suppressor/cytotoxic phenotype. The expansion of cells with these characteristics in the lung of these patients is likely to be related to a local immune response to the antigenic stimulus. In the lung of patients with AIDS we also found a discrete lymphocytic alveolitis bearing the CD8 cytotoxic-related phenotype. The role of cytotoxic events, related to the lymphocytes and macrophages, which are operative in the lung of AIDS patients, is being evaluated. The analysis of cells recovered from the lavage, mainly lymphocytes and macrophages, in terms of surface phenotype, functional in vitro evaluations and molecular analysis, has provided new insights into the pathogenesis of the above quoted interstitial lung disorders.
Article
To verify the hypothesis that alveolar macrophages (AMs) from patients infected with HIV-1 could synthesize and release TNF alpha, AMs recovered from the BAL fluid of 11 patients with seropositive HIV-1 (six with AIDS and five with ARC) were tested in vitro for their ability to destroy TNF alpha-susceptible targets. Furthermore, the presence of TNF alpha was assessed in AM-conditioned supernatants on the basis of their cytotoxic activity and by using an immunoenzymatic test and immunoblotting. Transcription of the TNF alpha gene in AMs was also studied by means of the Northern blot analysis. AMs freshly recovered from patients infected with HIV-1 exhibited high levels of cell-mediated cytotoxicity against U937 targets, and the addition of a polyclonal anti-TNF alpha antibody resulted in a significant inhibition of the target lysis. Cell-free supernatants conditioned by unstimulated AMs exerted high levels of cytotoxic activity against TNF alpha-sensitive targets, whereas duplicate, neutralization experiments performed in the presence of an anti-TNF alpha antibody proved that the observed cytotoxic activity was mostly mediated by TNF alpha. The presence of high amounts of TNF alpha in the conditioned media was confirmed by the immunoenzymatic test. In addition, the immunoblot analysis showed that the TNF alpha released by AMs has a Mr 17,000 band, identical to a standard preparation of recombinant TNF alpha. The Northern blot demonstrated that unstimulated AMs express detectable levels of mRNA transcripts for TNF alpha. Taken together, our data support the concept that AMs from patients with HIV-1 infection constitutively release TNF alpha.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
Immune responses in resting T cells are initiated by the presentation of antigen by bone marrow-derived dendritic cells (DC). Normal DC are susceptible to infection with human immunodeficiency virus (HIV) in vitro (Patterson & Knight, 1987) and this blocks their capacity to stimulate T-cell responses to other antigens (Macatonia, Patterson & Knight, 1989a). To study the relationship between HIV and DC in patients and its relevance to the pathogenesis of disease, DC have been isolated from the blood of individuals in the different clinical categories, counted, examined for the presence of virus genome and their antigen-presenting capacity measured. Infection, depletion and impaired function of DC occur in early HIV infection. HIV seropositive patients who were asymptomatic and those with symptoms of disease had significantly reduced numbers of DC, but patients with persistent generalized lymphadenopathy had normal numbers. Between 3% and 21% of DC, identified as large low-density cells not bearing monocyte, lymphocyte or natural killer cell markers, were infected with HIV, as indicated by in situ hybridization. Less than 0.12% of the lymphocytes or monocytes were infected. The DC from infected individuals were poor at enhancing responses to the mitogen concanavalin A (Con A). They also caused low levels of stimulation in allogeneic lymphocytes in mixed leucocyte cultures. By contrast, T cells from asymptomatic patients gave normal T-cell responses to uninfected allogeneic DC, although those from acquired immunodeficiency syndrome (AIDS) patients did show reduced responsiveness. Defects in DC thus precede both the appearance of symptoms and changes in T cells and may be instrumental in the development of AIDS. Furthermore, since DC numbers and function differ at different stages of disease, monitoring these may contribute to clinical assessment and lead to new therapeutic approaches.
Article
A CD8+ lymphocytic infiltration of the lungs is frequently observed in HIV-infected patients, even prior to the onset of opportunistic infections. In such patients, we could demonstrate that most of these CD8+ alveolar T lymphocytes displayed the D44 marker and were functional cytolytic T lymphocytes directed against autologous HIV-infected alveolar macrophages. This primary cytolytic activity was HLA-restricted and, at least partially, specific for the HIV envelope protein, since HLA-A2 alveolar T lymphocytes could specifically lyse cell lines expressing both the HLA-A2 and Env antigens. In contrast to data obtained in peripheral blood, no ADCC activity was observed against the Env antigen. HIV-specific alveolar T-lymphocyte cytolytic activity decreased with progression towards AIDS as shown by studies of a series of 40 patients. Functional abnormalities of the lung epithelium could be associated with the specific lysis of alveolar macrophages, suggesting that local tissue injury could result from the in vivo immune conflict between alveolar HIV-specific CTL and HIV-infected macrophages.